Atty Docket No.23887-2000140 USE OF THERMAL BIOSWITCHES FOR TREATING AUTOIMMUNE DISEASES CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of, and priority to, U.S. Provisional Application 63/610,542, filed on December 15, 2023, the contents of which are hereby incorporated herein by reference in their entirety. REFERENCE TO AN ELECTRONIC SEQUENCE LISTING [0002] The contents of the electronic sequence listing (238872000140SEQLIST.xml; Size: 126,037 bytes; and Date of Creation: December 13, 2024) is herein incorporated by reference in its entirety. BACKGROUND Field [0003] The present disclosure relates generally to the field of T-cell therapy for treating diseases or disorders in a subject (e.g., an autoimmune disease or disorder). Description of the Related Art [0004] Autoimmune diseases often have a strong local component (e.g., inflammatory bowel disease/ulcerative colitis, dermatomyositis/IBM, psoriasis, rheumatoid arthritis, type I diabetes, and lupus nephritis). Cell-based therapy is a major area of development. [0005] Regulatory T-cells (Tregs) can suppress pathogenic immune activity in tissue and lymph nodes, as validated in preclinical models and emerging clinical data. Cytotoxic CAR-T cells can ablate pathogenic immune cells (e.g., B-cells, T-cells). However, cell-based therapies in autoimmune disease face challenges. Tregs are generally considered safe but have limited persistence. Efforts have been made to enhance Treg proliferation and persistence by expressing T-cell receptors (TCRs) or chimeric antigen receptors (CARs) against disease-specific antigens, but these are challenging to identify and may cover only a subset of patients. There is a need for methods and compositions for activation and/or expansion of Treg cell therapies at disease site(s) to boost efficacy without systemic side-effects. SUMMARY [0006] Disclosed herein include compositions for treating an autoimmune disease or disorder in a subject. In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of -1-
ny-2833630
Atty Docket No.23887-2000140 inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product upon thermal stimulation. [0007] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter operably linked to a first polynucleotide comprising a transactivator gene, and a second promoter operably linked to a second polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the transactivator gene to generate a transactivator transcript in the presence of thermal stimulation, wherein the transactivator transcript is capable of being translated to generate a transactivator; and wherein, in the presence of the transactivator and; optionally, a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0008] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter and a second promoter each operably linked to a first polynucleotide comprising an immunostimulatory agent gene and to a second polynucleotide comprising a transactivator gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript upon thermal stimulation, wherein, in the presence of the transactivator and a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript, and wherein the polycistronic transcript is capable of being translated to generate a transactivator and an immunostimulatory agent protein. [0009] In some embodiments, the second promoter comprises one or more copies of a transactivator recognition sequence the transactivator is capable of binding to induce transcription. In some embodiments, the transactivator is incapable of binding the transactivator recognition sequence in the absence of the transactivator-binding compound. In some embodiments, the one or more copies of a transactivator recognition sequence comprise one or more copies of a tet operator (TetO). In some embodiments, the transactivator comprises reverse tetracycline-controlled transactivator (rtTA). In some embodiments, the transactivator comprises tetracycline-controlled transactivator (tTA). In some embodiments, the transactivator- binding compound comprises tetracycline, doxycycline or a derivative thereof. In some embodiments, the first polynucleotide and the second polynucleotide are operably linked to a -2-
ny-2833630
Atty Docket No.23887-2000140 tandem gene expression element. In some embodiments, the tandem gene expression element is an internal ribosomal entry site (IRES), foot-and-mouth disease virus 2A peptide (F2A), equine rhinitis A virus 2A peptide (E2A), porcine teschovirus 2A peptide (P2A) or Thosea asigna virus 2A peptide (T2A), or any combination thereof. [0010] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a first polynucleotide comprising a recombinase gene, wherein the first inducible promoter is capable of inducing transcription of the recombinase gene to generate a recombinase transcript upon thermal stimulation, and wherein the recombinase transcript is capable of being translated to generate a recombinase; a second promoter and a second polynucleotide comprising an immunostimulatory agent gene, wherein, in the absence of a recombination event, the second promoter and the second polynucleotide are not operably linked, wherein the recombinase is capable of catalyzing the recombination event, and a the second promoter and the second polynucleotide are operably linked after the recombination event such that the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0011] In some embodiments, the recombination event comprises removal of a sequence flanked by recombinase target sites or an inversion of a sequence flanked by recombinase target sites. In some embodiments, the second polynucleotide is flanked by recombinase target sites. In some embodiments, prior to the recombination event, the sequence of the immunostimulatory agent gene is inverted relative to the promoter. The composition can comprise at least one stop cassette situated between the second promoter and the immunostimulatory agent gene. In some embodiments, the stop cassette comprises one or more stop sequences In some embodiments, the one or more stop cassettes are flanked by recombinase target sites. In some embodiments, the immunostimulatory agent gene product is an mRNA transcript and/or protein. In some embodiments, the immunostimulatory agent transcript is capable of being translated to generate the immunostimulatory agent protein. In some embodiments, the at least one stop cassette is configured to prevent transcription of the immunostimulatory agent gene and/or translation of the immunostimulatory agent transcript. In some embodiments, the one or more stop sequences comprise a polyadenylation signal, a stop codon, a frame-shifting mutation, or any combination thereof. In some embodiments, the second promoter comprises a promoter selected from the group consisting of a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus-forming virus (SFFV), a Moloney -3-
ny-2833630
Atty Docket No.23887-2000140 murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock protein 90 kDa beta, member 1 (HSP90B1), heat shock -
-KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1 (PGK) promoter, 3-phosphoglycerate kinase promoter, a -
-actin (CBA) promoter, a CAG promoter, a CBH promoter, any variant thereof, or any combination thereof. In some , derivatives thereof, or any combination thereof. In some embodiments, the recombinase is a Flp recombinase and the recombinase target sites are FRT sites or the recombinase is a Cre recombinase and the recombinase target sites are loxP sites. [0012] In some embodiments, the immunostimulatory agent increases the persistence and/or activity of the genetically modified Treg cell or population thereof. In some embodiments, the immunostimulatory agent comprises a cytokine. In some embodiments, the cytokine is a chemokine, an interferon, an interleukin (IL), or a tumor necrosis factor (TNF). In some embodiments, the cytokine is selected from the group consisting of interleukin-1 (IL-1), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, and IL-35. In some embodiments, the cytokine is selected from the group consisting of CCL1, CCL2, CCL3, CCR4, CCL5, CCL7, CCL8/MCP-2, CCL11, CCL13/MCP- 4, HCC- 1/CCL14, CTAC/CCL17, CCL19, CCL22, CCL23, CCL24, CCL26, CCL27, VEGF, PDGF, lymphotactin (XCL1), Eotaxin, FGF, EGF, IP- 10, TRAIL, GCP-2/CXCL6, NAP- 2/CXCL7, CXCL8, CXCL10, ITAC/CXCL11, CXCL12, CXCL13, and CXCL15. In some embodiments, the cytokine is selected from the group consisting of TNF-alpha, TNF-beta, TNF- gamma, CD252, CD154, CD178, CD70, CD153, or 4-1BBL. In some embodiments, the cytokine is selected from the group consisting of interferon alpha, interferon beta, or interferon gamma. In some embodiments, the immunostimulatory agent is selected from the group consisting of granulocyte macrophage colony stimulating factor (GM-CSF), M-CSF, SCF, TSLP, oncostatin M, leukemia-inhibitory factor (LIF), CNTF, Cardiotropin-1, NNT-1/BSF-3, growth hormone, Prolactin, Erythropoietin, Thrombopoietin, Leptin, and G-CSF; or a receptor or ligand thereof. -4-
ny-2833630
Atty Docket No.23887-2000140 [0013] In some embodiments, the immunostimulatory agent comprises an antibody or fragment thereof. In some embodiments, the antibody or fragment thereof is a single-chain variable fragment (scFv), a single-domain antibody, a nanobody, an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab fragment, a Fab’ fragment, a F(ab’)2 fragment, an Fv fragment, a disulfide linked Fv, an scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, or a functionally active epitope-binding fragment thereof. In some embodiments, the antibody or fragment thereof is capable of binding PD-1, PD- L1, IL-2, CD47, 4-1BB, OX40, CD40, IL-2, IL-6, IL-10, BTLA, CD3, and/or CD4. [0014] In some embodiments, the immunostimulatory agent comprises a programmable DNA- binding protein. In some embodiments, the programmable DNA-binding protein comprises a programmable DNA-binding endonuclease selected from the group consisting of Casl, CaslB, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csnl and Csxl2), Casl00, Csyl, Csy2, Csy3, Csel, Cse2, Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, Csxl0, Csxl6, CsaX, Csx3, Csxl, Csxl5, Csfl, Csf2, Csf3, Csf4, or Cpfl endonuclease, or a homolog thereof. In some embodiments, the endonuclease comprises one or more mutations. In some embodiments, the one or more mutations converts the endonuclease to a nickase or renders the endonuclease catalytically dead. In some embodiments, the programmable DNA-binding protein comprises a fusion protein comprising the nickase or the catalytically dead endonuclease. In some embodiments, the fusion protein comprises the programmable DNA-binding protein fused to an enzyme selected from the group consisting of a deaminase, a polymerase, a histone deactylase (HDAC), a histone acetyl transferase (HAT), a recombinase, and an isomerase. In some embodiments, the Treg further comprises a polynucleotide encoding a gRNA capable of targeting the programmable DNA-binding protein to a target sequence. In some embodiments, the polynucleotide encoding the gRNA and the polynucleotide encoding the programmable DNA-binding protein are within the same nucleic acid molecule or separate nucleic acid molecules. In some embodiments, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25,
CD122, IL- 5, CCR8, SIRT1, GITR, OC40, TNFR2, -1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor. In some embodiments, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25, CD122, IL- -5-
ny-2833630
Atty Docket No.23887-2000140 and the programmable DNA-binding protein and the gRNA are configured to cause a reduction- of-function or knockout mutation in the gene. In some embodiments, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of Regnase-1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor and the programmable DNA-binding protein and the gRNA are configured to cause a gain-of-function mutation in the gene. [0015] In some embodiments, the genetically modified Treg cell comprises: (ii) a second nucleic acid comprising a constitutive promoter operably linked to a polynucleotide encoding a chimeric antigen receptor (CAR) or a T-cell receptor (TCR). In some embodiments, the constitutive promoter is a ubiquitous promoter or a tissue-specific promoter. In some embodiments, the constitutive promoter is selected from the group comprising a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus-forming virus (SFFV), a Moloney murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock protein 90 kDa beta, member -
-KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1 (PGK) promoter, 3-
phosphoglycerate kinase pr -actin (HBA) promoter, -actin (CBA) promoter, a CAG promoter, a CBH promoter, a variant thereof, or any combination thereof. [0016] In some embodiments, the CAR comprises a binding domain, a hinge region, a transmembrane domain, and an intracellular signaling domain. In some embodiments, the binding domain is capable of binding a target selected from the group consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6-trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage- gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), a heat shock protein, and a protein of the human leukocyte antigen (HLA) system. In some embodiments, the HLA protein is selected from the group
consisting of HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA- B2M), and serotypes or variants thereof. In some embodiments, the HLA protein comprises the HLA-A2 -6-
ny-2833630
Atty Docket No.23887-2000140 serotype. In some embodiments, the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. [0017] In some embodiments, the intracellular signaling domain comprises a primary signaling domain, a costimulatory domain, or both. In some embodiments, the primary signaling domain comprises a functional signaling domain of one or more proteins selected from the group consisting of CD3 zeta, CD3 gamma, CD3 delta, CD3 epsilon, common FcR gamma (FCER1G), FcR beta (Fc Epsilon R1b), CD79a, CD79b, Fcgamma RIIa, DAP10, and DAP12, or a functional variant thereof. In some embodiments, the costimulatory domain comprises a functional domain of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD28-OX40, CD28-4-1BB, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CD5, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA--6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D, or a functional variant thereof. In some embodiments, the transmembrane domain comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, IL2R beta, IL2R gamma, -6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and NKG2C, or a functional variant thereof. [0018] In some embodiments, the TCR further comprises a constant region and a variable region. In some embodiments, the TCR is capable of binding an antigen selected from the group -7-
ny-2833630
Atty Docket No.23887-2000140 consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6-trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage-gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), and a heat shock protein. In some embodiments, the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. [0019] In some embodiments, the inducible promoter comprises a core promoter and at least one heat shock element (HSE). In some embodiments, the core promoter comprises the heat shock protein transcription start site of a gene selected from the group consisting of HSPA1A, HSPH1, HSPB1, HSPA6, and YB. In some embodiments, the core promoter comprises the sequence of any one of SEQ ID NOs: 9-12 and 35-46 or a sequence having at least 85% identity to any one of SEQ ID NOs: 9-12 and 35-46. In some embodiments, the core promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 9-12 and 35-46. In some embodiments, the inducible promoter comprises seven HSEs. In some embodiments, each of the at least one HSE comprises the sequence of 5’- nGAAnnTTCnnGAAn-3’. In some embodiments, the at least one HSE comprises the sequence of any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence having at least 85% identity to any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence differing by one or two nucleotide mismatches relative to any one of SEQ ID NOs: 1-8, 53, and 112-116. In some embodiments, the at least one HSE comprises a sequence selected from the group consisting of SEQ ID NOs: 1-8, 53, and 112-116. In some embodiments, the inducible promoter comprises the sequence of any one of SEQ ID NOs: 13-20, 58-80, and 88-94 or a sequence having at least 85% identity to any one of SEQ ID NOs: 13-20, 58-80, and 88-94. In some embodiments, the inducible promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 13-20, 58-80, and 88-94. In some embodiments, the inducible promoter comprises the sequence of any one of SEQ ID NOs: 21-34 or a sequence having at least 85% identity to any one of SEQ ID NOs: 21-34. In some embodiments, the inducible promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 21-34. [0020] In some embodiments, the thermal stimulation comprises heating to an activating temperature; wherein the activating temperature is greater than 37°C. In some embodiments, the activating temperature comprises a temperature of at least 37°C to at most 70°C. In some embodiments, the activating temperature is about 37.0ºC, 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 -8-
ny-2833630
Atty Docket No.23887-2000140 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC. In some embodiments, the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC. In some embodiments, the activating temperature comprises a temperature of about 42°C to about 43°C. In some embodiments, the activating temperature is about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. [0021] In some embodiments, the genetically modified Treg cell expresses one or more of the markers selected from the group consisting of CD4, CD25, and forkhead box P3 (FOXP3). In some embodiments, the genetic modification comprises integration of the first nucleic acid into the genome of the Treg cell. In some embodiments, the first nucleic acid is integrated within a safe harbor locus. In some embodiments, the safe harbor locus comprises AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. In some embodiments, the genetic modification comprises integration of the second nucleic acid into the genome of the Treg cell. In some embodiments, the second nucleic acid is integrated within a safe harbor locus. In some embodiments, the safe harbor locus comprises AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. In some embodiments, the genetically modified Treg cell is allogenic or autologous to the subject. In some embodiments, the genetically modified Treg cell was differentiated from a genetically modified stem cell. In some embodiments, the genetically modified stem cell comprises an embryonic stem cell or an induced pluripotent stem cell. In some embodiments, the genetically modified stem cell comprises a hematopoietic stem cell. The composition can comprise one or more pharmaceutically acceptable excipients. Provided herein are compositions for use in treating an autoimmune disease in a subject. [0022] Disclosed herein include methods of treating an autoimmune disease in a subject. In some embodiments, the method comprises: administering any of the compositions disclosed herein to the subject; and applying thermal energy to a target site of the subject sufficient to -9-
ny-2833630
Atty Docket No.23887-2000140 increase local temperature of the target site to an activating temperature, thereby inducing the expression of the immunostimulatory agent gene product. [0023] In some embodiments, the method comprises administering the composition at a dose of about 1 × 10
5 to about 1 × 10
15 Treg cells. In some embodiments, the method comprises administering the composition at a dose of at least about 1 × 10
5, 1 × 10
6, 1 × 10
7, 1 × 10
8, 1 × 10
9, 1 × 10
10, 1 × 10
11, 1 × 10
12, 1 × 10
13, 1 × 10
14, or 1 × 10
15 Treg cells. [0024] In some embodiments, the method comprises a single administration of the composition to the subject. In some embodiments, the method comprises administering the composition to the subject two or more times. In some embodiments, each of the two or more administrations are one month, two months, 6 months, 1 year, or more, apart. In some embodiments, the period of time between the administering the composition and applying thermal energy is about 1 week, about 6 days, about 5 days, about 4 days, about 3 days, about 48 hours, about 44 hours, about 40 hours, about 35 hours, about 30 hours, about 25 hours, 20 hours, 15 hours, 10 hours, about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes, about 10 minutes, or about 5 minutes. [0025] In some embodiments, the activating temperature is greater than 37°C. In some embodiments, the activating temperature comprises a temperature of at least 37.5°C to at most 70.0°C. In some embodiments, the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC. In some embodiments, the thermal energy is applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, or about 1 minute. In some embodiments, the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, -10-
ny-2833630
Atty Docket No.23887-2000140 about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC. In some embodiments, the thermal energy is applied to the target site for a duration of time comprising about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. In some embodiments, the activating temperature comprises a temperature of about 42°C to about 43°C. In some embodiments, the activating temperature is about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. [0026] In some embodiments, the thermal energy is applied to the target site for a duration of time comprising about 1 second to about 20 minutes. In some embodiments, the thermal energy is applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. [0027] In some embodiments, the temperature at the target site is elevated to about 42°C to about 43°C after the applying. In some embodiments, the temperature at the target site is elevated to about 42°C to about 43°C after the applying. within at most 30 minutes from the applying. In some embodiments, the temperature at the target site is elevated to about 42°C to about 43°C within 10 minutes or less after the applying. In some embodiments, the temperature at the target site is elevated to about 42°C to about 43°C within about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, or about 10 minutes after the applying. In some embodiments, the temperature at the target site is maintained at about 42°C to about 43°C for at least about 15 minutes to at least about 120 minutes. In some embodiments, the temperature at the target site is maintained at 42°C to 43°C for at least 15 minutes, at least 20 minutes, at least 25 minutes, at least 30 minutes, at least 35 minutes, at least 40 minutes, at least 45 minutes, at least 50 minutes, at least 55 minutes, at least 60 minutes, at least 90 minutes, or at least 120 minutes. In some embodiments, the temperature at the target site is maintained at 42°C to 43°C for at least 20 minutes. In some embodiments, the temperature at the target site is maintained at about 42°C to -11-
ny-2833630
Atty Docket No.23887-2000140 about 43°C for about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. [0028] In some embodiments, the method comprises applying thermal energy to the subject two or more times. In some embodiments, each of the two or more applications of thermal energy are 1 week apart, 2 weeks apart, 3 weeks apart, 4 weeks apart, 5 weeks apart, or more. [0029] In some embodiments, the target site comprises one or more of muscle, skin, joints, the rectum, and the colon. In some embodiments, the target site is an internal organ. In some embodiments, the muscle is skeletal, cardiac, or smooth muscle. In some embodiments, the internal organ is liver, kidney, lung, pancreas, or heart. [0030] In some embodiments, applying thermal energy to a target site of the subject comprises the application of one or more of magnetic hyperthermia, microwave hyperthermia, inductive diathermy, contact hyperthermia, ultrasonic diathermy, deep hyperthermia ingestible hyperthermia, and rectal hyperthermia. In some embodiments, applying thermal energy to a target site of the subject comprises use of a device selected from the group consisting of: BSD- 500 Superficial Hyperthermia, BSD-2000 Deep Regional Hyperthermia, TheraTouch® DX2 Shortwave Diathermy, Dynatronics Solaris Plus, and Hill Laboratories HF54+. The method can comprise monitoring the temperature of the target region. In some embodiments, the monitoring is performed by magnetic resonance imaging (MRI). In some embodiments, the application of thermal energy to a target site of the subject is guided spatially by magnetic resonance imaging (MRI). [0031] In some embodiments, at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the population of thermally actuated genetically modified Treg cells at the target site express the immunostimulatory agent gene product after applying thermal energy to the target site. In some embodiments, less than about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%, of the population of thermally actuated genetically modified Treg cells at a site other than the target site express the immunostimulatory agent gene product. In some embodiments, upon applying thermal energy to the target site, the transcription of the immunostimulatory gene, the transactivator gene, the recombinase gene, or any combination thereof, increases by at least 1.1-fold. In some -12-
ny-2833630
Atty Docket No.23887-2000140 embodiments, upon applying thermal energy to the target site, the levels of the of the immunostimulatory gene product, the transactivator gene product, the recombinase gene product, or any combination thereof, increases by at least 1.1-fold. In some embodiments, the gene product comprises RNA transcribed from the gene. In some embodiments, the gene product comprises an mRNA, a protein translated from the mRNA, or both. [0032] In some embodiments, the autoimmune disease comprises type I diabetes, psoriasis, Lupus nephritis, multiple sclerosis, neuromyelitis optica, chronic inflammatory demyelinating polyneuropathy, polymyositis, dermatomyositis, inclusion body myositis, Crohn’s Disease, ulcerative colitis, primary sclerosing cholangitis, ankylosing spondylitis, rheumatic arthritis, spondyloarthritis, or juvenile idiopathic arthritis. In some embodiments, the method treats or prevents at least one symptom of the autoimmune disease. [0033] In some embodiments, the autoimmune disease is ulcerative colitis and the target site comprises the rectum and/or the colon. In some embodiments, (a) the method treats or prevents at least one symptom of the ulcerative colitis. In some embodiments, the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof; (b) the method reduces the frequency, duration, and/or intensity of flares in the subject. In some embodiments, a flare is characterized by one or more symptoms of (a); and/or (c) the method reduces the levels of anti- saccharomyces cerevisiae antibody (ASCA) in the subject. In some embodiments, the reduction of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. [0034] In some embodiments, the autoimmune disease is Crohn’s Disease. In some embodiments, target site comprises the rectum, colon, small intestine, or a combination thereof. In some embodiments, (a) the method treats or prevents at least one symptom of the Crohn’s disease. In some embodiments, the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof; (b) the method reduces the frequency, duration, and/or intensity of flares in the subject. In some embodiments, a flare is characterized by one or more symptoms of (a); and/or (c) the method reduces the levels of perinuclear anti-neutrophil antibodies (pANCA) in the subject. In some embodiments, the reduction of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. -13-
ny-2833630
Atty Docket No.23887-2000140 [0035] In some embodiments, the autoimmune disease is psoriasis. In some embodiments, the target site comprises the skin. In some embodiments, (a) the method treats or prevents at least one symptom of psoriasis. In some embodiments, the at least one symptom comprises skin rash, skin bleeding, skin itch, skin burning, skin soreness, or any combination thereof, and/or (b) the method reduces the frequency, duration, and/or intensity of flares in the subject. In some embodiments, a flare is characterized by one or more of the symptoms of (a). In some embodiments, the reduction of (b) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. [0036] In some embodiments, the autoimmune disease is rheumatoid arthritis (RA). In some embodiments, the target site comprises one or more joints. In some embodiments, the method treats or prevents at least one symptom of RA, where the at least one symptom comprises swelling and/or stiffness in one or more joints. In some embodiments, the at least one symptom comprises swelling and/or stiffness in one or more of a knee, finger, elbow, ankle, hip, wrist, and shoulder joint. [0037] In some embodiments, the autoimmune disease is type I diabetes (T1D). In some embodiments, the target site comprises the pancreas. In some embodiments, the method treats or prevents at least one symptom of T1D, where the at least one symptom comprises thirst, frequent urination, weight loss, fatigue, blurred vision, or any combination thereof. In some embodiments, (a) the method increases the level of C-peptide in the serum of the subject. In some embodiments, the C-peptide level in the serum of the subject is about 1.1 ng/mL to about 4.4 ng/mL following at least one application of thermal energy to the target site. In some embodiments, (b) the method decreases the A1C percentage in the blood of the subject. In some embodiments, the A1C percentage in the blood of the subject is less than 6.5% following at least one application of thermal energy to the target site. In some embodiments, (c) the method decreases fasting plasma glucose (FPG) level in the serum and/or plasma of the subject. In some embodiments, the FPG level in the serum and/or plasma of the subject is less than 125 mg/dL following at least one application of thermal energy to the target site. In some embodiments, the increase of (a) and the decrease of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. [0038] In some embodiments, the autoimmune disease is lupus nephritis. In some embodiments, the target site comprises the kidney. In some embodiments, the method treats or prevents at least one symptom of lupus nephritis, where the at least one symptom comprises joint pain or -14-
ny-2833630
Atty Docket No.23887-2000140 swelling, muscle pain, fever, rash, or any combination thereof. In some embodiments, (a) the method decreases the albumin-to-creatinine ratio (UACR) in the urine of the subject. In some embodiments, the UACR of the subject is less than 30 mg/g following at least one application of thermal energy to the target site, and/or (b) the method increases glomerular filtration rate (GFR) in the blood of the subject. In some embodiments, the GFR in the blood of the subject is greater than 60 following at least one application of thermal energy to the target site. In some embodiments, the decrease of (a) and the increase of (b) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. [0039] In some embodiments, the autoimmune disease is inclusion body myositis (IBM). In some embodiments, the target site is skeletal muscle. In some embodiments, the method treats or prevents at least one symptom of IBM, where the at least one symptom comprises muscle weakness in a wrist, finger, front of the thigh, muscles that lift the front of the foot, or any combination thereof. BRIEF DESCRIPTION OF THE DRAWINGS [0040] FIG.1 depicts a non-limiting exemplary schematic of methods and compositions provided herein for treating autoimmune diseases. [0041] FIGS.2A-2D depict an experimental design (FIG.2A) and results (FIGS.2B-2D) of studies determining duration and temperature of applications of thermal energy. FIG.2A provides a schematic representation of a construct where the thermal switch (TS) drives expression of a Gaussia luciferase (Gluc). FIG.2B provides a schematic representation of the temperature ramp profile in an in vitro heating assay, wherein transduced primary human T cells were heated with ramp times from 2 seconds to 180 minutes, including a 2 second heat shock- like ramp time, a 10-minute ramp time, and a 3-hr ramp time. FIG.2C provides the percentage of maximum Gluc luminescence and FIG.2D the relative luminescence, both 24 hrs post- heating. In FIG.1D, the dashed line represents the line-of-best-fit of the heated condition (red), fit to a 1-phase exponential decay with a half-life of 12.8 minutes. [0042] FIGS.3A-3B depict data related to expanded characterization of thermal switch activation. FIG.3A provides schematic representations of the circuit of the thermal switch driving expression of a Gaussia luciferase (Gluc) reporter and accompanying graphical representation of a transduced primary human T cell transduced with the circuit, wherein these transduced primary human T cells were heated to temperatures ranging from 37–67.3ºC, durations ranging from 1s–20 min, and then reporter output and viability were measured. FIG. 3B provides luminescence plots 24 hrs post-heating. -15-
ny-2833630
Atty Docket No.23887-2000140 [0043] FIGS.4A-4B show the results from evaluating T cell viability following heating at various thermal doses. The viability of primary human T cells was evaluated 24 post-heating at different thermal doses, as calculated by the cumulative equivalent minutes at 43°C (CEM43°C) model, as introduced by Sapareto and Dewey (Rad. Oncol.10(6):787-800, 1984), is a simple concept that translates all different temperature–time histories to a single number representing a “thermal isoeffect dose” CEM43ºC. FIG.4A provides the line of best fit, and FIG.4B shows the scatter as broken down by temperature. [0044] FIG.5 provides a schematic overview of exemplary heat shock element orientations and configurations. [0045] FIGS.6A-6C show the results from evaluating thermal bioswitches with different HSE architectures. The kinetics of expression of a Gaussia luciferase reporter following heating (30 min, 66% duty cycle) after varying HSE spacing (FIG.6A), the number of HSEs (FIG.6B), and the orientation of the HSEs (FIG.6C) were evaluated. [0046] FIGS.7A-7D show the results from evaluating thermal switch activity with different core promoters. FIG.7A shows a schematic representation of the tested exemplary constructs, wherein core from YB, CMVa, and CMVb were paired with 7HSEs, and placed upstream of a Gaussia luciferase reporter. FIG.7C shows the same, wherein instead of a Gaussia luciferase reporter, the payload gene is an EGFR chimeric antigen receptor (CAR) to demonstrate proof of principle using two core promoter conditions selected based on FIG.7B results. FIG.7B provides the luminescence signal 24 hrs post-heating. FIG.7D provides the mean fluorescence intensity of CAR expression as analyzed by FACS. [0047] FIGS.8A-8D provide schematic overviews of various exemplary thermal bioswitches. FIG.8A provides a schematic of the overall designs, where the core promoters are defined as
either including the native 5 UTR or not. FIG. 8B shows the variations in the core promoter and number of HSEs. FIG.8C shows the variations in the Kozak sequence upstream of the
exemplary transgene (e.g., CAR). FIG. 8D shows the variations in the 5 UTR sequence. [0048] FIG.9 demonstrates the kinetics of cytolysis of EGFR+ (Huh7) tumor cells following a single thermal treatment of two different thermally controlled Panitumumab-based EGFR CAR constructs (either using a 28z or BBz signaling domain; “TS.EGFR CAR” constructs) at 42ºC for 40 minutes with a 0.5ºC/min ramp time as measured by impedance-based measurements in real time (xCELLigence). [0049] FIG.10 demonstrates the kinetics of expression for thermally controlled EGFR CAR (Pani scFv; 28z) expression with either the S7YB (red) or new thermal bioswitches (all in gray) (Left column = unheated, right column = heated). -16-
ny-2833630
Atty Docket No.23887-2000140 [0050] FIGS.11A-11D show the kinetics of thermally controlled EGFR CAR expression and accompanying in vitro tumor cell killing activity of TS.EGFR-CAR constructs expressed by primary human T cells. FIG.11A shows the kinetics expression for thermally controlled EGFR CAR (Pani scFv; 28z) expression with either the S7YB (red) or new thermal bioswitches identified to have improved expression profiles over S7YB. FIG.11B provides the accompanying in vitro EGFR+ (Huh7) tumor cell killing data for primary human T cells comprising the constructs of FIG.11A and co-cultured at a 5:1 E:T ratio. FIG.11C shows the kinetics expression for thermally controlled EGFR CAR (Pani scFv; 28z) expression with either the S7YB (red) or new thermal bioswitches comprising CMVb core promoters identified to have improved expression profiles over S7YB. FIG.11D provides the accompanying in vitro EGFR+ (Huh7) tumor cell killing data for primary human T cells comprising the constructs of FIG.11C and co-cultured at a 5:1 E:T ratio. [0051] FIGS.12A-12B show the kinetics of expression for thermally controlled EGFR CAR (Pani scFv; 28z) expression with either the S7YB (red) or new thermal bioswitches identified to have baseline expression in the unheated state and that can be transiently boosted to higher levels upon heating (FIG.12A) and accompanying in vitro EGFR+ (Huh7) tumor cell killing data for primary human T cells comprising these constructs (FIG.12B) and co-cultured at 2:1 E:T ratio. [0052] FIGS.13A-13E depict a non-limiting exemplary circuit design (FIG.13A) and data (FIGS.13B-13E) related to studies thereof. DETAILED DESCRIPTION [0053] In the following detailed description, reference is made to the accompanying drawings, which form a part hereof. In the drawings, similar symbols typically identify similar components, unless context dictates otherwise. The illustrative embodiments described in the detailed description, drawings, and claims are not meant to be limiting. Other embodiments may be utilized, and other changes may be made, without departing from the spirit or scope of the subject matter presented herein. It will be readily understood that the aspects of the present disclosure, as generally described herein, and illustrated in the Figures, can be arranged, substituted, combined, separated, and designed in a wide variety of different configurations, all of which are explicitly contemplated herein and made part of the disclosure herein. [0054] All patents, published patent applications, other publications, and sequences from GenBank, and other databases referred to herein are incorporated by reference in their entirety with respect to the related technology. [0055] Disclosed herein include compositions for treating an autoimmune disease or disorder in a subject. In some embodiments, the composition comprises: a genetically modified T-regulatory -17-
ny-2833630
Atty Docket No.23887-2000140 (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product upon thermal stimulation. [0056] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter operably linked to a first polynucleotide comprising a transactivator gene, and a second promoter operably linked to a second polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the transactivator gene to generate a transactivator transcript in the presence of thermal stimulation, wherein the transactivator transcript is capable of being translated to generate a transactivator; and wherein, in the presence of the transactivator and; optionally, a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0057] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter and a second promoter each operably linked to a first polynucleotide comprising an immunostimulatory agent gene and to a second polynucleotide comprising a transactivator gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript upon thermal stimulation, wherein, in the presence of the transactivator and a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript, and wherein the polycistronic transcript is capable of being translated to generate a transactivator and an immunostimulatory agent protein. [0058] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a first polynucleotide comprising a recombinase gene, wherein the first inducible promoter is capable of inducing transcription of the recombinase gene to generate a recombinase transcript upon thermal stimulation, and wherein the recombinase transcript is capable of being translated to generate a recombinase; a second promoter and a second polynucleotide comprising an immunostimulatory -18-
ny-2833630
Atty Docket No.23887-2000140 agent gene, wherein, in the absence of a recombination event, the second promoter and the second polynucleotide are not operably linked, wherein the recombinase is capable of catalyzing the recombination event, and a the second promoter and the second polynucleotide are operably linked after the recombination event such that the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0059] Disclosed herein include methods of treating an autoimmune disease in a subject. In some embodiments, the method comprises: administering any of the compositions disclosed herein to the subject; and applying thermal energy to a target site of the subject sufficient to increase local temperature of the target site to an activating temperature, thereby inducing the expression of the immunostimulatory agent gene product. Definitions [0060] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present disclosure belongs. See, e.g. Singleton et al., Dictionary of Microbiology and Molecular Biology 2nd ed., J. Wiley & Sons (New York, NY 1994); Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press (Cold Spring Harbor, NY 1989). For purposes of the present disclosure, the following terms are defined below. [0061] As used herein, the terms “nucleic acid” and “polynucleotide” are interchangeable and refer to any nucleic acid, whether composed of phosphodiester linkages or modified linkages such as phosphotriester, phosphoramidate, siloxane, carbonate, carboxymethylester, acetamidate, carbamate, thioether, bridged phosphoramidate, bridged methylene phosphonate, bridged phosphoramidate, bridged phosphoramidate, bridged methylene phosphonate, phosphorothioate, methylphosphonate, phosphorodithioate, bridged phosphorothioate or sultone linkages, and combinations of such linkages. The terms “nucleic acid” and “polynucleotide” also specifically include nucleic acids composed of bases other than the five biologically occurring bases (adenine, guanine, thymine, cytosine and uracil). [0062] The term “vector” as used herein, can refer to a vehicle for carrying or transferring a nucleic acid. Non-limiting examples of vectors include plasmids and viruses (for example, AAV viruses). [0063] The term “construct,” as used herein, refers to a recombinant nucleic acid that has been generated for the purpose of the expression of a specific nucleotide sequence(s), or that is to be used in the construction of other recombinant nucleotide sequences. -19-
ny-2833630
Atty Docket No.23887-2000140 [0064] As used herein, the term “plasmid” refers to a nucleic acid that can be used to replicate recombinant DNA sequences within a host organism. The sequence can be a double stranded DNA. [0065] The term “element” refers to a separate or distinct part of something, for example, a nucleic acid sequence with a separate function within a longer nucleic acid sequence. The term “regulatory element” and “expression control element” are used interchangeably herein and refer to nucleic acid molecules that can influence the expression of an operably linked coding sequence in a particular host organism. These terms are used broadly to and cover all elements that promote or regulate transcription, including promoters, core elements required for basic interaction of RNA polymerase and transcription factors, upstream elements, enhancers, and response elements (see, e.g., Lewin, “Genes V” (Oxford University Press, Oxford) pages 847- 873). Exemplary regulatory elements in prokaryotes include promoters, operator sequences and a ribosome binding sites. Regulatory elements that are used in eukaryotic cells can include, without limitation, transcriptional and translational control sequences, such as promoters, enhancers, splicing signals, polyadenylation signals, terminators, protein degradation signals, internal ribosome-entry element (IRES), 2A sequences, and the like, that provide for and/or regulate expression of a coding sequence and/or production of an encoded polypeptide in a host cell. [0066] As used herein, the term “promoter” is a nucleotide sequence that permits binding of RNA polymerase and directs the transcription of a gene. Typically, a promoter is located in the 5’ non-coding region of a gene, proximal to the transcriptional start site of the gene. Sequence elements within promoters that function in the initiation of transcription are often characterized by consensus nucleotide sequences. Examples of promoters include, but are not limited to, promoters from bacteria, yeast, plants, viruses, and mammals (including humans). A promoter can be inducible, repressible, and/or constitutive. Inducible promoters initiate increased levels of transcription from DNA under their control in response to some change in environmental conditions, such as a change in temperature. In some embodiments, the inducible promoter is a “heat-shock promoter” that is activated in response to heat (e.g., thermal energy). [0067] The term “thermally-inducible” refers to the ability to turn ON a system, e.g., to initiate transcription of a transgene, by application of heat, heat transfer, heat energy, kinetic energy, etc. at sufficient levels to increase the temperature of the system or organism (e.g., immune cell) into which the transgene has been introduced to surpass a specific temperature threshold, thereby turning ON the system, e.g., initiating transient transcription of thermally controlled genes. If the -20-
ny-2833630
Atty Docket No.23887-2000140 temperature remains below the threshold, the system remains OFF, e.g., transcription remains silent or at baseline levels. [0068] The term “core promoter” refers to a region of DNA that is essential for initiating gene transcription. The core promoter serves as the binding site for RNA polymerase and transcription factors that are necessary for the initiation of transcription. They typically drive basal levels of transcription independent from any other regulatory elements. See, e.g., Ede, et al. (2016) ACS Synthetic Biol. [0069] As used herein, the term “enhancer” refers to a type of regulatory element that can increase the efficiency of transcription, regardless of the distance or orientation of the enhancer relative to the start site of transcription. [0070] As used herein, the term “operably linked” is used to describe the connection between regulatory elements and a gene or its coding region. Typically, gene expression is placed under the control of one or more regulatory elements, for example, without limitation, constitutive or inducible promoters, tissue-specific regulatory elements, and enhancers. A gene or coding region is said to be “operably linked to” or “operatively linked to” or “operably associated with” the regulatory elements, meaning that the gene or coding region is controlled or influenced by the regulatory element. For instance, a promoter is operably linked to a coding sequence if the promoter effects transcription or expression of the coding sequence. [0071] As used herein, a “gene product” is the biochemical material, e.g., RNA or protein, resulting from expression of a gene. As used herein, “gene expression” is the process by which information from a gene is used in the synthesis of a functional gene product. [0072] As used herein, a “subject” refers to an animal that is the object of treatment, observation or experiment. “Animal” includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles, and in particular, mammals. “Mammal,” as used herein, refers to an individual belonging to the class Mammalia and includes, but not limited to, humans, domestic and farm animals, zoo animals, sports and pet animals. Non-limiting examples of mammals include mice; rats; rabbits; guinea pigs; dogs; cats; sheep; goats; cows; horses; primates, such as monkeys, chimpanzees and apes, and, in particular, humans. In some embodiments, the mammal is a human. However, in some embodiments, the mammal is not a human. [0073] As used herein, the term “treatment” refers to an intervention made in response to a disease, disorder or physiological condition manifested by a patient. The aim of treatment may include, but is not limited to, one or more of the alleviation or prevention of symptoms, slowing or stopping the progression or worsening of a disease, disorder, or condition and the remission of the disease, disorder or condition. The term “treat” and “treatment” includes, for example, -21-
ny-2833630
Atty Docket No.23887-2000140 therapeutic treatments, prophylactic treatments, and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses embodiments in which one reduces symptoms or underlying risk factors. In some embodiments, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already affected by a disease or disorder or undesired physiological condition as well as those in which the disease or disorder or undesired physiological condition is to be prevented. As used herein, the term “prevention” refers to any activity that reduces the burden of the individual later expressing those symptoms. This can take place at primary, secondary and/or tertiary prevention levels, wherein: a) primary prevention avoids the development of symptoms/disorder/condition; b) secondary prevention activities are aimed at early stages of the condition/disorder/symptom treatment, thereby increasing opportunities for interventions to prevent progression of the condition/disorder/symptom and emergence of symptoms; and c) tertiary prevention reduces the negative impact of an already established condition/disorder/symptom by, for example, restoring function and/or reducing any condition/disorder/symptom or related complications. The term “prevent” does not require the 100% elimination of the possibility of an event. Rather, it denotes that the likelihood of the occurrence of the event has been reduced in the presence of the compound or method. [0074] As used herein, the term “effective amount” refers to an amount sufficient to effect beneficial or desirable biological and/or clinical results. [0075] “Pharmaceutically acceptable” carriers are ones which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. “Pharmaceutically acceptable” carriers can be, but not limited to, organic or inorganic, solid or liquid excipients which is suitable for the selected mode of application such as oral application or injection, and administered in the form of a conventional pharmaceutical preparation, such as solid such as tablets, granules, powders, capsules, and liquid such as solution, emulsion, suspension and the like. Often the physiologically acceptable carrier is an aqueous pH buffered solution such as phosphate buffer or citrate buffer. The physiologically acceptable carrier may also comprise one or more of the following: antioxidants including ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, such as serum albumin, gelatin, immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone, amino acids, carbohydrates including glucose, mannose, or dextrins, chelating agents such as EDTA, sugar alcohols such as mannitol or sorbitol, salt-forming counterions such as sodium, and nonionic surfactants such as Tween™, polyethylene glycol (PEG), and Pluronics™. Auxiliary, stabilizer, emulsifier, lubricant, binder, -22-
ny-2833630
Atty Docket No.23887-2000140 pH adjustor controller, isotonic agent and other conventional additives may also be added to the carriers. [0076] The term “antibody fragment” shall be given its ordinary meaning, and shall also refers to at least one portion of an antibody, that retains the ability to specifically interact with (e.g., by binding, steric hindrance, stabilizing/destabilizing, spatial distribution) an epitope of an antigen. 2, Fv fragments, scFv antibody fragments, disulfide-linked Fvs (sdFv), a Fd fragment consisting of the VH and CH1 domains, linear antibodies, single domain antibodies such as sdAb (either VL or VH), camelid VHH domains, multi-specific antibodies formed from antibody fragments such as a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region, and an isolated CDR or other epitope binding fragments of an antibody. An antigen binding fragment can also be incorporated into single domain antibodies, maxibodies, minibodies, nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NAR and bis-scFv (see, e.g., Hollinger and Hudson, Nature Biotechnology 23:1126-1136, 2005). Antigen binding fragments can also be grafted into scaffolds based on polypeptides such as a fibronectin type III (Fn3) (see U.S. Pat. No.6,703,199, which describes fibronectin polypeptide minibodies). [0077] The term “autologous” shall be given its ordinary meaning, and shall also refer to any material derived from the same individual to whom it is later to be re-introduced into the individual. [0078] The term “allogeneic” shall be given its ordinary meaning, and shall also refer to any material derived from a different animal of the same species as the individual to whom the material is introduced. Two or more individuals are said to be allogeneic to one another when the genes at one or more loci are not identical. In some aspects, allogeneic material from individuals of the same species may be sufficiently unlike genetically to interact antigenically. [0079] As used herein, the term “within or near a gene” refers to a site or region of genomic DNA that is an intronic or exonic component of said gene or is flanking said gene. In some embodiments, a site of genomic DNA is within a gene if it comprises at least a portion of an intron or exon of said gene. In some embodiments, a site of genomic DNA located near a gene may be at the 5 ’ or 3 ’ end of said gene (e.g., the 5’ or 3’ end of the coding region of said gene). In some embodiments, a site of genomic DNA located near a gene may be a promoter region or repressor region that modulates the expression of said gene. In some embodiments, a site of genomic DNA located near a gene may be on the same chromosome as said gene. In some embodiments, a site or region of genomic DNA is near a gene if it is within 50 kb, 40 kb, 30 kb, -23-
ny-2833630
Atty Docket No.23887-2000140 20 kb, 10 kb, 5 kb, 1 kb, or closer to the 5 ’ or 3 ’ end of said gene (e.g., the 5 ’ or 3 ’ end of the coding region of said gene). [0080] It is understood that wherever embodiments are described herein with the term “comprising” otherwise analogous embodiments described in terms of “consisting of” and/or “consisting essentially of” are also provided. It is also understood that wherever embodiments are described herein with the phrase “consisting essentially of” otherwise analogous embodiments described in terms of “consisting of” are also provided. Thermal control of T-cell activation [0081] Genetically engineered T cells are being developed to perform a variety of therapeutic functions. However, no robust mechanisms exist to externally control the activity of T cells at specific locations within the body. Such spatiotemporal control could help mitigate potential off- target toxicity due to incomplete molecular specificity in applications such as T-cell immunotherapy. Temperature is a versatile external control signal that can be delivered to target tissues in vivo using techniques such as focused ultrasound and magnetic hyperthermia. As demonstrated herein, heat shock promoters can mediate thermal actuation of genetic circuits in primary human T cells in the well-tolerated temperature range of, e.g., 37–42ºC. Disclosed herein are genetic architectures enabling the tuning of the amplitude and duration of thermal activation. Provided herein are uses of these circuits to control the expression of immunomodulating agents (e.g., cytokines) thereby increasing the persistence and/or activity of the T cells. The methods and compositions disclosed herein provide a critical tool to direct the activity of T cells after they are deployed inside the body. [0082] Provided herein are cellular engineering approaches to regulate the activity of therapeutic T cells (e.g., T-regulatory cells) with greater specificity through a combination of molecular and physical actuation. In some embodiments, this approach takes advantage of the ability of technologies such as focused ultrasound (FUS) and magnetic hyperthermia to non-invasively deposit heat at precise locations in deep tissue. By engineering thermal bioswitches that allow T cells to sense small changes in temperature and use them as inputs for the actuation of genetic circuits, these penetrant forms of energy are enabled to spatially control T-cell activity with the disclosed compositions and methods. The circuits provided herein can incorporate feed-forward amplification, positive feedback and/or recombinase-based state switches. Also provided herein are uses of these circuits to control the secretion of an immunostimulatory agent to promote the persistence and/or activity of therapeutic Treg cells. -24-
ny-2833630
Atty Docket No.23887-2000140 [0083] There are provided, in some embodiments, compositions for treating an autoimmune disease or disorder in a subject. In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises(i) a first nucleic acid comprising an inducible promoter operably linked to a polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product upon thermal stimulation. [0084] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises(i) a first nucleic acid comprising a first inducible promoter operably linked to a first polynucleotide comprising a transactivator gene, and a second promoter operably linked to a second polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the transactivator gene to generate a transactivator transcript in the presence of thermal stimulation, wherein the transactivator transcript is capable of being translated to generate a transactivator; and wherein, in the presence of the transactivator and; optionally, a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0085] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises(i) a first nucleic acid comprising a first inducible promoter and a second promoter each operably linked to a first polynucleotide comprising an immunostimulatory agent gene and to a second polynucleotide comprising a transactivator gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript upon thermal stimulation, wherein, in the presence of the transactivator and a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript, and wherein the polycistronic transcript is capable of being translated to generate a transactivator and an immunostimulatory agent protein. [0086] The second promoter can comprise one or more copies of a transactivator recognition sequence the transactivator is capable of binding to induce transcription, and wherein the transactivator is incapable of binding the transactivator recognition sequence in the absence of the transactivator-binding compound. In some embodiments, the one or more copies of a transactivator recognition sequence comprise one or more copies of a tet operator (TetO). The -25-
ny-2833630
Atty Docket No.23887-2000140 transactivator can comprise reverse tetracycline-controlled transactivator (rtTA). The transactivator can comprise tetracycline-controlled transactivator (tTA). The transactivator- binding compound can comprise tetracycline, doxycycline or a derivative thereof. The first polynucleotide and the second polynucleotide can be operably linked to a tandem gene expression element. The tandem gene expression element can be an internal ribosomal entry site (IRES), foot-and-mouth disease virus 2A peptide (F2A), equine rhinitis A virus 2A peptide (E2A), porcine teschovirus 2A peptide (P2A) or Thosea asigna virus 2A peptide (T2A), or any combination thereof. [0087] In some embodiments, the composition comprises: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a first polynucleotide comprising a recombinase gene, wherein the first inducible promoter is capable of inducing transcription of the recombinase gene to generate a recombinase transcript upon thermal stimulation, and wherein the recombinase transcript is capable of being translated to generate a recombinase; a second promoter and a second polynucleotide comprising an immunostimulatory agent gene, wherein, in the absence of a recombination event, the second promoter and the second polynucleotide are not operably linked, wherein the recombinase is capable of catalyzing the recombination event, and wherein the second promoter and the second polynucleotide are operably linked after the recombination event such that the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. [0088] The recombination event can comprise removal of a sequence flanked by recombinase target sites or an inversion of a sequence flanked by recombinase target sites. In some embodiments, the second polynucleotide can be flanked by recombinase target sites. Prior to the recombination event, the sequence of the immunostimulatory agent gene can be inverted relative to the promoter. The composition can comprise at least one stop cassette situated between the second promoter and the immunostimulatory agent gene. The stop cassette can comprise one or more stop sequences. The one or more stop cassettes can be flanked by recombinase target sites. The immunostimulatory agent gene product can be an mRNA transcript and/or protein, wherein the immunostimulatory agent transcript is capable of being translated to generate the immunostimulatory agent protein. The at least one stop cassette can be configured to prevent transcription of the immunostimulatory agent gene and/or translation of the immunostimulatory agent transcript. The one or more stop sequences can comprise a polyadenylation signal, a stop codon, a frame-shifting mutation, or any combination thereof. -26-
ny-2833630
Atty Docket No.23887-2000140 [0089] In some embodiments, the second promoter comprises a promoter selected from the group consisting of a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus-forming virus (SFFV), a Moloney murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock protein 90 kDa beta, member 1 (HSP90B1), heat shock -
-KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1 (PGK) promoter, 3-phosphoglycerate kinase promoter, a -
actin (HBA) promoter, ch -actin (CBA) promoter, a CAG promoter, a CBH promoter, any variant thereof, or any combination thereof. The Bxb1, R4, derivatives thereof, or any combination thereof. The recombinase can be a Flp recombinase and the recombinase target sites can be FRT sites or the recombinase can be a Cre recombinase and the recombinase target sites can be loxP sites. Immunostimulatory agents [0090] The Tregs provided herein can comprise nucleic acids comprising an immunostimulatory agent gene. In some embodiments, the immunostimulatory agent increases the persistence and/or activity of the genetically modified Treg cell or population thereof. In some embodiments, the persistence of the genetically modified Treg cell or population thereof is increased by or by about 0.000000001%, 0.00000001%, 0.0000001%, 0.000001%, 0.00001%, 0.0001%, 0.001%, 0.01%, 0.1%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, or a number or a range between any two of these values. In some embodiments, the activity of the genetically modified Treg cell or population thereof is increased by or by about 0.000000001%, 0.00000001%, 0.0000001%, 0.000001%, 0.00001%, 0.0001%, 0.001%, -27-
ny-2833630
Atty Docket No.23887-2000140 0.01%, 0.1%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, or a number or a range between any two of these values. [0091] The immunostimulatory agent can comprise a cytokine. Cytokines are a broad category of small proteins (~5–25 kDa) important in cell signaling. Due to their size, cytokines cannot cross the lipid bilayer of cells to enter the cytoplasm and therefore typically exert their functions by interacting with specific cytokine receptors on the target cell surface. Cytokines have been shown to be involved in autocrine, paracrine and endocrine signaling as immunomodulating agents. [0092] Cytokines can include chemokines, interferons, interleukins, lymphokines, and tumor necrosis factors, but generally not hormones or growth factors (despite some overlap in the terminology). Cytokines are produced by a broad range of cells, including immune cells like macrophages, B lymphocytes, T lymphocytes and mast cells, as well as endothelial cells, fibroblasts, and various stromal cells; a given cytokine may be produced by more than one type of cell. They act through cell surface receptors and are especially important in the immune system; cytokines modulate the balance between humoral and cell-based immune responses, and they regulate the maturation, growth, and responsiveness of particular cell populations. Some cytokines enhance or inhibit the action of other cytokines in complex ways. They are different from hormones, which are also important cell signaling molecules. Hormones circulate in higher concentrations, and tend to be made by specific kinds of cells. Cytokines are important in health and disease, specifically in host immune responses to infection, inflammation, trauma, sepsis, cancer, and reproduction. [0093] In one non-limiting example, the immunostimulatory agent can comprise IL-2. IL-2 cytokine is produced by activated CD4-positive helper T-cells and to a lesser extent activated CD8-positive T-cells and natural killer (NK) cells and plays pivotal roles in the immune response and tolerance. This cytokine binds to a receptor complex composed of either the high- affinity trimeric IL-2R (IL2RA/CD25, IL2RB/CD122 and IL2RG/CD132) or the low-affinity dimeric IL-2R (IL2RB and IL2RG). Interaction with the receptor leads to oligomerization and conformation changes in the IL-2R subunits resulting in downstream signaling starting with -28-
ny-2833630
Atty Docket No.23887-2000140 phosphorylation of JAK1 and JAK3. In turn, JAK1 and JAK3 phosphorylate the receptor to form a docking site leading to the phosphorylation of several substrates including STAT5. This process leads to activation of several pathways including STAT, phosphoinositide-3-kinase/PI3K and mitogen-activated protein kinase/MAPK pathways. In some embodiments, IL-2 functions as a T-cell growth factor and can increase NK-cell cytolytic activity as well. IL-2 can promote strong proliferation of activated B-cells and subsequently immunoglobulin production. In some embodiments, IL-2 plays a pivotal role in regulating the adaptive immune system by controlling the survival and proliferation of regulatory T cells, which are required for the maintenance of immune tolerance. Moreover, IL-2 participates in the differentiation and homeostasis of effector T-cell subsets, including Th1, Th2, Th17 as well as memory CD8-positive T-cells. Therefore, stimulating the production of IL-2 can promote, e.g., Treg persistence and/or activity. The antibody or fragment thereof can be capable of binding PD-1, PD-L1, IL-2, CD47, 4-1BB, OX40, CD40, IL-2, IL-6, IL-10, BTLA, CD3, and/or CD4. In some embodiments, the immunostimulatory agent comprises a wildtype IL-2. In some embodiments, the immunostimulatory agent comprises a variant IL-2. In some embodiments, the immunostimulatory agent comprises an IL-2 comprising the amino acid sequence of SEQ ID NO:107. See, e.g., US 10,035,836, hereby incorporated by reference in its entirety. [0094] In some embodiments, the immunostimulatory agent can comprise CCL21, IL-15, and/or IL-18. In some embodiments, there is provided a nucleic acid construct comprising a nucleotide encoding CCL21 under the transcriptional activity of one of the thermal bioswitches described herein. In some embodiments, CCL21 comprises the amino acid sequence of SEQ ID NO:106. In some embodiments, there is provided a nucleic acid construct comprising a nucleotide encoding an IL-15SA under the transcriptional activity of one of the thermal bioswitches described herein. In some embodiments, the IL-15SA comprises the amino acid sequence of SEQ ID NO:105. In some embodiments, there is provided a nucleic acid construct comprising a nucleotide encoding an IL-18 under the transcriptional activity of one of the thermal bioswitches described herein. In some embodiments, the IL-18 comprises the amino acid sequence of SEQ ID NO:103 or 104. [0095] The immunostimulatory agent can comprise an antibody or fragment thereof. The antibody or fragment thereof can be a single-chain variable fragment (scFv), a single-domain antibody, a nanobody, an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab fragment, a Fab’ fragment, a F(ab’)2 fragment, an Fv fragment, a disulfide linked Fv, an scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, or a functionally active epitope-binding fragment thereof. -29-
ny-2833630
Atty Docket No.23887-2000140 [0096] The antibody can be a Bi-specific T-cell engager (BiTE™). BiTEs are a class of artificial bispecific monoclonal antibodies. They direct a host's immune system, more specifically the T cells' cytotoxic activity, against target cells. BiTEs are fusion proteins consisting of two single-chain variable fragments (scFvs) of different antibodies, or amino acid sequences from four different genes, on a single peptide chain of about 55 kilodaltons. One of the scFvs binds to T cells via the CD3 receptor, and the other to a target cell. For example, in some embodiments, the immunostimulatory agent comprises a bispecific T cell engager, wherein a first antigen-binding domain (e.g., scFv) specifically binds to CD3, and wherein a second antigen-binding domain (e.g., scFv) specifically binds to NKG2D ligand. In some embodiments, the immunostimulatory agent comprises a bispecific T cell engager comprising the amino acid sequence of SEQ ID NO:117 or 118. [0097] The immunostimulatory agent can comprise a programmable nucleic acid-binding protein. The immunostimulatory agent can comprise a programmable DNA-binding protein. The immunostimulatory agent can comprise a programmable RNA-binding protein. The programmable DNA-binding protein comprises a programmable DNA-binding endonuclease selected from the group consisting of Casl, CaslB, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csnl and Csxl2), Casl00, Csyl, Csy2, Csy3, Csel, Cse2, Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, Csxl0, Csxl6, CsaX, Csx3, Csxl, Csxl5, Csfl, Csf2, Csf3, Csf4, or Cpfl endonuclease, or a homolog thereof. [0098] The endonuclease can comprise one or more mutations. In some embodiments, the one or more mutations converts the endonuclease to a nickase or renders the endonuclease catalytically dead. The programmable DNA-binding protein can comprise a fusion protein comprising the nickase or the catalytically dead endonuclease. The fusion protein can comprise the programmable DNA-binding protein fused to an enzyme selected from the group consisting of a deaminase, a polymerase, a histone deactylase (HDAC), a histone acetyl transferase (HAT), a recombinase, and an isomerase. [0099] In some embodiments, the Treg further comprises a polynucleotide encoding a gRNA capable of targeting the programmable DNA-binding protein to a target sequence. In some embodiments, the polynucleotide encoding the gRNA and the polynucleotide encoding the programmable DNA-binding protein can be within the same nucleic acid molecule or separate nucleic acid molecules. In some embodiments, gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25, CD122, IL- -30-
ny-2833630
Atty Docket No.23887-2000140 -1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor. In some embodiments, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25, CD122, IL- and the programmable DNA-binding protein and the gRNA are configured to cause a reduction- of-function or knockout mutation in the gene. In some embodiments, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of Regnase-1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor and the programmable DNA-binding protein and the gRNA are configured to cause a gain-of-function mutation in the gene. [0100] In certain embodiments, the one or more modulating agents may be a genetic modifying agent. The genetic modifying agents may manipulate nucleic acids (e.g., genomic DNA or mRNA). The genetic modulating agent can be used to up- or downregulate expression of a gene either by targeting a nuclease or functional domain to a DNA or RNA sequence. The genetic modifying agent may comprise an RNA-guided nuclease system (e.g., CRISPR system), RNAi system, a zinc finger nuclease, a TALE, or a meganuclease. [0101] In some embodiments, the immunostimulatory agent comprises a CRISPR-Cas and/or Cas-based system. The nucleotide sequence may be or encode one or more components of a CRISPR-Cas system. For example, the nucleotide sequences may be or encode guide RNAs. The nucleotide sequences may also encode CRISPR proteins, variants thereof, or fragments thereof. Th CRISPR-Cas system can be used to modify a gene involved in immunity. [0102] In general, a CRISPR-Cas or CRISPR system can refer collectively to transcripts and other elements involved in the expression of or directing the activity of CRISPR-associated (“Cas”) gene products, including sequences encoding a Cas gene product, a tracr (trans- activating CRISPR) sequence (e.g., tracrRNA or an active partial tracrRNA), a tracr-mate sequence (encompassing a “direct repeat” and a tracrRNA-processed partial direct repeat in the context of an endogenous CRISPR system), a guide sequence (also referred to as a “spacer”), or “RNA(s)” as that term is herein used (e.g., RNA(s) to guide Cas, such as Cas9, e.g., CRISPR RNA and transactivating (tracr) RNA or a single guide RNA (sgRNA) (chimeric RNA)) or other sequences and transcripts. In general, a CRISPR system is characterized by elements that promote the formation of a CRISPR complex at the site of a target sequence (also referred to as a protospacer in the context of an endogenous CRISPR system). [0103] CRISPR-Cas systems can generally fall into two classes based on their architectures of their effector molecules, which are each further subdivided by type and subtype. The two classes -31-
ny-2833630
Atty Docket No.23887-2000140 are Class 1 and Class 2. Class 1 CRISPR-Cas systems have effector modules composed of multiple Cas proteins, some of which form crRNA-binding complexes, while Class 2 CRISPR- Cas systems include a single, multi-domain crRNA-binding protein. [0104] In some embodiments, the system is a Cas-based system that is capable of performing a specialized function or activity. For example, the Cas protein may be fused, operably coupled to, or otherwise associated with one or more functional domains. In certain example embodiments, the Cas protein may be a catalytically dead Cas protein (“dCas”) and/or have nickase activity. A nickase is a Cas protein that cuts only one strand of a double stranded target. In such embodiments, the dCas or nickase provide a sequence specific targeting functionality that delivers the functional domain to or proximate a target sequence. Example functional domains that may be fused to, operably coupled to, or otherwise associated with a Cas protein can be or include, but are not limited to, a nuclear localization signal (NLS) domain, a nuclear export signal (NES) domain, a translational activation domain, a transcriptional activation domain (e.g. VP64, p65, MyoDl, HSF1, RTA, and SET7/9), a translation initiation domain, a transcriptional repression domain (e.g., a KRAB domain, NuE domain, NcoR domain, and a SID domain such as a SID4X domain), a nuclease domain (e.g., FokI), a histone modification domain (e.g., a histone acetyltransferase), a light inducible/controllable domain, a chemically inducible/controllable domain, a transposase domain, a homologous recombination machinery domain, a recombinase domain, an integrase domain, and combinations thereof. Methods for generating catalytically dead Cas variants are known in the art. [0105] In some embodiments, the functional domains can have one or more of the following activities: methylase activity, demethylase activity, translation activation activity, translation initiation activity, translation repression activity, transcription activation activity, transcription repression activity, transcription release factor activity, histone modification activity, nuclease activity, single-strand RNA cleavage activity, double-strand RNA cleavage activity, single- strand DNA cleavage activity, double-strand DNA cleavage activity, molecular switch activity, chemical inducibility, light inducibility, and nucleic acid binding activity. In some embodiments, the one or more functional domains may comprise epitope tags or reporters. Non-limiting examples of epitope tags include histidine (His) tags, V5 tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags. Examples of reporters include, but are not limited to, glutathione-S-transferase (GST), horseradish peroxidase (HRP), chloramphenicol acetyltransferase (CAT) beta-galactosidase, beta-glucuronidase, luciferase, green fluorescent protein (GFP), HcRed, DsRed, cyan fluorescent protein (CFP), -32-
ny-2833630
Atty Docket No.23887-2000140 yellow fluorescent protein (YFP), and auto-fluorescent proteins including blue fluorescent protein (BFP). [0106] The one or more functional domain(s) may be positioned at, near, and/or in proximity to a terminus of the programmable nucleic acid binding protein (e.g., a Cas protein). In embodiments having two or more functional domains, each of the two can be positioned at or near or in proximity to a terminus of the effector protein (e.g., a Cas protein). In some embodiments, such as those where the functional domain is operably coupled to the effector protein, the one or more functional domains can be tethered or linked via a suitable linker (including, but not limited to, GlySer linkers) to the effector protein (e.g., a Cas protein). When there is more than one functional domain, the functional domains can be same or different. In some embodiments, all the functional domains are the same. In some embodiments, all of the functional domains are different from each other. In some embodiments, at least two of the functional domains are different from each other. In some embodiments, at least two of the functional domains are the same as each other. [0107] In some embodiments, the CRISPR-Cas system is a split CRISPR-Cas system. In certain embodiments, each part of a split CRISPR protein are attached to a member of a specific binding pair, and when bound with each other, the members of the specific binding pair maintain the parts of the CRISPR protein in proximity. In certain embodiments, each part of a split CRISPR protein is associated with an inducible binding pair. An inducible binding pair is one which is capable of being switched “on” or “off’ by a protein or small molecule that binds to both members of the inducible binding pair. In some embodiments, CRISPR proteins may preferably split between domains, leaving domains intact. In particular embodiments, said Cas split domains (e.g., RuvC and HNH domains in the case of Cas9) can be simultaneously or sequentially introduced into the cell such that said split Cas domain(s) process the target nucleic acid sequence in the cell. The reduced size of the split Cas compared to the wild-type Cas allows other methods of delivery of the systems to the cells, such as the use of cell penetrating peptides as described herein. [0108] In some embodiments, the CRISPR/Cas9 system comprises a base editor. Thus, in some embodiments the Cas-based system can be a base editing system. As used herein “base editing” refers generally to the process of polynucleotide modification via a CRISPR-Cas-based or Cas- based system that does not include excising nucleotides to make the modification. Base editing can convert base pairs at precise locations without generating excess undesired editing byproducts that can be made using traditional CRISPR-Cas systems. -33-
ny-2833630
Atty Docket No.23887-2000140 [0109] In certain example embodiments, the nucleotide deaminase may be a DNA base editor used in combination with a DNA binding Cas protein such as, but not limited to, Class 2 Type II and Type V systems. Two classes of DNA base editors are generally known: cytosine base editors (CBEs) and adenine base editors (ABEs). CBEs convert a C•G base pair into a T•A base pair and ABEs convert an A•T base pair to a G•C base pair. Collectively, CBEs and ABEs can mediate all four possible transition mutations (C to T, A to G, T to C, and G to A). In some embodiments, the base editing system includes a CBE and/or an ABE. In some embodiments, a polynucleotide of the present invention described elsewhere herein can be modified using a base editing system. Base editors also generally do not need a DNA donor template and/or rely on homology-directed repair. Upon binding to a target locus in the DNA, base pairing between the guide RNA of the system and the target DNA strand leads to displacement of a small segment of ssDNA in an “R-loop”. Nishimasu et al. Cell.156:935-949. DNA bases within the ssDNA bubble are modified by the enzyme component, such as a deaminase. In some systems, the catalytically disabled Cas protein can be a variant or modified Cas can have nickase functionality and can generate a nick in the non-edited DNA strand to induce cells to repair the non-edited strand using the edited strand as a template. Base editors may be further engineered to optimize conversion of nucleotides (e.g. A:T to G:C). [0110] In some embodiments, the CRISPR/Cas9 system is a prime editing system. Like base editing systems, prime editing systems can be capable of targeted modification of a polynucleotide without generating double stranded breaks and does not require donor templates. Further, prime editing systems can be capable of all 12 possible combination swaps. Prime editing can operate via a “search-and-replace” methodology and can mediate targeted insertions, deletions, all 12 possible base-to-base conversion, and combinations thereof. Generally, a prime editing systemcan include a reverse transcriptase fused or otherwise coupled or associated with an RNA-programmable nickase, and a prime-editing extended guide RNA (pegRNA) to facilitate direct copying of genetic information from the extension on the pegRNA into the target polynucleotide. Embodiments that can be used with the present invention include these and variants thereof. Prime editing can have the advantage of lower off-target activity than traditional CRISPR-Cas systems along with few byproducts and greater or similar efficiency as compared to traditional CRISPR-Cas systems. [0111] In some embodiments, the prime editing guide molecule can specify both the target polynucleotide information (e.g., sequence) and contain a new polynucleotide cargo that replaces target polynucleotides. To initiate transfer from the guide molecule to the target polynucleotide, the PE system can nick the target polynucleotide at a target side to expose a 3'hydroxyl group, -34-
ny-2833630
Atty Docket No.23887-2000140 which can prime reverse transcription of an edit-encoding extension region of the guide molecule (e.g., a prime editing guide molecule or peg guide molecule) directly into the target site in the target polynucleotide. [0112] In some embodiments, a prime editing system can be composed of a Cas polypeptide having nickase activity, a reverse transcriptase, and a guide molecule. The Cas polypeptide can lack nuclease activity. The guide molecule can include a target binding sequence as well as a primer binding sequence and a template containing the edited polynucleotide sequence. The guide molecule, Cas polypeptide, and/or reverse transcriptase can be coupled together or otherwise associate with each other to form an effector complex and edit a target sequence. In some embodiments, the Cas polypeptide is a Class 2, Type V Cas polypeptide. In some embodiments, the Cas polypeptide is a Cas9 polypeptide (e.g. is a Cas9 nickase). In some embodiments, the Cas polypeptide is fused to the reverse transcriptase. In some embodiments, the Cas polypeptide is linked to the reverse transcriptase. [0113] The CRISPR-Cas or Cas-Based system described herein can, in some embodiments, include one or more guide molecules. The terms guide molecule, guide sequence and guide polynucleotide, refer to polynucleotides capable of guiding Cas to a target genomic locus and are used interchangeably herein. In general, a guide sequence is any polynucleotide comprising a sequence having sufficient complementarity with a target polynucleotide sequence to hybridize with the target sequence and direct sequence-specific binding of a CRISPR complex to the target sequence. The guide molecule can be a polynucleotide. In some embodiments, the portion of the guide molecule having complementarity with a DNA strand in a target gene is referred to as a “spacer” or “spacer sequence.” [0114] The ability of a guide sequence (within a nucleic acid-targeting guide RNA) to direct sequence-specific binding of a nucleic acid-targeting complex to a target nucleic acid sequence may be assessed by any suitable assay. For example, the components of a nucleic acid- targeting CRISPR system sufficient to form a nucleic acid-targeting complex, including the guide sequence to be tested, may be provided to a host cell having the corresponding target nucleic acid sequence, such as by transfection with vectors encoding the components of the nucleic acid-targeting complex, followed by an assessment of preferential targeting (e.g., cleavage) within the target nucleic acid sequence, such as by Surveyor assay. Similarly, cleavage of a target nucleic acid sequence may be evaluated in vitro by providing the target nucleic acid sequence, components of a nucleic acid-targeting complex, including the guide sequence to be tested and a control guide sequence different from the test guide sequence, and comparing -35-
ny-2833630
Atty Docket No.23887-2000140 binding or rate of cleavage at the target sequence between the test and control guide sequence reactions. Other assays are possible and are known to those skilled in the art. [0115] In some embodiments, the guide molecule is an RNA. The guide molecule(s) (also referred to interchangeably herein as guide polynucleotide and guide sequence) that are included in the CRISPR-Cas or Cas based system can be any polynucleotide comprising a sequence having sufficient complementarity with a target nucleic acid sequence to hybridize with the target nucleic acid sequence and direct sequence-specific binding of a nucleic acid-targeting complex to the target nucleic acid sequence (e.g., a spacer sequence). In some embodiments, the degree of complementarity, when optimally aligned using a suitable alignment algorithm, can be about or more than about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99%, or more. Optimal alignment may be determined with the use of any suitable algorithm for aligning sequences, non- limiting examples of which include the Smith-Waterman algorithm, the Needleman-Wunsch algorithm, algorithms based on the Burrows- Wheeler Transform (e.g., the Burrows Wheeler Aligner), ClustalW, Clustal X, BLAT, Novoalign (Novocraft Technologies; available at www.novocraft.com), ELAND (Illumina, San Diego, CA), SOAP (available at soap.genomics.org.cn), and Maq (available at maq.sourceforge.net). [0116] A guide or spacer sequence, and hence a nucleic acid-targeting guide, may be selected to target any target nucleic acid sequence. The target sequence may be DNA. The target sequence may be any RNA sequence. In some embodiments, a nucleic acid-targeting guide is selected to reduce the degree secondary structure within the nucleic acid-targeting guide. In some embodiments, about or less than about 75%, 50%, 40%, 30%, 25%, 20%, 15%, 10%, 5%, 1%, or fewer of the nucleotides of the nucleic acid-targeting guide participate in self- complementary base pairing when optimally folded. Optimal folding may be determined by any suitable polynucleotide folding algorithm. Some programs are based on calculating the minimal Gibbs free energy. An example of one such algorithm is mFold. Another example folding algorithm is the online webserver RNAfold, developed at Institute for Theoretical Chemistry at the University of Vienna, using the centroid structure prediction algorithm. [0117] In certain embodiments, a guide RNA or crRNA may comprise, consist essentially of, or consist of a direct repeat (DR) sequence and a guide sequence or spacer sequence. In certain embodiments, the guide RNA or crRNA may comprise, consist essentially of, or consist of a direct repeat sequence fused or linked to a guide sequence or spacer sequence. In certain embodiments, the direct repeat sequence may be located upstream (i.e., 5') from the guide sequence or spacer sequence. In other embodiments, the direct repeat sequence may be located downstream (i.e., 3') from the guide sequence or spacer sequence. In certain embodiments, the -36-
ny-2833630
Atty Docket No.23887-2000140 crRNA comprises a stem loop, preferably a single stem loop. In certain embodiments, the direct repeat sequence forms a stem loop, preferably a single stem loop. [0118] In certain embodiments, the spacer length of the guide RNA is from 15 to 35 nt. In certain embodiments, the spacer length of the guide RNA is at least 15 nucleotides. In certain embodiments, the spacer length is from 15 to 17 nt, e.g., 15, 16, or 17 nt, from 17 to 20 nt, e.g., 17, 18, 19, or 20 nt, from 20 to 24 nt, e.g., 20, 21, 22, 23, or 24 nt, from 23 to 25 nt, e.g., 23, 24, or 25 nt, from 24 to 27 nt, e.g., 24, 25, 26, or 27 nt, from 27 to 30 nt, e.g., 27, 28, 29, or 30 nt, from 30 to 35 nt, e.g., 30, 31, 32, 33, 34, or 35 nt, or 35 nt or longer. [0119] The “tracrRNA” sequence or analogous terms includes any polynucleotide sequence that has sufficient complementarity with a crRNA sequence to hybridize. In some embodiments, the degree of complementarity between the tracrRNA sequence and crRNA sequence along the length of the shorter of the two when optimally aligned is about or more than about 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97.5%, 99%, or higher. In some embodiments, the tracr sequence is about or more than about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, or more nucleotides in length. In some embodiments, the tracr sequence and crRNA sequence are contained within a single transcript, such that hybridization between the two produces a transcript having a secondary structure, such as a hairpin. [0120] In general, degree of complementarity is with reference to the optimal alignment of the sequence and tracr sequence, along the length of the shorter of the two sequences. Optimal alignment may be determined by any suitable alignment algorithm and may further account for secondary structures, such as self-complementarity within either the sea sequence or tracr sequence. In some embodiments, the degree of complementarity between the tracr sequence and sea sequence along the length of the shorter of the two when optimally aligned is about or more than about 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97.5%, 99%, or higher. [0121] In some embodiments, the degree of complementarity between a spacer (e.g., guide) sequence and its corresponding target sequence can be about or more than about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99%, or 100%. A guide RNA or sgRNA can be about or more than about 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75, 80, 85, 90, 95, 100 or more nucleotides in length. A guide RNA or sgRNA can be less than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12, or fewer nucleotides in length. Tracr RNA can be 30 or 50 nucleotides in length. In some embodiments, the degree of complementarity between a guide sequence (e.g., the spacer) and its corresponding target sequence is greater than 94.5% or 95% or 95.5% or 96% or 96.5% or 97% or 97.5% or 98% or 98.5% or 99% or 99.5% or 99.9%, or 100%. Off target is less than 100% or 99.9% or 99.5% or -37-
ny-2833630
Atty Docket No.23887-2000140 99% or 99% or 98.5% or 98% or 97.5% or 97% or 96.5% or 96% or 95.5% or 95% or 94.5% or 94% or 93% or 92% or 91% or 90% or 89% or 88% or 87% or 86% or 85% or 84% or 83% or 82% or 81% or 80% complementarity between the sequence and the guide, with it advantageous that off target is 100% or 99.9% or 99.5% or 99% or 99% or 98.5% or 98% or 97.5% or 97% or 96.5% or 96% or 95.5% or 95% or 94.5% complementarity between the sequence and the guide. [0122] In some embodiments, the guide RNA (capable of guiding Cas to a target locus) may comprise (1) a spacer sequence capable of hybridizing to a genomic target locus in the cell; (2) a tracr sequence; and (3) a tracr mate sequence. All (1) to (3) may reside in a single RNA, i.e., an sgRNA (arranged in a 5’ to 3’ orientation), or the tracr RNA may be a different RNA than the RNA containing the spacer and tracr sequence. The tracr hybridizes to the tracr mate sequence and directs the CRISPR/Cas complex to the target sequence. Where the tracr RNA is on a different RNA than the RNA containing the guide and tracr sequence, the length of each RNA may be optimized to be shortened from their respective native lengths, and each may be independently chemically modified to protect from degradation by cellular RNase or otherwise increase stability. [0123] Many modifications to guide sequences are known in the art and are further contemplated within the context of the present disclosure. Various modifications may be used to increase the specificity of binding to the target sequence and/or increase the activity of the Cas protein and/or reduce off-target effects. [0124] In the context of formation of a CRISPR complex, “target sequence” refers to a sequence to which a spacer sequence is designed to have complementarity, where hybridization between a target sequence and a guide sequence promotes the formation of a CRISPR complex. A target sequence may comprise DNA or RNA polynucleotides. The term “target sequence” can refer to a polynucleotide being or comprising the target sequence. In other words, the target polynucleotide can be a polynucleotide or a part of a polynucleotide to which a part of the guide sequence is designed to have complementarity with and to which the effector function mediated by the complex comprising the CRISPR effector protein and a guide molecule is to be directed. In some embodiments, a target sequence is located in the nucleus or cytoplasm of a cell. [0125] The guide sequence can specifically bind a target sequence in a target polynucleotide. The target polynucleotide may be DNA. The target polynucleotide may be RNA. The target polynucleotide can have one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, etc. or more) target sequences. The target polynucleotide can be on a vector. The target polynucleotide can be genomic DNA. The target polynucleotide can be episomal. -38-
ny-2833630
Atty Docket No.23887-2000140 [0126] Protospacer adjacent motifs (PAMs) are sequences that can be recognized and bound by Cas proteins. Cas proteins/effector complexes can then unwind the dsDNA at a position adjacent to the PAM element. It will be appreciated that Cas proteins and systems that include them that target RNA do not require PAM sequences Instead, many rely on PFSs (protospacer flanking sequence or site). In certain embodiments, the target sequence should be associated with a PAM (protospacer adjacent motif) or PFS (protospacer flanking sequence or site), that is, a short sequence recognized by the CRISPR complex. Depending on the nature of the CRISPR-Cas protein, the target sequence should be selected, such that its complementary sequence in the DNA duplex is upstream or downstream of the PAM. In some embodiments, the complementary sequence of the target sequence is downstream or 3’ of the PAM or upstream or 5’ of the PAM. The precise sequence and length requirements for the PAM differ depending on the Cas protein used, but PAMs are typically 2-5 base pair sequences adjacent to the protospacer (that is, the target sequence). Examples of the natural PAM sequences for different Cas proteins are provided herein below and the skilled person will be able to identify further PAM sequences for use with a given Cas protein. The ability to recognize different PAM sequences depends on the Cas polypeptide(s) included in the system. Therefore, in some embodiments, the spacer sequence and sequence adjacent to the PAM are the same. [0127] PAM sequences can be identified in a polynucleotide using an appropriate design tool, which are commercially available as well as online. Such freely available tools include, but are not limited to, CRISPRFinder and CRISPRTarget. Experimental approaches to PAM identification can include, but are not limited to, plasmid depletion assays, screened by a high- throughput in vivo model called PAM-SCNAR, and negative screening. [0128] As previously mentioned, CRISPR-Cas systems that target RNA do not typically rely on PAM sequences. Instead, such systems typically recognize protospacer flanking sites (PFSs) instead of PAMs Thus, Type VI CRISPR-Cas systems typically recognize protospacer flanking sites (PFSs) instead of PAMs. PFSs represents an analogue to PAMs for RNA targets. Type VI CRISPR-Cas systems employ a Cas13. Some Cast 3 proteins analyzed to date, such as Cast 3a (C2c2) identified from Leptotrichia shahii (LShCAs13a) have a specific discrimination against G at the 3'end of the target RNA. The presence of a C at the corresponding crRNA repeat site can indicate that nucleotide pairing at this position is rejected. However, some Cas13 proteins (e.g., LwaCAs13a and PspCas13b) do not seem to have a PFS preference. -39-
ny-2833630
Atty Docket No.23887-2000140 Chimeric Antigen Receptors and Engineered T Cell Receptors [0129] The genetically modified Treg cell can comprise: (ii) a second nucleic acid comprising a constitutive promoter operably linked to a polynucleotide encoding a chimeric antigen receptor (CAR) or a T-cell receptor (TCR). [0130] The term “Chimeric Antigen Receptor” or alternatively a “CAR” refers to a set of polypeptides, typically two in the simplest embodiments, which when in an immune effector cell, provides the cell with specificity for a target cell, with intracellular signal generation. The terms “CAR” and “CAR molecule” are used interchangeably. In some embodiments, a CAR comprises at least an extracellular antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (also referred to herein as “an intracellular signaling domain”) comprising a functional signaling domain derived from a stimulatory molecule and/or costimulatory molecule as defined below. In some embodiments, the set of polypeptides are in the same polypeptide chain (e.g., comprise a chimeric fusion protein). In some aspects, the set of polypeptides are contiguous with each other. In some embodiments, the set of polypeptides are not contiguous with each other, e.g., are in different polypeptide chains. In some embodiments, the set of polypeptides include a dimerization switch that, upon the presence of a dimerization molecule, can couple the polypeptides to one another, e.g., can couple an antigen binding domain to an intracellular signaling domain. In one aspect, the stimulatory molecule is the zeta chain associated with the T cell receptor complex. In one aspect, the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below. In some embodiments, the costimulatory molecule is chosen from the costimulatory molecules described herein, e.g., 4-1BB (i.e., CD137), CD27 and/or CD28. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising a functional signaling domain derived from a costimulatory molecule and a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an intracellular signaling domain comprising two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In some embodiments, the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain and an -40-
ny-2833630
Atty Docket No.23887-2000140 intracellular signaling domain comprising at least two functional signaling domains derived from one or more costimulatory molecule(s) and a functional signaling domain derived from a stimulatory molecule. In some embodiments the CAR comprises an optional leader sequence at the amino-terminus (N-terminus) of the CAR fusion protein. In some embodiments, the CAR further comprises a leader sequence at the N-terminus of the extracellular antigen binding domain, wherein the leader sequence is optionally cleaved from the antigen binding domain (e.g., a scFv) during cellular processing and localization of the CAR to the cellular membrane. [0131] The CAR and/or TCR can comprise one or more of an antigen binding domain, a transmembrane domain, and an intracellular signaling domain. The CAR or TCR further can comprise a leader peptide. The TCR further can comprise a constant region and/or CDR4. The term “signaling domain” refers to the functional portion of a protein which acts by transmitting information within the cell to regulate cellular activity via defined signaling pathways by generating second messengers or functioning as effectors by responding to such messengers. An “intracellular signaling domain,” as the term is used herein, refers to an intracellular portion of a molecule. The intracellular signaling domain generates a signal that promotes an immune effector function of the CAR containing cell, e.g., a CAR-T cell. Examples of immune effector function, e.g., in a CAR-T cell, include cytolytic activity and helper activity, including the secretion of cytokines. In an embodiment, the intracellular signaling domain can comprise a primary intracellular signaling domain. Exemplary primary intracellular signaling domains include those derived from the molecules responsible for primary stimulation, or antigen dependent simulation. In an embodiment, the intracellular signaling domain can comprise a costimulatory intracellular domain. Exemplary costimulatory intracellular signaling domains include those derived from molecules responsible for costimulatory signals, or antigen independent stimulation. For example, in the case of a CAR-T cell, a primary intracellular signaling domain can comprise a cytoplasmic sequence of a T-cell receptor, and a costimulatory intracellular signaling domain can comprise cytoplasmic sequence from co-receptor or costimulatory molecule. A primary intracellular signaling domain can comprise a signaling motif which is known as an immunoreceptor tyrosine-based activation motif or ITAM. Examples of ITAM containing primary cytoplasmic signaling sequences include, but are not limited to, those derived from CD3 zeta, common FcR gamma (FCER1G), Fc gamma RIIa, FcR beta (Fc Epsilon R1b), CD3 gamma, CD3 delta, CD3 epsilon, CD79a, CD79b, DAP10, and DAP12. [0132] The intracellular signaling domain can comprise a primary signaling domain, a costimulatory domain, or both of a primary signaling domain and a costimulatory domain. The cytoplasmic domain or region of the CAR includes an intracellular signaling domain. An -41-
ny-2833630
Atty Docket No.23887-2000140 intracellular signaling domain is generally responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR has been introduced. The term “effector function” refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines. Thus the term “intracellular signaling domain” refers to the portion of a protein which transduces the effector function signal and directs the cell to perform a specialized function. While usually the entire intracellular signaling domain can be employed, in many cases it is not necessary to use the entire chain. To the extent that a truncated portion of the intracellular signaling domain is used, such truncated portion may be used in place of the intact chain as long as it transduces the effector function signal. The term intracellular signaling domain is thus meant to include any truncated portion of the intracellular signaling domain sufficient to transduce the effector function signal. [0133] The term “costimulatory molecule” refers to a cognate binding partner on a T cell that specifically binds with a costimulatory ligand, thereby mediating a costimulatory response by the T cell, such as, but not limited to, proliferation. Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that contribute to an efficient immune response. Costimulatory molecules include, but are not limited to an MHC class I molecule, BTLA and a Toll ligand receptor, as well as OX40, CD27, CD28, CD5, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). Further examples of such costimulatory molecules include CD5, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, CD19a, and a ligand that specifically binds with CD83. A costimulatory intracellular signaling domain can be the intracellular portion of a costimulatory molecule. A costimulatory molecule can be represented in the following protein families: TNF receptor proteins, Immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocytic activation molecules (SLAM proteins), and activating NK cell receptors. The intracellular signaling domain can comprise the entire intracellular portion, or the entire native -42-
ny-2833630
Atty Docket No.23887-2000140 intracellular signaling domain, of the molecule from which it is derived, or a functional fragment or derivative thereof. [0134] Examples of intracellular signaling domains for use in a CAR include the cytoplasmic sequences of the T-cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction following antigen receptor engagement, as well as any derivative or variant of these sequences and any recombinant sequence that has the same functional capability. It is known that signals generated through the TCR alone are insufficient for full activation of the T cell and that a secondary and/or costimulatory signal is also required. Thus, T cell activation can be said to be mediated by two distinct classes of cytoplasmic signaling sequences: those that initiate antigen- dependent primary activation through the TCR (primary intracellular signaling domains) and those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic domain, e.g., a costimulatory domain). A primary signaling domain regulates primary activation of the TCR complex either in a stimulatory way, or in an inhibitory way. Primary intracellular signaling domains that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine-based activation motifs or ITAMs. The primary signaling domain can comprise a functional signaling domain of one or more proteins selected from the group consisting of CD3 zeta, CD3 gamma, CD3 delta, CD3 epsilon, common FcR gamma (FCER1G), FcR beta (Fc Epsilon R1b), CD79a, CD79b, Fcgamma RIIa, DAP10, and DAP12, or a functional variant thereof. [0135] In some embodiments, the intracellular signaling domain is designed to comprise two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains. In an embodiment, the two or more, e.g., 2, 3, 4, 5, or more, costimulatory signaling domains, are separated by a linker molecule, e.g., a linker molecule described herein. In one embodiment, the intracellular signaling domain comprises two costimulatory signaling domains. In some embodiments, the linker molecule is a glycine residue. In some embodiments, the linker is an alanine residue. The costimulatory domain can comprise a functional domain of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD28-OX40, CD28-4-1BB, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CD5, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), -43-
ny-2833630
Atty Docket No.23887-2000140 CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D, or a functional variant thereof. [0136] The portion of the CAR comprising an antibody or antibody fragment thereof may exist in a variety of forms where the antigen binding domain is expressed as part of a contiguous polypeptide chain including, for example, a single domain antibody fragment (sdAb), a single chain antibody (scFv), a humanized antibody, or bispecific antibody (Harlow et al., 1999, In: Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, In: Antibodies: A Laboratory Manual, Cold Spring Harbor, N.Y.; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426). In some embodiments, the antigen binding domain of a CAR comprises an antibody fragment. In a further aspect, the CAR comprises an antibody fragment that comprises a scFv. [0137] In some embodiments, the CAR comprises a target-specific binding element otherwise referred to as an antigen binding domain. The choice of moiety depends upon the type and number of ligands that define the surface of a target cell. For example, the antigen binding domain may be chosen to recognize a ligand that acts as a cell surface marker on target cells associated with a particular disease state. Thus, examples of cell surface markers that may act as ligands for the antigen binding domain in a CAR of the invention include those associated with viral, bacterial and parasitic infections, autoimmune disease and cancer cells. [0138] In some embodiments, the CAR-mediated T-cell response can be directed to an antigen of interest by way of engineering an antigen binding domain that specifically binds a desired antigen into the CAR. In some embodiments, the portion of the CAR comprising the antigen binding domain comprises an antigen binding domain that targets an antigen associated with autoimmune disease. The antigen binding domain can be any domain that binds to the antigen including but not limited to a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a human antibody, a humanized antibody, and a functional fragment thereof, including but not limited to a single-domain antibody such as a heavy chain variable domain (VH), a light chain variable domain (VL) and a variable domain (VHH) of camelid derived nanobody, and to an alternative scaffold known in the art to function as antigen binding domain, such as a recombinant fibronectin domain, a T cell receptor (TCR), or a fragment there of, e.g., single chain TCR, and the like. In some instances, it is beneficial for the antigen binding domain to be derived from the same species in which the CAR will ultimately be used in. For example, for use in humans, it may be beneficial for the antigen binding domain of the CAR to comprise human -44-
ny-2833630
Atty Docket No.23887-2000140 or humanized residues for the antigen binding domain of an antibody or antibody fragment. In some embodiments, the antigen binding domain comprises a humanized antibody or an antibody fragment. In some aspects, a non-human antibody is humanized, where specific sequences or regions of the antibody are modified to increase similarity to an antibody naturally produced in a human or fragment thereof. In some embodiments, the antigen binding domain is humanized. [0139] The antigen binding domain can comprise an antibody, an antibody fragment, an scFv, a domain, a Fab, a Fab', a F(ab')2, a Fv, a scFv, a dsFv, a diabody, a triabody, a tetrabody, a multispecific antibody formed from antibody fragments, a single-domain antibody (sdAb), a single chain comprising cantiomplementary scFvs (tandem scFvs) or bispecific tandem scFvs, an Fv construct, a disulfide-linked Fv, a dual variable domain immunoglobulin (DVD-Ig) binding protein or a nanobody, an aptamer, an affibody, an affilin, an affitin, an affimer, an alphabody, an anticalin, an avimer, a DARPin, a Fynomer, a Kunitz domain peptide, a monobody, or any combination thereof. [0140] In some embodiments, the antigen binding domain is a T cell receptor (“TCR”), or a fragment thereof, for example, a single chain TCR (scTCR). Methods to make such TCRs are known in the art. See, e.g., Willemsen R A et al, Gene Therapy 7: 1369-1377 (2000); Zhang T et al, Cancer Gene Ther 11: 487-496 (2004); Aggen et al, Gene Ther.19(4):365-74 (2012) (references are incorporated herein by its entirety). For example, scTCR can be engineered that ker (e.g., a flexible peptide). [0141] In some embodiments, the antigen binding domain is a multispecific antibody molecule. In some embodiments, the multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody has specificity for no more than two antigens. A bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope. In an embodiment the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein). In an embodiment the first and second epitopes overlap. In an embodiment the first and second epitopes do not overlap. In an embodiment the first and second epitopes are on different antigens, e.g., different proteins (or different subunits of a multimeric protein). In an embodiment a bispecific antibody molecule comprises a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a first epitope and a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody having -45-
ny-2833630
Atty Docket No.23887-2000140 binding specificity for a first epitope and a half antibody having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody, or fragment thereof, having binding specificity for a first epitope and a half antibody, or fragment thereof, having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a scFv, or fragment thereof, have binding specificity for a first epitope and a scFv, or fragment thereof, have binding specificity for a second epitope. [0142] The antigen binding domain can be connected to the transmembrane domain by a hinge region. In some instances, the transmembrane domain can be attached to the extracellular region of the CAR, e.g., the antigen binding domain of the CAR, via a hinge, e.g., a hinge from a human protein. For example, in one embodiment, the hinge can be a human Ig (immunoglobulin) hinge (e.g., an IgG4 hinge, an IgD hinge), a GS linker, a KIR2DS2 hinge or a CD8a hinge. [0143] With respect to the transmembrane domain, in various embodiments, a CAR can be designed to comprise a transmembrane domain that is attached to the extracellular domain of the CAR. A transmembrane domain can include one or more additional amino acids adjacent to the transmembrane region, e.g., one or more amino acid associated with the extracellular region of the protein from which the transmembrane was derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the extracellular region) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 up to 15 amino acids of the intracellular region). In some embodiments, the transmembrane domain is one that is associated with one of the other domains of the CAR e.g., in one embodiment, the transmembrane domain may be from the same protein that the signaling domain, costimulatory domain or the hinge domain is derived from. In some embodiments, the transmembrane domain is not derived from the same protein that any other domain of the CAR is derived from. In some instances, the transmembrane domain can be selected or modified by amino acid substitution to avoid binding of such domains to the transmembrane domains of the same or different surface membrane proteins, e.g., to minimize interactions with other members of the receptor complex. In some embodiments, the transmembrane domain is capable of homodimerization with another CAR on the cell surface of a CAR-expressing cell. In a different aspect, the amino acid sequence of the transmembrane domain may be modified or substituted so as to minimize interactions with the binding domains of the native binding partner present in the same CAR-expressing cell. [0144] The transmembrane domain can comprise a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, -46-
ny-2833630
Atty Docket No.23887-2000140 CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and NKG2C, or a functional variant thereof. The transmembrane domain may be derived either from a natural or from a recombinant source. Where the source is natural, the domain may be derived from any membrane-bound or transmembrane protein. In some embodiments the transmembrane domain is capable of signaling to the intracellular domain(s) whenever the CAR has bound to a target. [0145] The CAR can comprise a binding domain, a hinge region, a transmembrane domain, and an intracellular signaling domain. In some embodiments, the binding domain is capable of binding a target selected from the group consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6-trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage-gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), a heat shock protein, and a protein of the human leukocyte antigen (HLA) system. In some embodiments, the HLA protein is selected from the group consisting of HLA-A, HLA-B, HLA-
C, HLA-E, HLA-F, HLA- s or variants thereof. The HLA protein can comprise the HLA-A2 serotype. In some embodiments, the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. In some embodiments, the TCR further comprises a constant region and a variable region. [0146] In some embodiments, the TCR is capable of binding an antigen selected from the group consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6-trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage-gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), and a heat shock protein. In some embodiments, the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. [0147] In some embodiments, the second nucleic acid comprises a constitutive promoter operably linked to a polynucleotide encoding a chimeric antigen receptor (CAR) or a T-cell -47-
ny-2833630
Atty Docket No.23887-2000140 receptor (TCR). The constitutive promoter can be a ubiquitous promoter or a tissue-specific promoter. In some embodiments, the constitutive promoter is selected from the group comprising a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus-forming virus (SFFV), a Moloney murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock -
- KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1
(PGK) promoter, 3-phospho - -actin (CBA) promoter, a CAG promoter, a CBH promoter, a variant thereof, or any combination thereof. Engineered Immune Cells [0148] Disclosed herein include thermally actuated immune cells. The immune cell can be a mammalian cell (e.g., a human cell). The immune cell can be derived from blood, cord blood, bone marrow, or iPSC. The immune cell can be a T cell. The T cell can be a primary T cell. The T cell can be an autologous T cell or an allogeneic T cell. A single thermal stimulus can be sufficient to initiate a positive feedback loop of immune cell activation-driven expression of the immunostimulatory agent product. [0149] Regulatory T cells (Tregs or T
reg cells), also known as suppressor T cells, are a subpopulation of T cells that modulate the immune system, maintain tolerance to self-antigens, and prevent autoimmune disease. Treg cells are immunosuppressive and generally suppress or downregulate induction and proliferation of effector T cells (e.g., cytotoxic, helper T cells). T
reg cells express the biomarkers CD4, FOXP3, and CD25 and are thought to be derived from the same lineage as naïve CD4+ cells. Research has found that the cytokine transforming
growth factor beta (TGF- reg cells to differentiate from naïve CD4 + cells and is important in maintaining T
reg cell homeostasis. Modulation of T
reg cells can treat autoimmune disease and cancer and can facilitate organ transplantation and wound healing. In some embodiments, the immune cell is a T regulatory cell. In some embodiments, the genetically modified Treg cell expresses one or more of the markers selected from the group consisting of CD4, CD25, and forkhead box P3 (FOXP3). -48-
ny-2833630
Atty Docket No.23887-2000140 [0150] The genetic modification can comprise integration of the first nucleic acid into the genome of the Treg cell. The first nucleic acid can be integrated within a safe harbor locus. The safe harbor locus can comprise AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. The genetic modification can comprise integration of the second nucleic acid into the genome of the Treg cell. The second nucleic acid can be integrated within a safe harbor locus. The safe harbor locus can comprise AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. [0151] The genetically modified Treg cell can be allogenic or autologous to the subject. In some embodiments, the method comprises obtaining cells (e.g., hematopoietic cells) from a subject, genetically modifying the cells according to the methods and compositions disclosed herein, and administering the genetically modified cells to the subject. The genetically modified Treg cell can be differentiated from a genetically modified stem cell. The genetically modified stem cell can comprise an embryonic stem cell or an induced pluripotent stem cell. The genetically modified stem cell can comprise a hematopoietic stem cell. In some embodiments, the allogenic Treg cell comprises one or more mutations in gene(s), or one or more nucleic acids encoding gene products that reduce allogenic immune response in a subject. [0152] Disclosed herein include populations of the thermally actuated immune cells. In some embodiments, the population of the thermally actuated immune cells comprises: a plurality of the thermally actuated immune cells disclosed herein. Thermally actuated immune cells disclosed herein can be actuated (e.g., induction of expression from an inducible promoter) by T cell activity (e.g., immune cell stimulation) and/or thermal stimulation. [0153] Disclosed herein include methods of generating a thermally actuated immune cell. In some embodiments, the method comprises: introducing a nucleic acid disclosed herein or a nucleic acid composition disclosed herein into an immune cell, thereby generating a thermally actuated immune cell. The introducing step can comprise calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, electrical nuclear transport, chemical transduction, electrotransduction, Lipofectamine-mediated transfection, Effectene-mediated transfection, lipid nanoparticle (LNP)-mediated transfection, or any combination thereof. [0154] In embodiments described herein, the immune effector cell can be an allogeneic immune effector cell, e.g., T cell or NK cell. For example, the cell can be an allogeneic T cell, e.g., an allogeneic T cell lacking expression of a functional T cell receptor (TCR) and/or human leukocyte antigen (HLA), e.g., HLA class I and/or HLA class II. A T cell lacking a functional -49-
ny-2833630
Atty Docket No.23887-2000140 TCR can be, e.g., engineered such that it does not express any functional TCR on its surface, engineered such that it does not express one or more subunits that comprise a functional TCR or engineered such that it produces very little functional TCR on its surface. Alternatively, the T cell can express a substantially impaired TCR, e.g., by expression of mutated or truncated forms of one or more of the subunits of the TCR. The term “substantially impaired TCR” means that this TCR will not elicit an adverse immune reaction in a host. Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA. For example, the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN). [0155] In certain embodiments, the T cells are obtained from a donor subject. In some embodiments, the donor subject is human patient afflicted with an autoimmune disease or disorder. In other embodiments, the donor subject is a human patient not afflicted with an autoimmune disease or disorder. [0156] The immune cells of the present invention may be obtained through any source known in the art. For example, T cells can be differentiated in vitro from a hematopoietic stem cell population, or T cells can be obtained from a subject. T cells can be obtained from, e.g., peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In addition, the T cells can be derived from one or more T cell lines available in the art. T cells can also be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as FICOLL™ separation and/or apheresis. In certain embodiments, the cells collected by apheresis are washed to remove the plasma fraction, and placed in an appropriate buffer or media for subsequent processing. In some embodiments, the cells are washed with PBS. As will be appreciated, a washing step can be used, such as by using a semiautomated flowthrough centrifuge, e.g., the Cobe™ 2991 cell processor, the Baxter CytoMate™, or the like. In some embodiments, the washed cells are resuspended in one or more biocompatible buffers, or other saline solution with or without buffer. In certain embodiments, the undesired components of the apheresis sample are removed. Additional methods of isolating T cells for a T cell therapy are disclosed in U.S. Patent Publication No.2013/0287748, which is herein incorporated by references in its entirety. [0157] In certain embodiments, T cells are isolated from PBMCs by lysing the red blood cells and depleting the monocytes, e.g., by using centrifugation through a PERCOLL™ gradient. In -50-
ny-2833630
Atty Docket No.23887-2000140 some embodiments, a specific subpopulation of T cells, such as CD4
+, CD8
+, CD28
+, CD45RA
+, and CD45RO
+ T cells is further isolated by positive or negative selection techniques known in the art. For example, enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells. In some embodiments, cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected can be used. For example, to enrich for CD4
+ cells by negative selection, a monoclonal antibody cocktail typically includes antibodies to CD8, CD11b, CD14, CD16, CD20, and HLA-DR. In certain embodiments, flow cytometry and cell sorting are used to isolate cell populations of interest for use in the present invention. [0158] T regulatory cells are a component of the immune system that suppress immune responses of other cells. This is an important "self-check" built into the immune system to prevent excessive reactions. Regulatory T cells come in many forms with the most well- understood being those that express CD4, CD25, and FOXP3 (CD4+CD25+ regulatory T cells). These Treg cells are different from helper T cells. Another regulatory T cell subset is Treg17 cells. Regulatory T cells are involved in shutting down immune responses after they have successfully eliminated invading organisms, and also in preventing autoimmunity. [0159] In some embodiments, the Tregs are differentiated from stem cells. Stem cells, such as hematopoietic progenitor cells, are capable of proliferation and giving rise to more progenitor cells having the ability to generate a large number of mother cells that can in turn give rise to differentiated or differentiable daughter cells. The daughter cells themselves can be induced to proliferate and produce progeny that subsequently differentiate into one or more mature cell types, while also retaining one or more cells with parental developmental potential. The term “stem cell” refers then, to a cell with the capacity or potential, under particular circumstances, to differentiate to a more specialized or differentiated phenotype, and which retains the capacity, under certain circumstances, to proliferate without substantially differentiating. In one embodiment, the term progenitor or stem cell refers to a generalized mother cell whose descendants (progeny) specialize, often in different directions, by differentiation, e.g., by acquiring completely individual characters, as occurs in progressive diversification of embryonic cells and tissues. Cellular differentiation is a complex process typically occurring through many cell divisions. A differentiated cell may derive from a multipotent cell which itself is derived from a multipotent cell, and so on. While each of these multipotent cells may be considered stem cells, the range of cell types each can give rise to may vary considerably. Some differentiated -51-
ny-2833630
Atty Docket No.23887-2000140 cells also have the capacity to give rise to cells of greater developmental potential. Such capacity may be natural or may be induced artificially upon treatment with various factors. In many biological instances, stem cells are also “multipotent” because they can produce progeny of more than one distinct cell type, but this is not required for “stem-ness.” Self-renewal is the other classical part of the stem cell definition, and it is essential as used in this document. In theory, self-renewal can occur by either of two major mechanisms. Stem cells may divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype. Alternatively, some of the stem cells in a population can divide symmetrically into two stems, thus maintaining some stem cells in the population as a whole, while other cells in the population give rise to differentiated progeny only. Generally, “progenitor cells” have a cellular phenotype that is more primitive (i.e., is at an earlier step along a developmental pathway or progression than is a fully differentiated cell). Often, progenitor cells also have significant or very high proliferative potential. Progenitor cells can give rise to multiple distinct differentiated cell types or to a single differentiated cell type, depending on the developmental pathway and on the environment in which the cells develop and differentiate. [0160] In the context of cell ontogeny, the adjective “differentiated”, or “differentiating” is a relative term. A “differentiated cell” is a cell that has progressed further down the developmental pathway than the cell it is being compared with. Thus, stem cells can differentiate to lineage- restricted precursor cells (such as a hematopoietic progenitor cell), which in turn can differentiate into other types of precursor cells further down the pathway (such as an erythrocyte precursor), and then to an end-stage differentiated cell, such as an erythrocyte, which plays a characteristic role in a certain tissue type, and may or may not retain the capacity to proliferate further. [0161] In some embodiments, the genetically engineered cells described herein are derived from induced pluripotent stem cells (iPSCs). An advantage of using iPSCs is that the cells can be derived from the same subject to which the genetically modified cells (e.g., the genetically modified Tregs) are to be administered. That is, a somatic cell can be obtained from a subject, reprogrammed to an induced pluripotent stem cell, and then re-differentiated into an immune cell to be administered to the subject (e.g., autologous cells). Since the progenitors are essentially derived from an autologous source, the risk of engraftment rejection or allergic responses is reduced compared to the use of cells from another subject or group of subjects. In some embodiments, the hematopoietic progenitors are derived from non-autologous sources. In -52-
ny-2833630
Atty Docket No.23887-2000140 addition, the use of iPSCs negates the need for cells obtained from an embryonic source. Thus, in one embodiment, the stem cells used in the disclosed methods are not embryonic stem cells. [0162] Although differentiation is generally irreversible under physiological contexts, several methods have been recently developed to reprogram somatic cells to induced pluripotent stem cells. Exemplary methods are known to those of skill in the art and are described briefly herein below. [0163] As used herein, the term “reprogramming” refers to a process that alters or reverses the differentiation state of a differentiated cell (e.g., a somatic cell). Stated another way, reprogramming refers to a process of driving the differentiation of a cell backwards to a more undifferentiated or more primitive type of cell. It should be noted that placing many primary cells in culture can lead to some loss of fully differentiated characteristics. Thus, simply culturing such cells included in the term differentiated cells does not render these cells non- differentiated cells (e.g., undifferentiated cells) or pluripotent cells. The transition of a differentiated cell to pluripotency requires a reprogramming stimulus beyond the stimuli that lead to partial loss of differentiated character in culture. Reprogrammed cells also have the characteristic of the capacity of extended passaging without loss of growth potential, relative to primary cell parents, which generally have capacity for only a limited number of divisions in culture. [0164] The cell to be reprogrammed can be either partially or terminally differentiated prior to reprogramming. In some embodiments, reprogramming encompasses complete reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to a pluripotent state or a multipotent state. In some embodiments, reprogramming encompasses complete or partial reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to an undifferentiated cell (e.g., an embryonic-like cell). Reprogramming can result in expression of particular genes by the cells, the expression of which further contributes to reprogramming. In certain embodiments described herein, reprogramming of a differentiated cell (e.g., a somatic cell) causes the differentiated cell to assume an undifferentiated state (e.g., is an undifferentiated cell). The resulting cells are referred to as “reprogrammed cells,” or “induced pluripotent stem cells (iPSCs or iPS cells).” [0165] Reprogramming can involve alteration, e.g., reversal, of at least some of the heritable patterns of nucleic acid modification (e.g., methylation), chromatin condensation, epigenetic changes, genomic imprinting, etc., that occur during cellular differentiation. Reprogramming is distinct from simply maintaining the existing undifferentiated state of a cell that is already pluripotent or maintaining the existing less than fully differentiated state of a cell that is already -53-
ny-2833630
Atty Docket No.23887-2000140 a multipotent cell (e.g., a hematopoietic stem cell). Reprogramming is also distinct from promoting the self-renewal or proliferation of cells that are already pluripotent or multipotent, although the compositions and methods described herein can also be of use for such purposes, in some embodiments. The specific approach or method used to generate pluripotent stem cells from somatic cells (broadly referred to as “reprogramming”) is not critical to the claimed invention. Thus, any method that re-programs a somatic cell to the pluripotent phenotype would be appropriate for use in the methods described herein. [0166] Reprogramming methodologies for generating pluripotent cells using defined combinations of transcription factors have been described. In one example, somatic cells can be de-differentiated to ES cell-like cells with expanded developmental potential by the direct transduction of the so-called “Yamanaka factors”: Oct4, Sox2, Klf4, and c-Myc. iPSCs resemble ES cells as they restore the pluripotency-associated transcriptional circuitry and much of the epigenetic landscape. In addition, mouse iPSCs satisfy all the standard assays for pluripotency: specifically, in vitro differentiation into cell types of the three germ layers, teratoma formation, contribution to chimeras, germline transmission, and tetraploid complementation. In some embodiments, the transcription factor trio, OCT4, SOX2, and NANOG, has been established as the core set of transcription factors that govern pluripotency. The production of iPS cells can be achieved by the introduction of nucleic acid sequences encoding stem cell-associated genes into an adult, somatic cell, e.g. through using viral vectors. [0167] iPS cells can be generated or derived from terminally differentiated somatic cells, as well as from adult stem cells, or somatic stem cells. That is, a non-pluripotent progenitor cell can be rendered pluripotent or multipotent by reprogramming. In such instances, it may not be necessary to include as many reprogramming factors as required to reprogram a terminally differentiated cell. Further, reprogramming can be induced by the non-viral introduction of reprogramming factors, e.g., by introducing the proteins themselves, or by introducing nucleic acids that encode the reprogramming factors, or by introducing messenger RNAs that upon translation produce the reprogramming factors. Reprogramming can be achieved by introducing a combination of nucleic acids encoding stem cell-associated genes including, for example Oct-4 (also known as Oct-3/4 or Pouf51), Sox1, Sox2, Sox3, Sox 15, Sox 18, NANOG, Klf1, Klf2, Klf4, Klf5, NR5A2, c-Myc, 1-Myc, n-Myc, Rem2, Tert, and LIN28. In one embodiment, reprogramming can further comprise introducing one or more of Oct-3/4, a member of the Sox family, a member of the Klf family, and a member of the Myc family to a somatic cell. In one embodiment, the methods and compositions described herein further comprise introducing one or more of each of Oct 4, Sox2, Nanog, c-MYC and Klf4 for reprogramming. As noted above, -54-
ny-2833630
Atty Docket No.23887-2000140 the exact method used for reprogramming is not necessarily critical to the methods and compositions described herein. However, where cells differentiated from the reprogrammed cells are to be used in, e.g., human therapy, in one embodiment the reprogramming is not effected by a method that alters the genome. Thus, in such embodiments, reprogramming is achieved, e.g., without the use of viral or plasmid vectors. [0168] To confirm the induction of pluripotent stem cells for use with the methods described herein, isolated clones can be tested for the expression of a stem cell marker. Such expression in a cell derived from a somatic cell identifies the cells as induced pluripotent stem cells. Stem cell markers can be selected from the non-limiting group including SSEA3, SSEA4, CD9, Nanog, Fbx15, Ecat1, Esg1, Eras, Gdf3, Fgf4, Cripto, Dax1, Zpf296, Slc2a3, Rex1, Utf1, and Nat1. In one embodiment, a cell that expresses Oct4 or Nanog is identified as pluripotent. Methods for detecting the expression of such markers can include, for example, RT-PCR and immunological methods that detect the presence of the encoded polypeptides, such as Western blots or flow cytometric analyses. In some embodiments, detection does not involve only RT-PCR, but also includes detection of protein markers. Intracellular markers may be best identified via RT-PCR, while cell surface markers are readily identified, e.g., by immunocytochemistry. [0169] The pluripotent stem cell character of isolated cells can be confirmed by tests evaluating the ability of the iPSCs to differentiate to cells of each of the three germ layers. As one example, teratoma formation in nude mice can be used to evaluate the pluripotent character of the isolated clones. The cells are introduced to nude mice and histology and/or immunohistochemistry is performed on a tumor arising from the cells. The growth of a tumor comprising cells from all three germ layers, for example, further indicates that the cells are pluripotent stem cells. [0170] Somatic cells, as that term is used herein, refer to any cells forming the body of an organism, excluding germline cells. Every cell type in the mammalian body—apart from the sperm and ova, the cells from which they are made (gametocytes) and undifferentiated stem cells—is a differentiated somatic cell. For example, internal organs, skin, bones, blood, and connective tissue are all made up of differentiated somatic cells. Additional somatic cell types for use with the compositions and methods described herein include: a fibroblast (e.g., a primary fibroblast), a muscle cell (e.g., a myocyte), a cumulus cell, a neural cell, a mammary cell, an hepatocyte and a pancreatic islet cell. In some embodiments, the somatic cell is a primary cell line or is the progeny of a primary or secondary cell line. In some embodiments, the somatic cell is obtained from a human sample, e.g., a hair follicle, a blood sample, a biopsy (e.g., a skin biopsy or an adipose biopsy), a swab sample (e.g., an oral swab sample), and is thus a human somatic cell. Some non-limiting examples of differentiated somatic cells include, but are not -55-
ny-2833630
Atty Docket No.23887-2000140 limited to, epithelial, endothelial, neuronal, adipose, cardiac, skeletal muscle, immune cells, hepatic, splenic, lung, circulating blood cells, gastrointestinal, renal, bone marrow, and pancreatic cells. In some embodiments, a somatic cell can be a primary cell isolated from any somatic tissue including, but not limited to brain, liver, gut, stomach, intestine, fat, muscle, uterus, skin, spleen, endocrine organ, bone, etc. Further, the somatic cell can be from any mammalian species, with non-limiting examples including a murine, bovine, simian, porcine, equine, ovine, or human cell. In some embodiments, the somatic cell is a human somatic cell. [0171] When reprogrammed cells are used for generation of hematopoietic progenitor cells to be used in the therapeutic treatment of disease, it is desirable, but not required, to use somatic cells isolated from the patient being treated. For example, somatic cells involved in diseases, and somatic cells participating in therapeutic treatment of diseases and the like can be used. In some embodiments, a method for selecting the reprogrammed cells from a heterogeneous population comprising reprogrammed cells and somatic cells they were derived or generated from can be performed by any known means. For example, a drug resistance gene or the like, such as a selectable marker gene can be used to isolate the reprogrammed cells using the selectable marker as an index. [0172] Reprogrammed somatic cells as disclosed herein can express any number of pluripotent cell markers, including: alkaline phosphatase (AP); ABCG2; stage specific embryonic antigen-1 (SSEA-1); SSEA-3; SSEA-4; TRA-1-60; TRA-1-81; Tra-2-49/6E; ERas/ECAT5, E-cadherin; I
II-tubulin; - -SMA); fibroblast growth factor 4 (Fgf4), Cripto, Dax1; zinc finger protein 296 (Zfp296); N-acetyltransferase-1 (Nat1); (ES cell associated transcript 1 (ECAT1); ESG1/DPPA5/ECAT2; ECAT3; ECAT6; ECAT7; ECAT8; ECAT9; ECAT10; ECAT15-1; ECAT15-2; Fth117; Sal14; undifferentiated embryonic cell transcription factor (Utf1); Rex1; p53; G3PDH; telomerase, including TERT; silent X chromosome genes; Dnmt3a; Dnmt3b; TRIM28; F-box containing protein 15 (Fbx15); Nanog/ECAT4; Oct3/4; Sox2; Klf4; c- Myc; Esrrb; TDGF1; GABRB3; Zfp42, FoxD3; GDF3; CYP25A1; developmental pluripotency- associated 2 (DPPA2); T-cell lymphoma breakpoint 1 (Tcl1); DPPA3/Stella; DPPA4; other general markers for pluripotency, etc. Other markers can include Dnmt3L; Sox15; Stat3; Grb2; -catenin, and Bmi1. Such cells can also be characterized by the down-regulation of markers characteristic of the somatic cell from which the induced pluripotent stem cell is derived. [0173] As used herein, the term “genome editing” refers to a reverse genetics method using artificially engineered nucleases to cut and create specific double-stranded breaks at a desired location(s) in the genome, which are then repaired by cellular endogenous processes such as, homologous recombination (HR), homology directed repair (HDR) and non-homologous end- -56-
ny-2833630
Atty Docket No.23887-2000140 joining (NHEJ). NHEJ directly joins the DNA ends in a double-stranded break, while HDR utilizes a homologous sequence as a template for regenerating the missing DNA sequence at the break point. [0174] Genome editing cannot be performed using traditional restriction endonucleases since most restriction enzymes recognize a few base pairs on the DNA as their target and the probability is very high that the recognized base pair combination will be found in many locations across the genome resulting in multiple cuts (i.e., not limited to a desired location). To overcome this challenge and create site-specific double-stranded breaks, several distinct classes of nucleases have been discovered and bioengineered to date. These are the meganucleases, Zinc finger nucleases (ZFNs), Cas9/CRISPR system, and transcription-activator like effector nucleases (TALENs). [0175] Meganucleases are found commonly in microbial species and have the unique property of having very long recognition sequences (>14 bp) thus making them naturally very specific for cutting at a desired location. This can be exploited to make site-specific double-stranded breaks in genome editing. One of skill in the art can use these naturally occurring meganucleases, however the number of such naturally occurring meganucleases is limited. To overcome this challenge, mutagenesis and high throughput screening methods have been used to create meganuclease variants that recognize unique sequences. For example, various meganucleases have been fused to create hybrid enzymes that recognize a new sequence. Alternatively, DNA interacting amino acids of the meganuclease can be altered to design sequence specific meganucleases (see e.g., U.S. Pat. No.8,021,867). Meganucleases can be designed using the methods described in e.g., Certo, M T et al. Nature Methods (2012) 9:073-975; U.S. Pat. Nos. 8,304,222; 8,021,867; 8,119,381; 8,124,369; 8,129,134; 8,133,697; 8,143,015; 8,143,016; 8,148,098; or 8,163,514, the contents of each are incorporated herein by reference in their entirety. Alternatively, meganucleases with site specific cutting characteristics can be obtained using commercially available technologies e.g., Precision BioSciences' Directed Nuclease Editor™ genome editing technology. [0176] ZFNs and TALENs restriction endonuclease technology utilizes a non-specific DNA cutting enzyme which is linked to a specific DNA sequence recognizing peptide(s) such as zinc fingers and transcription activator-like effectors (TALEs). Typically an endonuclease whose DNA recognition site and cleaving site are separate from each other is selected and the its cleaving portion is separated and then linked to a sequence recognizing peptide, thereby yielding an endonuclease with very high specificity for a desired sequence. An exemplary restriction enzyme with such properties is Fold. Additionally Fold has the advantage of requiring -57-
ny-2833630
Atty Docket No.23887-2000140 dimerization to have nuclease activity and this means the specificity increases dramatically as each nuclease partner recognizes a unique DNA sequence. To enhance this effect, Fold nucleases have been engineered that can only function as heterodimers and have increased catalytic activity. The heterodimer functioning nucleases avoid the possibility of unwanted homodimer activity and thus increase specificity of the double-stranded break. [0177] Although the nuclease portions of both ZFNs and TALENs have similar properties, the difference between these engineered nucleases is in their DNA recognition peptide. ZFNs rely on Cys2-His2 zinc fingers and TALENs on TALEs. Both of these DNA recognizing peptide domains have the characteristic that they are naturally found in combinations in their proteins. Cys2-His2 Zinc fingers typically happen in repeats that are 3 bp apart and are found in diverse combinations in a variety of nucleic acid interacting proteins such as transcription factors. TALEs on the other hand are found in repeats with a one-to-one recognition ratio between the amino acids and the recognized nucleotide pairs. Because both zinc fingers and TALEs happen in repeated patterns, different combinations can be tried to create a wide variety of sequence specificities. Approaches for making site-specific zinc finger endonucleases include, e.g., modular assembly (where Zinc fingers correlated with a triplet sequence are attached in a row to cover the required sequence), OPEN (low-stringency selection of peptide domains vs. triplet nucleotides followed by high-stringency selections of peptide combination vs. the final target in bacterial systems), and bacterial one-hybrid screening of zinc finger libraries, among others. ZFNs for use with the methods and compositions described herein can be obtained commercially from e.g., Sangamo Biosciences™ (Richmond, Calif.). [0178] It is contemplated herein that the Cas9/CRISPR system of genome editing be employed with the methods and compositions described herein. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems is useful for RNA- programmable genome editing (see e.g., Jinek, M. et al. Science (2012) 337(6096):816-821). [0179] Trans-activating crRNA (tracrRNA) is a small trans-encoded RNA. It was first discovered in the human pathogen Streptococcus pyogenes. (see Deltcheva E, et al. (2011). Nature 471 (7340): 602-7). In bacteria and archaea, CRISPR/Cas (clustered, regularly interspaced short palindromic repeats/CRISPR-associated proteins) constitute an RNA-mediated defense system which protects against viruses and plasmids. This defensive pathway has three steps. First a copy of the invading nucleic acid is integrated into the CRISPR locus. Next, CRISPR RNAs (crRNAs) are transcribed from this CRISPR locus. The crRNAs are then incorporated into effector complexes, where the crRNA guides the complex to the invading nucleic acid and the Cas proteins degrade this nucleic acid. (See Terns MP and Terns RM -58-
ny-2833630
Atty Docket No.23887-2000140 (2011). Curr Opin Microbiol 14 (3): 321-7). There are several pathways of CRISPR activation, one of which requires a tracrRNA which plays a role in the maturation of crRNA. TracrRNA is complementary to and base pairs with a pre-crRNA forming an RNA duplex. This is cleaved by RNase III, an RNA-specific ribonuclease, to form a crRNA/tracrRNA hybrid. This hybrid acts as a guide for the endonuclease Cas9, which cleaves the invading nucleic acid. (see Deltcheva E, et al. supra; Jinek M, et al. (2012), Science 337 (6096): 816-21; and Brouns S J (2012), Science 337 (6096): 808-9). [0180] Alternatively, genome editing can be performed using recombinant adeno-associated virus (rAAV) based genome engineering, which is a genome-editing platform centered around the use of rAAV vectors that enables insertion, deletion or substitution of DNA sequences into the genomes of live mammalian cells. The rAAV genome is a single-stranded deoxyribonucleic acid (ssDNA) molecule, either positive- or negative-sensed, which is about 4.7 kilobase long. These single-stranded DNA viral vectors have high transduction rates and have a unique property of stimulating endogenous homologous recombination in the absence of causing double strand DNA breaks in the genome. One of skill in the art can design a rAAV vector to target a desired genomic locus and perform both gross and/or subtle endogenous gene alterations in a cell, such as a deletion. rAAV genome editing has the advantage in that it targets a single allele and does not result in any off-target genomic alterations. rAAV genome editing technology is commercially available, for example, the rAAV GENESIS™ system from Horizon™ (Cambridge, UK). [0181] In some embodiments, the immune cells, e.g., T cells, are genetically modified following isolation using known methods, or the immune cells are activated and expanded (or differentiated in the case of progenitors) in vitro prior to being genetically modified. In another embodiment, the immune cells, e.g., T cells, are genetically modified with the chimeric antigen receptors described herein (e.g., transduced with a viral vector comprising one or more nucleotide sequences encoding a CAR) and then are activated and/or expanded in vitro. Methods for activating and expanding T cells are known in the art and are described, e.g., in U.S. Pat. Nos.6,905,874; 6,867,041; and 6,797,514; and PCT Publication No. WO 2012/079000, the contents of which are hereby incorporated by reference in their entirety. Generally, such methods include contacting PBMC or isolated T cells with a stimulatory agent and costimulatory agent, such as anti-CD3 and anti-CD28 antibodies, generally attached to a bead or other surface, in a culture medium with appropriate cytokines, such as IL-2. Anti-CD3 and anti-CD28 antibodies attached to the same bead serve as a “surrogate” antigen presenting cell (APC). One example is the Dynabeads® system, a CD3/CD2S activator/stimulator system for physiological activation of -59-
ny-2833630
Atty Docket No.23887-2000140 human ‘I’ cells. In other embodiments, the T cells are activated and stimulated to proliferate with feeder cells and appropriate antibodies and cytokines using methods such as those described in U.S. Pat. Nos.6,040,177 and 5,827,642 and PCT Publication No. WO 2012/129514, the contents of which are hereby incorporated by reference in their entirety. [0182] Physical methods for introducing a polynucleotide into an immune cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. Methods for producing cells comprising vectors and/or exogenous nucleic acids are well-known in the art. See, for example, Sambrook et al., 2012, MOLECULAR CLONING: A LABORATORY MANUAL, volumes 1-4, Cold Spring Harbor Press, NY). In some embodiments, the method for the introduction of a polynucleotide into an immune cell is calcium phosphate transfection [0183] Chemical means for introducing a polynucleotide into an immune cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle). Other methods of state-of-the-art targeted delivery of nucleic acids are available, such as delivery of polynucleotides with targeted nanoparticles or other suitable sub-micron sized delivery system. [0184] In the case where a non-viral delivery system is utilized, an exemplary delivery vehicle is a liposome. The use of lipid formulations is contemplated for the introduction of the nucleic acids into an immune cell (in vitro, ex vivo or in vivo). In some embodiments, the nucleic acid may be associated with a lipid. The nucleic acid associated with a lipid may be encapsulated in the aqueous interior of a liposome, interspersed within the lipid bilayer of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the oligonucleotide, entrapped in a liposome, complexed with a liposome, dispersed in a solution containing a lipid, mixed with a lipid, combined with a lipid, contained as a suspension in a lipid, contained or complexed with a micelle, or otherwise associated with a lipid. Lipid, lipid/DNA or lipid/expression vector associated compositions are not limited to any particular structure in solution. [0185] Nucleic acids described herein can be introduced into immune cells using any of a number of different methods, for instance, commercially available methods which include, but are not limited to, electroporation (Amaxa Nucleofector-II (Amaxa Biosystems, Cologne, Germany)), (ECM 830 (BTX) (Harvard Instruments, Boston, Mass.) or the Gene Pulser II (BioRad, Denver, Colo.), Multiporator (Eppendorf, Hamburg Germany), cationic liposome -60-
ny-2833630
Atty Docket No.23887-2000140 mediated transfection using lipofection, polymer encapsulation, peptide mediated transfection, or biolistic particle delivery systems such as “gene guns” (see, for example, Nishikawa, et al. Hum Gene Ther., 12(8):861-70 (2001). [0186] In some aspects, non-viral methods can be used to deliver a nucleic acid described herein into an immune cell. In some embodiments, the non-viral method includes the use of a transposon (also called a transposable element). In some embodiments, a transposon is a piece of DNA that can insert itself at a location in a genome, for example, a piece of DNA that is capable of self-replicating and inserting its copy into a genome, or a piece of DNA that can be spliced out of a longer nucleic acid and inserted into another place in a genome. For example, a transposon comprises a DNA sequence made up of inverted repeats flanking genes for transposition. Exemplary methods of nucleic acid delivery using a transposon include a Sleeping Beauty transposon system (SBTS) and a piggyBac (PB) transposon system. In some embodiments, thermally actuated immune cells described herein are generated by using a combination of gene insertion using the SBTS and genetic editing using a nuclease (e.g., Zinc finger nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs), the CRISPR/Cas system, or engineered meganuclease re-engineered homing endonucleases). In some embodiments, use of a non-viral method of delivery permits reprogramming of cells, e.g., T cells, and direct infusion of the thermally actuated immune cells described herein into a subject. Advantages of non-viral vectors include but are not limited to the ease and relatively low cost of producing sufficient amounts required to meet a patient population, stability during storage, and lack of immunogenicity. Pharmaceutical compositions [0187] Disclosed herein are compositions. The compositions can be pharmaceutical compositions. The composition can comprise one or more pharmaceutically acceptable excipients. A “pharmaceutical composition” refers to a composition that usually contains an excipient, such as a pharmaceutically acceptable carrier that is conventional in the art and that is suitable for administration to cells or to a subject. The term “pharmaceutically acceptable” as used throughout this specification is consistent with the art and means compatible with the other ingredients of a pharmaceutical composition and not deleterious to the recipient thereof. [0188] As used herein, “carrier” or “excipient” includes any and all solvents, diluents, buffers (such as, e.g., neutral buffered saline or phosphate buffered saline), solubilisers, colloids, dispersion media, vehicles, fillers, chelating agents (such as, e.g., EDTA or glutathione), amino acids (such as, e.g., glycine), proteins, disintegrants, binders, lubricants, wetting agents, -61-
ny-2833630
Atty Docket No.23887-2000140 emulsifiers, sweeteners, colorants, flavourings, aromatisers, thickeners, agents for achieving a depot effect, coatings, antifungal agents, preservatives, stabilisers, antioxidants, tonicity controlling agents, absorption delaying agents, and the like. The use of such media and agents for pharmaceutical active components is well known in the art. Such materials should be non- toxic and should not interfere with the activity of the cells or active components. The precise nature of the carrier or excipient or other material will depend on the route of administration. For example, the composition may be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has suitable pH, isotonicity and stability. For general principles in medicinal formulation, the reader is referred to Cell Therapy: Stem Cell Transplantation, Gene Therapy, and Cellular Immunotherapy, by G. Morstyn & W. Sheridan eds., Cambridge University Press, 1996; and Hematopoietic Stem Cell Therapy, E. D. Ball, J. Lister & P. Law, Churchill Livingstone, 2000. [0189] In some embodiments, the pharmaceutical composition may be used for intravenous administration. In some embodiments, the pharmaceutical composition according to the invention is intended to be used as an infusion. The skilled person will understand that compositions which are to be administered orally or topically will usually not comprise cells, although it may be envisioned for oral compositions to also comprise cells, for example when gastro-intestinal tract indications are treated. Each of the cells or active components (e.g., immunomodulants) as discussed herein may be administered by the same route or may be administered by a different route. By means of example, and without limitation, cells may be administered parenterally and other active components may be administered orally. [0190] Liquid pharmaceutical compositions may generally include a liquid carrier such as water or a pharmaceutically acceptable aqueous solution. For example, physiological saline solution, tissue or cell culture media, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included. [0191] The composition may include one or more cell protective molecules, cell regenerative molecules, growth factors, anti-apoptotic factors or factors that regulate gene expression in the cells. Such substances may render the cells independent of their environment. [0192] Such pharmaceutical compositions may contain further components ensuring the viability of the cells therein. For example, the compositions may comprise a suitable buffer system (e.g., phosphate or carbonate buffer system) to achieve desirable pH, more usually near neutral pH, and may comprise sufficient salt to ensure isoosmotic conditions for the cells to prevent osmotic stress. For example, suitable solution for these purposes may be phosphate-buffered saline (PBS), sodium chloride solution, Ringer's Injection or Lactated Ringer's Injection, as known in -62-
ny-2833630
Atty Docket No.23887-2000140 the art. Further, the composition may comprise a carrier protein, e.g., albumin (e.g., bovine or human albumin), which may increase the viability of the cells. [0193] Further suitably pharmaceutically acceptable carriers or additives are well known to those skilled in the art and for instance may be selected from proteins such as collagen or gelatine, carbohydrates such as starch, polysaccharides, sugars (dextrose, glucose and sucrose), cellulose derivatives like sodium or calcium carboxymethylcellulose, hydroxypropyl cellulose or hydroxypropylmethyl cellulose, pregeletanized starches, pectin agar, carrageenan, clays, hydrophilic gums (acacia gum, guar gum, arabic gum and xanthan gum), alginic acid, alginates, hyaluronic acid, polyglycolic and polylactic acid, dextran, pectins, synthetic polymers such as water-soluble acrylic polymer or polyvinylpyrrolidone, proteoglycans, calcium phosphate and the like. [0194] In certain embodiments, a pharmaceutical cell preparation as taught herein may be administered in a form of liquid composition. In embodiments, the cells or pharmaceutical composition comprising such can be administered systemically. The compositions disclosed herein can advantageously be administered systemically, and activated locally, e.g., via localized thermal activation. [0195] The pharmaceutical compositions may comprise a therapeutically effective amount of the specified immune cells and/or other active components (e.g., immunomodulants). The term “therapeutically effective amount” refers to an amount which can elicit a biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, and in particular can prevent or alleviate one or more of the local or systemic symptoms or features of a disease or condition being treated. Inducible promoters and thermal circuits [0196] There are provided, in some embodiments, promoters capable of inducing transcription upon thermal stimulation and/or immune cell stimulation (e.g., inducible promoters). The inducible promoters provided herein can, in some embodiments, sense T cell activity. The first inducible promoter can comprise or can be derived from a mammalian heat shock promoter (HSP) or a C. elegans HSP. The mammalian HSP can be a human HSP or mouse HSP. The first inducible promoter can comprise a nucleotide sequence that is at least 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to SEQ ID NOs: 1-83, 88-102, and 108-116. The first inducible promoter can comprise one or more AP-1 sites. In some embodiments, the first inducible promoter does not comprise an AP-1 site. The first inducible promoter can comprise a bidirectional promoter and/or a minimal bidirectional promoter. The first inducible promoter can -63-
ny-2833630
Atty Docket No.23887-2000140 comprise one or more heat shock element (HSE) binding sites (e.g., four HSE binding sites). In some embodiments, the first inducible promoter does not comprise a human transcription factor binding site other than one or more HSE binding sites. In some embodiments, the first inducible promoter comprises one or more of a TATA box, GC-Box, CAAT signal, and AP-1 site. Nucleic acids provided herein can comprise a portion of a promoter, an enhancer, positive or negative cis-acting sequences, inducible or repressible control element, 5’ UTR sequences that are upstream of a gene, or any combination thereof. A disclosed promoter (e.g., first inducible promoter) can comprise 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 HSE binding sites. The inducible promoter can comprise a promoter sequence as shown, e.g., in Table 1. [0197] A disclosed promoter (e.g., first inducible promoter) can be derived from the heat shock promoter (HSP) of one or more species selected from the group comprising: Arabidopsis thaliana; Aspergillus nidulans; Bombyx mori; Candida albicans; Caenorhabditis elegans; Chlamydomonas rheinhardtii; Cricetulus griseus; Cyanophora paradoxa; Cylindrotheca fusiformis; Danio rerio; Dictyostelium discoideum; Drosophila melanogaster; Drosophila yakuba; Gallus gallus; Homo Sapiens; Leishmania chagasi; Leishmania major; Loligo pealii; Lymantria dispar; Monodelphis domestica; Morone saxatilis; Mus musculus; Nectria haematococca; Neurospora crassa; Nicotiana tabacum; Oryza sativa; Paracentrotus lividus; Plasmodium falciparum; Rattus norvegicus; Saccharomyces cerevisiae; Schizosaccharomyces pombe; Solanum tuberosum; Strongylocentrotus purpuratus; Syncephalastrum racemosum; Tetrahymena thermophila; Trypanosoma brucei; Ustilago maydis; Volvox carteri; and Xenopus laevis. [0198] The length of the promoters provided herein (e.g., first inducible promoter) can vary. In some embodiments, a disclosed promoter is, or is about, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 128, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 1100, 1150, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 2000, 2050, 2100, 2150, 2200, 2250, 2300, 2350, 2400, 2450, 2500, 2550, 2600, 2650, 2700, 2750, 2800, 2850, 2900, 2950, 3000, or 4000, or a number or a range between any two of these values, nucleotides in length. In some -64-
ny-2833630
Atty Docket No.23887-2000140 embodiments, a disclosed promoter is at least, or is at most, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 128, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 1100, 1150, 1000, 1050, 1100, 1150, 1200, 1250, 1300, 1350, 1400, 1450, 1500, 1550, 1600, 1650, 1700, 1750, 1800, 1850, 1900, 1950, 2000, 2050, 2100, 2150, 2200, 2250, 2300, 2350, 2400, 2450, 2500, 2550, 2600, 2650, 2700, 2750, 2800, 2850, 2900, 2950, 3000, or 4000, nucleotides in length. [0199] In some embodiments, the sequence identity between a disclosed promoter (e.g., first inducible promoter) and the sequence of any one of SEQ ID NOs: 1-83, 88-102, and 108-116 can be, or be about, 0.000000001%, 0.00000001%, 0.0000001%, 0.000001%, 0.00001%, 0.0001%, 0.001%, 0.01%, 0.1%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, or a number or a range between any two of these values. In some embodiments, the sequence identity between a disclosed promoter (e.g., first promoter) and the sequence of any one of SEQ ID NOs: 1-83, 88-102, and 108-116 can be at least, or at most, 0.000000001%, 0.00000001%, 0.0000001%, 0.000001%, 0.00001%, 0.0001%, 0.001%, 0.01%, 0.1%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. [0200] A disclosed promoter (e.g., first inducible promoter) can comprise at least about 20 consecutive nucleotides (e.g., about 20 nt, 25 nt, 30 nt, 35 nt, 40 nt, 45 nt, 50 nt, 60 nt, 70 nt, 80 nt, 90 nt, 100 nt, 110 nt, 120 nt, 128 nt, 130 nt, 140 nt, 150 nt, 160 nt, 170 nt, 180 nt, 190 nt, 200 -65-
ny-2833630
Atty Docket No.23887-2000140 nt, 210 nt, 220 nt, 230 nt, 240 nt, 250 nt, 260 nt, 270 nt, 280 nt, 290 nt, 300 nt, 310 nt, 320 nt, 330 nt, 340 nt, 350 nt, 360 nt, 370 nt, 380 nt, 390 nt, 400 nt, 410 nt, 420 nt, 430 nt, 440 nt, 450 nt, 460 nt, 470 nt, 480 nt, 490 nt, 500 nt, 510 nt, 520 nt, 530 nt, 540 nt, 550 nt, 560 nt, 570 nt, 580 nt, 590 nt, 600 nt, 610 nt, 620 nt, 630 nt, 640 nt, 650 nt, 660 nt, 670 nt, 680 nt, 690 nt, 700 nt, 710 nt, 720 nt, 730 nt, 740 nt, 750 nt, 760 nt, 770 nt, 780 nt, 790 nt, 800 nt, 810 nt, 820 nt, 830 nt, 840 nt, 850 nt, 860 nt, 870 nt, 880 nt, 890 nt, 900 nt, 910 nt, 920 nt, 930 nt, 940 nt, 950 nt, 960 nt, 970 nt, 980 nt, 990 nt, 1000 nt, or a number or a range between any two of these values) of a sequences described by SEQ ID NOS: 1-83, 88-102, and 108-116. [0201] Provided herein are Treg cells comprising nucleic acids comprising a polynucleotide encoding a immunostimulatory agent operably linked to an inducible promoter. The inducible promoter can comprise a core promoter and at least one heat shock element (HSE). In some embodiments, the core promoter comprises the heat shock protein transcription start site of a gene selected from the group consisting of HSPA1A, HSPH1, HSPB1, HSPA6, CMVa, CMVb, and YB. The core promoter can comprise or consist of the sequence of any one of SEQ ID NOs: 9-12 and 35-46 or a sequence having at least, about, or at least about 85% identity (e.g., 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or a number or a range between any two of these values) to any one of SEQ ID NOs: 9-12 and 35-46. The core promoter can comprise a sequence selected from the group consisting of SEQ ID NOs: 9-12 and 35-46. [0202] The inducible promoter can comprise seven HSEs. Each of the at least one HSE can comprise the sequence of 5’- nGAAnnTTCnnGAAn-3’. The at least one HSE can comprise or consist of the sequence of any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence having at least 85% identity (e.g., 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or a number or a range between any two of these values) to any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence differing by one or two nucleotide mismatches relative to any one of SEQ ID NOs: 1-8, 53, and 112-116. The at least one HSE can comprise a sequence selected from the group consisting of SEQ ID NOs: 1-8, 53, and 112-116. [0203] The inducible promoter can comprise or consist of the sequence of any one of SEQ ID NOs: 13-20, 58-80, and 88-94 or a sequence having at least 85% identity (e.g., 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or a number or a range between any two of these values) to any one of SEQ ID NOs: 13-20, 58-80, and 88- 94. The inducible promoter can comprise a sequence selected from the group consisting of SEQ ID NOs: 13-20, 58-80, and 88-94. The inducible promoter can comprise or consist of the sequence of any one of SEQ ID NOs: 21-34 or a sequence having at least 85% identity (e.g., -66-
ny-2833630
Atty Docket No.23887-2000140 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100% identity, or a number or a range between any two of these values) to any one of SEQ ID NOs: 21-34. The inducible promoter can comprise or consist of a sequence selected from the group consisting of SEQ ID NOs: 21-34. Table 1 displays exemplary HSE and inducible promoters of the present disclosure. Certain embodiments of the thermal circuits described herein are also described in PCT publications WO 2022/272102 and WO 2021/211776, the contents of which are hereby incorporated by reference in their entireties. TABLE 1: EXEMPLARY INDUCIBLE PROMOTER SEQUENCES
-67- n
y-2833630
Atty Docket No.23887-2000140
-68- n
y-2833630
Atty Docket No.23887-2000140
-69- n
y-2833630
Atty Docket No.23887-2000140

[0204] The thermal stimulation can comprise heating to an activating temperature. In some embodiments, the activating temperature is greater than 37°C. The activating temperature can comprise a temperature of at least 37°C to at most 70°C. The activating temperature can be about 37.0ºC, 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC. [0205] The activating temperature can be about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC. The activating temperature can comprise a temperature of about 42°C to about 43°C. The activating temperature can be about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. -70- n
y-2833630
Atty Docket No.23887-2000140 Expression Levels and Tuning [0206] In some embodiments, in the absence of thermal stimulation, the immunostimulatory agent reaches unstimulated steady state levels in an immune cell and/or in the surrounding tissue. Unstimulated steady state levels of the immunostimulatory agent product can be insufficient to exert a phenotypic effect and/or therapeutic effect. In some embodiments, upon thermal stimulation transcription of the immunostimulatory agent gene product, transactivator gene, oscillator gene, and/or recombinase gene from the first inducible promoter is increased by at least 1.1-fold fold (e.g., 1.1-fold, 1.3-fold, 1.5-fold, 1.7-fold, 1.9-fold, 2-fold, 3-fold, 4-fold, 5- fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, or a number or a range between any of these values). In some embodiments, increasing transactivator-binding compound concentration increases stimulated steady state levels. [0207] In some embodiments, the steady-state levels of the immunostimulatory agent transcript, the steady-state levels of transactivator transcript, the steady-state levels of recombinase transcript, the steady-state levels of oscillator transcript, and/or the steady-state levels of the polycistronic transcript are at least 1.1-fold (e.g., 1.1-fold, 1.3-fold, 1.5-fold, 1.7-fold, 1.9-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, or a number or a range between any of these values) higher upon thermal stimulation and/or immune cell stimulation. In some embodiments, upon thermal stimulation and/or immune cell stimulation, the immunostimulatory agent product reaches stimulated steady state immunostimulatory agent product levels in an immune cell. In some embodiments, the immunostimulatory agent product does not return to unstimulated steady state immunostimulatory agent product levels. [0208] Stimulated steady state immunostimulatory gene product levels can be at least 1.1-fold fold (e.g., 1.1-fold, 1.3-fold, 1.5-fold, 1.7-fold, 1.9-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7- fold, 8-fold, 9-fold, 10-fold, or a number or a range between any of these values) higher than unstimulated steady state immunostimulatory gene product levels. [0209] In some embodiments, after a first duration of time, the immunostimulatory gene product levels returns to unstimulated steady state immunostimulatory agent product levels from stimulated steady state immunostimulatory agent product levels, wherein the first duration of time is about 250 hours, about 200 hours, about 150 hours, about 96 hours, about 48 hours, about 44 hours, about 40 hours, about 35 hours, about 30 hours, about 25 hours, 20 hours, 15 hours, 10 hours, about 8 hours, about 8 hours, 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes, about 10 minutes, about 5 minutes, or a number or a range between any two of these values. -71-
ny-2833630
Atty Docket No.23887-2000140 [0210] In some embodiments, the nucleic acid composition comprises: a transcript stabilization element. The transcript stabilization element can comprise woodchuck hepatitis post- translational regulatory element (WPRE), bovine growth hormone polyadenylation (bGH-polyA) signal sequence, human growth hormone polyadenylation (hGH-polyA) signal sequence, or any combination thereof. The immunostimulatory agent gene can comprise a 5’UTR and/or a 3’UTR. The transactivator gene can comprise a 5’UTR and/or a 3’UTR. The recombinase gene can comprise a 5’UTR and/or a 3’UTR. The oscillator gene can comprise a 5’UTR and/or a 3’UTR. The 5’ UTR can comprise a Kozak sequence. In some embodiments, stimulated steady state immunostimulatory agent product levels, unstimulated steady state immunostimulatory agent product levels, the lower tuned threshold, and/or the upper tuned threshold can be tuned by adjusting the presence and/or sequence of the Kozak sequence. The 5’ UTR can comprise one or more micro open reading frames. In some embodiments, stimulated steady state immunostimulatory agent product levels, unstimulated steady state immunostimulatory agent product levels, the lower tuned threshold, and/or the upper tuned threshold can be tuned by adjusting the presence and/or sequence of the one or more micro open reading frames. Methods of treating autoimmune diseases [0211] Provided herein are compositions for use in treating an autoimmune disease in a subject. [0212] Disclosed herein include methods of treating an autoimmune disease in a subject. In some embodiments, the method comprises: administering any of the compositions disclosed herein to the subject; and applying thermal energy to a target site of the subject sufficient to increase local temperature of the target site to an activating temperature, thereby inducing the expression of the immunostimulatory agent gene product. [0213] The method can comprise administering the composition at a dose of about 1 × 10
5 to about 1 × 10
15 Treg cells. The method can comprise administering the composition at a dose of about, at least, or at least about 1 × 10
5, 1 × 10
6, 1 × 10
7, 1 × 10
8, 1 × 10
9, 1 × 10
10, 1 × 10
11, 1 × 10
12, 1 × 10
13, 1 × 10
14, or 1 × 10
15 Treg cells or a number or range between any two of these values. [0214] The method can comprise a single administration of the composition to the subject. The method can comprise administering the composition to the subject two or more times. In some embodiments, each of the two or more administrations are one month, two months, 6 months, 1 year, or more, apart. [0215] The period of time between the administering the composition and applying thermal energy can be about 1 week, about 6 days, about 5 days, about 4 days, about 3 days, about 48 -72-
ny-2833630
Atty Docket No.23887-2000140 hours, about 44 hours, about 40 hours, about 35 hours, about 30 hours, about 25 hours, 20 hours, 15 hours, 10 hours, about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes, about 10 minutes, or about 5 minutes. [0216] The activating temperature can be greater than 37°C. The activating temperature can comprise a temperature of at least 37.5°C to at most 70.0°C. The activating temperature can be about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC. The thermal energy can be applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, or about 1 minute. [0217] The activating temperature can be about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC. The thermal energy can be applied to the target site for a duration of time comprising about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. [0218] The activating temperature can comprise a temperature of about 42°C to about 43°C. The activating temperature can be about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. The thermal energy can be applied to the target site for a duration of time comprising about 1 second to about 20 minutes. -73-
ny-2833630
Atty Docket No.23887-2000140 [0219] The thermal energy can be applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. [0220] The temperature at the target site can be elevated to about 42°C to about 43°C after the applying. The temperature at the target site can be elevated to about 42°C to about 43°C after the applying. within at most 30 minutes from the applying. The temperature at the target site can be elevated to about 42°C to about 43°C within 10 minutes or less after the applying. [0221] The temperature at the target site can be elevated to about 42°C to about 43°C within about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, or about 10 minutes after the applying. [0222] The temperature at the target site can be maintained at about 42°C to about 43°C for at least about 15 minutes to at least about 120 minutes. The temperature at the target site can be maintained at 42°C to 43°C for at least 15 minutes, at least 20 minutes, at least 25 minutes, at least 30 minutes, at least 35 minutes, at least 40 minutes, at least 45 minutes, at least 50 minutes, at least 55 minutes, at least 60 minutes, at least 90 minutes, or at least 120 minutes. The temperature at the target site can be maintained at 42°C to 43°C for at least 20 minutes. The temperature at the target site can be maintained at about 42°C to about 43°C for about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. [0223] The method can comprise applying thermal energy to the subject two or more times. The method can comprise applying thermal energy to the subject two or more times, e.g., after a single administration of the composition to the subject. The method can comprise applying thermal energy to the subject two or more times, e.g., after each of one, two or three, administrations of the composition to the subject. In some embodiments, each of the two or more applications of thermal energy are 1 week apart, 2 weeks apart, 3 weeks apart, 4 weeks apart, 5 weeks apart, or more. In some embodiments, the presently disclosed method advantageously -74-
ny-2833630
Atty Docket No.23887-2000140 allows for repeated (e.g., monthly) activation, stimulation, and/or proliferation of the Treg cells through thermal stimulation, without requirement of additional administrations of the composition to the subject. [0224] The target site can comprise one or more of muscle, skin, joints, the rectum, and the colon. In some embodiments, the target site can be an internal organ. The muscle can be skeletal, cardiac, or smooth muscle. The internal organ can be liver, kidney, lung, pancreas, or heart. [0225] Applying thermal energy to a target site of the subject can comprise the application of one or more of magnetic hyperthermia, microwave hyperthermia, inductive diathermy, contact hyperthermia, ultrasonic diathermy, deep hyperthermia ingestible hyperthermia, and rectal hyperthermia. Applying thermal energy to a target site of the subject can comprise use of a device selected from the group consisting of: BSD-500 Superficial Hyperthermia, BSD-2000 Deep Regional Hyperthermia, TheraTouch® DX2 Shortwave Diathermy, Dynatronics Solaris Plus, and Hill Laboratories HF54+. The method can comprise monitoring the temperature of the target region. The monitoring can be performed by magnetic resonance imaging (MRI). The application of thermal energy to a target site of the subject can be guided spatially by magnetic resonance imaging (MRI). [0226] In some embodiments, at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the population of thermally actuated genetically modified Treg cells at the target site express the immunostimulatory agent gene product after applying thermal energy to the target site. [0227] In some embodiments, less than about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%, of the population of thermally actuated genetically modified Treg cells at a site other than the target site express the immunostimulatory agent gene product. [0228] In some embodiments, upon applying thermal energy to the target site, the transcription of the immunostimulatory gene, the transactivator gene, the recombinase gene, or any combination thereof, increases by at least 1.1-fold (e.g., 1.1-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, or a number or a range between any of these values). [0229] In some embodiments, upon applying thermal energy to the target site, the levels of the of the immunostimulatory gene product, the transactivator gene product, the recombinase gene product, or any combination thereof, increases by at least 1.1-fold (e.g., 1.1-fold, 1.5-fold, 2-fold, -75-
ny-2833630
Atty Docket No.23887-2000140 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, or a number or a range between any of these values). The gene product can comprise RNA transcribed from the gene. The gene product can comprise an mRNA, a protein translated from the mRNA, or both. [0230] The autoimmune disease can comprise type I diabetes, psoriasis, Lupus nephritis, multiple sclerosis, neuromyelitis optica, chronic inflammatory demyelinating polyneuropathy, polymyositis, dermatomyositis, inclusion body myositis, Crohn’s Disease, ulcerative colitis, primary sclerosing cholangitis, ankylosing spondylitis, rheumatic arthritis, spondyloarthritis, or juvenile idiopathic arthritis. In some embodiments, the method treats or prevents at least one symptom of the autoimmune disease. [0231] The autoimmune disease can be ulcerative colitis and the target site comprises the rectum and/or the colon. In some embodiments, the method treats or prevents at least one symptom of the ulcerative colitis, wherein the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof. In some embodiments, the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more symptoms of diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof. In some embodiments, the method reduces the levels of anti-saccharomyces cerevisiae antibody (ASCA) in the subject. In some embodiments, the reduction of the frequency, duration, and/or intensity of flares and/or the reduction of the levels of ASCA in a subject is relative to: a subject that has not been administered the composition; prior to administering the composition to the subject; and/or prior to the applying thermal energy to the subject. [0232] The autoimmune disease can be Crohn’s Disease. In some embodiments, the target site comprises the rectum, colon, small intestine, or a combination thereof. In some embodiments, the method treats or prevents at least one symptom of the Crohn’s disease, wherein the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof. In some embodiments, the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more symptoms comprising diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof. In some embodiments, the method reduces the levels of perinuclear anti- neutrophil antibodies (pANCA) in the subject. In some embodiments, the reduction of the frequency, duration, and/or intensity of flares and/or levels of pANCA in the subject is relative -76-
ny-2833630
Atty Docket No.23887-2000140 to: a subject that has not been administered the composition; prior to administering the composition to the subject; and/or prior to the applying thermal energy to the subject. [0233] The autoimmune disease can be psoriasis. The target site can comprise the skin. In some embodiments, the method treats or prevents at least one symptom of psoriasis, wherein the at least one symptom comprises skin rash, skin bleeding, skin itch, skin burning, skin soreness, or any combination thereof. In some embodiments, the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more of the symptoms comprising skin rash, skin bleeding, skin itch, skin burning, skin soreness, or any combination thereof. In some embodiments, the reduction of the frequency, duration, and/or intensity of flares in the subject is relative to: a subject that has not been administered the composition; prior to administering the composition to the subject; and/or prior to the applying thermal energy to the subject. [0234] The autoimmune disease can be rheumatoid arthritis (RA). The target site can comprise one or more joints. In some embodiments, the method treats or prevents at least one symptom of RA, where the at least one symptom comprises swelling and/or stiffness in one or more joints. The at least one symptom can comprise swelling and/or stiffness in one or more of a knee, finger, elbow, ankle, hip, wrist, and shoulder joint. [0235] The autoimmune disease can be type I diabetes (T1D). The target site can comprise the pancreas. In some embodiments, the method treats or prevents at least one symptom of T1D, where the at least one symptom comprises thirst, frequent urination, weight loss, fatigue, blurred vision, or any combination thereof. In some embodiments, the method increases the level of C- peptide in the serum of the subject. In some embodiments, the C-peptide level in the serum of the subject is about 1.1 ng/mL to about 4.4 ng/mL following at least one application of thermal energy to the target site. In some embodiments, the method decreases the A1C percentage in the blood of the subject. In some embodiments, the A1C percentage in the blood of the subject is less than 6.5% following at least one application of thermal energy to the target site. In some embodiments, the method decreases fasting plasma glucose (FPG) level in the serum and/or plasma of the subject. In some embodiments, the FPG level in the serum and/or plasma of the subject is less than 125 mg/dL following at least one application of thermal energy to the target site. In some embodiments, the increase of C-peptide levels and the decrease of A1C percentage and/or FPG level is relative to: a subject that has not been administered the composition; prior to administering the composition to the subject; and/or prior to the applying thermal energy to the subject. -77-
ny-2833630
Atty Docket No.23887-2000140 [0236] The autoimmune disease can be lupus nephritis. The target site can comprise the kidney. In some embodiments, the method treats or prevents at least one symptom of lupus nephritis, where the at least one symptom comprises joint pain or swelling, muscle pain, fever, rash, or any combination thereof. In some embodiments, the method decreases the albumin-to-creatinine ratio (UACR) in the urine of the subject. In some embodiments, the UACR of the subject is less than 30 mg/g following at least one application of thermal energy to the target site. In some embodiments, the method increases glomerular filtration rate (GFR) in the blood of the subject. In some embodiments, the GFR in the blood of the subject is greater than 60 following at least one application of thermal energy to the target site. In some embodiments, the decrease of UACR and/or the increase of GFR is relative to: a subject that has not been administered the composition; prior to administering the composition to the subject; and/or prior to the applying thermal energy to the subject. [0237] The autoimmune disease can be inclusion body myositis (IBM). The target site can be skeletal muscle. In some embodiments, the method treats or prevents at least one symptom of IBM, where the at least one symptom comprises muscle weakness in a wrist, finger, front of the thigh, muscles that lift the front of the foot, or any combination thereof. EXEMPLARY EMBODIMENTS 1. A composition comprising: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product upon thermal stimulation. 2. A composition comprising: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter operably linked to a first polynucleotide comprising a transactivator gene, and a second promoter operably linked to a second polynucleotide comprising an immunostimulatory agent gene, wherein the inducible promoter is capable of inducing transcription of the transactivator gene to generate a transactivator transcript in the presence of thermal stimulation, -78-
ny-2833630
Atty Docket No.23887-2000140 wherein the transactivator transcript is capable of being translated to generate a transactivator; and wherein, in the presence of the transactivator and; optionally, a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. 3. A composition comprising: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising a first inducible promoter and a second promoter each operably linked to a first polynucleotide comprising an immunostimulatory agent gene and to a second polynucleotide comprising a transactivator gene, wherein the inducible promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript upon thermal stimulation, wherein, in the presence of the transactivator and a transactivator-binding compound, the second promoter is capable of inducing transcription of the immunostimulatory agent gene and the transactivator gene to generate a polycistronic transcript, and wherein the polycistronic transcript is capable of being translated to generate a transactivator and an immunostimulatory agent protein. 4. The composition of any one of embodiments 2-3, wherein the second promoter comprises one or more copies of a transactivator recognition sequence the transactivator is capable of binding to induce transcription, and wherein the transactivator is incapable of binding the transactivator recognition sequence in the absence of the transactivator-binding compound, optionally the one or more copies of a transactivator recognition sequence comprise one or more copies of a tet operator (TetO). 5. The composition of any one of embodiments 2-4, wherein the transactivator comprises reverse tetracycline-controlled transactivator (rtTA). 6. The composition of any one of embodiments 2-4, wherein the transactivator comprises tetracycline-controlled transactivator (tTA). 7. The composition of any one of embodiments 2-6, wherein the transactivator-binding compound comprises tetracycline, doxycycline or a derivative thereof. -79-
ny-2833630
Atty Docket No.23887-2000140 8. The composition of any one of embodiments 2-7, wherein the first polynucleotide and the second polynucleotide are operably linked to a tandem gene expression element, optionally the tandem gene expression element is an internal ribosomal entry site (IRES), foot-and-mouth disease virus 2A peptide (F2A), equine rhinitis A virus 2A peptide (E2A), porcine teschovirus 2A peptide (P2A) or Thosea asigna virus 2A peptide (T2A), or any combination thereof. 9. A composition comprising: a genetically modified T-regulatory (Treg) cell or a population thereof, wherein the genetically modified Treg cell comprises (i) a first nucleic acid comprising an inducible promoter operably linked to a first polynucleotide comprising a recombinase gene, wherein the first inducible promoter is capable of inducing transcription of the recombinase gene to generate a recombinase transcript upon thermal stimulation, and wherein the recombinase transcript is capable of being translated to generate a recombinase; a second promoter and a second polynucleotide comprising an immunostimulatory agent gene, wherein, in the absence of a recombination event, the second promoter and the second polynucleotide are not operably linked, wherein the recombinase is capable of catalyzing the recombination event, and wherein the second promoter and the second polynucleotide are operably linked after the recombination event such that the second promoter is capable of inducing transcription of the immunostimulatory agent gene to generate an immunostimulatory agent gene product. 10. The composition of embodiment 9, wherein the recombination event comprises removal of a sequence flanked by recombinase target sites or an inversion of a sequence flanked by recombinase target sites; optionally the second polynucleotide is flanked by recombinase target sites. 11. The composition of any one of embodiments 9-10, wherein, prior to the recombination event, the sequence of the immunostimulatory agent gene is inverted relative to the promoter. -80-
ny-2833630
Atty Docket No.23887-2000140 12. The composition of any one of embodiments 9-11, comprising at least one stop cassette situated between the second promoter and the immunostimulatory agent gene, wherein the stop cassette comprises one or more stop sequences, and wherein the one or more stop cassettes are flanked by recombinase target sites. 13. The composition of any one of embodiments 1-12, wherein the immunostimulatory agent gene product is an mRNA transcript and/or protein, wherein the immunostimulatory agent transcript is capable of being translated to generate the immunostimulatory agent protein. 14. The composition of embodiment 13, wherein the at least one stop cassette is configured to prevent transcription of the immunostimulatory agent gene and/or translation of the immunostimulatory agent transcript. 15. The composition of any one of embodiments 12-14, wherein the one or more stop sequences comprise a polyadenylation signal, a stop codon, a frame-shifting mutation, or any combination thereof. 16. The composition of any one of embodiments 2-15, wherein the second promoter comprises a promoter selected from the group consisting of a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus- forming virus (SFFV), a Moloney murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3- phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock protein 90 kDa beta, member 1 (HSP90B1), heat -
-KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1 (PGK) promoter, 3-phosphoglycerate kinase
promoter, a cytomegalovirus enhance - -actin (CBA) promoter, a CAG promoter, a CBH promoter, any variant thereof, or any combination thereof. 17. The composition of any one of embodiments 9-16, wherein the recombinase is Cre, Dre, , derivatives thereof, or any combination thereof; optionally the recombinase is a Flp recombinase and the recombinase target sites are FRT sites or the recombinase is a Cre recombinase and the recombinase target sites are loxP sites. -81-
ny-2833630
Atty Docket No.23887-2000140 18. The composition of any one of embodiments 1-17, wherein the immunostimulatory agent increases the persistence and/or activity of the genetically modified Treg cell or population thereof. 19. The composition of any one of embodiments 1-18, wherein the immunostimulatory agent comprises a cytokine. 20. The composition of embodiment 19, wherein the cytokine is a chemokine, an interferon, an interleukin (IL), or a tumor necrosis factor (TNF). 21. The composition of any one of embodiments 19-20, wherein the cytokine is selected from the group consisting of interleukin-1 (IL-1), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, IL-34, and IL-35. 22. The composition of any one of embodiments 19-20, wherein the cytokine is selected from the group consisting of CCL1, CCL2, CCL3, CCR4, CCL5, CCL7, CCL8/MCP-2, CCL11, CCL13/MCP-4, HCC- 1/CCL14, CTAC/CCL17, CCL19, CCL22, CCL23, CCL24, CCL26, CCL27, VEGF, PDGF, lymphotactin (XCL1), Eotaxin, FGF, EGF, IP- 10, TRAIL, GCP- 2/CXCL6, NAP- 2/CXCL7, CXCL8, CXCL10, ITAC/CXCL11, CXCL12, CXCL13, and CXCL15. 23. The composition of any one of embodiments 19-20, wherein the cytokine is selected from the group consisting of TNF-alpha, TNF-beta, TNF-gamma, CD252, CD154, CD178, CD70, CD153, or 4-1BBL. 24. The composition of any one of embodiments 19-20, wherein the cytokine is selected from the group consisting of interferon alpha, interferon beta, or interferon gamma. 25. The composition of any one of embodiments 1-18, wherein the immunostimulatory agent is selected from the group consisting of granulocyte macrophage colony stimulating factor (GM- CSF), M-CSF, SCF, TSLP, oncostatin M, leukemia-inhibitory factor (LIF), CNTF, Cardiotropin- 1, NNT-1/BSF-3, growth hormone, Prolactin, Erythropoietin, Thrombopoietin, Leptin, and G- CSF; or a receptor or ligand thereof. 26. The composition of any one of embodiments 1-18, wherein the immunostimulatory agent comprises an antibody or fragment thereof. -82-
ny-2833630
Atty Docket No.23887-2000140 27. The composition of embodiment 26, wherein the antibody or fragment thereof is a single- chain variable fragment (scFv), a single-domain antibody, a nanobody, an immunoglobulin molecule, a monoclonal antibody, a chimeric antibody, a CDR-grafted antibody, a humanized antibody, a Fab fragment, a Fab’ fragment, a F(ab’)2 fragment, an Fv fragment, a disulfide linked Fv, an scFv, a single domain antibody, a diabody, a multispecific antibody, a dual specific antibody, an anti-idiotypic antibody, a bispecific antibody, or a functionally active epitope-binding fragment thereof. 28. The composition of any one of embodiments 26-27, wherein the antibody or fragment thereof is capable of binding PD-1, PD-L1, IL-2, CD47, 4-1BB, OX40, CD40, IL-2, IL-6, IL-10, BTLA, CD3, and/or CD4. 29. The composition of any one of embodiments 1-18, wherein the immunostimulatory agent comprises a programmable DNA-binding protein. 30. The composition of embodiment 29, wherein the programmable DNA-binding protein comprises a programmable DNA-binding endonuclease selected from the group consisting of Casl, CaslB, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csnl and Csxl2), Casl00, Csyl, Csy2, Csy3, Csel, Cse2, Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmrl, Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, Csxl0, Csxl6, CsaX, Csx3, Csxl, Csxl5, Csfl, Csf2, Csf3, Csf4, or Cpfl endonuclease, or a homolog thereof. 31. The composition of embodiment 30, wherein the endonuclease comprises one or more mutations, wherein the one or more mutations converts the endonuclease to a nickase or renders the endonuclease catalytically dead. 32. The composition of embodiment 31, wherein the programmable DNA-binding protein comprises a fusion protein comprising the nickase or the catalytically dead endonuclease. 33. The composition of embodiment 32, wherein the fusion protein comprises the programmable DNA-binding protein fused to an enzyme selected from the group consisting of a deaminase, a polymerase, a histone deactylase (HDAC), a histone acetyl transferase (HAT), a recombinase, and an isomerase. 34. The composition of any one of embodiments 29-33, wherein the Treg further comprises a polynucleotide encoding a gRNA capable of targeting the programmable DNA-binding protein to a target sequence, wherein the polynucleotide encoding the gRNA and the polynucleotide -83-
ny-2833630
Atty Docket No.23887-2000140 encoding the programmable DNA-binding protein are within the same nucleic acid molecule or separate nucleic acid molecules. 35. The composition of embodiment 34, wherein the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25, CD122, IL- -1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor; optionally the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of FOXP3, CD25, CD122, IL- able DNA-binding protein and the gRNA are configured to cause a reduction-of-function or knockout mutation in the gene; further optionally, the gRNA comprises a spacer sequence capable of hybridizing to a sequence within or near a gene selected from the group consisting of Regnase-1, Roquin-1, IL-6, a TNF receptor, a TLR receptor, and IL-1 Receptor and the programmable DNA-binding protein and the gRNA are configured to cause a gain-of-function mutation in the gene. 36. The composition of any one of embodiments 1-35, wherein the genetically modified Treg cell comprises: (ii) a second nucleic acid comprising a constitutive promoter operably linked to a polynucleotide encoding a chimeric antigen receptor (CAR) or a T-cell receptor (TCR). 37. The composition of embodiment 25, wherein the constitutive promoter is a ubiquitous promoter or a tissue-specific promoter. 38. The composition of any one of embodiments 25-37, wherein the constitutive promoter is selected from the group comprising a cytomegalovirus (CMV) immediate early promoter, a CMV promoter, a viral simian virus 40 (SV40) (e.g., early or late), spleen focus-forming virus (SFFV), a Moloney murine leukemia virus (MoMLV) LTR promoter, a Rous sarcoma virus (RSV) LTR, an RSV promoter, a herpes simplex virus (HSV) (thymidine kinase) promoter, H5, P7.5, and P11 promoters from vaccinia virus, an elongation factor 1-alpha (EF1a) promoter, early growth response 1 (EGR1), ferritin H (FerH), ferritin L (FerL), Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), eukaryotic translation initiation factor 4A1 (EIF4A1), heat shock 70 kDa protein 5 (HSPA5), heat shock protein 90 kDa beta, member 1 (HSP90B1), heat shock protein -84-
ny-2833630
Atty Docket No.23887-2000140 -
-KIN), the human ROSA 26 locus, a Ubiquitin C promoter (UBC), a phosphoglycerate kinase-1 (PGK) promoter, 3-phosphoglycerate kinase promoter, a -
-actin (CBA) promoter, a CAG promoter, a CBH promoter, a variant thereof, or any combination thereof. 39. The composition of any one of embodiments 25-38, wherein the CAR comprises a binding domain, a hinge region, a transmembrane domain, and an intracellular signaling domain. 40. The composition of embodiment 39, wherein the binding domain is capable of binding a target selected from the group consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6-trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage-gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), a heat shock protein, and a protein of the human leukocyte antigen (HLA) system. 41. The composition of embodiment 40, wherein the HLA protein is selected from the group consisting of HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA- serotypes or variants thereof. 42. The composition of embodiment 41, wherein the HLA protein comprises the HLA-A2 serotype. 43. The composition of embodiment 40, wherein the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. 44. The composition of any one of embodiments 39-40, wherein the intracellular signaling domain comprises a primary signaling domain, a costimulatory domain, or both. 45. The composition of embodiment 44, wherein the primary signaling domain comprises a functional signaling domain of one or more proteins selected from the group consisting of CD3 zeta, CD3 gamma, CD3 delta, CD3 epsilon, common FcR gamma (FCER1G), FcR beta (Fc Epsilon R1b), CD79a, CD79b, Fcgamma RIIa, DAP10, and DAP12, or a functional variant thereof. 46. The composition of any one of embodiments 44-45, wherein the costimulatory domain comprises a functional domain of one or more proteins selected from the group consisting of CD27, CD28, 4-1BB (CD137), OX40, CD28-OX40, CD28-4-1BB, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand -85-
ny-2833630
Atty Docket No.23887-2000140 that specifically binds with CD83, CD5, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA--6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO- 3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, and NKG2D, or a functional variant thereof. 47. The composition of any one of embodiments 39-46, wherein the transmembrane domain comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA- 1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and NKG2C, or a functional variant thereof. 48. The composition of any one of embodiments 25-47, wherein the TCR further comprises a constant region and a variable region. 49. The composition of embodiment 48, wherein the TCR is capable of binding an antigen selected from the group consisting of natural killer group 2D (NKG2D), CD19, Insulin, 2,4,6- trinitrophenol, carcinoembryonic antigen (CEA), myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), chloride voltage-gated channel 1 (CLCN1), a nicotinic acetylcholine receptor (nAChR), a muscarinic acetylcholine receptor (mAChR), and a heat shock protein. 50. The composition of embodiment 49, wherein the heat shock protein is selected from the group consisting of Hsp60, Hsp90, Hsp40, and Hsp70. -86-
ny-2833630
Atty Docket No.23887-2000140 51. The composition of any one of embodiments 1-50, wherein the inducible promoter comprises a core promoter and at least one heat shock element (HSE). 52. The composition of embodiment 51, wherein the core promoter comprises the heat shock protein transcription start site of a gene selected from the group consisting of HSPA1A, HSPH1, HSPB1, HSPA6, and YB. 53. The composition of any one of embodiments 51-52, wherein the core promoter comprises the sequence of any one of SEQ ID NOs: 9-12 and 35-46 or a sequence having at least 85% identity to any one of SEQ ID NOs: 9-12 and 35-46. 54. The composition of any one of embodiments 51-52, wherein the core promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 9-12 and 35-46. 55. The composition of any one of embodiments 51-54, wherein the inducible promoter comprises seven HSEs. 56. The composition of any one of embodiments 51-55, wherein each of the at least one HSE comprises the sequence of 5’- nGAAnnTTCnnGAAn-3’. 57. The composition of any one of embodiments 51-56, wherein the at least one HSE comprises the sequence of any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence having at least 85% identity to any one of SEQ ID NOs: 1-8, 53, and 112-116 or a sequence differing by one or two nucleotide mismatches relative to any one of SEQ ID NOs: 1-8, 53, and 112-116. 58. The composition of any one of embodiments 51-56, wherein the at least one HSE comprises a sequence selected from the group consisting of SEQ ID NOs: 1-8, 53, and 112-116. 59. The composition of any one of embodiments 1-50, wherein the inducible promoter comprises the sequence of any one of SEQ ID NOs: 13-20, 58-80, and 88-94 or a sequence having at least 85% identity to any one of SEQ ID NOs: 13-20, 58-80, and 88-94. 60. The composition of any one of embodiments 1-50, wherein the inducible promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 13-20, 58-80, and 88- 94. 61. The composition of any one of embodiments 1-50, wherein the inducible promoter comprises the sequence of any one of SEQ ID NOs: 21-34 or a sequence having at least 85% identity to any one of SEQ ID NOs: 21-34. -87-
ny-2833630
Atty Docket No.23887-2000140 62. The composition of any one of embodiments 1-50, wherein the inducible promoter comprises a sequence selected from the group consisting of SEQ ID NOs: 21-34. 63. The composition of any one of embodiments 1-62, wherein the thermal stimulation comprises heating to an activating temperature; wherein the activating temperature is greater than 37°C. 64. The composition of embodiment 63, wherein the activating temperature comprises a temperature of at least 37°C to at most 70°C. 65. The composition of embodiment 64, wherein the activating temperature is about 37.0ºC, 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC. 66. The composition of any one of embodiments 63-65, wherein the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC. 67. The composition of any one of embodiments 63-66, wherein the activating temperature comprises a temperature of about 42°C to about 43°C. 68. The composition of embodiment 67, wherein the activating temperature is about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. 69. The composition of any one of embodiments 1-68, wherein the genetically modified Treg cell expresses one or more of the markers selected from the group consisting of CD4, CD25, and forkhead box P3 (FOXP3). -88-
ny-2833630
Atty Docket No.23887-2000140 70. The composition of any one of embodiments 1-69, wherein the genetic modification comprises integration of the first nucleic acid into the genome of the Treg cell. 71. The composition of embodiment 70, wherein the first nucleic acid is integrated within a safe harbor locus; optionally, the safe harbor locus comprises AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. 72. The composition of any one of embodiments 25-71, wherein the genetic modification comprises integration of the second nucleic acid into the genome of the Treg cell. 73. The composition of embodiment 72, wherein the second nucleic acid is integrated within a safe harbor locus; optionally, the safe harbor locus comprises AAVS1 (PPP1 R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), G6PC, Gys2, HGD, Lpa, Pcsk9, Serpina1, TF, or TTR. 74. The composition of any one of embodiments 1-73, wherein the genetically modified Treg cell is allogenic or autologous to the subject. 75. The composition of any one of embodiments 1-74, wherein the genetically modified Treg cell was differentiated from a genetically modified stem cell; optionally, the genetically modified stem cell comprises an embryonic stem cell or an induced pluripotent stem cell; further optionally, the genetically modified stem cell comprises a hematopoietic stem cell. 76. The composition of any one of embodiments 1-75, further comprising one or more pharmaceutically acceptable excipients. 77. The composition of any one of embodiments 1-76, for use in treating an autoimmune disease in a subject. 78. A method of treating an autoimmune disease in a subject, the method comprising: administering the composition of any one of embodiments 1-77 to the subject; and applying thermal energy to a target site of the subject sufficient to increase local temperature of the target site to an activating temperature, thereby inducing the expression of the immunostimulatory agent gene product. 79. The method of embodiment 78, wherein the method comprises administering the composition at a dose of about 1 × 10
5 to about 1 × 10
15 Treg cells. -89-
ny-2833630
Atty Docket No.23887-2000140 80. The method of embodiment 78, wherein the method comprises administering the composition at a dose of at least about 1 × 10
5, 1 × 10
6, 1 × 10
7, 1 × 10
8, 1 × 10
9, 1 × 10
10, 1 × 10
11, 1 × 10
12, 1 × 10
13, 1 × 10
14, or 1 × 10
15 Treg cells. 81. The method of any one of embodiments 78-80, wherein the method comprises a single administration of the composition to the subject. 82. The method of any one of embodiments 78-80, wherein the method comprises administering the composition to the subject two or more times; optionally each of the two or more administrations are one month, two months, 6 months, 1 year, or more, apart. 83. The method of any one of embodiments 78-82, wherein the period of time between the administering the composition and applying thermal energy is about 1 week, about 6 days, about 5 days, about 4 days, about 3 days, about 48 hours, about 44 hours, about 40 hours, about 35 hours, about 30 hours, about 25 hours, 20 hours, 15 hours, 10 hours, about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 30 minutes, about 15 minutes, about 10 minutes, or about 5 minutes. 84. The method of any one of embodiments 78-83, wherein the activating temperature is greater than 37°C. 85. The method of embodiment 84, wherein the activating temperature comprises a temperature of at least 37.5°C to at most 70.0°C. 86. The method of embodiment 85, wherein the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, about 46.0ºC, about 46.5ºC, about 47.0ºC, about 47.5ºC, about 48.0ºC, about 48.5 ºC, about 49.0ºC, about 49.5ºC, about 50.0ºC, about 50.5ºC, about 51.0ºC, about 51.5ºC, about 52.0ºC, about 52.5ºC, about 53.0ºC, about 53.5ºC, about 54.0ºC, about 54.5ºC, about 55.0ºC, about 55.5ºC, about 56.0ºC, about 56.5ºC, about 57.0ºC, about 57.5ºC, about 58.0ºC, about 58.5ºC, about 59.0ºC, about 59.5ºC, about 60.0ºC, about 60.5ºC, about 61.0ºC, about 61.5ºC, about 62.0ºC, about 62.5ºC, about 63.0ºC, about 63.5ºC, about 64.0ºC, about 64.5ºC, about 65.0ºC, about 65.5ºC, about 66.0ºC, about 66.5ºC, about 67.0ºC, about 67.3ºC, about 67.5ºC, about 68.0ºC, about 68.5ºC, about 69.0ºC, about 69.5ºC, or about 70.0ºC; optionally, wherein the thermal energy is applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, -90-
ny-2833630
Atty Docket No.23887-2000140 about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, or about 1 minute. 87. The method of any one of embodiments 84-86, wherein the activating temperature is about 37.5ºC, about 38.0ºC, about 38.5ºC, about 39.0ºC, about 39.5ºC, about 40.0ºC, about 40.5ºC, about 41.0ºC, about 41.5ºC, about 42.0ºC, about 42.5ºC, about 43.0ºC, about 43.5ºC, about 44.0ºC, about 44.5ºC, about 45.0ºC, about 45.5ºC, or about 46.0ºC; optionally, wherein the thermal energy is applied to the target site for a duration of time comprising about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. 88. The method of any one of embodiments 84-87, wherein the activating temperature comprises a temperature of about 42°C to about 43°C. 89. The method of embodiment 88, wherein the activating temperature is about 42.0ºC, about 42.1ºC, about 42.2ºC, about 42.3ºC, about 42.4ºC, about 42.5ºC, about 42.6ºC, about 42.7ºC, about 42.8ºC, about 42.9ºC, or about 43.0ºC. 90. The method of any one of embodiments 78-89, wherein the thermal energy is applied to the target site for a duration of time comprising about 1 second to about 20 minutes. 91. The method of embodiment 90, wherein the thermal energy is applied to the target site for a duration of time comprising about 1 second, about 5 seconds, about 10 seconds, about 15 seconds, about 20 seconds, about 25 seconds, about 30 seconds, about 35 seconds, about 40 seconds, about 45 seconds, about 50 seconds, about 55 seconds, about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. 92. The method of any one of embodiments 78-91, wherein the temperature at the target site is elevated to about 42°C to about 43°C after the applying; optionally, within at most 30 minutes from the applying. -91-
ny-2833630
Atty Docket No.23887-2000140 93. The method of embodiment 92, wherein the temperature at the target site is elevated to about 42°C to about 43°C within 10 minutes or less after the applying. 94. The method of any one of embodiments 92-93, wherein the temperature at the target site is elevated to about 42°C to about 43°C within about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, or about 10 minutes after the applying. 95. The method of any one of embodiments 92-94, wherein the temperature at the target site is maintained at about 42°C to about 43°C for at least about 15 minutes to at least about 120 minutes. 96. The method of any one of embodiments 92-95, wherein the temperature at the target site is maintained at 42°C to 43°C for at least 15 minutes, at least 20 minutes, at least 25 minutes, at least 30 minutes, at least 35 minutes, at least 40 minutes, at least 45 minutes, at least 50 minutes, at least 55 minutes, at least 60 minutes, at least 90 minutes, or at least 120 minutes. 97. The method of embodiment 96, wherein the temperature at the target site is maintained at 42°C to 43°C for at least 20 minutes. 98. The method of embodiment 96, wherein the temperature at the target site is maintained at about 42°C to about 43°C for about 1 minute, about 2 minutes, about 3 minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. 99. The method of any one of embodiments 78-98, wherein the method comprises applying thermal energy to the subject two or more times. 100. The method of embodiment 99, wherein each of the two or more applications of thermal energy are 1 week apart, 2 weeks apart, 3 weeks apart, 4 weeks apart, 5 weeks apart, or more. 101. The method of any one of embodiments 78-100, wherein the target site comprises one or more of muscle, skin, joints, the rectum, and the colon; optionally, the target site is an internal organ. 102. The method of embodiment 101, wherein the muscle is skeletal, cardiac, or smooth muscle. -92-
ny-2833630
Atty Docket No.23887-2000140 103. The method of embodiment 101, wherein the internal organ is liver, kidney, lung, pancreas, or heart. 104. The method of any one of embodiments 78-103, wherein applying thermal energy to a target site of the subject comprises the application of one or more of magnetic hyperthermia, microwave hyperthermia, inductive diathermy, contact hyperthermia, ultrasonic diathermy, deep hyperthermia ingestible hyperthermia, and rectal hyperthermia. 105. The method of any one of embodiments 78-104, wherein applying thermal energy to a target site of the subject comprises use of a device selected from the group consisting of: BSD- 500 Superficial Hyperthermia, BSD-2000 Deep Regional Hyperthermia, TheraTouch® DX2 Shortwave Diathermy, Dynatronics Solaris Plus, and Hill Laboratories HF54+. 106. The method of any one of embodiments 78-105, comprising monitoring the temperature of the target region, optionally the monitoring is performed by magnetic resonance imaging (MRI). 107. The method of any one of embodiments 78-106, wherein the application of thermal energy to a target site of the subject is guided spatially by magnetic resonance imaging (MRI). 108. The method of any one of embodiments 78-107, wherein at least about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% of the population of thermally actuated genetically modified Treg cells at the target site express the immunostimulatory agent gene product after applying thermal energy to the target site. 109. The method of any one of embodiments 78-108, wherein less than about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%, of the population of thermally actuated genetically modified Treg cells at a site other than the target site express the immunostimulatory agent gene product. 110. The method of any one of embodiments 78-109, wherein upon applying thermal energy to the target site, the transcription of the immunostimulatory gene, the transactivator gene, the recombinase gene, or any combination thereof, increases by at least 1.1-fold. 111. The method of any one of embodiments 78-110, wherein upon applying thermal energy to the target site, the levels of the of the immunostimulatory gene product, the transactivator gene -93-
ny-2833630
Atty Docket No.23887-2000140 product, the recombinase gene product, or any combination thereof, increases by at least 1.1-fold, wherein the gene product comprises RNA transcribed from the gene. 112. The method of embodiment 111, wherein the gene product comprises an mRNA, a protein translated from the mRNA, or both. 113. The method of any one of embodiments 78-112, wherein the autoimmune disease comprises type I diabetes, psoriasis, Lupus nephritis, multiple sclerosis, neuromyelitis optica, chronic inflammatory demyelinating polyneuropathy, polymyositis, dermatomyositis, inclusion body myositis, Crohn’s Disease, ulcerative colitis, primary sclerosing cholangitis, ankylosing spondylitis, rheumatic arthritis, spondyloarthritis, or juvenile idiopathic arthritis. 114. The method of any one of embodiments 78-113, wherein the method treats or prevents at least one symptom of the autoimmune disease. 115. The method of embodiment 114, wherein the autoimmune disease is ulcerative colitis and the target site comprises the rectum and/or the colon, and wherein (a) the method treats or prevents at least one symptom of the ulcerative colitis, wherein the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof; (b) the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more symptoms of (a); and/or (c) the method reduces the levels of anti-saccharomyces cerevisiae antibody (ASCA) in the subject, wherein the reduction of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. 116. The method of embodiment 114, wherein the autoimmune disease is Crohn’s Disease, wherein the target site comprises the rectum, colon, small intestine, or a combination thereof; and wherein (a) the method treats or prevents at least one symptom of the Crohn’s disease, wherein the at least one symptom comprises diarrhea or constipation, abdominal pain, rectal bleeding, fever, reduced appetite, fatigue, weight loss, night sweats, or any combination thereof; (b) the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more symptoms of (a); and/or -94-
ny-2833630
Atty Docket No.23887-2000140 (c) the method reduces the levels of perinuclear anti-neutrophil antibodies (pANCA) in the subject, wherein the reduction of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. 117. The method of embodiment 114, wherein the autoimmune disease is psoriasis, wherein the target site comprises the skin, and wherein (a) the method treats or prevents at least one symptom of psoriasis, wherein the at least one symptom comprises skin rash, skin bleeding, skin itch, skin burning, skin soreness, or any combination thereof, and/or (b) the method reduces the frequency, duration, and/or intensity of flares in the subject, wherein a flare is characterized by one or more of the symptoms of (a); wherein the reduction of (b) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. 118. The method of embodiment 114, wherein the autoimmune disease is rheumatoid arthritis (RA), wherein the target site comprises one or more joints, and wherein the method treats or prevents at least one symptom of RA, where the at least one symptom comprises swelling and/or stiffness in one or more joints; optionally in one or more of a knee, finger, elbow, ankle, hip, wrist, and shoulder joint. 119. The method of embodiment 114, wherein the autoimmune disease is type I diabetes (T1D), wherein the target site comprises the pancreas, and wherein the method treats or prevents at least one symptom of T1D, where the at least one symptom comprises thirst, frequent urination, weight loss, fatigue, blurred vision, or any combination thereof; optionally (a) the method increases the level of C-peptide in the serum of the subject; optionally, wherein the C-peptide level in the serum of the subject is about 1.1 ng/mL to about 4.4 ng/mL following at least one application of thermal energy to the target site, (b) the method decreases the A1C percentage in the blood of the subject, optionally, the A1C percentage in the blood of the subject is less than 6.5% following at least one application of thermal energy to the target site, -95-
ny-2833630
Atty Docket No.23887-2000140 (c) the method decreases fasting plasma glucose (FPG) level in the serum and/or plasma of the subject, optionally, the FPG level in the serum and/or plasma of the subject is less than 125 mg/dL following at least one application of thermal energy to the target site, wherein the increase of (a) and the decrease of (b)-(c) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. 120. The method of embodiment 114, wherein the autoimmune disease is lupus nephritis, wherein the target site comprises the kidney, and wherein, the method treats or prevents at least one symptom of lupus nephritis, where the at least one symptom comprises joint pain or swelling, muscle pain, fever, rash, or any combination thereof; optionally (a) the method decreases the albumin-to-creatinine ratio (UACR) in the urine of the subject; optionally, wherein the UACR of the subject is less than 30 mg/g following at least one application of thermal energy to the target site, and/or (b) the method increases glomerular filtration rate (GFR) in the blood of the subject, optionally, the GFR in the blood of the subject is greater than 60 following at least one application of thermal energy to the target site, wherein the decrease of (a) and the increase of (b) is relative to (i) a subject that has not been administered the composition; (ii) prior to administering the composition to the subject; and/or (iii) prior to the applying thermal energy to the subject. 121. The method of embodiment 114, wherein the autoimmune disease is inclusion body myositis (IBM), wherein the target site is skeletal muscle, and wherein the method treats or prevents at least one symptom of IBM, where the at least one symptom comprises muscle weakness in a wrist, finger, front of the thigh, muscles that lift the front of the foot, or any combination thereof. Embodiment 1A. An expression system comprising: a first polynucleotide comprising: (a) a thermally-inducible promoter, and (b) a first nucleic acid encoding a payload; wherein the thermally-inducible promoter comprises: (i) at least 6 heat shock elements (HSE), and -96-
ny-2833630
Atty Docket No.23887-2000140 (ii) a core promoter comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:9-12 and 35-46. Embodiment 2A. The expression system of embodiment 1A, wherein at least one of the HSEs is inverted relative to the core promoter. Embodiment 3A. An expression system comprising: a first polynucleotide comprising: (a) a thermally-inducible promoter, and (b) a first nucleic acid encoding a payload; wherein the thermally-inducible promoter comprises: (i) a core promoter, and (ii) at least 6 heat shock elements (HSE), wherein at least 1 HSE is inverted relative to the core promoter. Embodiment 4A. The expression system of any one of embodiments 1A-3A, wherein the thermally-inducible promoter comprises about 6 HSEs to about 15 HSEs. Embodiment 5A. The expression system of any one of embodiments 1A-4A, wherein the thermally-inducible promoter comprises 7 HSEs or 8 HSEs. Embodiment 6A. The expression system of any one of embodiments 1A-5A, wherein each heat shock element comprises a sequence set forth in nGAAnnTTCnnGAAn, wherein “n” is a nucleotide selected from the group consisting of guanine (“G”), cytosine (“C”), adenosine (“A”), and thymine (“T”). Embodiment 7A. The expression system of any one of embodiments 2A-6A, wherein the at least one inverted heat shock element comprises a sequence set forth in nTTCnnGAAnnTTCn, wherein “n” is a nucleotide selected from the group consisting of guanine (“G”), cytosine (“C”), adenosine (“A”), and thymine (“T”). Embodiment 8A. The expression system of any one of embodiments 1A-7A, wherein the number of nucleotides located between each HSE is about 1 to about 50 nucleotides. Embodiment 9A. The expression system of any one of embodiments 1A-8A, wherein each HSE comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:47- 52, 85-87, and 95-102. -97-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 10A. The expression system of any one of embodiments 2A-9A, wherein alternating HSEs are inverted. Embodiment 11A. The expression system of any one of embodiments 2A-10A, wherein the core promoter is selected from the group consisting of a YB core promoter, a CMVa core
promoter, a CMVb core promoter, a CMVd core promoter, an SV40 core promoter, an EF1 core promoter, a CAG core promoter, a PGK1 core promoter, an SFFV core promoter, and an hUBC core promoter. Embodiment 12A. The expression system of any one of embodiments 3A-11A, wherein the core promoter is a YB core promoter, a CMVa core promoter, a CMVb core promoter, a CMVd
core promoter, or an EF1 core promoter. Embodiment 13A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 7 HSEs; and (ii) a YB core promoter. Embodiment 14A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 7 HSEs; and (ii) any one of CMVa core promoter, CMVb core promoter, or CMVd core promoter. Embodiment 15A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 7 HSEs; and (
ii) an EF1 core promoter. Embodiment 16A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 8 HSEs; and (ii) a YB core promoter. Embodiment 17A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 8 HSEs; and (ii) any one of CMVa core promoter, CMVb core promoter, or CMVd core promoter. -98-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 18A. The expression system of any one of embodiments 1A-12A, wherein the thermally-inducible promoter comprises: (i) 8 HSEs; and (
ii) an EF1 core promoter. Embodiment 19A. The expression system of any one of embodiments 1A-18A, wherein the
core promoter comprises a 5 untranslated region (5 UTR), wherein the 5 UTR is located at the the core promoter.
Embodiment 20A. The expression system of embodiment 19A, wherein the 5 UTR and a TATA box of the core promoter are derived from the same promoter sequence.
Embodiment 21A. The expression system of embodiment 19A, wherein the 5 UTR and a TATA box of the core promoter are derived from different promoters. Embodiment 22A. The expression system of any one of embodiments 19A-21A, wherein the selected from the group consisting of any one of SEQ ID NOs:54-57. Embodiment 23A. The expression system of any one of embodiments 19A-21A, wherein the ID NOs:54 and 108-111. Embodiment 24A. The expression system of any one of embodiments 19A-23A, wherein the thermally-inducible promoter further comprises a Kozak sequence set forth in any one of SEQ
ID NOs:81- Embodiment 25A. The expression system of any one of embodiments 1A-24A, wherein the thermally-inducible promoter comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:13-20, 58-80, and 88-94. Embodiment 26A. The expression system of any one of embodiments 1A-25A, further comprising a second polynucleotide comprising a second nucleic acid encoding one or more additional payloads. Embodiment 27A. The expression system of any one of embodiments 1A-26A, wherein the first payload and/or the one or more additional payloads is selected from the group consisting of -99-
ny-2833630
Atty Docket No.23887-2000140 a cytokine, a T-cell engager molecule, a chimeric receptor, a gene editing system, a fluorescent protein, a detection tag, and any combination thereof. Embodiment 28A. A vector comprising the first polynucleotide and/or the second polynucleotide of the expression system of any one of embodiments 1A-27A. Embodiment 29A. A cell comprising the expression system of any one of embodiments 1A- 27A or the vector of embodiment 28A. Embodiment 30A. A cell comprising a first polynucleotide comprising: (a) a thermally-inducible promoter, and (b) a nucleic acid encoding a payload; wherein the thermally-inducible promoter comprises: (i) at least 6 heat shock elements (HSE), and (ii) a core promoter comprising a nucleic acid sequence selected from the group consisting of SEQ ID NOs:9-12 and 35-46. Embodiment 31A. The cell of embodiment 30A, wherein at least one of the HSEs is inverted relative to the core promoter. Embodiment 32A. A cell comprising a first polynucleotide comprising: (a) a thermally-inducible promoter, and (b) a nucleic acid encoding a payload; wherein the thermally-inducible promoter comprises: (i) a core promoter, and (ii) at least 6 heat shock elements (HSE), wherein at least one HSE is inverted relative to the core promoter. Embodiment 33A. The cell of any one of embodiments 30A-32A, wherein the thermally- inducible promoter comprises about 6 HSEs to about 15 HSEs. Embodiment 34A. The cell of any one of embodiments 30A-33A, wherein the thermally- inducible promoter comprises 7 HSEs or 8 HSEs. Embodiment 35A. The cell of any one of embodiments 30A-34A, wherein each heat shock element comprises a sequence set forth in nGAAnnTTCnnGAAn, wherein “n” is a nucleotide -100-
ny-2833630
Atty Docket No.23887-2000140 selected from the group consisting of guanine (“G”), cytosine (“C”), adenosine (“A”), and thymine (“T”). Embodiment 36A. The cell of any one of embodiments 31A-35A, wherein the at least one inverted heat shock element comprises a sequence set forth in nTTCnnGAAnnTTCn, wherein “n” is a nucleotide selected from the group consisting of guanine (“G”), cytosine (“C”), adenosine (“A”), and thymine (“T”). Embodiment 37A. The cell of any one of embodiments 30A-36A, wherein the number of nucleotides located between each HSE is about 1 to about 50 nucleotides. Embodiment 38A. The cell tem of any one of embodiments 30A-37A, wherein each HSE comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:47-52, 85- 87, and 95-102. Embodiment 39A. The cell of any one of embodiments 31A-38A, wherein alternating HSEs are inverted. Embodiment 40A. The cell of any one of embodiments 31A-39A, wherein the core promoter is selected from the group consisting of a YB core promoter, a CMVa core promoter, a CMVb
core promoter, a CMVd core promoter, an SV40 core promoter, an EF1 core promoter, a CAG core promoter, a PGK1 core promoter, an SFFV core promoter, and an hUBC core promoter. Embodiment 41A. The cell of any one of embodiments 32A-40A, wherein the core promoter is a YB core promoter, a CMVa core promoter, a CMVb core promoter, a CMVd core promoter,
or an EF1 core promoter. Embodiment 42A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 7 HSEs; and (ii) a YB core promoter. Embodiment 43A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 7 HSEs; and (ii) any one of CMVa core promoter, CMVb core promoter, or CMVd core promoter. -101-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 44A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 7 HSEs; and (
ii) an EF1 core promoter. Embodiment 45A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 8 HSEs; and (ii) a YB core promoter. Embodiment 46A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 8 HSEs; and (ii) any one of CMVa core promoter, CMVb core promoter, or CMVd core promoter. Embodiment 47A. The cell of any one of embodiments 30A-41A, wherein the thermally- inducible promoter comprises: (i) 8 HSEs; and (
ii) an EF1 core promoter. Embodiment 48A. The cell of any one of embodiments 30A-47A, wherein the core promoter
comprises a 5 untranslated region (5 UTR), wherein the 5 UTR is located the core promoter.
Embodiment 49A. The cell of embodiment 48A, wherein the 5 UTR and a TATA box of the core promoter are derived from the same promoter sequence.
Embodiment 50A. The cell of embodiment 48A, wherein the 5 UTR and a TATA box of the core promoter are derived from different promoters. Embodiment 51A. The cell of any one of embodiments 48A- from the group consisting of any one of SEQ ID NOs:54-57. Embodiment 52A. The cell of any one of embodiments 48A- comprises a nucleic acid sequence selected from the group consisting of any one of SEQ ID NOs:54 and 108-111. -102-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 53A. The cell of any one of embodiments 48A-52A, wherein the thermally- inducible promoter further comprises a Kozak sequence set forth in any one of SEQ ID NOs:81- Embodiment 54A. The cell of any one of embodiments 30A-53A, wherein the thermally- inducible promoter comprises a nucleic acid sequence selected from the group consisting of SEQ ID NOs:13-20, 58-80, and 88-94. Embodiment 55A. The cell of any one of embodiments 30A-54A, further comprising a second polynucleotide comprising a second nucleic acid encoding one or more additional payloads. Embodiment 56A. The cell of any one of embodiments 30A-55A, wherein the first payload and/or the one or more additional payloads is selected from the group consisting of a cytokine, a T-cell engager molecule, a chimeric receptor, a gene editing system, a fluorescent protein, a detection tag, and any combination thereof. Embodiment 57A. The cell of any one of embodiments 29A-56A, wherein the cell is a primary cell or a cell line. Embodiment 58A. The cell of embodiment 57A, wherein the primary cell is selected from the group consisting of a T cell, a B cell, an NK cell, a macrophage, a monocyte, a dendritic cell, a granulocyte, an innate lymphoid cell, a hematopoietic progenitor cell, a myeloid progenitor cell, a lymphoid progenitor cell, and a hematopoietic stem cell. Embodiment 59A. The cell of embodiment 58A, wherein the T cell is selected from the group consisting of an effector T cell, a memory T cell, a regulatory T (“Treg”) cell, a naïve T cell, a tissue-resident T cell, a tumor-infiltrating T cell, and an immature T cell. Embodiment 60A. The cell of embodiment Embodiment 61A. A method of tunable thermal activation of a thermally-inducible promoter, comprising applying thermal energy to the cell of any one of embodiments 29A-60A, or to an individual comprising the cell of any one of embodiments 29A-60A, wherein the thermal energy is applied over a gradient of temperatures for at least about 1 second (1s) before reaching a maximum temperature. -103-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 62A. The method of embodiment 61A, wherein the thermal stimulation is applied over a gradient of temperatures for about 10 minutes to about 120 minutes before reaching a maximum temperature; optionally wherein the maximum temperature is at least Embodiment 63A. The method of embodiment 62A, wherein the maximum temperature is from about Embodiment 64A. The method of any one of embodiments 61A-63A, wherein applying thermal energy over a gradient of temperatures for a period of time of at least about 10 minutes to at least about 120 minutes before reaching a maximum temperature of about 42°C to about 43°C induces a lower level of payload gene expression by the thermally-inducible promoter compared to applying thermal energy over a gradient of temperatures for a period of time of about 1s to about 10 minutes before reaching a maximum temperature of about Embodiment 65A. A method of transiently activating a thermally-inducible promoter by thermal induction, comprising applying thermal energy to the cell of any one of embodiments 29A-60A, or to an individual comprising the cell of any one of embodiments 29A-60A, wherein the thermal energy is applied at least two times, wherein thermal induction is reversed after about 6 hours to about 24 hours following the applying thermal energy. Embodiment 66A. The method of embodiment 65A, wherein each time of applying the thermal energy is separated by a rest period of at least about 24 hours. Embodiment 67A. The method of any one of embodiments 61A-66A, wherein the thermal energy is applied for at least about 5 minutes. Embodiment 68A. The method of any one of embodiments 61A-67A, wherein the thermal energy is applied for about 5 minutes to about 60 minutes. Embodiment 69A. The method of any one of embodiments 65A-68A, wherein the maximum temperature of the thermal stimulation is Embodiment 70A. The method of any one of embodiments 63A-69A, wherein the maximum
temperature of the thermal stimulation is about 42 4 -104-
ny-2833630
Atty Docket No.23887-2000140 Embodiment 71A. The method of any one of embodiments 61A-70A, wherein the thermal activation of a thermally-inducible promoter induces expression of a payload gene. Embodiment 72A. The method of embodiments 71A, wherein the payload is not expressed or is expressed at an undetectable level in the absence of applying thermal energy. Embodiment 73A. The method of any one of embodiments 61A-70A, wherein the thermal energy is applied by radiofrequency, laser, ultrasound, heating pad, heating gel, or any combination thereof. Embodiment 74A. The method of any one of embodiments 65A-73A, wherein the expression level of the payload peaks by at least about 6 to about 12 hours. Embodiment 75A. The method of any one of embodiments 61A-74A, wherein the thermal energy is applied at a thermal dose of about 382 at 50% T cell viability 24 hrs post-heating as calculated by cumulative equivalent minutes at 43ºC (CEM43ºC). Embodiment 76A. The method of any one of embodiments 61A-74A, wherein the thermal energy is applied at a thermal dose of about 20 at 80% T cell viability 24 hrs post-heating as calculated by cumulative equivalent minutes at 43ºC (CEM43ºC). SEQUENCE LISTING

-105-
ny-2833630
Atty Docket No.23887-2000140
-106-
ny-2833630
Atty Docket No.23887-2000140
-107-
ny-2833630
Atty Docket No.23887-2000140
-108-
ny-2833630
Atty Docket No.23887-2000140
-109-
ny-2833630
Atty Docket No.23887-2000140
-110-
ny-2833630
Atty Docket No.23887-2000140
-111-
ny-2833630
Atty Docket No.23887-2000140
-112-
ny-2833630
Atty Docket No.23887-2000140
-113-
ny-2833630
Atty Docket No.23887-2000140
-114-
ny-2833630
Atty Docket No.23887-2000140

EXAMPLES [0238] Some aspects of the embodiments discussed above are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the present disclosure. Example 1 Expanded characterization of thermal switch activation [0239] This Example provides data related to parameters for timing and temperatures for activating the thermal switches described herein. [0240] Background: To evaluate the potential for inadvertent thermal switch activation by fever, the thermal switch activity was evaluated when exposed to temperatures higher than 41.5ºC (106.7 ºF), a level of fever known as hyperpyrexia. Clinically, the body takes several hours to reach peak fever temperatures. Additionally, ramp up time to achieve target temperatures also vary depending on the source of energy or hyperthermia device used. Laser and ultrasound can generally reach target temperatures within seconds, whereas radiofrequency devices can take up to 10–15 minutes to reach temperatures around, e.g., 42ºC, especially when heating larger deep -115-
ny-2833630
Atty Docket No.23887-2000140 structures in abdomen and pelvis. To mimic possible clinical scenarios, how ramp time to peak temperature affects thermal switch activity was evaluated. [0241] Results: When primary human T cells transduced with thermally controlled Gaussia Luciferase (hereafter referred to as “TS.Gluc”) and a constitutive HER2 CAR were heated to 42ºC for 20 minutes, the thermal switch activates expression of the luminescent reporter maximally following a short temperature ramp time of 2 seconds, as measured by a Gaussia Luciferase Glow Assay Kit (Thermo Fisher Scientific 16161). Gluc reporter activity is reduced by ~50% for every ~12.8 minutes that the ramp time is extended (see FIGS.2C–2D). When ramp time was extended to 3 hours, <3% reporter activity was detected (see FIG.2C). [0242] To quantify thermal switch activity at temperatures ranging from 37–67.3ºC and durations ranging from 1s–20 min, primary human T cells transduced with TS.Gluc were heated instantly (<10 seconds) to target temperature at indicated conditions in a thermal cycler. Using a Gaussia Luciferase Glow Assay Kit (Thermo Fisher Scientific 16161), at a temperature range of approximately 41–45ºC, the luminescent reporter activity was detected maximally at heating durations of 1.6 min and above, while temperatures 45º–67.3ºC only required seconds for thermal switch activation (see FIGS.3A-3B). As shown in, e.g., FIGS.3A-3B, primary human T cells were transduced with a genetic circuit containing the S7YB thermal switch (SEQ ID NO: 19) driving the expression of a Gaussia luciferase reporter. Cells were heated at temperatures ranging from 37–67.3ºC and heating durations from 1s – 20 min. Expression of the Gluc (glucose) reporter was measured by luminescence 24 hrs post-heating and is displayed in the 3D and 2D plots as shown in FIGS.3A-3B. [0243] These data demonstrate that the thermal switch can be activated when heated at mild temperatures (~40–45ºC) for durations in the order of minutes and at high temperatures (~45– 70ºC) for short durations in the order of seconds. [0244] These results demonstrate that the S7YB TS will not activate transgene expression if exposed to fever, even with high temperatures in the hyperpyrexia range. These results also provide insight into how temperature ramp time affects thermal switch activity and provide a control parameter to alter the output of the thermal switch. [0245] The cumulative equivalent minutes at 43ºC (CEM43ºC) model translates all different temperature-time heating parameters into a single number representing a thermal dose. To determine the effect of thermal dose on viability of primary human T cells, all heating parameters from FIGS.3A-3B were converted to CEM43ºC thermal doses. The CEM43ºC dose across all conditions that led to 50% T cell viability 24 hrs post-heating was 382, while the CEM43ºC dose that led to >80% viability was 20 (FIG.4A). When broken up by the -116-
ny-2833630
Atty Docket No.23887-2000140 temperature delivered (FIG.4B), the LD50 of the thermal dose shifts to higher CEM43ºC values. These data show that higher thermal doses can be well tolerated in primary human T cells. Example 2 New Thermal Bioswitch Designs [0246] Background: Heat shock elements (HSEs) are key drivers of the activity of synthetic thermal bioswitches because they function as binding sites for the heat shock transcription factor HSF1. Upon exposure to heat, HSF1 becomes activated, trimerizes, and binds to HSEs to drive transcriptional activation of the synthetic promoter. Thus, it was reasoned that the architecture of the HSEs within the synthetic thermal bioswitch could alter responses to heat. The number and spacing of heat shock elements in a synthetic thermal bioswitch affect its activity [0247] To test how the HSE architecture affects thermal bioswitch activity, panels of synthetic thermal gene switches containing combinations of HSEs upstream of a YB core promoter (e.g., SEQ ID NO:12) were constructed and screened. HSEs were (1) arranged in alternating inverted position (e.g., “i”, such as “S7YBi”), (2) spaced out by up to 30 bps (e.g., “-0S” to “-30S”, such as “S7YB-0S” to “S7YB-30S”), and (3) the number of HSEs increased from 1 to 15 (e.g., “S1”- “S15”, such as “S1YB”-“S15YB”) (FIG.5). It was found that increasing the HSE spacing from 4 bps to 30 bps decreased reporter expression at every measured timepoint, while increasing the HSE spacing to 15 bps showed insignificant changes to reporter expression up to 12 hrs. (FIG. 6A). Increasing the number of HSEs from 7 to 15 was further tested. See Tables 2-4 for exemplary sequences. Table 2. Exemplary heat shock element (HSE) sequences.

-117-
ny-2833630
Atty Docket No.23887-2000140
Table 3. Exemplary core promoter sequences.
-118-
ny-2833630
Atty Docket No.23887-2000140
-119-
ny-2833630
Atty Docket No.23887-2000140
Table 4. Exemplary thermally-inducible promoter sequences.
-120-
ny-2833630
Atty Docket No.23887-2000140
-121-
ny-2833630
Atty Docket No.23887-2000140
-122-
ny-2833630
Atty Docket No.23887-2000140
-123-
ny-2833630
Atty Docket No.23887-2000140

[0248] It was found that all thermal bioswitches with more than 7 HSEs had greater activity than S7YB. Unexpectedly, thermal bioswitch activity peaked at 8 HSEs (~2-fold greater than S7YB), and subsequent additions of HSEs did not further improve thermal bioswitch activity (see FIG. 6B). The impact of inverting alternate HSEs on thermal bioswitch activity with 7 HSEs was then evaluated. Improved thermal bioswitch activity in T cells derived from one donor was improved, but no differences were seen in T cells engineered with cells from a second donor (FIG.6C). [0249] First was tested 7 heat shock elements (HSEs) in combination with different core promoters (YB, CMVa, and CMVb; i.e., “S7YB” (SEQ ID NO:19), “S7CMVa” (SEQ ID NO:58), “S7CMVb” (SEQ ID NO:59), and S7CMVd” (SEQ ID NO:60)). YB is a synthetic core promoter described previously in Ede et al. (2016) ACS Synth. Biol.5, 395–404 and Hansen et al. (2014) Proc. Natl Acad. Sci. USA 111, 15705–15710, while the CMVa and CMVb core promoter are different truncations of the native CMV promoter described previously in Fonseca et al. (2019) ACS Synthetic Biology 8 (11), 2593-2606. Each thermal switch was placed upstream of either a Gaussia luciferase reporter (FIGS.7A-7B) or an EGFR-targeting CAR (FIGS.7C-7D). -124-
ny-2833630
Atty Docket No.23887-2000140
Testing core promoters, number of HSEs, Kozak, and 5 UTR elements in the thermal bioswitch. [0250] 27 thermal bioswitches were tested, wherein were varied the core promoter, number of HSEs (7 or 8 HSEs), or the Kozak sequence following the S7YB promoter. [0251] A core promoter is a specific DNA sequence located immediately upstream of the transcription start site (TSS) of a gene, which serves as the minimal essential region required for the assembly of the transcription machinery and the initiation of transcription. It typically spans a region of approximately 50-100 base pairs and contains specific motifs, such as the TATA box. The core promoter is necessary for the basal transcriptional activity of the gene. Here, core
promoter variants were tested one of two ways: either (1) removing the native 5 UTR from the indicated core promoter and appending it to the YB 5 UTR (e.g., CMVa core includes the 5 UTR of the YB promoter, e.g., SEQ ID NO:39), or (2) including the 5 UTR native to the indicated same core promoter (e.g., CMVa core includes the 5 UTR of the CMVa promoter, e.g., SEQ ID NO:40) (FIG.8A). [0252] The next component to be tested was the number of heat shock elements (HSEs) in the thermal bioswitch. Either 7 or 8 repeats of the HSE nGAAnnTTCnnGAAn were tested, both of which were shown to have superior transgene expression over constructs with other number of
HSEs (FIGS. 6A-6C), in combination with either the YB, CMVa, CMVb, CMVd, or EF1 core promoters (FIG.8B). Lastly were tested alternative Kozak sequences (SEQ ID NOs:81-83) (see
FIG. 8C) and alternate synthetic 5 UTRs (SEQ ID NOs:55-57) (FIG. 8D) combined with 7 HSEs and the YB core promoter (i.e., “S7YB”; e.g., SEQ ID NO:19). [0253] Each of the 27 thermal switches were placed upstream of an EGFR-targeting CAR. These were packaged into lentivirus and used to transduce primary human T cells. [0254] Results: T cells were isolated, transduced, and cultured as described above. To test thermal switch activation, engineered T cells were heated from 37ºC to 42ºC over 10 minutes (~0.5ºC/min) and maintained at 42ºC continuously for 40 min. EGFR CAR expression was assessed by staining with a soluble EGFR-Fc chimera protein conjugated to Alexa Fluor 647 or using an anti (G4S)3 linker antibody conjugated to APC (FIGS.6A-6C). Flow cytometry plots are gated on transduced (GFP+) T cells. [0255] Two EGFR CARs were tested, wherein one utilized a CD28 signaling domain and the other a 4-1BB signaling domain. Expression of an EGFR CAR under the control of the S7YB thermal switch enabled killing of EGFR+ tumor cells in a dose dependent manner (FIG.9). Thermal control of either EGFR CAR variant led to tumor killing. [0256] All thermal bioswitches tested led to CAR expression upon heating (FIG.10), some designs which are characterized by increased expression over S7YB upon heating and no -125-
ny-2833630
Atty Docket No.23887-2000140 expression while unheated (FIGS.11A-11D) and others which show baseline expression that is enhanced upon heating (FIGS.12A-12B).
[0257] S7YB designed with alternate synthetic 5 UTRs paired with alternate Kozak sequences (i.e., NEO UTR #2 + Kozak 1 and NEO UTR #3 + Kozak 3) led to a similar lack of expression in the unheated state compared to S7YB, but higher CAR expression upon heating (FIG.11A). This improved expression led to more potent killing over S7YB-engineered T cells (FIG.11B). Similarly, constructs with the CMVb core promoter that showed improved expression in the heated state were identified, showed a similar lack of activity in the unheated state, and subsequently resulted in higher killing over S7YB-transduced cells. (FIGS.11C-11D). Example 3 Heat-inducible Treg potentiation [0258] A subject can be administered any of the compositions disclose herein (e.g., as outlined in FIG.1). In some embodiments, the composition comprises Tregs that comprise a pHSP- controlled booster (e.g., IL-2). A constitutively expressed CAR targets, e.g., a stress-induced or tissue-abundant antigen. Induced IL-2 combined with CAR expression leads to Treg proliferation at site of heating. The administered Tregs can reside in tissues, for example tissues affected by an autoimmune disease. Periodic heating of major disease sites can drive Treg proliferation (e.g., monthly). [0259] The methods and compositions of the present disclosure address major efficacy-driving issues of proliferation and persistence in tissues. IL-2, for example, is an established proliferation signal supported by clinical data but has systemic toxicity. In the methods described herein, IL-2 is released only at the user-defined site(s) and only if Tregs are present. Example 4 Thermally Induced Production of IL-2 from Tregs [0260] This example demonstrates the thermally induced production of IL-2 from Tregs employing the methods and compositions provided herein. FIGS.13A-13E depict a non-limiting exemplary circuit design (FIG.13A) and data (FIGS.13B-13E) related to studies thereof. Circuit design comprised of the S7YB synthetic thermal switch (TS) driving the thermally controlled expression of IL-2 and EF1alpha promoter driving constitutive expression of a chimeric antigen receptor (FIG.13A). Engineered T
regs expressed CD25 and FOXP3, markers associated with Treg function and phenotype as measured by flow cytometry (FIG.13B). FIG. 13C depicts representative flow plots for GFP and NKG2D CAR expression on engineered Tregs. In engineered T
regs, the thermal switch (TS.IL-2) remained silent at temperatures up to 40.2ºC and produced IL-2 when cells were heated to temperatures above 40.2ºC for 30 min, as measured -126-
ny-2833630
Atty Docket No.23887-2000140 24 hrs post heating (FIG.13D). NKG2D CAR Tregs expressed IL-10 upon engagement with their target antigen expression on ARPE-19 (NKG2DL+) tumor cells (FIG.13E). When heated to 42ºC, higher levels of IL-10 were detected in cell supernatant detected by ELISA. [0261] In at least some of the previously described embodiments, one or more elements used in an embodiment can interchangeably be used in another embodiment unless such a replacement is not technically feasible. It will be appreciated by those skilled in the art that various other omissions, additions and modifications may be made to the methods and structures described above without departing from the scope of the claimed subject matter. All such modifications and changes are intended to fall within the scope of the subject matter, as defined by the appended claims. [0262] With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated. [0263] It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases “at least one” and “one or more” to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles “a” or “an” limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases “one or more” or “at least one” and indefinite articles such as “a” or “an” (e.g., “a” and/or “an” should be interpreted to mean “at least one” or “one or more”); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at -127-
ny-2833630
Atty Docket No.23887-2000140 least the recited number (e.g., the bare recitation of “two recitations,” without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to “at least one of A, B, and C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “ a system having at least one of A, B, and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to “at least one of A, B, or C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “ a system having at least one of A, B, or C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. [0264] In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. [0265] As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible sub-ranges and combinations of sub-ranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like include the number recited and refer to ranges which can be subsequently broken down into sub-ranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 articles refers to groups having 1, 2, or 3 articles. Similarly, a group having 1-5 articles refers to groups having 1, 2, 3, 4, or 5 articles, and so forth. [0266] While various aspects and embodiments have been disclosed herein, other aspects and embodiments will be apparent to those skilled in the art. The various aspects and embodiments disclosed herein are for purposes of illustration and are not intended to be limiting, with the true scope and spirit being indicated by the following claims. -128-
ny-2833630