Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR ENHANCED LYMPHOCYTE-MEDIATED IMMUNOTHERAPY
Document Type and Number:
WIPO Patent Application WO/2020/163953
Kind Code:
A1
Abstract:
Lymphocytes having a suppressed autophagy gene useful in immunotherapy are disclosed. The lymphocytes can express an antigen targeting receptor such as a chimeric antigen receptor (CAR) or endogenous or engineered T-cell receptor to target cells expressing a tumor-specific antigen. Methods of making and using such lymphocytes are disclosed. Some such lymphocytes are useful in conducting CAR-T or TCR-T therapy.

Inventors:
LUM JULIAN J (CA)
DEVORKIN LINDSAY (CA)
DOYON YANNICK (CA)
CARLETON GILLIAN (CA)
Application Number:
PCT/CA2020/050185
Publication Date:
August 20, 2020
Filing Date:
February 12, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PROVINCIAL HEALTH SERVICES AUTHORITY (CA)
UNIVERSITÉ LAVAL (CA)
International Classes:
C12N15/113; A61K35/17; A61P35/00; A61P35/02; A61P37/02; C07K14/705; C07K19/00; C12N5/10; C12N15/09; C12N15/12; C12N15/62; C12N15/85; C12N15/864; C12N15/90
Other References:
PAN HONGMING, CHEN LIUXI, XU YINGHUA, HAN WEIDONG, LOU FANG, FEI WEIQIANG, LIU SHUIPING, JING ZHAO, SUI XINBING: "Autophagy-associated immune responses and cancer immunotherapy", ONCOTARGET, vol. 7, no. 16, 13 January 2016 (2016-01-13), pages 21235 - 21246, XP055733149, ISSN: 1949-2553
CARTELLIERI ET AL.: "Chimeric antigen receptor-engineered T cells for immunotherapy of cancer", JOURNAL OF BIOMEDICINE AND BIOTECHNOLOGY, vol. 2010, 5 May 2010 (2010-05-05), pages 1 - 13, XP002673541, ISSN: 1110-7243, DOI: 10.1155/2010/956304
AMITA SEHGAL: "Biomembranes Part R: Transport theory : cells and model membranes; IN: Methods in Enzymology", vol. 588, 12 January 2016, ACADEMIC PRESS, ISBN: 978-0-12-182072-5, ISSN: 0076-6879, article OPREY, SAKAMAKI, BAUDOT, NEW, VAN ACKER, TOOZE, LONG, RYAN: "Chapter 6: Application of CRISPR/Cas9 to autophagy research", pages: 79 - 108, XP009529993, DOI: 10.1016/bs.mie.2016.09.076
DEVORKIN LINDSAY, PAVEY NILS, CARLETON GILLIAN, COMBER ALEXANDRA, HO CALLY, LIM JUNGHYUN, MCNAMARA ERIN, HUANG HAOCHU, KIM PAUL, Z: "Autophagy regulation of metabolism is requiredfor CD 8+ T cell anti-tumor immunity", CELL REPORTS, vol. 27, 4 September 2019 (2019-09-04), pages 502 - 513, XP055733153, ISSN: 2211-1247
See also references of EP 3924495A4
Attorney, Agent or Firm:
MARLES, Jennifer A. et al. (CA)
Download PDF:
Claims:
CLAIMS:

1 A lymphocyte comprising an antigen targeting receptor and a suppressed autophagy gene.

2 A lymphocyte as defined in claim 1 , comprising a nucleic acid encoding the antigen targeting receptor inserted within a locus of the autophagy gene to disrupt expression of the autophagy gene.

3 A lymphocyte as defined in the claim 2, wherein the nucleic acid encoding the

antigen targeting receptor is inserted within the locus of the autophagy gene so that expression of the antigen targeting receptor is regulated by an endogenous promoter of the autophagy gene.

4 A lymphocyte as defined in any one of claims 1 to 3, wherein the nucleic acid

encoding the antigen targeting receptor is inserted within an exon of the autophagy gene.

5 A lymphocyte as defined in any one of claims 1 to 3, wherein the nucleic acid

encoding the antigen targeting receptor is inserted within an intron of the autophagy gene, optionally wherein the autophagy gene is ATG5 and the nucleic acid encoding the antigen targeting receptor is inserted within intron 2 of the ATG5 gene, and further optionally where the nucleic acid used to insert the antigen targeting receptor has the sequence of one of SEQ ID NO:8 or SEQ ID NO:9. 6. A lymphocyte as defined in claim 1 , wherein the autophagy gene is at a first locus of a genome of the lymphocyte and the lymphocyte comprises a nucleic acid encoding the antigen targeting receptor at a second locus of the genome, the first locus being different from the second locus.

7. A lymphocyte as defined in claim 6, wherein expression of the antigen targeting

receptor is regulated by a heterologous promoter.

8. A lymphocyte for use in immunotherapy comprising a suppressed autophagy gene.

29

RECTIFIED SHEET (RULE 91.1)

9. A lymphocyte as defined in any one of claims 1 to 8, wherein the autophagy gene is ablated, or wherein the autophagy gene is suppressed by RNAi.

10. A method of conducting immunotherapy comprising administering a lymphocyte as defined in any one of claims 1 to 9 to a subject.

1 1 . A method of conducting immunotherapy using lymphocytes, the method comprising administering to a subject lymphocytes that have been modified to suppress an autophagy gene.

12. A method of conducting immunotherapy using lymphocytes, the method comprising administering to a subject lymphocytes that have been modified to express an antigen targeting receptor and to suppress an autophagy gene.

13. A method of treating cancer, the method comprising a step of conducting

immunotherapy as defined in any one of claims 10-12.

14. A method as defined in claim 13, wherein the cancer is B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), B-cell lymphoma or other lymphoid malignancy, liver, pancreatic, brain, breast, ovarian, colorectal, acute myeloid leukemia (AML), multiple myeloma, lung, gastric, glioma, EGFR-positive solid tumor, glioblastoma, glioblastoma multiforme, stomach, nasopharyngeal, esophageal, prostate, neuroblastoma, hepatocellular, squamous cell lung, MSLN- positive solid tumor, non-small-cell lung (NSCLC), triple-negative breast cancer (TNBC), sarcoma, advanced solid tumor, renal cell, central nervous system, or an ROR1 -positive malignancy; and optionally wherein the cancer is ovarian cancer, breast cancer, or lung cancer.

15. A method of making a lymphocyte for use in immunotherapy, the method comprising a step of modifying the lymphocyte to suppress an autophagy gene.

16. A method as defined in claim 15, further comprising modifying the lymphocyte to express an antigen targeting receptor.

17. A method as defined in any one of claims 15 or 16, wherein one or both of the steps of modifying the lymphocyte to suppress the autophagy gene and modifying the lymphocyte to express the antigen targeting receptor is conducted using a gene editing method or using RNAi, wherein the gene-editing method optionally comprises CRISPR-Cas.

18. A method as defined in claim 17, wherein the gene editing method is used to insert a nucleic acid encoding the antigen targeting receptor at a locus of the autophagy gene.

19. A method as defined in claim 18, wherein the gene editing method is used to insert the nucleic acid encoding the antigen targeting receptor so that expression of the antigen targeting receptor is regulated by an endogenous promoter of the autophagy gene.

20. A method as defined in any one of claims 17 to 19, wherein the gene editing method is used to insert the nucleic acid encoding the antigen targeting receptor in an intron of the autophagy gene, optionally wherein the autophagy gene is ATG5 and the nucleic acid encoding the antigen targeting receptor is inserted into intron 2 of the ATG5 gene, further optionally wherein the nucleic acid construct that is used to insert the antigen targeting receptor comprises the sequence of one of SEQ ID NO:8 or SEQ ID NO:9, and further optionally wherein the gene editing method comprises CRISPR-Cas and the sgRNAs used to conduct the CRISPR-Cas gene editing comprise one of SEQ ID NOs:1-7. 21 . A method as defined in any one of claims 15-17, wherein the step of modifying the lymphocyte to express the antigen targeting receptor comprises inserting a nucleic acid encoding the antigen targeting receptor within a genome of the lymphocyte at a first locus.

22. A method as defined in claim 21 , wherein the step of modifying the lymphocyte to suppress an autophagy gene comprises knocking out the autophagy gene at a second locus of the genome, the second locus being different from the first locus.

23. A method as defined in either one of claims 21 or 22, wherein the step of modifying the lymphocyte to express the antigen targeting receptor comprises inserting a nucleic acid encoding a heterologous promoter to drive expression of the antigen targeting receptor.

24. A method as defined in any one of claims 15 to 22, wherein the autophagy gene is suppressed by knocking out the autophagy gene, optionally wherein the autophagy gene is ATG5 and further optionally wherein the step of knocking out the autophagy gene comprises disrupting ATG5 at exon 4 or exon 5.

25. A method as defined in any one of claims 15 to 24, wherein the step of modifying the lymphocyte to suppress the autophagy gene comprises using CRISPR-Cas, zinc- finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), Sleeping Beauty (SB), RNAi, meganucleases, or megaTALs.

26. A method of conducting immunotherapy comprising administering to a subject a lymphocyte made by the method of any one of claims 15 to 25.

27. An sgRNA molecule having a targeting domain complementary to an autophagy gene. 28. An sgRNA molecule as defined in claim 27, having the nucleotide sequence of any one of SEQ ID NOs: 1 -7, 24 or 25.

29. A nucleic acid encoding an sgRNA molecule as defined in any one of claims 27 or

28.

30. A lymphocyte or method as defined in any one of claims 1 to 26, wherein the

lymphocyte comprises a lymphocyte with activity in killing tumor cells.

31 . A lymphocyte or method as defined in any one of claims 1 to 26 or 30, wherein the lymphocyte comprises a cytotoxic lymphocyte.

32. A lymphocyte or method as defined in any one of claims 1 to 26, 31 or 31 , wherein the lymphocyte comprises a T-cell, a natural killer (NK) cell, or a B-cell.

33. A lymphocyte or method as defined in claim 32, wherein the T-cell comprises an induced pluripotent stem cell, a CD34+ T-cell, a CD4+ T-cell, a CD8+ T-cell, a Treg cell, a tissue-resident memory T-cell (TRM), or a natural killer T-cell (NKT).

34. A lymphocyte, method or sgRNA as defined in any one of claims 1 to 33, wherein the autophagy gene is ULK1, ULK2, ULK3, FIP200 , Vps34, Beclin-1, p150, UVRAG, ATG1, ATG4, ATG5, ATG7, ATG8, ATG9, ATG10, ATG12, ATG13, ATG14L, ATG16L, ATG16L1, ATG18, VMP1, or GABARAP, and optionally wherein the autophagy gene is ATG5 or ATG14.

35. A lymphocyte or method as defined in any one of claims 1 to 26 or 30 to 34, wherein the antigen targeting receptor comprises a chimeric antigen receptor (CAR) or an endogenous or engineered T-cell receptor.

36. A lymphocyte or method as defined in any one of claims 1 to 26 or 30 to 35, wherein the antigen targeting receptor is specific for a tumor-specific antigen, optionally wherein the tumor-specific antigen comprises the folate receptor (FR), the a-folate receptor, the b-folate receptor, the g-folate receptor, CD19, CD20, CD133, CD138, CEA, Claudin 18.2, EGFR, EGFRvlll, EphA2, EpCAM, GD2, GPC3, HER2, MSLN, MG7, MUC1 , NY-ESO-1 , LMP1 , prostate specific membrane antigen (PSMA), Fra, NKG2DI, BCMA, IL13Ralpha2, LeY, CD70, B7-H3, ROR1 , or PSCA.

37. A lymphocyte or method as defined in claim 36, wherein the antigen targeting

receptor comprises a chimeric antigen receptor (CAR), and wherein an antigen binding fragment of the CAR is specific for the tumor-specific antigen.

38. A lymphocyte or method as defined in any one of claims 1 to 26 or 30 to 36, wherein the antigen targeting receptor comprises a chimeric antigen receptor (CAR), and wherein the CAR is an a-folate receptor CAR, and wherein optionally the nucleotide construct used to insert the CAR in the lymphocyte has the nucleic acid sequence of either one of SEQ ID NO:8 or SQ ID NO:9.

39. A lymphocyte or method as defined in any one claims 1 to 26 or 30 to 38, wherein the subject is a mammal, and optionally wherein the subject is a human.

Description:
COMPOSITIONS AND METHODS FOR ENHANCED

LYMPHOCYTE-MEDIATED IMMUNOTHERAPY

Reference to Related Applications

[0001] This application claims priority to, and the benefit of, United States provisional patent application No. 62/804658 filed 12 February 2019, the entirety of which is incorporated by reference herein in its entirety for all purposes

Government License Rights

[0002] This invention was made, in part, with government support under Grant Number OC170169 made under Award Number W81XWH- 18- 1-0264 awarded by the U.S. Army Medical Research and Development Command. The government has certain rights in the invention.

Field of the Invention

[0003] Some embodiments of this invention relate to genetically engineered lymphocytes having improved efficacy as immunotherapeutics. Some embodiments of this invention relate to methods and compositions to modify the metabolism of lymphocytes in order to improve their efficacy as immunotherapeutics. Some embodiments relate to methods for the genetic manipulation of autophagy in chimeric antigen receptor T-cells (CAR-T) or engineered or endogenous T cell receptor T-cells (TCR-T) to enhance their effectiveness for immunotherapy of cancer.

Background

[0004] CAR-T-cells are engineered with an antigen-binding domain such as an antibody single chain variable fragment (scFv) to bind antigens expressed on the cell surface of tumor cells. In standard therapy, a CAR construct is transduced into autologous CD3+ T- cells, expanded ex vivo and then infused into a patient.“New generation” CAR-T-cells contain optimized CD3z signalling domains fused in combination with co-stimulatory molecules such as CD28, 41 BBL and CD27 (3). Despite these modifications, the persistence of CAR-T-cells in solid tumors remains poor. Thus, other barriers besides pro survival signals control the function of CAR-T-cells in the tumor microenvironment.

[0005] Despite decades of research, treatments for ovarian cancer have not improved patient outcomes. High-grade serous carcinoma (HGSC), the most common histotype, is often diagnosed at stage 3-4 disease and patients receive debulking surgery followed by several rounds of chemotherapy. The 5-year survival rate for stage 4 disease is < 20%. Although vaccines, checkpoint blockade and T-cell therapy have been explored, early trials have not improved overall patient survival. However, there is unequivocal evidence that the presence and function of tumor infiltrating lymphocytes (TIL) is strongly associated with improved survival implying that the immune system is beneficial in this disease (4).

[0006] Folate receptors (FR) exist as 3 isoforms, a, b, and y, and function in folic acid and folate uptake. High levels of aFR have been observed in ovarian, breast and lung cancers but are low in normal tissue (5). In one trial, 14 ovarian cancer patients who received infusions of aFR-CAR-T-cells with or without I L-2 showed no evidence of clinical response despite treatment being well-tolerated. Using radiolabel tracer imaging, the authors concluded that the aFR-CAR-T-cells were unable to persist shortly after infusion (2).

Therefore, the current approaches to use aFR-CAR-T in ovarian cancer have not been successful, in part, due to the lack of persistence of cells after infusion. However, this work provides an established safety profile for aFR-CAR-T therapy.

[0007] Manufacturing CAR-T-cells requires specialized infrastructure, patients must undergo lymphodepletion prior to T-cell infusions and acute life-threatening immune adverse events can arise. Moreover, CAR-T-cells have shown limited efficacy in solid tumors (6). One possibility to explain the lack of observed efficacy for solid tumors is a metabolic barrier imposed by the tumor ecosystem due to the high metabolic demands of rapidly proliferating tumor cells. Ultimately, this may cause a loss of T-cell function and persistence. Thus, it appears that metabolism can significantly impact T-cell behaviour providing a unique opportunity to modify T-cell metabolism and achieve better therapeutic success in solid cancers.

[0008] Deregulation of central metabolism is a universal hallmark of cancers, an observation reported for ovarian cancer (7). There is widespread appreciation that cancers use glucose and glutamine at high rates to serve as the main biosynthetic precursor for cell growth and proliferation (8). Recent work demonstrates that T-cells have adaptive metabolism depending on their state of activation and differentiation (7,9-12). For example, the transition from a naive T-cell to an activated effector cell is accompanied by switch from oxidative metabolism to a more glycolytic phenotype (9). The metabolic similarities of proliferating T-cells and tumor cells is striking and implies that insufficiencies in glucose and other nutrients that are essential to support proliferation can contribute to reduced T-cell function and exhaustion (10).

[0009] Metabolic competition could suppress T-cells that have been infused as part of T-cell therapy. One report found that CD8+ tumor infiltrating lymphocytes (TIL) in murine tumors were more functional in tumors with reduced glucose consumption compared to tumors with high rates of glycolysis suggesting that glucose consumption by tumors directly impaired T- cell activity (10). This defect was rescued by expressing metabolic enzymes that restore T- cell glycolysis. In another report, loss of mitochondrial mass in tumor-infiltrating lymphocytes (TIL) was associated with a reduction in type 1 cytokines, increased checkpoint inhibitor expression and loss of anti-tumor activity (13). However, enforced expression of PGC1 a, a transcription factor involved in mitochondria biogenesis, restored anti-tumor immunity. In the face of metabolic stress, reprogramming TIL to use alternative fuels may help sustain their anti-tumor activity. Despite these studies, there are no reports examining the role of metabolism on human TIL known to the inventors.

[0010] Autophagy is a form of catabolic metabolism where cells engulf portions of the cytosol and degrade cellular contents in the lysosome for metabolite recycling, protein quality control or destruction of damaged organelles (14). Autophagy is largely a survival pathway activated by nutrient and growth factor deprivation (14). T-cells lacking autophagy genes such as Atg5 or Atg7 have impaired thymocyte development and a reduction in peripheral T-cells (15). In line with this, it has been reported that autophagy is essential for CD8+ effector T-cell survival and memory development (16, 17).

[0011] Given the fundamental role that metabolism plays in organismal homeostasis and cancer, one might expect deleterious or pro-oncogenic phenotypes associated with manipulating the autophagy pathway. In the case of autophagy deficiency,

haploinsufficency of one autophagy gene, Beclin-1 , has been found to promote tumor formation and monoallelic deletion is observed in 40-75% of sporadic breast, ovarian and prostate cancer (18). Moreover, aged Atg5 or Atg7 liver-specific knockouts develop spontaneous liver tumors (19). It is believed that loss of Atg5 n T-cells does not lead to oncogenesis based on the inventors’ own observations. Moreover, gain- or loss-of-function in metabolic genes is required but not sufficient for tumorigenesis. For instance, T-cell specific Glutl transgenic mice do not spontaneously develop tumors (20).

[0012] There have been reports using CRISPR/Cas9 for gene-editing in primary human T- cells (21 , 22). A Chinese group has successfully knocked out PD-1 in T-cells, though the details of their strategy have yet to be reported. Another group was able to target the CD19 CAR to the T-cell receptor-a locus and cause enhanced tumor rejection in a mouse model (22). Most recently, a group at the University of Pennsylvania led by Carl June successfully conducted a first-in-human phase 1 trial testing the safety and feasibility of multiplex CRISPR-Cas9 editing in T-cells (24).

[0013] Many types of cells are involved in killing tumor cells, including NK cells; T-cells including CD34+, CD4+ or CD8+ T-cells, Treg cells, tissue-resident memory T cells (TRM cells), natural killer T-cells (NKT); B-cells, and the like. These cells act through a similar mechanism of recognizing an antigen such as a tumor-specific antigen on the surface of the tumor cell and acting to kill such cell.

[0014] Some embodiments of the present invention address an unmet need in the field for improved treatments using lymphocytes for immunotherapy, including CAR-T therapy, especially for treatment of solid cancers, for example those of the ovary, breast or lung.

[0015] The foregoing examples of the related art and limitations related thereto are intended to be illustrative and not exclusive. Other limitations of the related art will become apparent to those of skill in the art upon a reading of the specification and a study of the drawings.

Summary

[0016] The following embodiments and aspects thereof are described and illustrated in conjunction with systems, tools and methods which are meant to be exemplary and illustrative, not limiting in scope. In various embodiments, one or more of the above- described problems have been reduced or eliminated, while other embodiments are directed to other improvements. [0017] In one aspect, a lymphocyte having an antigen targeting receptor and a suppressed autophagy gene is provided. In some aspects, the lymphocyte is provided with a nucleic acid encoding the antigen targeting receptor inserted within a locus of the autophagy gene to disrupt expression of the autophagy gene. In some aspects, the autophagy gene is located at a first locus of the genome and the nucleic acid encoding the antigen targeting receptor is inserted at a second locus of the genome that is different from the first locus. In some such aspects, the autophagy gene is knocked out or disrupted at the first locus or suppressed in some other manner, for example using RNAi.

[0018] In one aspect, a method of conducting immunotherapy is provided that involves administering to a subject an engineered lymphocyte as described in this specification. In one aspect, a method of conducting immunotherapy is provided in which lymphocytes that have been modified to suppress an autophagy gene are administered to a subject. In some aspects, a method of conducting immunotherapy is provided in which lymphocytes that have been modified to both suppress an autophagy gene and express an antigen targeting receptor are administered to a subject. In some aspects, the immunotherapy is used to treat cancer.

[0019] In one aspect, a method of making a lymphocyte for use in immunotherapy is provided and involves modifying the lymphocyte to suppress an autophagy gene. In some such aspects, the lymphocyte is further modified to express a desired antigen targeting receptor.

[0020] In some aspects, any suitable technique may be used to suppress expression of the autophagy gene, including using CRISPR-Cas, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), Sleeping Beauty (SB), RNAi, meganucleases, or megaTALs. In some aspects, the autophagy gene is disrupted or knocked out, for example using CRISPR-Cas, zinc-finger nucleases (ZFN), transcription activator-like effector nucleases (TALEN), Sleeping Beauty (SB), meganucleases, or megaTALs.

[0021] In some aspects, methods and compositions to improve the anti-tumor efficacy of lymphocytes are provided. In some aspects, methods and compositions to improve the anti-tumor efficacy of cytotoxic lymphocytes are provided. In some aspects, methods and compositions to improve the anti-tumor efficacy of cytotoxic cells are provided. In some aspects, methods and compositions to improve the anti-tumor efficacy of lymphocytes including NK cells; T-cells including CD34+, CD4+ or CD8+ T-cells, Treg cells, tissue- resident memory T cells (TRM cells), natural killer T-cells (NKT); B-cells; or the like are provided.

[0022] In some aspects, the genetically engineered lymphocytes express an antigen targeting receptor such as a chimeric antigen receptor (CAR) or an endogenous or engineered T-cell receptor which targets a desired tumor-specific antigen.

[0023] In some aspects, methods and compositions to improve the anti-tumor efficacy of T- cells (T-lymphocytes) for use in adoptive cellular cancer immunotherapy, comprising suppression of autophagy in said T-cells are provided.

[0024] In one aspect, methods to improve the anti-tumor efficacy of chimeric antigen receptor (CAR) T-cells (CAR-T) for therapy of cancer by suppressing autophagy in said CAR-T-cells are provided.

[0025] In one aspect, the novel use of gene-editing methods (including but not limited to CRISPR-Cas9) to insert CAR nucleic-acid sequences at the loci of autophagy ( ATG ) genes in primary human autologous T-cells to generate‘knock-in’ CAR-T-cells deficient in autophagy is provided. These CAR-T-cells are deficient in expression of the ATG gene targeted by the CAR sequences using CRISPR-Cas9 and are believed to have improved anti-tumor efficacy (against tumors specifically targeted by the CAR-T) by virtue of the ablated or suppressed ATG gene.

[0026] In one aspect, the T-cell autophagy (ATG) gene targeted for‘knock-in’ of the CAR sequences using CRISPR-Cas9 gene editing (or targeted for knock-out or suppression in other aspects) may be any autophagy-related (ATG) gene known by those skilled in the art and may include but are not limited to ATG \ , ATG4, ATG5, ATG7, ATG8, ATG13, ATG4, ATG18, ATG16L1, and GABARP. In some aspects, the autophagy gene that is suppressed in the lymphocyte is one or more of ULK1, ULK2, ULK3 , FIP200 , Vps34, Beclin-1 , p150, UVRAG, ATG1, ATG4, ATG5, ATG7, ATG8, ATG9, ATG10, ATG12, ATG13, ATG14L, ATG16L, ATG16L1, ATG18, VMP1, GABARAP, or the like.

[0027] In one aspect, the CAR sequences used in the CRISPR-Cas9 knock-in at the chosen ATG gene locus of the T-cell may be directed to (specific for) any desired tumor (or disease/target) antigen. In some aspects, the antigen targeted by the antigen targeting receptor is a tumor-specific antigen including but not limited to CD19, CD20, BCMA, Her2, EGFRvlll, PSMA (prostate specific membrane antigen) and the FR (folate receptor) for example. In some aspects, the tumor-specific antigen is the folate receptor (FR), the o folate receptor, the b-folate receptor, the g-folate receptor, CD19, CD20, CD133, CD138, CEA, Claudin 18.2, EGFR, EGFRvlll, EphA2, EpCAM, GD2, GPC3, HER2, MSLN, MG7, MUC1 , NY-ESO-1 , LMP1 , PSMA, Fra, NKG2DI, BCMA, IL13Ralpha2, LeY, CD70, B7-H3, ROR1 , PSCA, or the like.

[0028] In one aspect, an engineered CAR-T-cell in which CAR sequences specific for the alpha (a) folate receptor (aFR) are‘knocked-in’ to the ATG5 gene locus of T-cells using CRISPR-Cas9 gene-editing. This generates a novel engineered CAR-T-cell (termed aFR- CAR-T) with ablated ATG5 gene activity/function that has improved anti-tumour activity (relative to ATG wild-type T-cells) for treatment of a-FR-expressing cancers including but not limited to ovarian, breast and lung cancer. Expression of the aFR-CAR following targeted integration into the ATG5 locus may be driven by the endogenous gene promoter or using heterologous promoters. Methods for nuclease and donor delivery are known by persons skilled in the art and can include electroporation of nucleic acids or

ribonucleoprotein (RNP) complexes or recombinant virus-mediated delivery.

[0029] In one aspect, an engineered CAR-T-cell in which CAR sequences specific for the alpha (a) folate receptor (aFR) are‘knocked-in’ to the ATG14 gene locus of T-cells using CRISPR-Cas9 gene-editing. This generates an engineered CAR-T-cell (termed aFR-CAR- T) with ablated ATG 14 gene activity/function that has improved anti-tumour activity (relative to ATG wild-type T-cells) for treatment of a-FR-expressing cancers including but not limited to ovarian, breast and lung cancer.

[0030] In one aspect, the aFR-CAR sequences for the knock-in are targeted to intron 2 of the ATG5 locus. In one aspect, the aFR-CAR is introduced by any suitable method (for example, via electroporation or suitable lentiviral vector or retroviral vector), and then ATG5 is knocked out. In some such aspects, ATG5 is knocked out at exon 4 or exon 5 of the ATG5 locus.

[0031] In one aspect, the sgRNA construct design for the CRISPR-Cas9-mediated knock-in to the ATG5 locus has the nucleotide sequence of SEQ ID NOs: 1-7 OR 24-25. [0032] In one aspect, the primary T-cells used to generate CAR-T knock-ins at various ATG gene loci of the invention, may be induced pluripotent stem cells, CD34+, CD4+ or CD8+ T- cells.

[0033] In one aspect, more than one ATG loci/gene (i.e. two or more different ATG genes) may be simultaneously edited for knock-in by a particular CAR sequence/construct for improved anti-tumour efficacy.

[0034] In one aspect, the aFR-CAR sequences for the knock-in are targeted to introns or exons (i.e. exons 1 , 2, 3, 4 etc.) other than intron 2 of the A7G5 locus/gene.

[0035] Further aspects of the invention will become apparent from consideration of the ensuing description of preferred embodiments of the invention. A person skilled in the art will realise that other embodiments of the invention are possible and that the details of the invention can be modified in a number of respects, all without departing from the inventive concept. Thus, the following drawings, descriptions and examples are to be regarded as illustrative in nature and not restrictive.

Brief Description of the Drawings

[0036] Exemplary embodiments are illustrated in referenced figures of the drawings. It is intended that the embodiments and figures disclosed herein are to be considered illustrative rather than restrictive.

[0037] FIG. 1 is a schematic diagram showing schematically an example embodiment of a chimeric antigen receptor (CAR).

[0038] FIG. 2 is a schematic diagram of an embodiment in which the DNA encoding the CAR has been inserted at a locus of the genome of the cytotoxic lymphocyte cell at a location other than the locus of the autophagy gene that has been disrupted.

[0039] FIG. 3 is a schematic diagram of an embodiment in which the DNA encoding the CAR has been inserted at the locus of an autophagy gene within the genome of a cytotoxic lymphocyte cell.

[0040] FIG. 4 shows an example embodiment of a method for producing genetically engineered lymphocytes for use in immunotherapy. [0041] FIG. 5 shows another example embodiment of a method for producing genetically engineered lymphocytes for use in immunotherapy.

[0042] FIG. 6 shows another example embodiment of a method for producing genetically engineered lymphocytes for use in immunotherapy.

[0043] FIG. 7, Panels A-E, show the results of an example demonstrating that autophagy enhances anti-tumor immunity and that this effect is dependent on T-cells.

[0044] FIG. 8, Panels A-K, shows the metabolomics of Atg5 /_ CD8+ T-cells.

[0045] FIG. 9, Panels A and B, show the results of an example demonstrating that Atg5 deficiency leads to increased effector memory CD8+ T-cells.

[0046] FIG. 10, Panels A-E, show the results of an example demonstrating that Atg5 _/

CD8+ T-cells have enhanced anti-tumor function.

[0047] FIG. 1 1 , Panels A-J, show the results of experiments demonstrating that Atg5 _/ T- cells have changes in histone trimethylation and increase in methylation at immune response gene loci.

[0048] FIG. 12, Panels A-l, show a CRISPR-Cas9 strategy and validation of its successful implementation for gene-editing at the ATG5 locus to target the CAR to the ATG5 locus with a concomitant functional knock out of ATG5 and targeted integration of a gene trap vector using CRISPR-Cas9.

[0049] FIG. 13 shows the results of experiments demonstrating the delivery of Cas9 RNP targeting ATG5 in primary T-cells.

[0050] FIG. 14 shows the expression of an example aFR-CAR construct on human T-cells.

[0051] FIG. 15, Panels A-E, show the results of experiments demonstrating efficient targeting of ATG5 and AAVS1 in CD34+ hematopoietic stem cells and activated CD8+ T- cells.

[0052] FIG. 16 shows the incorporation of the desired aFR-CAR targeting ATG5 into intron 2 of A TG5 in CD8+ T-cells.

[0053] FIG. 17 shows the strategy for incorporating a desired aFR-CAR construct transduced by a lentiviral vector into the T-cell genome followed by electroporation of A TG5 sgRNA into exon 4 or exon 5 of the ATG5 gene to knock out ATG5. Panel A shows the strategy for incorporating the construct into T-cells and knocking out ATG5. Panel B shows the results of TIDE analysis showing the successful deletion at exon 4 of A TG5.

Description

[0054] Throughout the following description, specific details are set forth in order to provide a more thorough understanding of the invention. However, the invention may be practiced without these particulars. In other instances, well known elements have not been shown or described in detail to avoid unnecessarily obscuring the invention. Accordingly, the specification and drawings are to be regarded in an illustrative, rather than a restrictive sense.

[0055] The inventors have now found methods and compositions to improve the anti-tumor efficacy of lymphocytes that have application, for example, in chimeric antigen receptor (CAR) T-cell therapy or T-cell receptor-engineered T-cell therapy. Gene-editing methods for the ablation of autophagy gene expression in lymphocytes including CAR-T-cells have been developed to enhance the therapeutic efficacy of the cells, including against solid tumors. In some embodiments, the gene-editing methods cause the engineered

lymphocytes, such as CAR-T-cells, to specifically target the alpha-folate receptor (aFR) in order to improve their effectiveness for treatment of cancers that express the aFR such as ovarian, breast and lung cancer.

[0056] In one aspect, the inventors have designed a novel CRISPR-Cas9 gene-editing strategy to engineer autophagy-deficient lymphocytes, including CAR-T-cells, directed against the ovarian cancer antigen folate receptor alpha (aFR) as an exemplary tumor- specific antigen. By using a gene-trap approach to target the aFR chimeric antigen receptor (CAR) as an exemplary antigen targeting receptor into the locus of an exemplary autophagy gene, autophagy-related gene 5 ( ATG5 ), the inventors have disrupted autophagy while concurrently placing the CAR under control of the endogenous ATG5 promoter. Expression of the CAR is therefore upregulated in areas of hypoxia and stress, such as in the solid tumor microenvironment.

[0057] Without being bound by theory, it is believed that placing the antigen targeting receptor, e.g. CAR construct or engineered T-cell receptor (TCR) construct, under control of a promoter, such as the endogenous autophagy gene promoter, that induces expression in response to stress, means that the level of expression of the antigen targeting receptor (e.g. CAR or TCR) will be low in the periphery of the body, but will increase in the tumor microenvironment. This could potentially reduce the toxicity associated with a high dose of cells expressing the antigen targeting receptor, e.g. CAR-T-cells. As an example, ATG5 is constitutively expressed at a low level, but its level of expression increases in response to stress.

[0058] Further without being bound by theory, autophagy deficient mouse T-cells do not generate memory after in vivo infection (25). Without being bound by theory, this may be beneficial for the safety of treatment using the genetically engineered lymphocytes because after the modified cells reach the peak of response, they will die (i.e. because there is no memory formation), thereby clearing all of the genetically engineered lymphocytes from the subject. This provides a measure of safety in administering treatment to patients.

[0059] In one embodiment, the inventors have determined that Atg5 deficient mice have significantly enhanced anti-tumor activity against hormone insensitive prostate and estrogen positive breast tumor cell lines. This anti-tumor response is dependent on T-cells and can be fully replicated with animals defective in other Atg genes (e.g. Atg14, ATG16L1). When T-cells deficient in Atg5 were used in an adoptive T-cell therapy experiments, the inventors observed a therapeutic effect on tumors when compared to wild-type CD8+ T-cells. Thus, the loss of the T-cell intrinsic ATG5 or other autophagy genes has been demonstrated to result in enhanced anti-tumor immune responses.

[0060] Although there are defects of Atg5 loss in other immune subsets including antigen presentation cells, without being bound by theory, the specific loss of autophagy in T-cells may have several benefits for T-cell therapy. Atg5 A T-cells from tumor bearing mice have been found to have high rates of glycolysis and low oxidative metabolism. This could be a metabolic advantage as increased glucose metabolism could make tumor infiltrating lymphocytes (TIL) more competitive for glucose. These metabolic changes are associated with Atg5 A T-cells skewing towards effector cells with enhanced antigen specific T-cell responses. Without being bound by theory, the reduced proliferation of Atg5 T-cells may have unintended benefits. Since Atg5 T-cells are highly antigen specific, infusions would require fewer cells to achieve equivalent anti-tumor responses and recipient patients may not need to have lymphodepletion; a procedure that can have significant risk of adverse events. Another feature of Atg5 T-cells is the metabolic-dependent change in histone trimethylation at specific loci of immune response genes and increase in transcriptional expression of those targets.

[0061] Such benefits provided by the suppression of an autophagy gene can be extended to other lymphocytes and other cells involved in tumor killing. Examples of such cells include NK cells; various types of T-cells including CD34+, CD4+ or CD8+ T-cells, Treg cells, tissue-resident memory T cells (TRM cells), natural killer T (NKT) cells; B-cells, and the like. These cells act through a similar mechanism of recognizing an antigen such as a tumor-specific antigen on the surface of the tumor cell and acting to kill such tumor cell. Without being bound by theory, suppression of autophagy in such cells can enhance effector anti-tumor activity via metabolomics shifts to more glycolysis and greater IFNy secretion. Also in the case of Treg cells, suppression of autophagy could also result in loss of suppressor function against effector T cells.

[0062] In some embodiments, an engineered lymphocyte such as a CAR T-cell having one or more disruptions in an autophagy gene or an otherwise suppressed autophagy gene is provided. In some embodiments, the disruptions in the autophagy gene suppress or ablate expression of the autophagy gene. In some embodiments, the disruption in the autophagy gene is provided using a‘knock-in’ genetic engineering strategy. In some embodiments, the disruption in the autophagy gene is provided by using a knock-out genetic engineering strategy. In alternative embodiments, any desired strategy could be used to suppress one or more autophagy genes in the lymphocyte, e.g. a CAR T-cell. Likewise, any suitable genetic engineering strategy could be used to cause the engineered lymphocyte, e.g. CAR T-cell, to express the desired antigen targeting receptor, e.g. a chimeric antigen receptor (CAR) or an engineered T-cell receptor. For example, cells can be transduced with a viral vector (e.g. a lentiviral vector or a retroviral vector) to introduce a nucleotide construct encoding the CAR or engineered T-cell receptor into the cells. In some embodiments, a CRISPR-Cas9 gene editing system (which includes a CRISPR-Cas gene editing system using any suitable Cas protein, e.g. Cas9, Cas12a, or the like) or other similar gene editing technique can be used to disrupt the autophagy gene and/or introduce the antigen targeting receptor construct into the cell for expression. In some embodiments, a combination of different genetic engineering techniques could be used to both disrupt the autophagy gene and introduce the CAR construct into the T-cell for expression. In some embodiments, zinc- finger nucleases (ZFN), transcription activator-like effector nuclease (TALEN), Sleeping Beauty (SB), RNAi, meganucleases, megaTALs, or other gene knockout methods could be used to disrupt or suppress the autophagy gene.

[0063] In some embodiments, the engineered lymphocyte, e.g. CAR T-cell, expresses an antigen targeting receptor, e.g. a chimeric antigen receptor (CAR) or an endogenous or engineered T-cell receptor, that is selective for a tumor-specific antigen. In some embodiments, the tumor-specific antigen is the a-folate receptor (a-FR), which is an antigen that is highly expressed in certain kinds of cancer including ovarian cancer, breast cancer and lung cancer, but which is not expressed at a high level by normal cells. In some embodiments, the tumor-specific antigen is the folate receptor (FR), the b-folate receptor, the g-folate receptor, CD19, CD20, BCMA, Her2, EGFRvlll, or prostate specific membrane antigen (PSMA). In some embodiments, the tumor-specific antigen is the folate receptor (FR), the a-folate receptor, the b-folate receptor, the g-folate receptor, CD19, CD20, CD133, CD138, CEA, Claudin 18.2, EGFR, EGFRvlll , EphA2, EpCAM, GD2, GPC3, HER2, MSLN, MG7, MUC1 , NY-ESO-1 , LMP1 , PSMA, Fra, NKG2DI, BCMA, IL13Ralpha2, LeY, CD70, B7-H3, ROR1 , PSCA, or the like. In alternative embodiments, the tumor-specific antigen targeted by the antigen targeting receptor can be any desired tumor-specific antigen. A non-exhaustive list of exemplary tumor-specific antigens currently being evaluated for treatment of certain cancers is given in Table 1. In some embodiments, the cancer is B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), B-cell lymphoma or other lymphoid malignancy, liver, pancreatic, brain, breast, ovarian, colorectal, acute myeloid leukemia (AML), multiple myeloma, lung, gastric, glioma, EGFR-positive solid tumor, glioblastoma, glioblastoma multiforme, stomach, nasopharyngeal, esophageal, prostate, neuroblastoma, hepatocellular, squamous cell lung, MSLN-positive solid tumor, non-small-cell lung (NSCLC), triple-negative breast cancer (TNBC), sarcoma, advanced solid tumor, renal cell, central nervous system, or an ROR1-positive malignancy.

Table 1. Exemplary tumor-specific antigens that can be targeted with antigen targeting receptor constructs such as a CAR.

[0064] In some embodiments, the antigen targeting receptor, e.g. CAR construct or endogenous or engineered T-cell receptor (TCR) construct, is inserted at the locus of the autophagy gene that is to be suppressed using a knock-in strategy. This places the antigen targeting receptor, e.g. CAR construct or TCR construct, under control of the endogenous promoter of the autophagy gene. In some embodiments, the antigen targeting receptor, e.g. CAR construct or TCR construct, is inserted in the lymphocyte’s (e.g. T-cell) genome at a locus other than the autophagy gene that is to be suppressed. In some such

embodiments, expression of the antigen targeting receptor, e.g. CAR construct or TCR construct, is controlled by a heterologous promoter. In some embodiments, the antigen targeting receptor, e.g. CAR construct or TCR construct, is inserted together with a desired exogenous promoter to control expression of the antigen targeting receptor, e.g. CAR or TCR, by the lymphocyte (e.g. T-cell) under the control of an exogenous promoter. In some embodiments, the antigen targeting receptor (e.g. CAR or TCR construct) is placed under control of a promoter that increases expression of the antigen targeting receptor in response to stress. In some embodiments, the promoter that increases expression of the antigen targeting receptor in response to stress is a promoter of an autophagy gene.

[0065] In some embodiments, rather than expressing a CAR, the lymphocyte is engineered to express an endogenous or engineered T-cell receptor as the antigen targeting receptor. TCR-engineered T-cells are currently being developed for use in various types of immunotherapy, including the treatment of solid tumors, and can be used in a manner similar to CAR-T cells to specifically target and kill cells expressing a particular antigen, e.g. a tumor-specific antigen.

[0066] In some embodiments, the autophagy gene ( ATG ) that is suppressed in the lymphocyte is any gene that is now known or is later discovered to be essential for autophagy. In some embodiments, the autophagy gene is one or more of ATG†, ATG4, ATG5, ATG7, ATG8, ATG13, ATG18, ATG16L1, and GABARAP. In some embodiments, the autophagy gene is one or more of ULK1, ULK2, ULK3, FIP200, Vps34, Beclin-1, p150, UVRAG, ATG1, ATG4, ATG5, ATG7, ATG8, ATG9, ATG10, ATG12, ATG13, ATG14L, ATG16L, ATG16L1, ATG18, VMP1, GABARAP, or the like.

[0067] In some embodiments, the CAR construct has a targeting moiety, a transmembrane domain and a CD3z intracellular domain. In some embodiments, the targeting moiety is an antigen-binding fragment of an antibody. In some embodiments, the targeting moiety is an ScFV of an antibody. In alternative embodiments, any CAR construct now known or later developed could be used. [0068] In alternative embodiments, an engineered T-cell receptor is used as the antigen targeting receptor. An example of an engineered T-cell receptor has an a chain and a b chain, each containing a variable domain (v) and a constant domain (c), as well as a transmembrane domain and 6 CD3 chains for T-cell activation. In alternative embodiments, any T-cell receptor construct now known or later developed could be used.

[0069] An example embodiment of a CAR construct 30 for use in some embodiments is shown in FIG. 1 . The CAR construct has a targeting moiety 32, e.g. a suitable ScFV that targets a desired tumor-specific antigen, a transmembrane domain 34, and a CD3z intracellular domain 38. In some embodiments, CAR construct 30 can have other domains such as a suitable co-stimulatory domain 36 (e.g. CD27, CD28, 4-1 BB, ICOS, 0X40, MYD88, IL1 R1 , CD70, or the like), or other domains intended to enhance the characteristics of the CAR construct.

[0070] FIG. 2 shows schematically an example embodiment of an engineered lymphocyte 50. Engineered lymphocyte 50 has been genetically modified to suppress one or more genes essential for autophagy at a locus 52 of the genomic DNA of the lymphocyte. The gene essential for autophagy is under control of an endogenous promoter 56. Engineered lymphocyte 50 has also been genetically modified to express a desired antigen targeting receptor construct at a locus 54 of the genomic DNA of the lymphocyte which is different from locus 52. The expression of the antigen targeting receptor construct from locus 54 is under control of a promoter 58, which can be an endogenous promoter in some

embodiments or a heterologous promoter in some embodiments.

[0071] FIG. 3 shows schematically an example embodiment of an engineered lymphocyte 70. Engineered lymphocyte 70 has been genetically modified to both suppress one or more genes essential for autophagy and express a desired antigen targeting receptor construct at a locus 72 of the genomic DNA of the lymphocyte. For example, a knock-in strategy can be employed to both disrupt the gene essential for autophagy and insert the desired antigen targeting receptor construct at locus 72 of the genomic DNA of the lymphocyte. In this embodiment, expression of the desired antigen targeting receptor construct from locus 72 is under control of the endogenous promoter 76 of the gene essential for autophagy.

[0072] Engineered lymphocytes 50, 70, or modified lymphocytes, including engineered T- cells, according to any other embodiment, can be made via any suitable genetic engineering technique now known or later made available to one skilled in the art. In one example embodiment shown in FIG. 4, a method 100 of creating modified lymphocytes such as T-cells using a gene editing strategy to insert the desired antigen targeting receptor construct at the locus of the autophagy gene to be disrupted is shown. At step 102, the desired lymphocytes such as T-cells are obtained from a source of cells. The source of the cells may be any suitable source, for example, the subject to whom the immunotherapeutic lymphocytes are to be administered, a healthy donor, a pluripotent stem cell line, or the like. In some embodiments, the lymphocytes are autologous cells, i.e. the lymphocytes are acquired from the subject to be treated. In some embodiments, the lymphocytes are allogenic, i.e. obtained from source of cells other than the subject to be treated, such as a healthy donor or a cell line such as induced pluripotent stem cells. Any source of lymphocytes now known or later discovered can be used in certain embodiments.

[0073] At step 104, the lymphocytes are genetically engineered to insert DNA encoding the antigen targeting receptor construct in the genomic DNA of the lymphocytes and suppress the function of at least one autophagy gene. In some embodiments, step 104 is carried out using a CRISPR-Cas gene editing strategy, by devising suitable sgRNA to insert the antigen targeting receptor construct at an appropriate location within an autophagy gene so that the antigen targeting receptor construct can be inserted and the autophagy gene simultaneously suppressed using a knock-in strategy. The sgRNA, DNA encoding the antigen targeting receptor construct, and a Cas protein such as Cas9 are delivered to the lymphocyte in any suitable manner, for example via electroporation or chemical transfection techniques, or other suitable techniques as appropriate, for example using a delivery system based on a lentivirus, adenovirus, or adeno-associated virus.

[0074] At step 106, the genetically engineered lymphocytes in which the antigen targeting receptor construct has been successfully inserted at the correct locus are expanded. At step 108, the engineered lymphocytes are introduced into the subject as an

immunotherapeutic.

[0075] FIG. 5 shows an alternative embodiment of a method 150 of creating modified lymphocytes such as T-cells for immunotherapy. At step 152, lymphocytes are obtained from a source of cells in a similar manner as described for step 102. At step 154, the lymphocytes, e.g. T-cells, are genetically engineered in any suitable manner to suppress one or more autophagy genes. Examples of techniques that can be used to suppress the autophagy gene include ablation via gene knock out techniques using CRISPR-Cas,

TALEN, ZFN, SB, meganucleases, megaTALs, or suppression via gene knockdown via RNAi, or other suitable techniques.

[0076] At step 156, in some embodiments, the genetically engineered lymphocytes in which the autophagy gene has been suppressed are expanded. At step 158, the lymphocytes are introduced into the subject as an immunotherapeutic.

[0077] FIG. 6 shows an alternative embodiment of a method 170 of creating modified lymphocytes such as T-cells for immunotherapy. Steps 172 and 174 are carried out in like manner to steps 152 and 154 of method 150 to produce lymphocytes, e.g. T-cells, that are genetically engineered to suppress one or more autophagy genes. At step 176, the lymphocytes, e.g. T-cells, are further genetically engineered in any suitable manner to express a desired antigen targeting receptor, such as a CAR construct or engineered T-cell receptor construct. Examples of techniques that may be used to engineer the lymphocytes, e.g. T-cells, to express a desired antigen targeting receptor such as a CAR construct or engineered T-cell receptor include CRISPR-Cas editing systems or transduction with a suitable lentiviral or retroviral vector. At step 178, the lymphocytes, e.g. T-cells, that have been genetically engineered to have both a suppressed autophagy gene and to express the desired antigen targeting receptor, e.g. CAR construct or engineered T-cell receptor, are expanded. At step 180, the expanded cells are introduced into the subject as an immunotherapeutic. In alternative embodiments, the order in which steps 174 and 176 are carried out can be reversed, that is the lymphocytes, e.g. T-cells, can be first engineered to express the desired antigen targeting receptor, e.g. CAR construct or engineered T-cell receptor, and then engineered to suppress one or more autophagy genes.

[0078] In some embodiments, the genetically engineered lymphocytes, e.g. T-cells, are used as an immunotherapeutic to treat cancer. In some embodiments, the cancer is ovarian cancer, breast cancer, or lung cancer. In some embodiments, the cancer is B-cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), B-cell lymphoma or other lymphoid malignancy, liver, pancreatic, brain, breast, ovarian, colorectal, acute myeloid leukemia (AML), multiple myeloma, lung, gastric, glioma, EGFR-positive solid tumor, glioblastoma, glioblastoma multiforme, stomach, nasopharyngeal, esophageal, prostate, neuroblastoma, hepatocellular, squamous cell lung, MSLN-positive solid tumor, non-small-cell lung (NSCLC), triple-negative breast cancer (TNBC), sarcoma, advanced solid tumor, renal cell, central nervous system, or an ROR1-positive malignancy. The genetically engineered lymphocytes, e.g. T-cells, can be administered to the subject in any suitable manner, for example via intravenous diffusion.

[0079] In some embodiments in which a CRISPR-Cas strategy is used to introduce the antigen targeting receptor construct into the lymphocyte for expression, the CRISPR-Cas strategy is also used to simultaneously suppress expression of an autophagy gene.

Persons skilled in the art can develop appropriate single guide RNAs (sgRNAs) to effect the insertion of the antigen targeting receptor, e.g. CAR, at a desired location in the genome of the lymphocyte. In some embodiments, the CRISPR-Cas strategy is used to insert the antigen targeting receptor, e.g. CAR, within the locus of the ATG5 gene in the genome of the lymphocyte. In some embodiments, the CRISPR-Cas strategy is used to insert the antigen targeting receptor, e.g. CAR, within intron 2 of the ATG5 gene. In some

embodiments, the CRISPR-Cas strategy is used to disrupt the ATG5 gene at exon 4 or exon 5 of the ATG5 gene while the antigen targeting receptor is inserted at a different locus within the genome, for example using transduction with an appropriate lentiviral, retroviral or adeno-associated viral vector. In some embodiments, the CRISPR-Cas strategy is used to insert DNA encoding an engineered T-cell receptor rather than DNA encoding a CAR.

[0080] In one example embodiment, the sgRNA used to target intron 2 of A TG5 has the sequence of one of SEQ ID NOs:1-7 listed in Table 2. In one example embodiment, the sgRNA used to target exon 4 of ATG5 has the sequence of SEQ ID NO:24. In one example embodiment, the sgRNA used to target exon 5 of ATG5 has the sequence of SEQ ID NO:25.

[0081] In one example embodiment, the nucleotide construct used to insert the CAR into the T-cells has one of SEQ ID NO:8 or SEQ ID NO:9.

Examples

[0082] Certain embodiments are further described with reference to the following examples, which are intended to be illustrative and not limiting in nature. While the following examples demonstrate the suppression of an autophagy gene and expression of a CAR in T-cells, the techniques described below are equally applicable to conduct the genetic engineering of other types of lymphocytes to suppress expression of an autophagy gene and express a desired CAR or engineered T-cell receptor.

[0083] The inventors designed and tested several single guide RNAs (sgRNAs) targeted to the locus of autophagy-related gene 5 ( ATG5 ), and quantified indel formation by

sequencing and mismatch cleavage assay. The inventors optimized conditions for homology-directed repair (HDR) in K562 cells using a fluorescent reporter construct. Out- out polymerase chain reaction (PCR) and Western blot were used to evaluate construct integration and autophagy activity. To identify transfection parameters that would yield optimal editing and expansion in human T-cells, the inventors then performed a series of electroporations with sgRNA and recombinant Cas9 protein. The inventors used these electroporation parameters in combination with adeno-associated virus (AAV) vectors to target the aFR chimeric receptor into the locus of ATG5 in healthy donor T-cells.

[0084] The inventors observed loss of functional autophagy in clones with donor integration. Furthermore, the inventors confirmed that these results were due to on-target editing by delivering ATG5 cDNA to an intergenic locus and restoring autophagy function. After screening multiple transfection parameters (cell density, RNP concentration, electroporation pulse code, and addition of an anionic polymer) the inventors identified a set of conditions for RNP delivery that yielded upwards of 80% indel formation in healthy donor T-cells. Using these parameters in conjunction with an AAV vector encoding the aFR CAR, the inventors successfully generated 24.1 % ATG5 ~ ' ~ aFR-CAR-T-cells by CRISPR-mediated homology directed repair.

[0085] FIG. 7 shows the results of experiments demonstrating that deletion of autophagy enhances anti-tumor immunity and is dependent on T-cells. The inventors made an inducible Cre-ERT2 mouse to delete Atg5 in all tissues upon tamoxifen injection. 4 days post tamoxifen treatment, e0771 breast (Panel A) or TRAMP-C2 prostate (Panel B) tumor cell lines were injected subcutaneously and tumor volume measured over time. n=12-13 mice per experimental group.

[0086] Panel C shows using a different autophagy gene, Atg14, that Atg14-dependent enhancement of anti-tumor immunity is cell-intrinsic to the T-cells. The inventors performed bone-marrow chimera experiments where wild type (WT) mice were reconstituted with Atg5 or Atg5 +/ bone marrow (BM). Atg5 /_ BMC mice show enhanced tumor rejection. n=10 mice per experimental group. Similar experiments were performed using Atg14 /_ or Atg14 +/ BM (data not shown).

[0087] Panel D shows that antibody depletion of CD8+ T-cells (achieved using an anti-CD8 antibody) results in loss of tumor control in Atg5-/- mice. n=4-7 mice per experimental group. ****p<0.0001 , two-way ANOVA. Error bars indicate +/- SEM. Panel E shows the adoptive transfer of naive CD8+ Atg5 deficient (AA) or Atg5 wildtype (Aa) T-cells into EG7 tumor bearing mice. Tumors were implanted and donor T-cells transferred on Day 1 1 (ACT). **** p <0.0001 , Student’s t test. Error bars indicated +/- SD.

[0088] FIG. 8 shows the results of experiments evaluating the metabolomics of Atg5 /_ CD8+ T-cells and showing that Atg5 /_ CD8+ T-cells are more glycolytic. *p<0.01 , **p<0.01 , ***p<0.001 , ****p<0.001. Panel A shows the relative lactate levels identified by

metabolomics. The error bars indicate +/- SD. Panel B shows oxygen consumption rate (OCR) results for CD8+ T-cells that were isolated from spleens of e0771 tumour bearing mice and were subjected to Seahorse Bioanalyzer in the presence or absence of oligomycin, FCCP and Antimycin/Rotenone (Ant/Rot). Data was normalized to protein concentration. Error bars indicate +/- SEM. OCR (Panel C), extracellular acidification rate (ECAR) (Panel D) and OCR:ECAR (Panel E) ratio of Atg5 +/ and Atg5 /_ CD8+ T-cells was examined at basal levels. Atg5 /_ T-cells exhibit an increased oxygen consumption rate and increased extracellular acidification rate.

[0089] Panel F on the left shows a representative flow cytometry plot showing fluorescent glucose analog 2-NBDG uptake in splenic Atg5 +/ (lower trace) and Atg5 /_ (upper trace) CD8+ T-cells from e0771 tumour bearing mice. Graph on the right half of Panel F represents the mean fluorescence intensity (MFI) of 2-NBDG +/- SEM, with Atg5 /_ exhibiting considerably higher fluorescence and therefore higher glucose uptake.

[0090] Panel G shows quantitative RT-PCR of Hxk2 expression in Atg5 +/ and Atg5 /_ CD8+ T-cells. Results are relative to Actb. Data are expressed as average +/- SEM. Panel H shows spare respiratory capacity, indicated by baseline OCR subtracted from maximal OCR, and Panel I shows mitochondrial mass as measured by MitoTracker Green, as determined in Atg5 +/ and Atg5 /_ CD8+ T-cells isolated from e0771 tumour bearing mice. Data are expressed as average +/- SEM. n=4-5 mice per group. [0091] Panel J shows T metabolomics analysis for cells from tumor bearing Atg5 /_ (n=5) or Atg5 +/ (n=4) mice that were harvested on day 14 post-tumor implantation and (10,000 cells). Panel K shows a volcano plot analysis which revealed a significant increase in glycolysis (e.g. lactate) with concomitant reductions in oxidative metabolism (e.g. glutamate, aspartate).

[0092] FIG. 9 shows the results of experiments demonstrating that Atg5 deficiency leads to increased effector memory CD8+ T-cells. Panel A shows representative flow cytometry plots showing naive (CD62LhiCD44lo), central memory (CD62LhiCD44hi) and effector memory (CD62LloCD44hi) CD8+ T-cells isolated from blood (left side), spleen (centre) and tumours (right side) of e0771 tumour bearing mice. Panel B shows the percentages of CD62LloCD44hi effector memory CD8+ T-cells in blood, spleen and tumours from e0771 tumour bearing mice. * p<0.05, ** p<0.01.

[0093] With reference to FIG. 10, the inventors have carried out experiments demonstrating that Atg5 _/ CD8+ T-cells have enhanced anti-tumor function. The left two images of Panel A show representative flow cytometry plots showing IFNy and TNFa expression following PMA/ionomycin stimulation of Atg5 +/ and Atg5 /_ CD8+ T-cells from Tramp-C2 tumour bearing mice. The graphs shown to the right of Panel A represent the percentage of IFNy+TNFa+ CD8+ T-cells in e0771 and Tramp-C2 tumour bearing mice +/- SEM, with the percentage being higher for Atg5 /_ cells. Panel B shows serum from e0771 or Tramp-C2 tumour bearing mice as analyzed by ELISA. Graphs represent the average +/- SEM.

Levels of IFNy were higher for Atg5 /_ mice. Panel C shows splenocytes harvested from Atg5 +/ and Atg5 _/ e0771 tumour bearing mice which were were stimulated with e0771 cells, and IFNy ELISPOT assays were performed. Data are expressed as average +/- SEM. In another experiment, splenocytes harvested from Tramp-C2 tumour bearing mice were stimulated with Tramp-C2 cells (Panel D) or Spas-1 peptide (Panel E), and IFNy ELISPOT assays were performed. Data are expressed as average +/- SEM. *p<0.05, **p<0.01 , ***p<0.001 , ****p<0.0001. Increased TNFa and IFNy are observed in Atg5 /_ T-cells.

[0094] With reference to FIG. 1 1 , the inventors conducted experiments to show that Atg5 _/ T-cells have changes in histone trimethylation and increase in methylation at immune response gene loci. Panel A shows pathway and gene ontology analysis of H3K4me3- marked promoters unique to Atg5 versus Atg5 +/~ CD8+ T-cells are strongly enriched in genes related to T-cell activation and adaptive immunity (Benjamini q value <10e-12).

Panel B shows differential normalized tagged density of H3K27me3 and H3K4m3 in a subset of immune response genes. Panels C and D show ChIP-Seq on Atg5 +/~ and Atg5 CD8+ T-cells for H3K4me3 for Ifng. Panel C shows normalized tag density of Ifng in knockout or control CD8+ T-cells. Panel D shows quantitative RT-PCR of Ifng expression in Atg14 +/~ and Atg14 CD8+ T-cells. Results are relative to Actb. Data are expressed as average +/- SEM of a triplicate experiment with at least 2 mice per group. Panels E-G show ChIP-Seq on Atg5 +/~ and Atg5 A CD8+ T-cells for H3K4me3 for Glutl. Panel E shows normalized tag density of Glutl in knockout or control CD8+ T-cells. Panel F shows quantitative RT-PCR of Glutl expression in Atg5 +/~ and Atg5 CD8+ T-cells or (Panel G) Atg14 +/~ and Atg14 CD8+ T-cells. Results are relative to Actb. Data are expressed as average +/- SEM of a triplicate experiment with at least 2 mice per group. Panels H-J show ChIP-Seq on Atg5 +/~ and Atg5 ~ CD8+ T-cells for H3K4me3 for Hk2. Panel H shows normalized tag density of Hk2 in knockout or control CD8+ T-cells. Panel I shows quantitative RT-PCR of Hk2 expression in Atg5 +/~ and Atg5 A CD8+ T-cells or (Panel J) Atg14 +/~ and Atg14 CD8+ T-cells. Results are relative to Actb. Data are expressed as average +/- SEM of a triplicate experiment with at least 2-3 mice per group. *p<0.01 , **p<0.01 , ****p<0.001 , n.s. not significant.

[0095] With reference to FIG. 12, a CRISPR-Cas9 strategy and validation for gene-editing at the ATG5 locus was developed for the targeting of the CAR to the ATG5 locus.

Concomitant functional knock out of ATG5 and targeted integration of a gene trap vector using CRISPR-Cas9 was confirmed. Panel A shows a schematic of the ATG5 locus and the regions screened to identify functional SpCas9-sgRNAs. Exons E2 and E3 of the ATG5 gene are shown as open boxes. Genomic structure and target regions within intron 2 (I2) of ATG5 are illustrated schematically. A, B, and C denote non-repetitive target DNA sequences used to design sgRNAs using the online CRISPOR tool. Intron 2 was selected as a preferential target region since all reported mRNA isoforms of ATG5 include this region (i.e. this ensures that all potential transcripts are captured).

[0096] Panel B of FIG. 12 shows the sgRNA sequences identified by CRISPOR targeting different regions of intron 2 to arrive at the sgRNA sequences shown in Table 2. Table 2. Example sgRNA sequences for ATG5 ablation via insertion in intron 2.

[0097] Panel C of FIG. 12 shows the identification of active sgRNAs using the surveyor nuclease assay. In this assay, a mismatch sensitive nuclease is used to determine the frequency of the small insertions and deletions (indels) indicative of nuclease activity. As an example, sgRNA 824 appears to be the most active nuclease for section I2A of intron 2 and was selected for further experiments.

[0098] Panel D of FIG. 12 shows a schematic of the ATG5 locus following CAR addition. The first and second schematics show the genomic structure of the CAR-T integration site and donor template. Shown are the locations of the splice acceptor site (SA), 2A self cleaving peptide sequence (2A), polyadenylation sequence (pA), and homology arms left and right (HA-L, HA-R). The third schematic shows the integrated CAR construct at the ATG5 locus.

[0099] Panel E of FIG. 12 shows out-out PCR showing integration of a test SA-2A- mScarlet-pA reporter sequence within intron 2 of ATG5. Complete integration at all alleles is observed for K562 clones 5,6,7,8, 10. Panel F is a Western blot showing loss of ATG5 in K562 clones correlates with complete integration of the mScarlet construct (i.e. loss of ATG5 is observed for clones 5,6,7,8,10. Panel G is an immunofluoresence image of K562 cells showing nuclear staining (left) and mScarlet expression (right), showing good expression of the mScarlet reporter construct. Panel H is a Western blot showing response to treatment with hydroxychloroquine and/or rapamycin. Loss of LC3-II indicating inhibited autophagic flux is observed in K562 clones 5,6,7 (clones 8 and 10 were not tested). Panel I is a Western blot showing that for clones for which an ATG5 cDNA was targeted to an intergenic locus (AAVS1) to restore expression of ATG5 (clones 5.1 , 5.3 and 5.6), autophagy was restored as confirmed by the return of LC3-II.

[0100] FIG. 13 shows the results of experiments demonstrating delivery of Cas9 RNP targeting ATG5 in primary T-cells. Purified CD3 cells stimulated with CD3/CD28 beads were electoporated with recombinant SpCas9 complexed with a chemically modified sgRNA (824) using the amaxa 4D Nucleofector. Two different pulses were tested. Genomic DNA was extracted 3 days post transfection and the Surveyor assay was performed to determine the frequency of indels.

[0101] FIG. 14 shows the expression of an exemplary aFR-CAR (CD3zBB) with a GFP reporter from Human CD8+ T-cells transduced with lentiviruses encoding the construct.

The left panel is empty vector. The right panel is the aFR-CAR construct showing expression of the desired protein on human T-cells.

[0102] The sequences of the CAR constructs used to insert the aFR-CAR at the ATG5 locus are given as SEQ ID NOs:8 and 9The sequences of the CAR constructs used to insert the aFR-CAR within the AAVS1 locus as a control are given as SEQ ID NOs:10 and 1 1. SEQ ID NO:8 encodes a CAR construct having an anti-folate receptor ScFv, a transmembrane domain, a CD27 co-stimulatory domain, and a CD3z intracellular domain. SEQ ID NO:9 encodes a CAR construct having an anti-folate receptor ScFv, a

transmembrane domain, a CD28 co-stimulatory domain, and a CD3z intracellular domain. SEQ ID NO: 10 encodes a CAR construct having an anti-folate receptor ScFv, a

transmembrane domain, a CD27 co-stimulatory domain, and a CD3z intracellular domain. SEQ ID NO: 1 1 encodes a CAR construction having an anti-folate receptor ScFv, a transmembrane domain, a CD28 co-stimulatory domain, and a CD3z intracellular domain.

[0103] FIG. 15 shows the efficient targeting of ATG5 and AAVS1 in CD34+ hematopoietic stem cells and activated CD8+ T-cells (no donor CAR construct was present in these experiments). Panel A is a histogram showing editing efficiency at ATG5 and AAVS1 in cord blood derived CD34+ HSCs after electroporation with Cas9 protein and 100-300 pmol sgRNA as indicated. Genomic DNA from each target region was amplified and sequenced, and the sequence traces analyzed using the decomposition algorithm TIDE. Panels B and C are flow cytometry dot plots showing 47.5% viability ( ATG5) and 31.6% viability ( AAVS1 ) in CD8+ T-cells three days after electroporation with a ribonucleoprotein (RNP) complex consisting of Cas9 protein and sgRNA. Panels D and E are histograms showing 54.1 % overall editing efficiency at ATG5 and 53.9% overall editing efficiency at AAVS1 in CD8+ T- cells as determined by TIDE analysis.

[0104] FIG. 16 shows the validation of the incorporation of the CAR construct into intron 2 of A TG5 in healthy donor T-cells. CD8+ T-cells were isolated from healthy donor PBMCs by magnetic bead separation, stimulated with TransAct (Miltenyi), and cultured in 100U/ml IL-2. On day 4 post-activation, the cells were electroporated with an RNP consisting of 100pmol sgRNA targeting ATG5 and 61 pmol Cas9 protein. 15 minutes post-electroporation, C4-CD28-CD3z AAV (having SEQ ID NO:9) was given at an MOI of 2e5. The cells were expanded for 13 days and then stained for flow cytometry on a Cytek Aurora. The results show that 24.1 % of the cells were the desired ATG5 _/ aFR-CAR-T-cells.

[0105] FIG. 17 shows the strategy for incorporating a desired aFR-CAR transduced by a lentiviral vector into the T-cell genome followed by electroporation of sgRNA targeting ATG5 into exon 4 or exon 5 of the ATG5 gene using the sgRNA sequences shown in Table 3. Panel A shows the strategy for incorporating the construct into T-cells and ablating ATG5 at exon 4 (Target 56) or exon 5 (Target 150). Panel B shows the results of TIDE analysis showing the successful deletion at exon 4 of ATG5 using an sgRNA having the sequence of SEQ ID NO:24 (Target 56).

Table 3. Example sgRNA sequences for ATG5 ablation via insertion in exon 4 or exon 5.

The foregoing example demonstrates that a desired antigen targeting receptor construct can be integrated into the genome of a lymphocyte such as a T-cell at one locus in the genome, while an autophagy gene such as ATG5 can be knocked out at a different locus using CRISPR-Cas gene editing.

References

[0106] The following references are of interest with respect to the subject matter described herein. Each one of the following references is incorporated by reference herein in its entirety.

1. Geyer MB, Brentjens RJ. 2016. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T-cells. Cytotherapy 18: 1393-409

2. Kershaw MH, et al. , 2006. A phase I study on adoptive immunotherapy using gene- modified T-cells for ovarian cancer. Clin Cancer Res 12: 6106-15

3. Zhao Z, et al., 2015. Structural Design of Engineered Costimulation Determines Tumor Rejection Kinetics and Persistence of CAR T-cells. Cancer Cell 28: 415-28

4. Milne K, et al., 2009. Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors. PLoS One 4: e6412

5. Cheung A, et al., 2016. Targeting folate receptor alpha for cancer treatment.

Oncotarget

6. Newick K, et al., 2017. CAR T-cell Therapy for Solid Tumors. Annu Rev Med 68:

139-52

7. Townsend KN, et al., 2013. Markers of T-cell infiltration and function associate with favorable outcome in vascularized high-grade serous ovarian carcinoma. PLoS One 8: e82406

8. Vander Heiden MG, DeBerardinis RJ. 2017. Understanding the Intersections

between Metabolism and Cancer Biology. Cell 168: 657-69

9. Chang CH, Pearce EL. 2016. Emerging concepts of T-cell metabolism as a target of immunotherapy. Nat Immunol 17: 364-8

10. Chang CH, et al., 2015. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Ce// 162: 1229-41

11. MacPherson S, et al., 2017. STAT3 regulation of citrate synthase is essential during the initiation of lymphocyte cell growth. Cell Rep 19(5):910-918

12. Ma EH, et al., 2017. Serine Is an Essential Metabolite for Effector T-cell Expansion.

Cell Metab 25: 345-57 13. Scharping NE, et al., 2016. The Tumor Microenvironment Represses T-cell

Mitochondrial Biogenesis to Drive Intratumoral T-cell Metabolic Insufficiency and Dysfunction. Immunity

14. Lum JJ, DeBerardinis RJ, Thompson CB. 2005. Autophagy in metazoans: cell

survival in the land of plenty. Nat Rev Mol Cell Biol 6: 439-48

15. Pua HH, He YW. 2009. Autophagy and lymphocyte homeostasis. Curr Top Microbiol Immunol 335: 85-105

16. Schlie K, et al., 2015. Survival of effector CD8+ T-cells during influenza infection is dependent on autophagy. J Immunol 194: 4277-86

17. Puleston DJ, et al., 2014. Autophagy is a critical regulator of memory CD8+ T-cell formation. Elite 3

18. Qu X, et al., 2003. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J Clin Invest 1 12: 1809-20

19. Takamura A, et al., 201 1. Autophagy-deficient mice develop multiple liver tumors.

Genes Dev 25: 795-800

20. Macintyre AN, et al., 2014. The glucose transporter Glutl is selectively essential for CD4 T-cell activation and effector function. Cell Metab 20: 61-72

21. Hultquist JF, et al., 2016. A Cas9 Ribonucleoprotein Platform for Functional Genetic Studies of HIV-Host Interactions in Primary Human T-cells. Cell Rep 17: 1438-52

22. Eyquem J, et al., 2017. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543: 1 13-7.

23. DeVorkin L, et al., 2019. Autophagy regulation of metabolism is required for CD8+ T-cell anti-tumour immunity. Cell Rep 27(2):502-513.

24. Stadtmauer, EA, et al. , 2020. CRISPR-engineered T cells in patients with refractory cancer. Science 06 Feb 2020. DOI: 10.1 126/science. aba7365.

25. Schlie et al., 2015. Survival of effector CD8+ T-Cells during influenza infection is dependent on autophagy. J. Immunol. 194(9):4277-4286.

[0107] While a number of exemplary aspects and embodiments have been discussed above, those of skill in the art will recognize certain modifications, permutations, additions and sub-combinations thereof. It is therefore intended that the following appended claims and claims hereafter introduced are interpreted to include all such modifications, permutations, additions and sub-combinations as are consistent with the broadest interpretation of the specification as a whole.