WO2023235422A2 - Engineered chimeric fusion protein compositions and methods of use thereof - Google Patents

Engineered chimeric fusion protein compositions and methods of use thereof Download PDF

Info

Publication number
WO2023235422A2
WO2023235422A2 PCT/US2023/024049 US2023024049W WO2023235422A2 WO 2023235422 A2 WO2023235422 A2 WO 2023235422A2 US 2023024049 W US2023024049 W US 2023024049W WO 2023235422 A2 WO2023235422 A2 WO 2023235422A2
Authority
WO
WIPO (PCT)
Prior art keywords
domain
sequence
cell
composition
cfp
Prior art date
Application number
PCT/US2023/024049
Other languages
French (fr)
Other versions
WO2023235422A3 (en
Inventor
Daniel Getts
Original Assignee
Myeloid Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Myeloid Therapeutics, Inc. filed Critical Myeloid Therapeutics, Inc.
Publication of WO2023235422A2 publication Critical patent/WO2023235422A2/en
Publication of WO2023235422A3 publication Critical patent/WO2023235422A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • Myeloid cells are a major cellular compartment of the immune system comprising monocytes, dendritic cells, tissue macrophages, and granulocytes. Their enormous plasticity and heterogeneity, during both homeostasis and disease can be harnessed to develop safe and efficacious therapeutics.
  • Myeloid cells can be engineered to specifically target and destroy a range of pathogenic agents or pathogenic cell types, mount a strong immunological response that can further induce a T cell response in vivo.
  • Engineered myeloid cells can also be short-lived in vivo, phenotypically diverse, sensitive, plastic, and are often found to be difficult to manipulate in vitro.
  • monocytes have been difficult compared to exogenous gene expression in non- hematopoietic cells.
  • myeloid cells e.g., monocytes/macrophages.
  • myeloid cells such as monocytes/macrophages, comprise many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process. This is especially true of activated macrophages which undergo a dramatic change in their physiology following exposure to immune or inflammatory stimuli.
  • the diverse functionality of myeloid cells makes them an ideal cell therapy candidate that can be engineered to have numerous therapeutic effects.
  • the present disclosure is related to immunotherapy using myeloid cells (e.g., CD14+ cells) of the immune system, particularly phagocytic cells.
  • myeloid cell immunotherapy could be exceedingly important in cancer, autoimmunity, fibrotic diseases and infections.
  • the present disclosure is related to immunotherapy using myeloid cells, including phagocytic cells of the immune system, particularly macrophages. It is an object of the invention disclosed herein to harness one or more of these functions of myeloid cells for therapeutic uses.
  • the present disclosure provides innovative methods and compositions that can successfully transfect or transduce a myeloid cell, or otherwise induce a genetic modification in a myeloid cell, with the purpose of augmenting a functional aspect of a myeloid cell, additionally, without compromising the cell’s differentiation capability, maturation potential, and/or its plasticity.
  • the present disclosure involves making and using engineered myeloid cells (e.g., CD14+ cells, such as macrophages or other phagocytic cells, which can attack and kill (ATAK) diseased cells directly and/or indirectly, such as cancer cells and infected cells.
  • engineered myeloid cells e.g., CD14+ cells, such as macrophages or other phagocytic cells, which can attack and kill (ATAK) diseased cells directly and/or indirectly, such as cancer cells and infected cells.
  • Engineered myeloid cells such as macrophages and other phagocytic cells, can be prepared by incorporating nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric fusion protein (CFP), that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology.
  • nucleic acid sequences e.g., mRNA, plasmids, viral constructs
  • CRISPR chimeric fusion protein
  • transduction e.g., using viral constructs
  • electroporation e.g., electroporation, or nucleofection, or in
  • myeloid cells can be engineered to have a broad and diverse range of activities.
  • myeloid cells can be engineered to express a chimeric fusion protein (CFP) containing an antigen binding domain to have a broad and diverse range of activities.
  • CFP chimeric fusion protein
  • myeloid cells can be engineered to have enhanced phagocytic activity such that upon binding of the CFP to an antigen on a target cell, the cell exhibits increased phagocytosis of the target cell.
  • myeloid cells can be engineered to promote T cell activation such that upon binding of the CFP to an antigen on a target cell, the cell promotes activation of T cells, such as T cells in the tumor microenvironment.
  • the engineered myeloid cells can be engineered to promote secretion of tumoricidal molecules such that upon binding of the CFP to an antigen on a target cell, the cell promotes secretion of tumoricidal molecules from nearby cells.
  • the engineered myeloid cells can be engineered to promote recruitment and trafficking of immune cells and molecules such that upon binding of the CFP to an antigen on a target cell, the cell promotes recruitment and trafficking of immune cells and molecules to the target cell or a tumor microenvironment.
  • the one or more polynucleic acids is an mRNA.
  • the recombinant polynucleic acid comprises a sequence encoding the CFP.
  • the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements, e.g., a LINE1 element, e.g. sequences encoding L1 -ORF1 and L1-ORF2.
  • the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements and a sequence for a guide RNA.
  • the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements in which the retrotransposable elements is fused with a heterologous protein or functional portion thereof.
  • the recombinant polynucleic acid comprises a sequence encodes for two CFPs (e.g., a CFP and a CAR) in tandem, separated by an autocleavable moiety (e.g., T2A, P2A etc).
  • the mRNA is 2kb - 20,000kb long.
  • the recombinant polynucleic acid comprises one or more elements for stabilizing structures. Particularly important for long mRNA is to have 5’ Cap protection.
  • the RNA products required for generating engineered myeloid cells of therapeutic interest are generated in vitro, 5’capped and 3’-polyA tail.
  • the 5’Cap is modified from a native mRNA 5’Cap.
  • One aspect of this disclosure involves having an efficient Cap structure using suitable capping analogs.
  • the RNA products prepared by IVT mRNA are co-transcriptional capped with m7GpppN-derived dinucleotides.
  • the 5’-Cap is a trinucleotide cap analog.
  • the first nucleotide base following a 5’-Cap plays a crucial role in mRNA stability.
  • the base is adenine (A).
  • the base is cytosine (C).
  • the base is guanine (G).
  • the base is uracil (U).
  • the Cap analog is m 7 GpppApG.
  • the Cap analog is m 7 GpppAmpG.
  • the Cap analog is m 7 Gpppm 6 ApG.
  • the Cap analog is m 7 Gpppm 6 A m pG.
  • the Cap analog is m 7 GpppCpG.
  • the Cap analog is m 7 GpppCmpG.
  • the Cap analog is m 7 GpppGpG.
  • the Cap analog is m 7 GpppG m pG. [0024] In some embodiments, the Cap analog is m 7 GpppUpG. [0025] In some embodiments, the Cap analog is m 7 GpppU m pG. [0026] In some embodiments, the 5’-Cap is anti-reverse cap analogs (ARCAs) having chemically modified 3 -O or 2 -O positions of m7G (m2 7,3 -O GpppG). [0027] In some embodiments, the 5’-Cap is N7-methylguanosine 5 -diphosphate (m7GDPIm).
  • the caping involves post-transcriptional enzymatic capping of 5 - triphosphates RNA using Vaccinia virus capping complex.
  • the 5’capping confers stability and translational efficiency to the mRNA.
  • cap-proximal sequence is crucial for mRNA stability and function.
  • the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer.
  • a method for treating a disease in a subject in need thereof comprising administering a therapeutically effective amount of a pharmaceutical composition described herein to the subject.
  • the disease is cancer.
  • the disease is an infection.
  • the subject is human.
  • a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a CD5 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least three intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (A) a first intracellular signaling domain from Fc epsilon receptor Ig (FCER1G), and (B) a second intracellular signaling domain comprising a PI3K recruitment domain.
  • FCER1G Fc epsilon receptor Ig
  • the CFP has the sequence according to SEQ ID NO:50.
  • the Cap structure is a trinucleotide cap analog.
  • a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a HER2 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least three intracellular signaling domains comprise: (A) a first intracellular signaling domain from CD40, (B)
  • the CFP has the sequence according to SEQ ID NO:51.
  • the Cap structure is a trinucleotide cap analog.
  • the Cap comprises a 2’-O-methyl modification.
  • the Cap comprises an N6 methyl modification.
  • the Cap comprises both a 2’-O-methyl modification and an N6 methyl modification.
  • the mRNA is in vitro transcribed. In some embodiments, the mRNA is HPLC purified.
  • composition comprising an mRNA comprising a sequence encoding a CFP; a 5’UTR, a 3’UTR, the 5’UTR terminating in a 5’cap analog represented by the formula 5’-m7GpppAmpG, wherein, G represents Guanine, A represents Adenine, p represents phosphate group, and m represents methylation; and wherein, the CFP comprises an antigen binding domain having a variable heavy chain sequence with at least 95% sequence identity to [0038] In some embodiments, the CFP comprises an antigen binding domain having a variable light chain sequence with at least 95% sequence identity to [0039] In some embodiments, the CFP comprises an scFv having a sequence that is at least 95% identical to the sequence [0040] In some embodiments, the CFP further comprises a transmembrane domain comprising a CD8 transmembrane domain, the sequence comprising at least 95% identity to IYIWAPLAGTC
  • the CFP further comprises a CD8 hinge domain having a sequence that has at least 95% sequence identity to the sequence
  • the CFP further comprises an intracellular signaling domain encoding an FCER1G intracellular signaling domain having a sequence: RRLKIQVRKAAITSYEKSDGVYTGLSTRNQETYETLKHEKPPQ.
  • the CFP further comprises an intracellular signaling domain encoding an intracellular signaling domain from PI3kinase recruitment domain, having the sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM.
  • the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
  • the CFP specifically binds to CD5 antigen.
  • the cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
  • a nucleotide in the second position of the 5’cap analog is methylated.
  • the nucleotide in the second position of the 5’cap is further modified at the N6 position.
  • expression of the sequence encoded by the mRNA in a cell is enhanced compared to an mRNA that does not comprise the 5’cap specified herein.
  • the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to [0044] In some embodiments, the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRF SGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKVEIK.
  • the CFP further comprises an antigen binding domain that is an scFv having a sequence that is at least 95% identical to the sequence
  • the CFP further comprises a CD8 hinge domain having a sequence ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLD.
  • the CFP further comprises a CD8 transmembrane domain having a sequence, IYIWAPLAGTCGVLLLSLVITLYC.
  • the intracellular signaling domain from CD40 comprises a sequence Q.
  • the intracellular signaling domain from FCER1G comprises a sequence .
  • the intracellular signaling domain from PI3K recruitment domain comprises a sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM.
  • a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; and wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a GPC3 binding domain and (ii) a CD89 transmembrane domain.
  • CFP chimeric fusion protein
  • the CD89 transmembrane domain is operably linked to the extracellular domain.
  • the CFP further comprises (iii) a CD89 intracellular domain.
  • the CFP further comprises an extracellular domain from CD89.
  • the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
  • the GPC3 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is FYSYTY.
  • the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence In some embodiments, the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence Q Q Q Q Q In some embodiments, wherein the CD89 transmembrane domain is LIRMAVAGLVLVALLAILV. In some embodiments, CFP comprises a CD89 sequence that is at least 95 % identical to the sequence SVC . In some embodiments, CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
  • composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a TROP2 binding domain and (ii) a CD89 transmembrane domain.
  • CFP chimeric fusion protein
  • the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV.
  • the HCDR1 sequence is TNYGM; and the HCDR2 sequence is WINTYTGEPTYTDDFKG.
  • the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLTF.
  • the the LCDR1 sequence is KASQDVSIA; and the LCDR2 sequence is SASYRYT.
  • the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence VSS. In some embodiments, the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence In some embodiments, the CD89 transmembrane domain is LIRMAVAGLVLVALLAILV. In some embodiments, the CFP comprises a CD89 extracellular sequence DSIHQDYTTQN. In some embodiments, the CFP comprises a CD89 intracellular sequence In some embodiments, the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
  • composition comprising: (a) a lipid nanoparticle comprising (i) a cationic lipid (ii) a non-cationic lipid (iii) a neutral lipid; the lipid nanoparticle encapsulating: (b) an mRNA encoding a chimeric fusion protein (CFP), wherein the mRNA comprises a 5’cap analog represented by the formula 5’-m7GpppAmpG; the CFP comprise an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain; wherein the transmembrane domain of the protein CD89, Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a).
  • CFP chimeric fusion protein
  • the 5’cap is a cap 1 structure.
  • a nucleotide in the second position of the 5’cap is methylated.
  • the nucleotide in the second position of the 5’cap is further modified.
  • the nucleotide in the second position of the 5’cap is modified at the N6 position.
  • the lipid nanoparticle further comprises cholesterol.
  • the lipid nanoparticle is less than 200 nM in diameter.
  • the chimeric antigen receptor comprises an antigen binding domain that binds to a cancer antigen.
  • the chimeric antigen receptor comprises an antigen binding domain that is an scFv, and that specifically binds to a cancer antigen. In some embodiments, the chimeric antigen receptor comprises an antigen binding domain that binds to TROP2. In some embodiments, the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV. In some embodiments, the HCDR1 sequence is TNYGM; and the HCDR2 sequence is WINTYTGEPTYTDDFKG.
  • VH variable heavy chain
  • the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLTF.
  • VL variable light chain
  • the LCDR1 sequence is KASQDVSIA; and the LCDR2 sequence is SASYRYT.
  • the chimeric antigen receptor comprises GPC3 binding variable heavy chain (VH), and a variable light chain (VL), wherein the variable heavy chain comprises a HCDR1, a HCDR2 and a HCDR3, the HCDR3 having a sequence of ATACADTTQYAYDY, ATACADTTLYEYDY, ATACVDTTQYEYDY, ATACADATQHEYDY, ATACADTTQYDYDY, ATACADTTQYEYDY, ATACADTTHYEYDY, ATACVITTLYEYDY, ATACAETTLYEYDY, ATACADTTQHEYDY, ATACVDTTHYEYDY, ATACASTTLYEYDY, ATACVVTTLYEYDY, ATACGGATGPYDY, ATACAGAIGPYDY, ATACVVVGDQNDY, ATACVVVGDRNDY, ATDCAGGTSTPYDY, ATDCAGGTATPYDY, ATACVVADRNEYDY, ATACVVADR
  • the chimeric antigen receptor comprises an antigen binding domain that is an variable heavy chain (VHH), and that specifically binds to a cancer antigen.
  • VHH variable heavy chain
  • a pharmaceutical composition comprising the composition described above, and a pharmaceutically acceptable excipient.
  • a cell comprising the recombinant mRNA.
  • the cell is a live cell.
  • the cell is a myeloid cell.
  • the cell is a CD14+ cell.
  • the cell is a CD14+ CD16- cell.
  • the cell is a CD14+ CD16+ cell.
  • the cell is a myeloid cell.
  • the cell is a monocyte.
  • the cell is a myeloid cell. In some embodiments, the cell is a monocyte precursor. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a macrophage cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a dendritic cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a precursor of a dendritic cell.
  • a method of expressing a chimeric fusion protein in a cell comprising in vitro transcribing a recombinant mRNA encoding the chimeric fusion protein, 5’capping the mRNA with a Cap structure selected from the Cap structures of Table 7, thereby generating a 5’-capped recombinant mRNA; contacting the cell with a composition comprising the 5’-capped recombinant mRNA encoding the chimeric fusion protein, wherein the mRNA is incorporated into the cell.
  • the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
  • a nucleotide in the second position of the 5’cap is methylated, and is further modified at the N6 position.
  • the mRNA expression is enhanced compared to a recombinant mRNA that does not comprise the 5’cap structure specified herein.
  • the composition further comprises a lipid nanoparticle encapsulating the 5’-capped recombinant mRNA.
  • the cell does not express CD16.
  • the cell is CD14+ and CD16+.
  • Provided herein is a method of treating cancer in a subject, the method comprising administering to the subject in need thereof any one of the compositions described above, an mRNA, a naked mRNA, an mRNA encapsulated in a lipid nanoparticle, or a cell comprising the mRNA, wherein the mRNA comprises a 5’cap as described herein at least at the time of uptake of the mRNA by a cell.
  • the cancer is ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer.
  • the cancer is breast cancer.
  • FIG. 1 outlines the ARCA versus Clean Cap processes of mRNA capping in vitro.
  • ARCA left
  • Clean Cap analog right
  • FIG. 2 depicts a diagram showing naturally occurring m6Am Cap (top left), and the structure of bases (top right); and four trinucleotide Cap analog structures.
  • FIG.3 depicts structures of an adenine Cap analogue.
  • FIG. 4 depicts graphical structures of different Cap analogs, ARCA (Cap0), and two other (Cap1) structures, termed herein as CC1 (Cap1) and EC1 (Cap 1), whose structural design are shown.
  • the polynucleotide chain in blue is mRNA terminating in the cap at it’s 5’ end.
  • FIG. 5 depicts a generalized workflow for evaluating the effect of the Cap analogs described in FIG.4. EP, electroporation.
  • FIG. 6 depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct on the expression level at 24 hours after introduction to primary monocytes derived from human donors.
  • FIG. 7 depicts data from experiment conducted as described for FIG. 6, showing the expression level of HER2 binding CAR over the time period indicated in the X axis after introduction of the mRNA in primary monocytes derived from human donors.
  • FIG.8 depicts a graphical representation of a workflow for evaluating the effect of the cap analogs for immunogenic response.
  • FIG. 9A depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct, interferon-alpha (IFN- ⁇ ) and interferon-beta (IFN- ⁇ ) release at 4h and 20h after introduction to primary monocytes derived from human donors. Data on commercial ARCA is boxed.
  • FIG. 9B depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct, CXCL-10 and IFITM1 generation at 4h and 20h after introduction to primary monocytes derived from human donors. Data on commercial ARCA is boxed.
  • FIG. 9A depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct, CXCL-10 and IFITM1 generation at 4h and 20h after introduction to primary monocytes derived from human donors. Data on commercial ARCA is boxed.
  • FIG. 10 depicts visualization of RNA products from a large scale production of capped mRNA, involving comparison studies of ARCA -vs – CC1 capped mRNA encoding a GP-75 binding CAR, gp-75-FcR-PI3K.
  • Left figure mRNA resolved in an electrophoresis, against a molecular size marker ladder (left-most well).
  • Right figure dot blot depiction of the prepared mRNA samples for detection of double-stranded RNA produced which are contaminant components in the preparation.
  • GP75-FcR-PI3K is a structural depiction of the domains of the CAR, having an anti-GP75 extracellular antigen binding domain comprising an anti-GP75 scFv; an FcR transmembrane domain, and a PI3Kinase intracellular domain.
  • FIG. 12 depicts experiment results, showing the effect of monocytes expressing the indicated anti-gp75 CAR mRNA (gp75-FLAG-FcR-PI3K) capped with ARCA or CC1 cap on tumor size in a syngeneic mouse tumor model.
  • FIG. 13 depicts a gel electrophoresis showing electrophoretic mobility of capped and uncapped mRNA of the indicated preparations. The bands for capped and uncapped mRNA are shown by arrows. The ratio of the amount of cap to the ATP or GTP amount in the preparation are indicated at the top.
  • FIG. 14A-14B represents data from an independent experiment, involving an assay using monocytes from a single donor (MYE101-0023B) using the protocol depicted in FIG. 5; mRNA encoding anti-HER2 CARs were tested for the effect of different cap structures in the mRNA constructs as depicted at the top of the figures. The data agrees with the data shown in FIG.6.
  • FIG. 15 depicts data showing binding of HER2, and an assay that determines the expression level of the anti-HER2 CAR in the cells from the experiment of FIGs.14A-14B. 0.1 microgram RNA was electroporated in 2x 10 ⁇ 6 cells in 20 microliters reaction volume, final RNA concentration 0.1 uM. [0076] FIG.
  • FIG. 16 depicts data from the same experiment of FIGs 14A, 14B and 15, the data showing duration of expression of the anti-HER2 CAR based on the cap structures.
  • FIG. 17 depicts data from experiment performed with cells from a donor PD-004-04 electroporated with 0.1 microM final concentration mRNA of any one of uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti-HER2 CAR-polyA, m6A-capped -anti-HER2 CAR-polyA, EC1- capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR-polyA mRNA constructs. Data showing flow cytometry of cells electroporated with the constructs.
  • FIG. 18 depicts data from the experiment described in FIG. 17, showing expression of anti-HER2 as determined by binding to HER2.
  • FIG. 19 depicts data from the experiment described in FIG. 17 and FIG. 18, showing expression of anti-HER2 over time.
  • FIG. 20 depicts data from a parallel experiment similar to that described in FIG.
  • the different mRNA sets are: uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti-HER2 CAR- polyA, m6A-capped -anti-HER2 CAR-polyA, EC1-capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR-polyA mRNA constructs. Data showing flow cytometry of cells transfected with the constructs. Transfection was done with 6x10 ⁇ 5 cells/24 well/0.5 microgram RNA. [0081] FIG. 21 depicts data showing anti-HER2 binder expression in the different experimental sets described in FIG.20.
  • FIG. 22 depicts data from the experiment described in FIG. 20 and FIG. 21, showing expression of anti-HER2 over time following the lipofection.
  • FIG. 23 depicts data from an experiment performed with cells from a donor MYE-101- 0024A electroporated with 0.1 microM of uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti- HER2 CAR-polyA, EC1-capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR- polyA mRNA constructs. Data showing flow cytometry of cells electroporated with the constructs. RNA was made with IVT buffer at pH 6.5 or ARCA kit. [0084] FIG.
  • FIG. 24 depicts data showing expression of anti-HER2 binder (CAR) in the experiment described in FIG.23.
  • FIG. 25 depicts data from the experiment described in FIG. 23 and FIG. 24, showing expression of anti-HER2 CAR over time following the lipofection.
  • FIG. 26 depicts data from an experiment where the monocyte donor is PD004-04. Monocytes were electroporated with mRNA that were not purified or purified by HPLC. 2x10 ⁇ 6 cells were electroporated with RNA, final concentration 0.5 uM.
  • FIG.27A depicts data indicating expression of anti-HER2 CAR in the different sets, in the experiment described for FIG.26 at 4h and 24hs.
  • FIG.27A depicts data indicating expression of anti-HER2 CAR in the different sets, in the experiment described for FIG.26 at 4h and 24hs.
  • FIG. 27B depicts data indicating expression of anti-HER2 CAR in the different sets, at 48h after electroporation.
  • FIG.28 depicts data showing the duration of the HER2 binder expression.
  • DETAILED DESCRIPTION All terms are intended to be understood as they would be understood by a person skilled in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains. [0091] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. [0092] Although various features of the present disclosure can be described in the context of a single embodiment, the features can also be provided separately or in any suitable combination.
  • the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open- ended and do not exclude additional, unrecited elements or method steps. It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method or composition of the disclosure, and vice versa. Furthermore, compositions of the disclosure can be used to achieve methods of the disclosure.
  • engineered myeloid cells including, but not limited to, neutrophils, monocytes, myeloid dendritic cells (mDCs), mast cells and macrophages
  • the engineered myeloid cells can attack and kill target cells directly (e.g., by phagocytosis) and/or indirectly (e.g., by activating T cells).
  • the target cell is a cancer cell.
  • cancer is one exemplary embodiment described in detail in the instant disclosure, the methods and technologies described herein are contemplated to be useful in targeting an infected or otherwise diseased cell inside the body. Similarly, therapeutic and vaccine compositions using the engineered cells are described herein.
  • compositions and methods for treating diseases or conditions such as cancer.
  • the compositions and methods provided herein utilize human myeloid cells, including, but not limited to, neutrophils, monocytes, myeloid dendritic cells (mDCs), mast cells and macrophages, to target diseased cells, such as cancer cells.
  • the compositions and methods provided herein can be used to eliminate diseased cells, such as cancer cells and or diseased tissue, by a variety of mechanisms, including T cell activation and recruitment, effector immune cell activation (e.g., CD8 T cell and NK cell activation), antigen cross presentation, enhanced inflammatory responses, reduction of regulatory T cells and phagocytosis.
  • the myeloid cells can be used to sustain immunological responses against cancer cells.
  • a composition comprising a recombinant polynucleic acids, wherein the one or more polynucleic acids encodes one or more chimeric fusion proteins, and wherein the recombinant polynucleic acid when introduced systemically, is expressed in specific cell types in vivo.
  • the recombinant polynucleic acid is an mRNA.
  • the recombinant polynucleic acid is an mRNA comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for myeloid cell-specific expression.
  • CFP chimeric fusion protein
  • the recombinant polynucleic acid is an mRNA.
  • the recombinant polynucleic acid is an mRNA comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for non-myeloid cell-specific expression.
  • the non-myeloid cell is a lymphocyte, e.g., B cell, a T cell, an NK cell etc.
  • the recombinant polynucleic acid comprises a first sequence comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for myeloid cell- specific expression, and a second sequence encoding a chimeric receptor that is designed for non- myeloid cell specific expression.
  • non-myeloid cell is a T cell.
  • a single polynucleotide is designed that comprises a first sequence that encodes a CFP, which predominantly expresses in a myeloid cell; and a second sequence that encodes a chimeric antigen receptor (CAR) that is predominantly expressed in a T cell.
  • the first and the second sequences are intervened by a self-cleavage sequence such as a T2A or P2A sequence, thereby post-translationally generating two peptides, one comprising the CFP, the other comprising the CAR.
  • a recombinant polypeptide translation product of the recombinant polynucleic acid described above comprising (1) a single polypeptide comprising a CFP that is expressed in a myeloid cell, and (2) a single polypeptide comprising a CAR that is expressed in a T cell.
  • a polynucleic acid construct comprising sequence encoding the CFP arm and the sequence encoding the CAR arm as described herein can be delivered in vivo in a suitable vehicle such that the CFP is expressed in a myeloid cell in vivo, and the CAR is expressed in a lymphoid cell in vivo.
  • the recombinant polynucleic acid is an mRNA comprising: (a) a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) a CFP extracellular domain comprising a first antigen binding domain, and (ii) a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and (b) a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: (i) an CAR extracellular domain comprising a second antigen binding domain, (ii) a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts
  • CFP chimeric
  • compositions comprising: (A). one or more lipid nanoparticles, (B) one or more polynucleic acids, the one or more polynucleic acids comprising: (a) a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) a CFP extracellular domain comprising a first antigen binding domain, and (ii) a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and (b) a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: (i) a CAR extracellular domain comprising a second antigen binding domain, (ii) a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR trans
  • the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence, wherein the first sequence and the second sequence are operatively linked by a linker sequence.
  • the linker sequence comprises a protease cleavage site such as a T2A cleavage site or a P2A cleavage site. The P2A or the T2A sequence can cleave the first arm and the second arm, releasing the CFP and the CAR arms in vivo for independent expression.
  • the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle.
  • the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle.
  • the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle.
  • the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle.
  • after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject.
  • the CAR after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject.
  • the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject.
  • the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR- beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR- delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • the CAR intracellular domain further comprises a costimulatory domain.
  • the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137).
  • the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain.
  • the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89.
  • CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain.
  • the CFP after administration of the pharmaceutical composition to a human subject the CFP is expressed or specifically expressed in myeloid cells, monocytes or macrophages of the human subject.
  • the CAR is expressed or specifically expressed in T cells of the human subject.
  • the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89.
  • the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain.
  • the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from Fc ⁇ R, Fc ⁇ R, Fc ⁇ R, CD40 or CD3zeta.
  • the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain.
  • PI3K phosphoinositide 3-kinase
  • the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4.
  • the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89.
  • the one or more polynucleic acids is an mRNA.
  • the nanoparticle delivery vehicle comprises a lipid nanoparticle.
  • the lipid nanoparticle comprises a polar lipid.
  • the lipid nanoparticle comprises a non-polar lipid.
  • the lipid nanoparticle is from 100 to 300 nm in diameter.
  • the first antigen binding domain and the second antigen binding domain bind to the same antigen.
  • the first antigen binding domain and the second antigen binding domain bind to different antigens.
  • the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75.
  • the T cell is a CD3+ T cell, a CD4+ T cell, a CD8+ T cell, an NK T cell, an ⁇ T cell or a ⁇ T cell.
  • the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer.
  • the present disclosure is based on the important finding that engineered myeloid cells overcome at least some of the limitations of CAR-T cells, including being readily recruited to solid tumors; having an engineerable duration of survival, therefore lowering the risk of prolonged persistence resulting in aplasia and immunodeficiency; myeloid cells cannot be contaminated with T cells; myeloid cells can avoidance of fratricide, for example because they do not express the same antigens as malignant T cells; and myeloid cells have a plethora of anti-tumor functions that can be deployed.
  • engineered myeloid derived cells can be safer immunotherapy tools to target and destroy diseased cells.
  • myeloid cells such as macrophages
  • TAE tumor environment
  • myeloid cells such as macrophages
  • the present invention relates too harnessing myeloid cell function and specifically for targeting, killing and directly and/or indirectly clearing diseased cells as well as the delivery payloads such as antigens and cytokines.
  • Engineered T cells and myeloid cells can be achieved through the in vivo incorporation of nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric fusion protein (CFP), that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, ex vivo using recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology.
  • nucleic acid sequences e.g., mRNA, plasmids, viral constructs
  • CRISPR chimeric fusion protein
  • transduction e.g., using viral constructs
  • electroporation e.g., electroporation, or nucleofection, or in viv
  • the present disclosure involves, in addition to the programming of myeloid cells in vivo, programming T cells in vivo. It has been found that the combination of T cell and myeloid cell engineering can have robust anti-tumor cell activity. For example, it has been found that T cells and myeloid cells can be engineered to express a chimeric antigen receptor (CAR) and chimeric fusion protein (CFP), respectively, each containing an antigen binding domain to have a broad and diverse range of activities. For example, it has been found that T cells can be engineered to have enhanced cytolytic activity such that upon binding of the CAR to an antigen on a target cell, the cell exhibits increased lysis of the target cell.
  • CAR chimeric antigen receptor
  • CFP chimeric fusion protein
  • Engineered T cells can be achieved through the in vivo incorporation of nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric antigen receptor (CAR) that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, ex vivo using recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology.
  • nucleic acid sequences e.g., mRNA, plasmids, viral constructs
  • CAR chimeric antigen receptor
  • Engineered myeloid cells can also be short-lived in vivo, phenotypically diverse, sensitive, plastic, and are often found to be difficult to manipulate in vitro. For example, exogenous gene expression in monocytes has been difficult compared to exogenous gene expression in non- hematopoietic cells. There are significant technical difficulties associated with transfecting myeloid cells (e.g., monocytes/macrophages). As professional phagocytes, myeloid cells, such as monocytes/macrophages, comprise many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process.
  • the present disclosure provides innovative methods and compositions that can successfully transfect or transduce a myeloid cell, or otherwise induce a genetic modification in a myeloid cell, with the purpose of augmenting a functional aspect of a myeloid cell, additionally, without compromising the cell’s differentiation capability, maturation potential, and/or its plasticity.
  • a pharmaceutical composition comprising: (A) a recombinant nucleic acid comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient; wherein the recombinant nucleic acid is a recombinant self-amplifying nucleic acid.
  • CFP chimeric fusion protein
  • PR phagocytic or tethering receptor subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transme
  • the recombinant self-amplifying nucleic acid is a recombinant self- amplifying RNA.
  • the recombinant self-amplifying nucleic acid is a recombinant self- amplifying mRNA.
  • the recombinant self-amplifying nucleic acid comprises a 5' Cap.
  • the recombinant self-amplifying nucleic acid comprises a 3' polyadenylated (polyA) tail sequence.
  • the recombinant self-amplifying nucleic acid comprises a subgenomic promoter upstream of the sequence encoding a CFP. In some embodiments, the recombinant self-amplifying nucleic acid comprises a sequence encoding one or more replication genes from a virus. In some embodiments, the sequence encoding one or more replication genes from a virus is upstream of the sequence encoding a CFP. In some embodiments, the sequence encoding one or more replication genes from the virus is upstream of the subgenomic promoter. In some embodiments, the recombinant self-amplifying nucleic acid comprises a promoter upstream of the sequence encoding one or more replication genes from a virus.
  • the one or more replication genes comprise one or more non-structural proteins (NSPs).
  • the one or more NSPs comprises one or more of NSP1, NSP2, NSP3 and/or NSP4.
  • the recombinant self-amplifying nucleic acid lacks one or more viral genes encoding viral structural proteins required to make an infectious viral particle.
  • the virus is an RNA virus.
  • the virus is a single stranded virus.
  • the virus is an alphavirus or a flavivirus, selected from Venezuelan equine encephalitis virus (VEE), Sindbis virus (SINV), vaccinia virus and Semliki forest virus (SFV).
  • the recombinant self-amplifying nucleic acid is encapsulated within a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • a pharmaceutical composition comprising: (A) a recombinant RNA comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient, wherein the recombinant RNA is an in vitro glycosylated RNA comprising at least one residue that comprises a glycan.
  • the glycan is an N-linked glycan.
  • the at least one residue that comprises a glycan is a guanosine (G).
  • the glycan is sialylated glycan.
  • the glycan is fucosylated.
  • the glycan is wherein the recombinant RNA is a recombinant mRNA.
  • the recombinant RNA is encapsulated within a lipid nanoparticle (LNP).
  • the in vitro glycosylated RNA is glycosylated at specific residues.
  • the recombinant mRNA comprises 1,2 ,3 ,4, 5, 6, 7, 8, 9, 10 or more glycans.
  • the recombinant mRNA is a self-amplifying mRNA.
  • a pharmaceutical composition comprising: (A) a recombinant nucleic acid comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient wherein the recombinant nucleic acid comprises a sequence complementary to an miRNA, wherein the miRNA: (i) is expressed in a myeloid cell and binding of the
  • the non-myeloid cell is a neuronal cell. In some embodiments, the non-myeloid cell is an epithelial cell. In some embodiments, the non-myeloid cell is a hepatocyte. In some embodiments, the non-myeloid cell is a cardiomyocyte. In some embodiments, the non- myeloid cell is an embryonic stem cell. In some embodiments, the non-myeloid cell is an endothelial cell.
  • the miRNA is hsa-mir-302a, hsa-miR-302b, hsa-miR-302c, hsa-miR- 302d, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373, miR-10a/b, miR-24/27, miR-125a, miR-126 or miR-221/222.
  • the myeloid cell is a CD14+ cell.
  • the CD14+ cell is CD16- or CD16low.
  • the composition further comprises one or more lipid and/or carbohydrate or one or more polymer components.
  • the recombinant mRNA that is at least 5 kb in length.
  • the pharmaceutical composition is formulated for systemic delivery.
  • the recombinant mRNA is encapsulated in a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • the lipid nanoparticle is about 100 nm in diameter.
  • the lipid nanoparticle comprises at least a cationic lipid, and/or a non-cationic lipid.
  • the cationic lipid is selected from a group consisting of: phosphatidylserines, phosphatidic acids, phosphatidylglycerols, phosphatidylinositols, sterol hemisuccinates, dialkyl trimethylammonium-propanes, (e.g. DOTAP, DOTMA), dialkyl dimethylaminopropanes, ethyl phosphocholines, dimethylaminoethane carbamoyl sterols (e.g. DC- Chol).
  • the non-cationic lipid is a neutral lipid.
  • the non-cationic lipid is selected from the group consisting of: DSPC, DPPC, DMPC, DOPC, POPC, DOPE and SM.
  • the molar ratio of the mRNA to the neutral lipid ranges from about 2:1 to about 8:1.
  • the lipid composition further comprises a pegylated lipid.
  • the PEGylated lipid comprises a 50-200 carbon linear chain.
  • the pegylated lipid comprises one or more of a pegylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a pegylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2′,3′-di(tetradecanoyloxy)propyl-1-O-(cw- methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), or, a pegylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as ⁇ -methoxy(polyethoxy)ethyl-N-(2,3- di(tetradecanoxy)
  • PEG-DAG
  • the molar ratio of the mRNA to the pegylated lipid ranges from about 100:1 to about 25:1.
  • the lipid nanoparticle further comprises a steroid or steroid analog.
  • the steroid or steroid analogue is cholesterol.
  • the molar ratio of the mRNA to cholesterol ranges from about 2:1 to 1:1.
  • the pharmaceutical composition further comprising one or more phospholipids.
  • the one or more phospholipids selected from the group consisting of phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, di stearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine.
  • the CFP further comprises an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell.
  • the intracellular signaling domain comprises a phagocytosis activation domain.
  • the intracellular signaling domain is derived from a receptor selected from the group consisting of the receptors listed in Table 2.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain.
  • the antigen is an antigen or a surface molecule that is expressed by a diseased cell or a pathogenic cell or a pathogen.
  • the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a V H domain, a V L domain, a VNAR domain, a V HH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof.
  • the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a V H domain, a V L domain, a VNAR domain, a V HH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof.
  • the antigen is selected from the group consisting of Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), Mesothelin, EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B-Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2,
  • TK1 Thym
  • the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56. [0127] In some embodiments, the antigen is human CD5. In some embodiments, the antigen is human HER2. [0128] In some embodiments, the antigen is human CD19. In some embodiments, the antigen is a viral antigen expressed on a mammalian cell. In some embodiments, the antigen is a bacterial antigen expressed on a mammalian cell.
  • the killing activity of a cell expressing the CFP is increased by at least greater than 20% compared to a cell not expressing the CFP, wherein killing or phagocytosis activity of a cell expressing the CFP is increased by at least 20% compared to a cell not expressing the CFP, wherein killing or phagocytosis activity is measured by flow cytometry.
  • the cell expressing the CFP exhibits at least a 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 1.6-fold, 1.7-fold, 1.8-fold, 1.9-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, or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP, wherein phagocytosis is measured by flow cytometry.
  • the target cell expressing the antigen is a diseased cell.
  • the cell expressing the CFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof.
  • a cell expressing the CFP exhibits an increase in effector activity compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in cross-presentation compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP. In some embodiments, the cell expressing the CFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP.
  • the pharmaceutical composition is aqueous.
  • the pharmaceutical composition comprises one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: an CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts
  • CFP chimeric fusion protein
  • a pharmaceutical composition comprising one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: a CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR
  • the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence. In some embodiments, the first sequence and the second sequence are operatively linked by a linker sequence. In some embodiments, the linker sequence comprises a protease cleavage site In some embodiments, the protease cleavage site is a T2A cleavage site or a P2A cleavage site. [0134] In some embodiments, the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle. In some embodiments, the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle.
  • the one or more polynucleic acids comprises a first polynucleic acid molecule comprising the first sequence and a second polynucleic acid molecule comprising the second sequence.
  • the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle.
  • the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle.
  • after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject.
  • the CAR after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject.
  • the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject.
  • the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • the CAR intracellular domain further comprises a costimulatory domain.
  • the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137).
  • the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain.
  • the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89.
  • the CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain.
  • the CFP after administration of the pharmaceutical composition to a human subject the CFP is specifically expressed in myeloid cells, monocytes or macrophages of the human subject.
  • the CAR is specifically expressed in T cells of the human subject.
  • the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89.
  • the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain.
  • the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from Fc ⁇ R, Fc ⁇ R, Fc ⁇ R, CD40 or CD3zeta.
  • the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain.
  • PI3K phosphoinositide 3-kinase
  • the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4.
  • the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89.
  • compositions comprising a recombinant nucleic acid encoding a chimeric fusion protein (CFP), such as a phagocytic receptor (PR) fusion protein (PFP), a scavenger receptor (SR) fusion protein (SFP), an integrin receptor (IR) fusion protein (IFP) or a caspase- recruiting receptor (caspase-CAR) fusion protein.
  • CFP chimeric fusion protein
  • PR phagocytic receptor
  • SFP scavenger receptor
  • IR integrin receptor
  • caspase-CAR caspase- recruiting receptor
  • the extracellular domain can be fused to a hinge domain or an extracellular domain derived from a receptor, such as CD2, CD8, CD28, CD64 or CD68, a phagocytic receptor, a scavenger receptor or an integrin receptor.
  • the CFP encoded by the recombinant nucleic acid can further comprise a transmembrane domain, such as a transmembrane domain derived from CD2, CD8, CD28, CD64 or CD68, a phagocytic receptor, a scavenger receptor or an integrin receptor.
  • a CFP encoded by the recombinant nucleic acid further comprises an intracellular domain comprising an intracellular signaling domain, such as an intracellular signaling domain derived from a phagocytic receptor, a scavenger receptor or an integrin receptor.
  • an intracellular signaling domain such as an intracellular signaling domain derived from a phagocytic receptor, a scavenger receptor or an integrin receptor.
  • the intracellular domain can comprise one or more intracellular signaling domains derived from a phagocytic receptor, a scavenger receptor or an integrin receptor.
  • the intracellular domain can comprise one or more intracellular signaling domains that promote phagocytic activity, inflammatory response, nitric oxide production, integrin activation, enhanced effector cell migration (e.g., via chemokine receptor expression), antigen presentation, and/or enhanced cross presentation.
  • the CFP is a phagocytic receptor fusion protein (PFP).
  • the CFP is a phagocytic scavenger receptor fusion protein (PFP).
  • the CFP is an integrin receptor fusion protein (IFP).
  • the CFP is an inflammatory receptor fusion protein.
  • a CFP encoded by the recombinant nucleic acid further comprises an intracellular domain comprising a recruitment domain.
  • the intracellular domain can comprise one or more PI3K recruitment domains, caspase recruitment domains or caspase activation and recruitment domains (CARDs).
  • composition comprising a recombinant nucleic acid encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising a phagocytic receptor intracellular signaling domain; and an extracellular antigen binding domain specific to an antigen, e.g., an antigen of or presented on a target cell; wherein the transmembrane domain and the extracellular antigen binding domain are operatively linked such that antigen binding to the target by the extracellular antigen binding domain of the fused receptor activated in the intracellular signaling domain of the phagocytic receptor.
  • PR phagocytic or tethering receptor
  • composition comprising a recombinant nucleic acid sequence encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the CFP to the antigen of the target cell, the killing or phagocytosis activity of a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%
  • a myeloid cell such as
  • composition comprising a recombinant nucleic acid sequence encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the CFP to the antigen of the target cell, the killing or phagocytosis activity of a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold,
  • a myeloid cell such as
  • a pharmaceutical composition comprising: (a) a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage cell comprising a recombinant polynucleic acid, wherein the recombinant polynucleic acid comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising an anti-CD5 binding domain, and (ii) a transmembrane domain operatively linked to the extracellular domain; and (b) a pharmaceutically acceptable carrier; wherein the myeloid cell expresses the CFP and exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing CD5 compared to a myeloid cell not expressing the CFP.
  • a myeloid cell such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage cell comprising a recombin
  • the CD5 binding domain is a CD5 binding protein that comprises an antigen binding fragment of an antibody, an Fab fragment, an scFv domain or an sdAb domain.
  • the CD5 binding domain comprises an scFv comprising: (i) a variable heavy chain (V H ) sequence of SEQ ID NO: 1 (see, Table 4) or with at least 90% sequence identity to SEQ ID NO: 1; and (ii) a variable light chain (V L ) sequence of SEQ ID NO: 2 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 2.
  • the CD5 binding domain comprises an scFv comprising SEQ ID NO: 33 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 33.
  • the HER2 binding domain comprises an scFv comprising: (i) a variable heavy chain (V H ) sequence of SEQ ID NO: 8 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 8; and (ii) a variable light chain (VL) sequence of SEQ ID NO: 9 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 9.
  • V H variable heavy chain
  • VL variable light chain
  • the CD5 binding domain comprises an scFv comprising SEQ ID NO: 32 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 32.
  • the CFP further comprises an intracellular domain, wherein the intracellular domain comprises one or more intracellular signaling domains, and wherein a wild-type protein comprising the intracellular domain does not comprise the extracellular domain.
  • the extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the transmembrane domain and the anti-CD5 binding domain.
  • the extracellular hinge domain comprises a sequence of SEQ ID NO: 7 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 7.
  • the CFP comprises an extracellular domain fused to a transmembrane domain of SEQ ID NO: 30 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 30.
  • the CFP comprises an extracellular domain fused to a transmembrane domain of SEQ ID NO: 31 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 31.
  • the transmembrane domain comprises a CD8 transmembrane domain.
  • the transmembrane domain comprises a sequence of SEQ ID NO: 6 or 29 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 6 or 29.
  • the transmembrane domain comprises a sequence of SEQ ID NO: 18 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 18.
  • the transmembrane domain comprises a sequence of SEQ ID NO: 34 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 34.
  • the transmembrane domain comprises a sequence of SEQ ID NO: 19 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 19.
  • the CFP comprises one or more intracellular signaling domains that comprise a phagocytic signaling domain.
  • the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, FcR ⁇ , and Bai1.
  • the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, an FcR, and Bai1. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than CD3 ⁇ . In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from FcR ⁇ , FcR ⁇ or FcR ⁇ . In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from CD3 ⁇ .
  • the CFP comprises an intracellular signaling domain of any one of SEQ ID NOs: 3, 20, 27 and 28 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any one of SEQ ID NOs: 3, 20, 27 and 28.
  • the one or more intracellular signaling domains further comprises a proinflammatory signaling domain.
  • the proinflammatory signaling domain comprises a PI3-kinase (PI3K) recruitment domain.
  • the proinflammatory signaling domain comprises a sequence of SEQ ID NO: 4 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 4.
  • the proinflammatory signaling domain is derived from an intracellular signaling domain of CD40.
  • the proinflammatory signaling domain comprises a sequence of SEQ ID NO: 5 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 5.
  • the CFP comprises an intracellular signaling domain of SEQ ID NO: 21 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 21.
  • the CFP comprises an intracellular signaling domain of SEQ ID NO: 23 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 23.
  • the CFP comprises a sequence of SEQ ID NO: 14 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 14.
  • the CFP comprises a sequence of SEQ ID NO: 15 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 15.
  • the CFP comprises a sequence of SEQ ID NO: 16 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 16.
  • the CFP comprises a sequence of SEQ ID NO: 24 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 24.
  • the CFP comprises a sequence of SEQ ID NO:25 or with at least 70%, 75%, 80%, 85%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 25.
  • the CFP comprises: (a) an extracellular domain comprising: (i) a scFv that specifically binds CD5, and (ii) a hinge domain derived from CD8; a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68; (b) a CD8 transmembrane domain, a CD28 transmembrane domain, a CD2 transmembrane domain or a CD68 transmembrane domain; and (c) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from FcR ⁇ , FcR ⁇ or FcR ⁇ , and (ii) a second intracellular signaling domain: (A) comprising a PI3K recruitment domain, or (B) derived from CD40.
  • an extracellular domain comprising: (i) a scFv that specifically binds CD5, and (ii) a hinge domain
  • the CFP comprises as an alternative (c) to the above: an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from a phagocytic receptor intracellular domain, and (ii) a second intracellular signaling domain derived from a scavenger receptor phagocytic receptor intracellular domain comprising: (A) comprising a PI3K recruitment domain, or (B) derived from CD40.
  • Exemplary scavenger receptors from which an intracellular signaling domain may be derived may be found in Table 2.
  • the CFP comprises and intracellular signaling domain derived from an intracellular signaling domain of an innate immune receptor.
  • the recombinant polynucleic acid is an mRNA. In some embodiments, the recombinant polynucleic acid is a circRNA. In some embodiments, the recombinant polynucleic acid is a viral vector. In some embodiments, the recombinant polynucleic acid is delivered via a viral vector.
  • the nanoparticle delivery vehicle comprises a lipid nanoparticle. In some embodiments, the lipid nanoparticle comprises a polar lipid.
  • the lipid nanoparticle comprises a non-polar lipid. In some embodiments, the lipid nanoparticle is from 100 to 300 nm in diameter. [0159] In some embodiments, the first antigen binding domain and the second antigen binding domain bind to the same antigen. In some embodiments, the first antigen binding domain and the second antigen binding domain bind to different antigens. In some embodiments, the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75.
  • an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75.
  • the myeloid cell is a CD14+ cell, a CD14+/CD16- cell, a CD14+/CD16+ cell, a CD14-/CD16+ cell, CD14-/CD16- cell, a dendritic cell, an M0 macrophage, an M2 macrophage, an M1 macrophage or a mosaic myeloid cell/macrophage/dendritic cell.
  • the T cell is a T cell precursor.
  • the T cell is an undifferentiated and/or unpolarized.
  • the T cell is a CD4 T cells.
  • T cell is a CD8 T cell.
  • T cell is gamma delta T cell.
  • the cell is a NK T cell.
  • a method of treating cancer in a human subject in need thereof comprising administering a pharmaceutical composition to the human subject, the pharmaceutical composition comprising: (a) a myeloid cell comprising a recombinant polynucleic acid sequence, wherein the polynucleic acid sequence comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising an anti-CD5 binding domain, and (ii) a transmembrane domain operatively linked to the extracellular domain; and (b) a pharmaceutically acceptable carrier; wherein the myeloid cell expresses the CFP.
  • CFP chimeric fusion protein
  • the cancer upon binding of the CFP to CD5 expressed by a target cancer cell of the subject killing or phagocytosis activity of the myeloid cell is increased by greater than 20% compared to a myeloid cell not expressing the CFP. In some embodiments, growth of a tumor is inhibited in the human subject.
  • the cancer is a CD5+ cancer. In some embodiments, the cancer is leukemia, T cell lymphoma, or B cell lymphoma.
  • the anti-CD5 binding domain is a CD5 binding protein that comprises an antigen binding fragment of an antibody, an scFv domain, an Fab fragment, or an sdAb domain.
  • the anti-CD5 binding domain is a protein or fragment thereof that binds to CD5, such as a ligand of CD5 (e.g., a natural ligand of CD5).
  • the CFP further comprises an intracellular domain, wherein the intracellular domain comprises one or more intracellular signaling domains, wherein the one or more intracellular signaling domains comprises a phagocytosis signaling domain and wherein a wild-type protein comprising the intracellular domain does not comprise the extracellular domain.
  • the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, FcR ⁇ and Bai1.
  • the phagocytosis signaling domain comprises an intracellular signaling domain derived from FcR ⁇ , FcR ⁇ or FcR ⁇ .
  • the one or more intracellular signaling domains further comprises a proinflammatory signaling domain.
  • the proinflammatory signaling domain comprises a PI3-kinase (PI3K) recruitment domain.
  • the transmembrane domain comprises a CD8 transmembrane domain.
  • the extracellular domain comprises a hinge domain derived from CD8, a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68.
  • the CFP comprises: (a) an extracellular domain comprising: (i) a scFv that specifically binds CD5, and (ii) a hinge domain derived from CD8, a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68; (b) a CD8 transmembrane domain, a CD28 transmembrane domain, a CD2 transmembrane domain or a CD68 transmembrane domain; and (c) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from FcR ⁇ or FcR ⁇ , and (ii) a second intracellular signaling domain that: (A) comprises a PI3K recruitment domain, or (B) is derived from CD40.
  • the recombinant nucleic acid is mRNA or circRNA.
  • the myeloid cell is a CD14+ cell, a CD14+/CD16- cell, a CD14+/CD16+ cell, a CD14-/CD16+ cell, CD14-/CD16- cell, a dendritic cell, an M0 macrophage, an M2 macrophage, an M1 macrophage or a mosaic myeloid cell/macrophage/dendritic cell.
  • the method further comprises administering an additional therapeutic agent selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity, an agent that promotes sequestration of lymphocytes in primary and/or secondary lymphoid organs, an agent that increases concentration of na ⁇ ve T cells and central memory T cells in secondary lymphoid organs, and any combination thereof.
  • an additional therapeutic agent selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits
  • the myeloid cell further comprises: (a) an endogenous peptide or protein that dimerizes with the CFP, (b) a non-endogenous peptide or protein that dimerizes with the CFP; and/or (c) a second recombinant polynucleic acid sequence, wherein the second recombinant polynucleic acid sequence comprises a sequence encoding a peptide or protein that interacts with the CFP; wherein the dimerization or the interaction potentiates phagocytosis by the myeloid cell expressing the CFP as compared to a myeloid cell that does not express the CFP.
  • the myeloid cell exhibits (i) an increase in effector activity, cross- presentation, respiratory burst, ROS production, iNOS production, inflammatory mediators, extra- cellular vesicle production, phosphatidylinositol 3,4,5-trisphosphate production, trogocytosis with the target cell expressing the antigen, resistance to CD47 mediated inhibition of phagocytosis, resistance to LILRB1 mediated inhibition of phagocytosis, or any combination thereof; and/or (ii) an increase in expression of a IL-1, IL3, IL-6, IL-10, IL-12, IL-13, IL-23, TNF ⁇ , a TNF family of cytokines, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL-17, IP-10, RANTES, an interferon, MHC class I protein,
  • the intracellular signaling domain is derived from a phagocytic or tethering receptor or wherein the intracellular signaling domain comprises a phagocytosis activation domain.
  • the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1.
  • the intracellular signaling domain is derived from a protein, such as receptor (e.g., a phagocytic receptor), selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc ⁇ receptor I, CR1, CD35, CD3 ⁇ , a complement receptor, CR3, CR4, Tim-1, Tim-4 and CD169.
  • receptor e.g., a phago
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. [0174] In some embodiments, the intracellular signaling domain is derived from an ITAM domain containing receptor.
  • composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR ⁇ , or Bai1.
  • a recombinant nucleic acid encoding a CFP such as a phagocytic or tethering receptor (PR) fusion protein (PFP)
  • PR phagocytic or tethering receptor
  • the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP.
  • the CFP functionally incorporates into a cell membrane of a cell when the CFP is expressed in the cell.
  • the killing activity of a cell expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 5.5-fold, 6- fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold,-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30-fold, 40-fold, 50-fold, 75-fold, or 100-fold compared to a cell not expressing the CFP.
  • the intracellular signaling domain is derived from a receptor, such as a phagocytic receptor, selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc ⁇ receptor I, CR1, CD35, CD3 ⁇ , CR3, CR4, Tim-1, Tim-4 and CD169.
  • a receptor such as a phagocytic receptor, selected from the group consisting of TN
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain.
  • a composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a receptor, such as a phagocytic receptor, selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB
  • a receptor such as a phag
  • the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP.
  • the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR ⁇ , or Bai1.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain.
  • the intracellular signaling domain comprises a PI3K recruitment domain, such as a PI3K recruitment domain derived from CD19.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain.
  • composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain.
  • a CFP such as a phagocytic or tethering receptor (PR) fusion protein (PFP)
  • PR phagocytic or tethering receptor
  • an engineered CFP such as a phagocytic receptor fusion protein
  • a cell such as a myeloid cell
  • an engineered myeloid cell that can target a target cell, such as a diseased cell.
  • a target cell is, for example, a cancer cell.
  • the engineered myeloid cell after engulfment of a cancer cell may present a cancer antigen on its cell surface to activate a T cell.
  • An “antigen” is a molecule capable of stimulating an immune response.
  • T helper (TH) cells helper T lymphocytes
  • CTLs cytotoxic T lymphocytes
  • MHC proteins such as class I or class II MHC proteins
  • the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP.
  • the CFP functionally incorporates into a cell membrane of a cell when the CFP is expressed in the cell.
  • the killing activity of a cell expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 5.5-fold, 6- fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold,-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30-fold, 40-fold, 50-fold, 75-fold, or 100-fold compared to a cell not expressing the CFP.
  • the target cell expressing the antigen is a cancer cell. In some embodiments, the target cell expressing the antigen is at least 0.8 microns in diameter. [0185] In some embodiments, a cell expressing the CFP exhibits an increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits at least a 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10- fold, 20-fold, 30-fold or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL- 23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP- 10, RANTES, an interferon and combinations thereof.
  • a cell expressing the CFP exhibits an increase in effector activity compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in cross-presentation compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TRAIL/TNF Family death receptors compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in expression of B7-H2 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of LIGHT compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of HVEM compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD40 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TL1A compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in expression of 41BBL compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of OX40L compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of GITRL death receptors compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD30L compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TIM4 compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in expression of TIM1 ligand compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of SLAM compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD48 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD58 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD155 compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in expression of CD112 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of PDL1 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of B7-DC compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in respiratory burst compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in ROS production compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in iNOS production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in iNOS production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in extra-cellular vesicle production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in trogocytosis with a target cell expressing the antigen compared to a cell not expressing the CFP.
  • a cell expressing the CFP exhibits an increase in resistance to CD47 mediated inhibition of phagocytosis compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production. [0186] In some embodiments, the extracellular domain of a CFP comprises an Ig binding domain.
  • the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, FcR ⁇ I, FcR ⁇ IIA, FcR ⁇ IIB, FcR ⁇ IIC, FcR ⁇ IIIA, FcR ⁇ IIIB, FcRn, TRIM21, FcRL5 binding domain.
  • the extracellular domain of a CFP comprises an FcR extracellular domain.
  • the extracellular domain of a CFP comprises an FcR ⁇ , FcR ⁇ , FcR ⁇ or FcR ⁇ extracellular domain.
  • the extracellular domain comprises an FcR ⁇ (FCAR) extracellular domain.
  • the extracellular domain comprises an FcR ⁇ extracellular domain. In some embodiments, the extracellular domain comprises an FCER1A extracellular domain. In some embodiments, the extracellular domain comprises an FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, or FCGR3B extracellular domain. In some embodiments, the extracellular domain comprises an integrin domain or an integrin receptor domain.
  • the extracellular domain comprises one or more integrin ⁇ 1, ⁇ 2, ⁇ IIb, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, ⁇ 10, ⁇ 11, ⁇ D, ⁇ E, ⁇ L, ⁇ M, ⁇ V, ⁇ X, ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, or ⁇ 8 domains.
  • the CFP further comprises an extracellular domain operatively linked to the transmembrane domain and the extracellular antigen binding domain.
  • the extracellular domain further comprises an extracellular domain of a receptor, a hinge, a spacer and/or a linker.
  • the extracellular domain comprises an extracellular portion of a phagocytic receptor. In some embodiments, the extracellular portion of the CFP is derived from the same receptor as the receptor from which the intracellular signaling domain is derived. In some embodiments, the extracellular domain comprises an extracellular domain of a scavenger receptor. In some embodiments, the extracellular domain comprises an immunoglobulin domain. In some embodiments, the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region. In some embodiments, the extracellular domain comprises a phagocytic engulfment domain. In some embodiments, the extracellular domain comprises a structure capable of multimeric assembly.
  • the extracellular domain comprises a scaffold for multimerization. In some embodiments, the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length. In some embodiments, the extracellular antigen binding domain specifically binds to the antigen of a target cell. In some embodiments, the extracellular antigen binding domain comprises an antibody domain.
  • the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof.
  • the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor.
  • the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen.
  • the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen.
  • the antigen is a cancer associated antigen, a lineage associated antigen, a pathogenic antigen or an autoimmune antigen.
  • the antigen comprises a viral antigen.
  • the antigen is a T lymphocyte antigen.
  • the antigen is an extracellular antigen.
  • the antigen is an intracellular antigen.
  • the antigen is selected from the group consisting of an antigen from Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B- Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2, a Natural Killer Group 2
  • the antigen is an antigen of a protein selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, and CD56.
  • the antigen is an ovarian cancer antigen or a T lymphoma antigen.
  • the antigen is an antigen of an integrin receptor.
  • the antigen is an antigen of an integrin receptor or integrin selected from the group consisting of ⁇ 1, ⁇ 2, ⁇ IIb, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, ⁇ 10, ⁇ 11, ⁇ D, ⁇ E, ⁇ L, ⁇ M, ⁇ V, ⁇ X, ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, and ⁇ 8.
  • the antigen is an antigen of an integrin receptor ligand.
  • the antigen is an antigen of fibronectin, vitronectin, collagen, or laminin.
  • the antigen binding domain can bind to two or more different antigens.
  • the antigen binding domain comprises an autoantigen or fragment thereof, such as Dsg1 or Dsg3.
  • the extracellular antigen binding domain comprises a receptor domain or an antibody domain wherein the antibody domain binds to an auto antigen, such as Dsg1 or Dsg3.
  • the transmembrane domain and the extracellular antigen binding domain are operatively linked through a linker.
  • the transmembrane domain and the extracellular antigen binding domain are operatively linked through a linker such as a hinge region of CD8 ⁇ , IgG1 or IgG4.
  • the extracellular domain comprises a multimerization scaffold.
  • the transmembrane domain comprises a CD8 transmembrane domain.
  • the transmembrane domain comprises a CD28 transmembrane domain.
  • the transmembrane domain comprises a CD68 transmembrane domain.
  • the transmembrane domain comprises a CD2 transmembrane domain.
  • the transmembrane domain comprises an FcR transmembrane domain.
  • the transmembrane domain comprises an FcR ⁇ transmembrane domain.
  • the transmembrane domain comprises an FcR ⁇ transmembrane domain. In some embodiments, the transmembrane domain comprises an FcR ⁇ transmembrane domain. In some embodiments, the transmembrane domain comprises an FcR ⁇ transmembrane domain. In some embodiments, the transmembrane domain comprises a transmembrane domain from a syntaxin, such as syntaxin 3 or syntaxin 4 or syntaxin 5. In some embodiments, the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the CFP is expressed in a cell.
  • the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an endogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain is derived from a protein that is different than the protein from which the intracellular signaling domain is derived.
  • the transmembrane domain is derived from a protein that is different than the protein from which the extracellular domain is derived. In some embodiments, the transmembrane domain comprises a transmembrane domain of a phagocytic receptor. In some embodiments, the transmembrane domain and the extracellular domain are derived from the same protein. In some embodiments, the transmembrane domain is derived from the same protein as the intracellular signaling domain. In some embodiments, the recombinant nucleic acid encodes a DAP12 recruitment domain. In some embodiments, the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12.
  • the transmembrane domain is at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. In some embodiments, the transmembrane domain is at most 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length.
  • the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor. In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR ⁇ , or Bai1.
  • the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I, CR1, CD35, CD3 ⁇ , CR3, CR4, Tim-1, Tim-4 and CD169.
  • a phagocytic receptor selected from the group consisting of TNFR1, MDA5,
  • the intracellular signaling domain comprises a PI3K recruitment domain. In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a scavenger receptor. In some embodiments, the intracellular domain comprises a CD47 inhibition domain. In some embodiments, the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain. In some embodiments, the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the PFP.
  • the intracellular domain comprises an F- actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain.
  • the intracellular domain comprises a phosphatase inhibition domain.
  • the intracellular domain comprises an ARP2/3 inhibition domain.
  • the intracellular domain comprises at least one ITAM domain. In some embodiments, the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more ITAM domains.
  • the intracellular domain comprises at least one ITAM domain select from an ITAM domain of CD3 zeta, CD3 epsilon, CD3 gamma, CD3 delta, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278, CD66d, functional fragments thereof, and amino acid sequences thereof having at least one but not more than 20 modifications thereto.
  • ITAM domain select from an ITAM domain of CD3 zeta, CD3 epsilon, CD3 gamma, CD3 delta
  • the at least one ITAM domain comprises a Src-family kinase phosphorylation site. In some embodiments, the at least one ITAM domain comprises a Syk recruitment domain. In some embodiments, the intracellular domain comprises an F-actin depolymerization activation domain. In some embodiments, the intracellular domain lacks enzymatic activity. [0195] In some embodiments, the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. In some embodiments, the intracellular domain does not comprise a domain derived from a MerTK intracellular domain. In some embodiments, the intracellular domain does not comprise a domain derived from a TLR4 intracellular domain.
  • the intracellular domain comprises a CD47 inhibition domain.
  • the intracellular signaling domain comprises a domain that activates integrin, such as the intracellular region of PSGL-1.
  • the intracellular signaling domain comprises a domain that activates Rap1 GTPase, such as that from EPAC and C3G.
  • the intracellular signaling domain is derived from paxillin.
  • the intracellular signaling domain activates focal adhesion kinase.
  • the intracellular signaling domain is derived from a single phagocytic receptor.
  • the intracellular signaling domain is derived from a single scavenger receptor.
  • the intracellular domain comprises a phagocytosis enhancing domain.
  • the intracellular domain comprises a pro-inflammatory signaling domain.
  • the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain.
  • the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain.
  • the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, STING, an NLRP family member, NLRP1-14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40, Tank1-binding kinase (TBK), a caspase domain, a procaspase binding domain or any combination thereof.
  • the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a connexin (Cx) protein.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Cx43, Cx46, Cx37, Cx40, Cx33, Cx50, Cx59, Cx62, Cx32, Cx26, Cx31, Cx30.3, Cx31.1, Cx30, Cx25, Cx45, Cx47, Cx31.3, Cx36, Cx31.9, Cx39, Cx40.1 or Cx23.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Cx43.
  • the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a SIGLEC protein.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Siglec-1 (Sialoadhesin), Siglec-2 (CD22), Siglec-3 (CD33), Siglec-4 (MAG), Siglec-5, Siglec-6, Siglec-7, Siglec-8, Siglec-9, Siglec-10, Siglec-11, Siglec-12, Siglec-13, Siglec-14, Siglec-15, Siglec- 16 or Siglec-17.
  • Siglec-1 Sialoadhesin
  • Siglec-2 CD22
  • Siglec-3 CD33
  • Siglec-4 MAG
  • Siglec-5 Siglec-6, Siglec-7, Siglec-8, Siglec-9, Siglec-10, Siglec-11, Siglec-12, Siglec-13, Siglec-14, Siglec-15, Siglec-
  • the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a C-type lectin protein.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from a mannose receptor protein.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an asialoglycoprotein receptor protein.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from macrophage galactose-type lectin (MGL), DC-SIGN (CLEC4L), Langerin (CLEC4K), Myeloid DAP12 associating lectin (MDL)-1 (CLEC5A), a DC associated C type lectin 1 (Dectin1) subfamily protein, dectin 1/CLEC7A, DNGR1/CLEC9A, Myeloid C type lectin like receptor (MICL) (CLEC12A), CLEC2 (CLEC1B), CLEC12B, a DC immunoreceptor (DCIR) subfamily protein, DCIR/CLEC4A, Dectin 2/CLEC6A, Blood DC antigen 2 (BDCA2) ( CLEC4C), Mincle (macrophage inducible C type lectin) (CLEC4E), a NOD-like receptor protein, NOD-like receptor, MHC Class II transactivator (CI
  • the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a cell adhesion molecule.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an IgCAMs, a cadherin, an integrin, a C-type of lectin-like domains protein (CTLD) and/or a proteoglycan molecule.
  • a signaling domain such as an intracellular signaling domain, derived from an IgCAMs, a cadherin, an integrin, a C-type of lectin-like domains protein (CTLD) and/or a proteoglycan molecule.
  • CTL C-type of lectin-like domains protein
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an E-cadherin, a P-cadherin, an N-cadherin, an R- cadherin, a B-cadherin, a T-cadherin, or an M-cadherin.
  • the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from a selectin, such as an E-selectin, an L-selectin or a P-selectin.
  • the CFP does not comprise a full length intracellular signaling domain.
  • the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length.
  • the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • the FcR ⁇ chain or the FcR ⁇ chain forms a complex with FcR ⁇ when expressed in a cell.
  • the FcR ⁇ chain or FcR ⁇ chain forms a complex with endogenous FcR ⁇ when expressed in a cell.
  • the FcR ⁇ chain or the FcR ⁇ chain does not incorporate into a cell membrane of a cell that does not express FcR ⁇ .
  • the CFP does not comprise an FcR ⁇ chain intracellular signaling domain.
  • the CFP does not comprise an FcR ⁇ chain intracellular signaling domain.
  • the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain.
  • the TREM is TREM1, TREM 2 or TREM 3.
  • the recombinant nucleic acid comprises a sequence encoding a pro- inflammatory polypeptide.
  • the composition further comprises a proinflammatory nucleotide or a nucleotide in the recombinant nucleic acid, for example, an ATP, ADP, UTP, UDP, and/or UDP-glucose.
  • the composition further comprises a pro-inflammatory polypeptide.
  • the pro-inflammatory polypeptide is a chemokine, cytokine.
  • the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL- 12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, and interferon.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, and interferon.
  • the myeloid cells are specifically targeted for delivery. Myeloid cells can be targeted using specialized biodegradable polymers, such as PLGA (poly(lactic-co- glycolic) acid and/or polyvinyl alcohol (PVA).
  • one or more compounds can be selectively incorporated in such polymeric structures to affect the myeloid cell function.
  • the targeting structures are multilayered, e.g., of one or more PLGA and one or more PVA layers.
  • the targeting structures are assembled in an order for a layered activity.
  • the targeted polymeric structures are organized in specific shaped components, such as labile structures that can adhere to a myeloid cell surface and deliver one or more components such as growth factors and cytokines, such as to maintain the myeloid cell in a microenvironment that endows a specific polarization.
  • the polymeric structures are such that they are not phagocytosed by the myeloid cell, but they can remain adhered on the surface.
  • the one or more growth factors may be M1 polarization factors, such as a cytokine.
  • the one or more growth factors may be an M2 polarization factor, such as a cytokine.
  • the one or more growth factors may be a macrophage activating cytokine, such as IFN ⁇ .
  • the polymeric structures are capable of sustained release of the one or more growth factors in an in vivo environment, such as in a solid tumor.
  • the recombinant nucleic acid comprises a sequence encoding a homeostatic regulator of inflammation.
  • the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA.
  • the sequence in the UTR is a sequence that binds to an RNA binding protein.
  • translation is inhibited or prevented upon binding of the RNA binding protein to the sequence in an untranslated region (UTR).
  • the sequence in the UTR comprises a consensus sequence of WWWU(AUUUA)UUUW, wherein W is A or U.
  • the recombinant nucleic acid is expressed on a bicistronic vector.
  • the target cell is a mammalian cell. In some embodiments, the target cell is a human cell. In some embodiments, the target cell comprises a cell infected with a pathogen. In some embodiments, the target cell is a cancer cell. In some embodiments, the target cell is a cancer cell that is a lymphocyte. In some embodiments, the target cell is a cancer cell that is an ovarian cancer cell. In some embodiments, the target cell is a cancer cell that is a breast cell. In some embodiments, the target cell is a cancer cell that is a pancreatic cell. In some embodiments, the target cell is a cancer cell that is a glioblastoma cell.
  • the recombinant nucleic acid is DNA. In some embodiments, the recombinant nucleic acid is RNA. In some embodiments, the recombinant nucleic acid is mRNA. In some embodiments, the recombinant nucleic acid is an unmodified mRNA. In some embodiments, the recombinant nucleic acid is a modified mRNA. In some embodiments, the recombinant nucleic acid is a circRNA. In some embodiments, the recombinant nucleic acid is a tRNA. In some embodiments, the recombinant nucleic acid is a microRNA.
  • a vector comprising a recombinant nucleic acid sequence encoding a CFP described herein.
  • the vector is viral vector.
  • the viral vector is retroviral vector or a lentiviral vector.
  • the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides.
  • the vector is polycistronic.
  • each of the at least one nucleic acid sequence is operably linked to a separate promoter.
  • the vector further comprises one or more internal ribosome entry sites (IRESs).
  • the vector further comprises a 5’UTR and/or a 3’UTR flanking the at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector further comprises one or more regulatory regions.
  • a cell comprising a composition described herein, a vector described herein or a polypeptide described herein. In some embodiments, the cell is a phagocytic cell.
  • the cell is a stem cell derived cell, a myeloid cell, a macrophage, a dendritic cell, a lymphocyte, a mast cell, a monocyte, a neutrophil, a microglia, or an astrocyte.
  • the cell is an autologous cell.
  • the cell is an allogeneic cell.
  • the cell is an M1 cell.
  • the cell is an M2 cell.
  • the cell is an M1 macrophage cell.
  • the cell is an M2 macrophage cell.
  • the cell is an M1 myeloid cell.
  • the cell is an M2 myeloid cell.
  • compositions described herein comprising a composition described herein, such as a recombinant nucleic acid described herein, a vector described herein, a polypeptide described herein or a cell described herein; and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition further comprises an additional therapeutic agent.
  • the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • the pharmaceutically acceptable excipient comprises serum free media, a lipid, or a nanoparticle.
  • the disease is cancer.
  • the cancer is a solid cancer.
  • the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer.
  • the cancer is a liquid cancer.
  • the liquid cancer is leukemia or a lymphoma.
  • the liquid cancer is a T cell lymphoma.
  • the disease is a T cell malignancy.
  • the method further comprises administering an additional therapeutic agent to the subject.
  • the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • administering comprises infusing or injecting. In some embodiments, administering comprises administering directly to the solid cancer. In some embodiments, administering comprises a circRNA-based delivery procedure, anon-particle encapsulated mRNA-based delivery procedure, an mRNA-based delivery procedure, viral-based delivery procedure, particle-based delivery procedure, liposome-based delivery procedure, or an exosome-based delivery procedure. In some embodiments, a CD4+ T cell response or a CD8+ T cell response is elicited in the subject.
  • Also provided herein is a method of preparing a cell, the method comprising contacting a cell with a composition described herein, a vector described herein or a polypeptide described herein.
  • contacting comprises transducing.
  • contacting comprises chemical transfection, electroporation, nucleofection, or viral infection or transduction.
  • Also provided herein is a method of preparing a pharmaceutical composition comprising contacting a lipid to a composition described herein or a vector described herein.
  • contacting comprises forming a lipid nanoparticle.
  • Also provided herein is a method of preparing a pharmaceutical composition comprising contacting an antibody to a composition described herein or the vector described herein.
  • contacting comprises forming a lipid nanoparticle.
  • An “agent” can refer to any cell, small molecule chemical compound, antibody or fragment thereof, nucleic acid molecule, or polypeptide.
  • An “alteration” or “change” can refer to an increase or decrease. For example, an alteration can be an increase or decrease of 1%, 2%, 3%, 4%, 5%, 10%, 20%, 30%, or by 40%, 50%, 60%, or even by as much as 70%, 75%, 80%, 90%, or 100%.
  • an alteration can be an increase or decrease of 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 30-fold, or by 40-fold, 50-fold, 60- fold, or even by as much as 70-fold, 75-fold, 80-fold, 90-fold, or 100-fold.
  • An “antigen presenting cell” or “APC” as used herein includes professional antigen presenting cells (e.g., B lymphocytes, macrophages, monocytes, dendritic cells, Langerhans cells), as well as other antigen presenting cells (e.g., keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes, thymic epithelial cells, thyroid epithelial cells, glial cells (brain), pancreatic beta cells, and vascular endothelial cells).
  • professional antigen presenting cells e.g., B lymphocytes, macrophages, monocytes, dendritic cells, Langerhans cells
  • other antigen presenting cells e.g., keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes, thymic epithelial cells, thyroid epithelial cells, glial cells (bra
  • An APC can express Major Histocompatibility complex (MHC) molecules and can display antigens complexed with MHC on its surface which can be recognized by T cells and trigger T cell activation and an immune response.
  • MHC Major Histocompatibility complex
  • Professional antigen- presenting cells notably dendritic cells, play a key role in stimulating naive T cells.
  • APCs can also cross-present peptide antigens by processing exogenous antigens and presenting the processed antigens on class I MHC molecules.
  • Antigens that give rise to proteins that are recognized in association with class I MHC molecules are generally proteins that are produced within the cells, and these antigens are processed and associate with class I MHC molecules.
  • a “biological sample” can refer to any tissue, cell, fluid, or other material derived from an organism.
  • the term “epitope” can refer to any protein determinant, such as a sequence or structure or amino acid residues, capable of binding to an antibody or binding fragment thereof, a T cell receptor, and/or an antibody-like molecule. Epitopic determinants typically consist of chemically active surface groups of molecules such as amino acids or sugar side chains and generally have specific three dimensional structural characteristics as well as specific charge characteristics.
  • a “T cell epitope” can refer to peptide or peptide-MHC complex recognized by a T cell receptor.
  • Chimeric fusion proteins may be used to designate a chimeric receptor made via recombinant DNA technology, and is exclusively detailed in the specification.
  • a CFP may be termed a phagocytic receptor fusion protein (PFP) and the terms can be used interchangeably.
  • a CAR construct may comprise a CD3z domain and a CFP construct may comprise a CD89 transmembrane domain.
  • An engineered cell such as an engineered myeloid cell, can refer to a cell that has at least one exogenous nucleic acid sequence in the cell, even if transiently expressed. Expressing an exogenous nucleic acid may be performed by various methods described elsewhere, and encompasses methods known in the art.
  • the present disclosure relates to preparing and using engineered cells, for example, engineered myeloid cells, such as engineered phagocytic cells.
  • the present disclosure relates to, inter alia, an engineered cell comprising an exogenous nucleic acid encoding, for example, a chimeric fusion protein (CFP).
  • CFP chimeric fusion protein
  • immune response includes, but is not limited to, T cell mediated, NK cell mediated and/or B cell mediated immune responses. These responses may be influenced by modulation of T cell costimulation and NK cell costimulation.
  • Exemplary immune responses include T cell responses, e.g., cytokine production, and cellular cytotoxicity.
  • immune responses include immune responses that are indirectly affected by NK cell activation, B cell activation and/or T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., macrophages.
  • Immune responses include adaptive immune responses. The adaptive immune system can react to foreign molecular structures, such as antigens of an intruding organism.
  • the adaptive immune system is highly specific to a pathogen. Adaptive immunity can also provide long-lasting protection. Adaptive immune reactions include humoral immune reactions and cell-mediated immune reactions. In humoral immune reactions, antibodies secreted by B cells into bodily fluids bind to pathogen-derived antigens leading to elimination of the pathogen through a variety of mechanisms, e.g. complement-mediated lysis. In cell-mediated immune reactions, T cells capable of destroying other cells are activated. For example, if proteins associated with a disease are present in a cell, they can be fragmented proteolytically to peptides within the cell.
  • a “ligand” can refer to a molecule which is capable of binding or forming a complex with another molecule, such as a receptor.
  • a ligand can include, but is not limited to, a protein, a glycoprotein, a carbohydrate, a lipoprotein, a hormone, a fatty acid, a phospholipid, or any component that binds to a receptor.
  • a receptor has a specific ligand.
  • a receptor may have promiscuous binding to a ligand, in which case it can bind to several ligands that share at least a similarity in structural configuration, charge distribution or any other physicochemical characteristic.
  • a ligand may be a biomolecule.
  • a ligand may be an abiotic material.
  • a ligand may be a negative charged particle that is a ligand for scavenger receptor MARCO.
  • a ligand may be TiO 2 , which is a ligand for the scavenger receptor SRA1.
  • MHC major histocompatibility complex
  • MHC molecule or MHC protein
  • HLA complex human leukocyte antigen (HLA)
  • HLA molecule or HLA protein
  • HLA proteins can be classified as HLA class I or HLA class II. The structures of the proteins of the two HLA classes are very similar; however, they have very different functions.
  • Class I HLA proteins are present on the surface of almost all cells of the body, including most tumor cells. Class I HLA proteins are loaded with antigens that usually originate from endogenous proteins or from pathogens present inside cells, and are then presented to na ⁇ ve or cytotoxic T-lymphocytes (CTLs). HLA class II proteins are present on antigen presenting cells (APCs), including but not limited to dendritic cells, B cells, and macrophages. They mainly present peptides which are processed from external antigen sources, e.g. outside of cells, to helper T cells.
  • APCs antigen presenting cells
  • phagocytes such as macrophages and immature dendritic cells can take up entities by phagocytosis into phagosomes – though B cells exhibit the more general endocytosis into endosomes – which fuse with lysosomes whose acidic enzymes cleave the uptaken protein into many different peptides.
  • Autophagy is another source of HLA class II peptides.
  • the most studied subclass II HLA genes are: HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, and HLA-DRB1.
  • HLA class II molecules are typically heterodimers of ⁇ -and ⁇ -chains that interact to form a peptide- binding groove that is more open than class I peptide-binding grooves.
  • HLA alleles are typically expressed in codominant fashion.
  • each person carries 2 alleles of each of the 3 class I genes, (HLA-A, HLA-B and HLA-C) and so can express six different types of class II HLA.
  • each person inherits a pair of HLA-DP genes (DPA1 and DPB1, which encode ⁇ and ⁇ chains), HLA-DQ (DQA1 and DQB1, for ⁇ and ⁇ chains), one gene HLA-DR ⁇ (DRA1), and one or more genes HLA-DR ⁇ (DRB1 and DRB3, -4 or- 5).
  • HLA ⁇ DRB1 has more than nearly 400 known alleles.
  • HLA genes are highly polymorphic; many different alleles exist in the different individuals inside a population. Genes encoding HLA proteins have many possible variations, allowing each person’s immune system to react to a wide range of foreign invaders. Some HLA genes have hundreds of identified versions (alleles), each of which is given a particular number.
  • the class I HLA alleles are HLA-A*02:01, HLA-B*14:02, HLA- A*23:01, HLA-E*01:01 (non-classical).
  • class II HLA alleles are HLA- DRB*01:01, HLA-DRB*01:02, HLA-DRB*11:01, HLA-DRB*15:01, and HLA-DRB*07:01.
  • a “myeloid cell” can refer broadly to cells of the myeloid lineage of the hematopoietic cell system, and can exclude, for example, the lymphocytic lineage.
  • Myeloid cells comprise, for example, cells of the granulocyte lineage and monocyte lineages. Myeloid cells are differentiated from common progenitors derived from the hematopoietic stem cells in the bone marrow.
  • myeloid cells may be characterized as cells having a level of plasticity, which may be described as the ability to further differentiate into terminal cell types based on extracellular and intracellular stimuli.
  • Myeloid cells can be rapidly recruited into local tissues via various chemokine receptors on their surface. Myeloid cells are responsive to various cytokines and chemokines.
  • a myeloid cell may be a cell that originates in the bone marrow from a hematopoietic stem cell under the influence of one or more cytokines and chemokines, such as G- CSF, GM-CSF, Flt3L, CCL2, VEGF and S100A8/9.
  • the myeloid cell is a precursor cell.
  • the myeloid cell may be a cell having characteristics of a common myeloid progenitor, or a granulocyte progenitor, a myeloblast cell, or a monocyte-dendritic cell progenitor or a combination thereof.
  • a myeloid can include a granulocyte or a monocyte or a precursor cell thereof.
  • a myeloid can include an immature granulocyte, an immature monocyte, an immature macrophage, an immature neutrophil, and an immature dendritic cell.
  • a myeloid can include a monocyte or a pre-monocytic cell or a monocyte precursor.
  • a myeloid cell as used herein may refer to a monocyte having an M0 phenotype, an M1 phenotype or an M2 phenotype.
  • a myeloid can include a dendritic cell (DC), a mature DC, a monocyte derived DC, a plasmacytoid DC, a pre-dendritic cell, or a precursor of a DC.
  • a myeloid can include a neutrophil, which may be a mature neutrophil, a neutrophil precursor, or a polymorphonucleocyte (PMN).
  • a myeloid can include a macrophage, a monocyte-derived macrophage, a tissue macrophage, a macrophage of an M0, an M1 or an M2 phenotype.
  • a myeloid can include a tumor infiltrating monocyte (TIM).
  • a myeloid can include a tumor associated monocyte (TAM).
  • a myeloid can include a myeloid derived suppressor cell (MDSC).
  • a myeloid can include a tissue resident macrophage.
  • a myeloid can include a tumor associated DC (TADC).
  • TADC tumor associated DC
  • a myeloid cell may express one or more cell surface markers, for example, CD11b, CD14, CD15, CD16, CD38, CCR5, CD66, Lox-1, CD11c, CD64, CD68, CD163, CCR2, CCR5, HLA-DR, CD1c, CD83, CD141, CD209, MHC-II, CD123, CD303, CD304, a SIGLEC family protein and a CLEC family protein.
  • a myeloid cell may be characterized by a high or a low expression of one or more of cell surface markers, for example, CD11b, CD14, CD15, CD16, CD66, Lox-1, CD11c, CD64, CD68, CD163, CCR2, CCR5, HLA-DR, CD1c, CD83, CD141, CD209, MHC-II, CD123, CD303, CD304 or a combination thereof.
  • “Phagocytosis” is used interchangeably with “engulfment” and can refer to a process by which a cell engulfs a particle, such as a cancer cell or an infected cell. This process can give rise to an internal compartment (phagosome) containing the particle.
  • This process can be used to ingest and or remove a particle, such as a cancer cell or an infected cell from the body.
  • a phagocytic receptor may be involved in the process of phagocytosis.
  • the process of phagocytosis can be closely coupled with an immune response and antigen presentation.
  • the processing of exogenous antigens follows their uptake into professional antigen presenting cells by some type of endocytic event. Phagocytosis can also facilitate antigen presentation.
  • antigens from phagocytosed cells or pathogens, including cancer antigens can be processed and presented on the cell surface of APCs.
  • a “polypeptide” can refer to a molecule containing amino acids linked together via a peptide bond, such as a glycoprotein, a lipoprotein, a cellular protein or a membrane protein.
  • a polypeptide may comprise one or more subunits of a protein.
  • a polypeptide may be encoded by a recombinant nucleic acid.
  • polypeptide may comprise more than one peptide sequence in a single amino acid chain, which may be separated by a spacer, a linker or peptide cleavage sequence.
  • a polypeptide may be a fused polypeptide.
  • a polypeptide may comprise one or more domains, modules or moieties.
  • a “receptor” can refer to a chemical structure composed of a polypeptide, which transduces a signal, such as a polypeptide that transduces an extracellular signal to a cell.
  • a receptor can serve to transmit information in a cell, a cell formation or an organism.
  • a receptor comprises at least one receptor unit and can contain two or more receptor units, where each receptor unit comprises a protein molecule, e.g., a glycoprotein molecule.
  • a receptor can contain a structure that binds to a ligand and can form a complex with the ligand. Signaling information can be transmitted by a conformational change of the receptor following binding with the ligand on the surface of a cell.
  • antibody refers to a class of proteins that are generally known as immunoglobulins, including, but not limited to IgGl, IgG2, IgG3, and IgG4), IgA (including IgAl and IgA2), IgD, IgE, IgM, and IgY,
  • antibody includes, but is not limited to, full length antibodies, single-chain antibodies, single domain antibodies (sdAb) and antigen-binding fragments thereof.
  • Antigen-binding antibody fragments include, but are not limited to, Fab, Fab’ and F(ab’)2, Fd (consisting of V H and C H 1), single-chain variable fragment (scFv), single-chain antibodies, disulfide-linked variable fragment (dsFv) and fragments comprising a VL and/or a VH domain.
  • Antibodies can be from any animal origin.
  • Antigen-binding antibody fragments, including single- chain antibodies can comprise variable region(s) alone or in combination with tone or more of a hinge region, a CH1 domain, a CH2 domain, and a CH3 domain. Also included are any combinations of variable region(s) and hinge region, CH1, CH2, and CH3 domains.
  • Antibodies can be monoclonal, polyclonal, chimeric, humanized, and human monoclonal and polyclonal antibodies which, e.g., specifically bind an HLA-associated polypeptide or an HLA-peptide complex.
  • the term “recombinant nucleic acid” refers a nucleic acid prepared, expressed, created or isolated by recombinant means. A recombinant nucleic acid can contain a nucleotide sequence that is not naturally occurring. A recombinant nucleic acid may be synthesized in the laboratory.
  • a recombinant nucleic acid may be prepared by using recombinant DNA technology, for example, enzymatic modification of DNA, such as enzymatic restriction digestion, ligation, and DNA cloning.
  • a recombinant nucleic acid can be DNA, RNA, analogues thereof, or a combination thereof.
  • a recombinant DNA may be transcribed ex vivo or in vitro, such as to generate a messenger RNA (mRNA).
  • mRNA messenger RNA
  • a recombinant mRNA may be isolated, purified and used to transfect a cell.
  • a recombinant nucleic acid may encode a protein or a polypeptide.
  • the process of introducing or incorporating a nucleic acid into a cell can be via transformation, transfection or transduction. Transformation is the process of uptake of foreign nucleic acid by a bacterial cell. This process is adapted for propagation of plasmid DNA, protein production, and other applications. Transformation introduces recombinant plasmid DNA into competent bacterial cells that take up extracellular DNA from the environment. Some bacterial species are naturally competent under certain environmental conditions, but competence is artificially induced in a laboratory setting. Transfection is the introduction of small molecules such as DNA, RNA, or antibodies into eukaryotic cells. Transfection may also refer to the introduction of bacteriophage into bacterial cells.
  • a vector can refer to a nucleic acid molecule capable of autonomous replication in a host cell, and which allow for cloning of nucleic acid molecules.
  • a vector includes, but is not limited to, a plasmid, cosmid, phagemid, viral vectors, phage vectors, yeast vectors, mammalian vectors and the like.
  • a vector for exogenous gene transformation may be a plasmid.
  • a vector comprises a nucleic acid sequence containing an origin of replication and other elements necessary for replication and/or maintenance of the nucleic acid sequence in a host cell.
  • a vector or a plasmid provided herein is an expression vector. Expression vectors are capable of directing the expression of genes and/or nucleic acid sequence to which they are operatively linked.
  • an expression vector or plasmid is in the form of circular double stranded DNA molecules.
  • a vector or plasmid may or may not be integrated into the genome of a host cell.
  • nucleic acid sequences of a plasmid are not integrated in a genome or chromosome of the host cell after introduction.
  • the plasmid may comprise elements for transient expression or stable expression of the nucleic acid sequences, e.g. genes or open reading frames harbored by the plasmid, in a host cell.
  • a vector is a transient expression vector.
  • a vector is a stably expressed vector that replicates autonomously in a host cell.
  • nucleic acid sequences of a plasmid are integrated into a genome or chromosome of a host cell upon introduction into the host cell.
  • Expression vectors that can be used in the methods as disclosed herein include, but are not limited to, plasmids, episomes, bacterial artificial chromosomes, yeast artificial chromosomes, bacteriophages or viral vectors.
  • a vector can be a DNA or RNA vector.
  • a vector provide herein is a RNA vector that is capable of integrating into a host cell’s genome upon introduction into the host cell (e.g., via reverse transcription), for example, a retroviral vector or a lentiviral vector.
  • Other forms of expression vectors known by those skilled in the art which serve the equivalent functions can also be used, for example, self-replicating extrachromosomal vectors or vectors capable of integrating into a host genome.
  • Exemplary vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked.
  • the terms “spacer” or “linker” as used in reference to a fusion protein refers to a peptide sequence that joins two other peptide sequences of the fusion protein.
  • a linker or spacer has no specific biological activity other than to join or to preserve some minimum distance or other spatial relationship between the proteins or RNA sequences.
  • the constituent amino acids of a spacer can be selected to influence some property of the molecule such as the folding, flexibility, net charge, or hydrophobicity of the molecule.
  • Suitable linkers for use in an embodiment of the present disclosure are well known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers.
  • a linker is used to separate two or more polypeptides, e.g. two antigenic peptides by a distance sufficient to ensure that each antigenic peptide properly folds.
  • Exemplary peptide linker sequences adopt a flexible extended conformation and do not exhibit a propensity for developing an ordered secondary structure.
  • Amino acids in flexible linker protein region may include Gly, Asn and Ser, or any permutation of amino acid sequences containing Gly, Asn and Ser.
  • Thr and Ala Other near neutral amino acids, such as Thr and Ala, also can be used in the linker sequence.
  • the terms “treat,” “treated,” “treating,” “treatment,” and the like are meant to refer to reducing, preventing, or ameliorating a disorder and/or symptoms associated therewith (e.g., a neoplasia or tumor or infectious agent or an autoimmune disease). “Treating” can refer to administration of the therapy to a subject after the onset, or suspected onset, of a disease (e.g., cancer or infection by an infectious agent or an autoimmune disease).
  • a disease e.g., cancer or infection by an infectious agent or an autoimmune disease.
  • Treating includes the concepts of “alleviating”, which can refer to lessening the frequency of occurrence or recurrence, or the severity, of any symptoms or other ill effects related to the disease and/or the side effects associated with therapy.
  • Treating also encompasses the concept of “managing” which refers to reducing the severity of a disease or disorder in a patient, e.g., extending the life or prolonging the survivability of a patient with the disease, or delaying its recurrence, e.g., lengthening the period of remission in a patient who had suffered from the disease. It is appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition, or symptoms associated therewith be completely eliminated.
  • treating a subject or a patient as described herein comprises administering a therapeutic composition, such as a drug, a metabolite, a preventive component, a nucleic acid, a peptide, or a protein that encodes or otherwise forms a drug, a metabolite or a preventive component.
  • treating comprises administering a cell or a population of cells to a subject in need thereof.
  • treating comprises administering to the subject one or more of engineered cells described herein, e.g. one or more engineered myeloid cells, such as phagocytic cells.
  • Treating comprises treating a disease or a condition or a syndrome, which may be a pathological disease, condition or syndrome, or a latent disease, condition or syndrome.
  • treating as used herein may comprise administering a therapeutic vaccine.
  • the engineered phagocytic cell is administered to a patient or a subject.
  • a cell administered to a human subject results in reduced immunogenicity.
  • an engineered phagocytic cell may lead to no or reduced graft versus host disease (GVHD) or fratricide effect.
  • GVHD graft versus host disease
  • an engineered cell administered to a human subject is immunocompatible to the subject (i.e. having a matching HLA subtype that is naturally expressed in the subject).
  • Subject specific HLA alleles or HLA genotype of a subject can be determined by any method known in the art.
  • the methods include determining polymorphic gene types that can comprise generating an alignment of reads extracted from a sequencing data set to a gene reference set comprising allele variants of the polymorphic gene, determining a first posterior probability or a posterior probability derived score for each allele variant in the alignment, identifying the allele variant with a maximum first posterior probability or posterior probability derived score as a first allele variant, identifying one or more overlapping reads that aligned with the first allele variant and one or more other allele variants, determining a second posterior probability or posterior probability derived score for the one or more other allele variants using a weighting factor, identifying a second allele variant by selecting the allele variant with a maximum second posterior probability or posterior probability derived score, the first and second allele variant defining the gene type for the polymorphic gene, and providing an output of the first and second allele variant.
  • a “fragment” can refer to a portion of a protein or nucleic acid. In some embodiments, a fragment retains at least 50%, 75%, or 80%, or 90%, 95%, or even 99% of the biological activity of a reference protein or nucleic acid.
  • isolated The terms “isolated,” “purified”, “biologically pure” and their grammatical equivalents refer to material that is free to varying degrees from components which normally accompany it as found in its native state. “Isolate” denotes a degree of separation from original source or surroundings. “Purify” denotes a degree of separation that is higher than isolation.
  • a “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of the present disclosure is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography. The term “purified” can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel.
  • neoplasia refers to any disease that is caused by or results in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both.
  • Glioblastoma is one non-limiting example of a neoplasia or cancer.
  • cancer or “tumor” or “hyperproliferative disorder” refer to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features.
  • Cancer cells are often in the form of a tumor, but such cells can exist alone within an animal, or can be a non- tumorigenic cancer cell, such as a leukemia cell.
  • the term “vaccine” is to be understood as meaning a composition for generating immunity for the prophylaxis and/or treatment of diseases (e.g., neoplasia/tumor/infectious agents/autoimmune diseases).
  • vaccines as used herein are medicaments which comprise recombinant nucleic acids, or cells comprising and expressing a recombinant nucleic acid and are intended to be used in humans or animals for generating specific defense and protective substance by vaccination.
  • a “vaccine composition” can include a pharmaceutically acceptable excipient, carrier or diluent. Aspects of the present disclosure relate to use of the technology in preparing a phagocytic cell-based vaccine.
  • pharmaceutically acceptable refers to approved or approvable by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • a “pharmaceutically acceptable excipient, carrier or diluent” refers to an excipient, carrier or diluent that can be administered to a subject, together with an agent, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent.
  • Nucleic acid molecules useful in the methods of the disclosure include, but are not limited to, any nucleic acid molecule with activity or that encodes a polypeptide. Polynucleotides having substantial identity to an endogenous sequence are typically capable of hybridizing with at least one strand of a double-stranded nucleic acid molecule.
  • Hybridize refers to when nucleic acid molecules pair to form a double-stranded molecule between complementary polynucleotide sequences, or portions thereof, under various conditions of stringency.
  • stringent salt concentration can ordinarily be less than about 750 mM NaCl and 75 mM trisodium citrate, less than about 500 mM NaCl and 50 mM trisodium citrate, or less than about 250 mM NaCl and 25 mM trisodium citrate.
  • Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high stringency hybridization can be obtained in the presence of at least about 35% formamide, or at least about 50% formamide.
  • Stringent temperature conditions can ordinarily include temperatures of at least about 30° C, at least about 37°C, or at least about 42°C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed.
  • hybridization can occur at 30° C in 750 mM NaCl, 75 mM trisodium citrate, and 1% SDS. In another exemplary embodiment, hybridization can occur at 37° C in 500 mM NaCl, 50 mM trisodium citrate, 1% SDS, 35% formamide, and 100 ⁇ g/ml denatured salmon sperm DNA (ssDNA). In another exemplary embodiment, hybridization can occur at 42° C in 250 mM NaCl, 25 mM trisodium citrate, 1% SDS, 50% formamide, and 200 ⁇ g/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art.
  • washing steps that follow hybridization can also vary in stringency.
  • Wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature.
  • stringent salt concentration for the wash steps can be less than about 30 mM NaCl and 3 mM trisodium citrate, or less than about 15 mM NaCl and 1.5 mM trisodium citrate.
  • Stringent temperature conditions for the wash steps can include a temperature of at least about 25°C, of at least about 42°C, or at least about 68°C. In exemplary embodiments, wash steps can occur at 25° C in 30 mM NaCl, 3 mM trisodium citrate, and 0.1% SDS.
  • wash steps can occur at 42° C in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS.
  • wash steps can occur at 68° C in 15 mM NaC1, 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art. Hybridization techniques are well known to those skilled in the art and are described, for example, in Benton and Davis (Science 196:180, 1977); Grunstein and Hogness (Proc. Natl. Acad. Sci., USA 72:3961, 1975); Ausubel et al.
  • substantially identical refers to a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein).
  • sequence can be at least 60%, 80% or 85%, 90%, 95%, 96%, 97%, 98%, or even 99% or more identical at the amino acid level or nucleic acid to the sequence used for comparison.
  • Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs).
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs.
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs.
  • Such software matches identical or similar sequence
  • Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • a BLAST program can be used, with a probability score between e-3 and e-m° indicating a closely related sequence.
  • a “reference” is a standard of comparison. It will be understood that the numbering of the specific positions or residues in the respective sequences depends on the particular protein and numbering scheme used.
  • Numbering might be different, e.g., in precursors of a mature protein and the mature protein itself, and differences in sequences from species to species may affect numbering.
  • One of skill in the art will be able to identify the respective residue in any homologous protein and in the respective encoding nucleic acid by methods well known in the art, e.g., by sequence alignment to a reference sequence and determination of homologous residues.
  • the term “subject” or “patient” refers to an organism, such as an animal (e.g., a human) which is the object of treatment, observation, or experiment.
  • a subject includes, but is not limited to, a mammal, including, but not limited to, a human or a non-human mammal, such as a non-human primate, murine, bovine, equine, canine, ovine, or feline.
  • a disorder e.g., a neoplasia, tumor, or infection by an infectious agent or an autoimmune disease
  • its associated pathology e.g., a neoplasia, tumor, or infection by an infectious agent or an autoimmune disease
  • “Therapeutically effective amount” refers to an amount of an agent which is effective, upon single or multiple dose administration to the cell or subject, in prolonging the survivability of the patient with such a disorder, reducing one or more signs or symptoms of the disorder, preventing or delaying, and the like beyond that expected in the absence of such treatment. “Therapeutically effective amount” is intended to qualify the amount required to achieve a therapeutic effect. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the “therapeutically effective amount” (e.g., ED50) of the pharmaceutical composition required.
  • ED50 e.g., ED50
  • compositions and methods for preparing targeted killer myeloid cells by leveraging the innate functional role in immune defense, ranging from properties related to detecting foreign bodies, particles, diseased cells, cellular debris, inflammatory signal, chemoattract; activating endogenous DAMP and PAMP signaling pathways; trigger myelopoiesis, extravasation; chemotaxis; phagocytes; pinocytosis; recruitment; engulfment; scavenging; activating intracellular oxidative burst and lysis or killing of pathogens, detecting, engulfing and killing diseased or damaged cells; removing unwanted cellular, tissue or acellular debris in vivo; antigen presentation and role in activating innate immunity; activating and modulating an immune response cascade; activating T cell repertoire; autophagy; inflammatory and non-inflammatory apoptosis; pyroptos
  • the one or more functions may be one or more of: detecting foreign bodies, particles, diseased cells, cellular debris, inflammatory signal, chemoattract; activating endogenous DAMP and PAMP signaling pathways; trigger myelopoiesis, extravasation; chemotaxis; phagocytosis; pinocytosis; recruitment; trogocytosis; engulfment; scavenging; activating intracellular oxidative burst and intracellular lysis or killing of pathogens, detecting, engulfing and killing diseased or damaged cells; removing unwanted cellular, tissue or acellular debris in vivo; antigen presentation and role in activating innate immunity; activating and modulating an immune response cascade; activating T cell repertoire; autophagy; inflammatory and non-inflammatory apoptosis; pyroptosis, immune editing to response to stress and restoration of tissue home
  • compositions and methods are also directed to augmenting the targeting, and killing function of certain myeloid cells, by genetic modification of these cells.
  • the compositions and methods described herein are directed to creating engineered myeloid cells, wherein the engineered myeloid cells comprise at least one genetic modification, and can be directed to recognize and induce effector functions against a pathogen, a diseased cell, such as a tumor or cancer cell, such that the engineered myeloid cell is capable of recognizing, targeting, phagocytosing, killing and/or eliminating the pathogen or the diseased cell or the cancer cell, and additionally, may activate a specific immune response cascade following the phagocytosis, killing and/or eliminating the pathogen or the diseased cell.
  • Myeloid cells appear to be the most abundant cells in a tumor (FIG. 1B). Myeloid cells are also capable of recognizing a tumor cell over a healthy normal cell of the body and mount an immune response to a tumor cell of the body. As sentinels of innate immune response, myeloid cells are able to sense non-self or aberrant cell types and clear them via a process called phagocytosis. This can be directed to a therapeutic advantage in driving myeloid cell mediated phagocytosis and lysis of tumor cells.
  • these naturally occurring tumor-infiltrating myeloid cells may be subjected to influence of the tumor microenvironment (TME). TIMs constitute a heterogeneous population of cells.
  • TIMs originate from circulating monocytes and granulocytes, which in turn stem from bone marrow-derived hematopoietic stem cells.
  • monocyte and granulocyte progenitors divert from their intrinsic pathway of terminal differentiation into mature macrophages, DCs or granulocytes, and may become tumor promoting myeloid cell types. Differentiation into pathological, alternatively activated immature myeloid cells is favored.
  • immature myeloid cells include tumor-associated DCs (TADCs), tumor-associated neutrophils (TANs), myeloid- derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs).
  • TADCs tumor-associated DCs
  • TANs tumor-associated neutrophils
  • MDSCs myeloid- derived suppressor cells
  • TAMs tumor-associated macrophages
  • TAMs may also originate from tissue-resident macrophages, which in turn can be of embryonic or monocytic origin. These tissue-resident macrophages undergo changes in phenotype and function during carcinogenesis, and proliferation may help to maintain TAMs derived from tissue-resident macrophages.
  • a tumor microenvironment may drive a tumor infiltrating myeloid cell to become myeloid derived suppressor cells and acquire the ability to suppress T cells.
  • innovative methods are necessary to create therapeutically effective TIMs that can infiltrate a tumor, and can target tumor cells for phagocytic uptake and killing.
  • engineered myeloid cells that are capable of targeting specific target cells, for example, tumor cells or pathogenic cells.
  • engineered myeloid cells provided herein are potent in infiltrating, targeting, and killing tumor cells.
  • An engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which encodes one or more proteins that help target the phagocytic cell to a target cell, for example a tumor cell or a cancer cell.
  • the engineered myeloid cell is capable of readily infiltrating a tumor.
  • the engineered myeloid cell has high specificity for the target cell, with none or negligible cross-reactivity to a non-tumor, non-diseased cell of the subject while in circulation.
  • the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which will help the cell to overcome/bypass the TME influence and mount a potent anti-tumor response.
  • the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which augments phagocytosis of the target cell.
  • the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which augments reduce or eliminate trogocytosis and/or enhance phagocytic lysis or of the target cell.
  • the compositions disclosed herein comprise a myeloid cell, comprising a recombinant nucleic acid encoding a chimeric receptor fusion protein (CFP), for example, a phagocytic receptor (PR) fusion protein (PFP).
  • CFP chimeric receptor fusion protein
  • PR phagocytic receptor
  • the recombinant nucleic acid can comprise a sequence encoding a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising a PR intracellular signaling domain; and an extracellular antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular antigen binding domain are operatively linked; wherein the PR intracellular signaling domain is derived from a receptor with a signal transduction domain.
  • the recombinant nucleic acid further encodes for one or more polypeptides that constitute one or more plasma membrane receptors that helps engage the phagocytic cell to the target cell, and enhance its phagocytic activity.
  • the myeloid cell described herein comprises one or more recombinant proteins comprising a chimeric receptor, wherein the chimeric receptor is capable of responding to a first phagocytic signal directed to a target cell, which may be a diseased cell, a tumor cell or a pathogen, and a second signal, which is an inflammatory signal, that augments the phagocytic and killing response to target initiated by the first signal.
  • a target cell which may be a diseased cell, a tumor cell or a pathogen
  • a second signal which is an inflammatory signal
  • PHAGOCYTIC CELLS, MYELOID CELLS, ENGINEERED MYELOID CELLS [0258]
  • methods and compositions for immunotherapy comprising ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a specific target, which can be a specific cell type or class of cells in a patient or a subject.
  • the subject is a patient having a disease.
  • the terms subject and patient may often be used interchangeably in this section.
  • the phagocytic cell is derived from the subject having a disease, wherein the disease is, for example, cancer.
  • the autologous cells from the subject may be modified in vitro and administered into the cell, where the modified phagocytic cell is redesigned to specifically attack and kill the cancer cells in the subject.
  • the subject has a disease that is not a cancer.
  • the subject has a disease that is an infection.
  • the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards an infection, for example an infected cell within the subject.
  • the subject has a disease that is a viral, a bacterial, a fungal or a protozoal infection.
  • the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a virus infected cell, a bacteria infected cell, a fungus infected cell or a protozoa infected cell inside the infected subject.
  • the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a virus, a bacteria, a fungus or any pathogen in a subject, such that the virus, the bacteria, the fungus or the pathogen in a subject is phagocytosed, and/or killed.
  • the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a viral antigen, a bacterial antigen, a fungal antigen or an antigen of a pathogen in a subject, such that there is at least one improved immune response within the subject to the virus, the bacteria, the fungus or the pathogen in the subject.
  • the myeloid cells such as phagocytic cells
  • the methods and compositions for immunotherapy provided herein comprises obtaining myeloid cells, such as phagocytic cells, derived from an allogeneic source.
  • the myeloid cells can thereafter be modified or engineered and introduced into a diseased subject, such that the modified or engineered cells from the allogeneic source are capable of attacking a diseased cell of the subject, phagocytose the diseased cell and/or kill the diseased cell directly or indirectly, or improve at least one immune response of the subject to the disease.
  • the allogeneic source is a human.
  • the allogeneic source is a healthy human.
  • Phagocytes are the natural sentinels of the immune system and form the first line of defense in the body.
  • phagocytes therefore can be activated by a variety of non-self (and self) elements and exhibit a level of plasticity in recognition of their “targets”.
  • MPS Mononuclear phagocytic system
  • monocytes comprised of monocytes, macrophages, and dendritic cells
  • the MPS is a cell lineage which originates from bone marrow progenitor cells and gives rise to blood monocytes, tissue macrophages and dendritic cells.
  • the process of generating a macrophage from the MPS begins with a promonocyte in the BM which undergoes a differentiation process into a monocyte that is ready to enter the systemic circulation.
  • phagocytosis clears and degrades disease-causing microbes, induces pro- inflammatory signaling through cytokine and chemokine secretion, and recruits immune cells to mount an effective inflammatory response.
  • This type of phagocytosis is often referred to as "inflammatory phagocytosis" (or “immunogenic phagocytosis”).
  • anti-inflammatory responses may follow microbial uptake.
  • Anti- microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells. Phagocytosis of damaged, apoptotic cells or cell is typically a non-inflammatory (also referred to as a "nonimmunogenic") process. Transformed or malignant cells (self-cells), and cells are phagocytosed and apoptotic cells are removed promptly without causing damage to the surrounding tissues or inducing a pro-inflammatory immune response.
  • DCs immature dendritic cells
  • macrophages by tissue-resident immune cells.
  • Phagocytosis of damaged, apoptotic cells or cell is typically a non-inflammatory (also referred to as a "nonimmunogenic") process. Transformed or malignant cells (self-cells), and cells are phagocytosed and apoptotic cells are removed promptly without causing damage to the surrounding tissues or
  • This type of apoptotic cell clearance is non-inflammatory and include release of "find me” signals from apoptotic cells to recruit phagocytes to the location of apoptotic cells; accompanied by "eat me” signals exposed on the surface of apoptotic cells are bound by phagocytes via specific receptors; cytoskeletal rearrangement to engulf the apoptotic cell; followed by the ingested apoptotic cell is digested and specific phagocytic responses are elicited (e.g., secretion of anti-inflammatory cytokines).
  • Phagocytosis the cellular uptake of particulates, e.g.
  • particles >0.5 ⁇ m within a plasma- membrane envelope is closely related to and partly overlaps the endocytosis of soluble ligands by fluid-phase macropinocytic and receptor pathways.
  • Variants associated with the uptake of apoptotic cells also known as efferocytosis, and that of necrotic cells arising from infection and inflammation (necroptosis and pyroptosis).
  • the uptake of exogenous particles has features in common with autophagy, an endogenous process of sequestration and lysosomal disposal of damaged intracellular organelles There is a spectrum of uptake mechanisms depending on the particle size, multiplicity of receptor-ligand interactions, and involvement of the cytoskeleton.
  • the phagosome vacuole can fuse selectively with primary lysosomes, or the product of the endoplasmic reticulum (ER) and Golgi complex, to form a secondary phagolysosome (Russell, D.G. (2011). Immunol. Rev. 240, 252–268).
  • This pathway is dynamic in that it undergoes fusion and fission with endocytic and secretory vesicles macrophages, DCs, osteoclasts, and eosinophils.
  • Anti- microbe phagocytosis clears and degrades disease-causing microbes, induces pro-inflammatory signaling through cytokine and chemokine secretion, and recruits immune cells to mount an effective inflammatory response.
  • This type of phagocytosis is often referred to as “inflammatory phagocytosis” (or “immunogenic phagocytosis”).
  • anti-inflammatory responses may follow microbial uptake.
  • Anti-microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells.
  • DCs immature dendritic cells
  • Phagocytosis of damaged, self-derived apoptotic cells or cell debris is typically a non-inflammatory (also referred to as a "nonimmunogenic") process.
  • Billions of damaged, dying, and unwanted cells undergo apoptosis each day.
  • Unwanted cells include, for example, excess cells generated during development, senescent cells, infected cells (intracellular bacteria or viruses), transformed or malignant cells, and cells irreversibly damaged by cytotoxic agents.
  • the bone marrow is the source of circulating neutrophils and monocytes that will replace selected tissue-resident macrophages and amplify tissue myeloid populations during inflammation and infection.
  • phagocytes are monocytes or cells of the monocyte lineage.
  • myeloid cells are phagocytic macrophages are MARCO+ SignR1+ and are found in the outer marginal zone rapidly clear capsulated bacteria. Similar CD169+ F4/80 _ macrophages line the subcapsular sinus in lymph nodes and have been implicated in virus infection. It was noted that endothelial macrophages, including Kupffer cells in the liver, clear microbial and antigenic ligands from blood and lymph nodes to provide a sinusoidal immune function comparable to but distinct from mucosal immunity. Not all tissue macrophages are constitutively phagocytic, even though they still express typical macrophage markers.
  • metallophilic macrophages which lack F4/80, strongly express CD169, sialic acid-binding immunoglobulin (Ig)-like lectin 1 (SIGLEC1 [sialoadhesin]), but are poorly phagocytic.
  • Non- professional phagocytes include epithelial cells, and fibroblasts. Fibroblasts are ‘‘working-class phagocytes” that clear apoptotic debris by using integrins other than CD11b-CD18 through adhesion molecules ICAM and vitronectin receptors. Astrocytes have also been reported to engulf, even if not efficiently degrade, apoptotic corpses.
  • Plasma-membrane receptors relevant to phagocytosis can be opsonic, FcRs (activating or inhibitory) for mainly the conserved domain of IgG antibodies, and complement receptors, such as CR3 for iC3b deposited by classical (IgM or IgG) or alternative lectin pathways of complement activation. CR3 can also mediate recognition in the absence of opsonins, perhaps by depositing macrophage-derived complement.
  • Anti-microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells.
  • DCs immature dendritic cells
  • cells that are used for engineering for use in immunotherapy are potently phagocytic.
  • cells that are used for engineering for use in immunotherapy are obtained from whole blood, peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue.
  • cells that are used for engineering for use in immunotherapy are obtained from peripheral blood.
  • the myeloid cells that are engineered for use in immunotherapy in the instant application comprise myeloid cells selected from the group consisting of macrophages, dendritic cells, mast cells, monocytes, neutrophils, microglia, and astrocytes.
  • the myeloid cells that are engineered for use in immunotherapy are phagocytic cells. In some embodiments, the phagocytic cells are monocytes.
  • the myeloid cells that are engineered for use in immunotherapy in the instant application are monocytes, monocyte derived macrophages, and/or dendritic cells. [0276] In some embodiments the myeloid cells that are engineered for use in immunotherapy in the instant application are monocytes or macrophages. [0277] In some embodiments the cells that myeloid cells obtained from the peripheral blood. [0278] In some embodiments, the myeloid cells are selected by selection marker CD14 + CD16 low . In some embodiments the myeloid cells are selected via elutriation. [0279] In some embodiments, the myeloid cells are isolated from leukapheresis column of the subject.
  • the subject is the same subject who is administered the pharmaceutical composition comprising engineered cells. [0280] In some embodiments the subject is not the same subject who is administered the pharmaceutical composition comprising engineered cells. [0281] In some embodiments, the leukapheresis is performed on the same subject once a week to collect more myeloid cells. In some embodiments, the leukapheresis is performed on the same subject more than once in a span of 8-10 days to collect more myeloid cells. In some embodiments, the leukapheresis is performed on the same subject more than twice in a span of one month to collect more myeloid cells. [0282] In some embodiments, myeloid cells are isolated from a leukapheresis sample or a peripheral blood sample.
  • the myeloid cell is a progenitor cell. In some embodiments, the myeloid cell is a monocyte precursor cell. In some embodiments, a myeloid cell described herein is not differentiated into a terminal cell and do not exhibit a terminal cell phenotype, such as tissue macrophages. In some embodiments, the myeloid cells comprise CD14+ cells. In some embodiments, the myeloid cells do not express CD16. In some embodiments the myeloid cells express low amounts of CD16. In some embodiments, the myeloid cells are pre- selected for the purpose of engineering from a biological sample, such as peripheral blood or an apheresis sample by selection of CD14+ cells.
  • the selection is performed without contacting with or engaging with the myeloid cell to be selected.
  • the myeloid cells are selected prior to engineering from a biological sample by sorting, for example a flow cytometry based cell sorter (FACS).
  • FACS flow cytometry based cell sorter
  • the myeloid cells expressing CD16 are captured by an antibody and the remaining myeloid cells were collected and used for engineering.
  • one or more other cell surface molecules are targeted for capturing in the negative selection process in addition to CD16, in order to obtain the myeloid cells, for example CD3, CD8, CD11c, CD40, or CD206.
  • myeloid cells comprising at least one exogenous recombinant nucleic acid that encodes for a fusion protein.
  • the fusion protein may be a chimeric protein comprising at least a transmembrane domain and an extracellular domain that comprises a region that can bind to a target cell.
  • the chimeric protein may bind to a target, e.g. a target antigen, an antigenic peptide, or a ligand on the target cell.
  • the target cell is a cancer cell.
  • a target is a cancer antigen.
  • the chimeric protein is expressed in the myeloid cell and activates the myeloid cell to overcome TME induced suppressive signal and act as an activated pro-inflammatory myeloid cell.
  • the chimeric protein that is expressed in the myeloid cell is capable of being responsive to a second signal other than the target (the first signal), wherein the second signal is a pro- inflammatory signal and an activating signal.
  • the chimeric protein that is expressed in the myeloid cell is capable of being responsive to multiple signals other than the target or the first signal.
  • the chimeric protein may be able to respond to one, two, three, four, five, or more signals besides the target or the first signal.
  • the chimeric protein that is expressed in the myeloid cell is specific for binding to a target.
  • the target is a cancer antigen. Expression of the chimeric protein endows target specificity to the myeloid cell.
  • the chimeric protein that is expressed in the myeloid cell is capable of multiplexing, for example, has multiple domains for activation and processing of more than one signal or signal types. In some embodiments, activation of the multiple domains simultaneously leads to an augmented effector response to the myeloid cell.
  • An effector response for the myeloid cell encompasses, for example, enhanced phagocytosis, pro-inflammatory activation, and killing of target cell.
  • the chimeric protein that is expressed in the myeloid cell, capable of multiplexing is capable of binding to more than one ligands, such as a target antigen and a helper molecule. In some embodiments, the chimeric protein is capable of binding to multiple target antigens on a cancer cell. In some embodiments, the chimeric protein is capable of multiplexing is capable of binding to multiple target antigens on multiple cells. In some embodiments, the chimeric protein may bind to a macrophage-monocyte inhibitory target on a cancer cell, and create a stimulatory signal upon contact using the pro-inflammatory domain fused to the intracellular end, a process termed as “signal switch”.
  • an extracellular domain of the chimeric protein may comprise a CD47-binding domain
  • the chimeric fusion protein lacks the transmembrane and/or intracellular domain of the native CD47 receptor, but comprises a PI3K recruiter domain at the intracellular region, thereby converting the macrophage-monocyte inhibitory signal from contact with the tumor cell to a pro-inflammatory phagocytosis enhancing signal.
  • the chimeric protein is capable of binding to multiple units of the expressed chimeric protein, for example, multimerizing.
  • Multimerizing comprises dimer, trimer, tetramer, pentamer, hexamer, heptamer, octamer, nonamer, or decamer formations.
  • multimerizing can occur via association of the transmembrane region, the extracellular region or the intracellular region or combinations thereof.
  • a chimeric protein comprising a region of the collagenous domain of the phagocytic receptor MARCO may form a trimer for its effective function.
  • the chimeric protein is capable of associating with other molecules for example, another receptor.
  • the chimeric protein comprises an Fc-alpha transmembrane domain that dimerizes with Fc ⁇ TM domain, wherein the Fc ⁇ may be an endogenous receptor.
  • the chimeric protein capable of multiplexing comprises multiple intracellular domains that can be activated by more than one signal and can in turn activate multiple intracellular signaling molecules.
  • the chimeric protein may comprise, a phagocytosis receptor domain and a pro-inflammatory domain.
  • the chimeric protein comprises a FcR signaling domain and an additional phosphorylation domain that recruits procaspases.
  • recombinant nucleic acid encoding a CFP that is phagocytic receptor (PR) fusion protein (PFP).
  • the PFP can comprise a PR subunit comprising: a transmembrane (TM) domain, and an intracellular domain (ICD) comprising a PR intracellular signaling domain.
  • TM transmembrane
  • ICD intracellular domain
  • the recombinant nucleic acid encoding the PFP when expressed in a cell the PFP functionally incorporates into the cell membrane of the cell.
  • the recombinant nucleic acid encodes for a transmembrane domain that specifically incorporates in the membrane of a myeloid cell, such as a phagocytic cell, e.g., a macrophage.
  • the suitable PR is selected after screening a library of membrane spanning proteins.
  • the PR subunit is fused at the extracellular domain with a cancer cell binding antibody.
  • the PR may be fused with one or more additional domains at the intracellular end.
  • INTRACELLULAR DOMAIN OF CFP FUSION PROTEIN [0290]
  • the CFP subunit comprises a TM domain of a phagocytic receptor.
  • the CFP subunit comprises an ICD domain of a phagocytic receptor.
  • the phagocytic receptor is a scavenger receptor. Whilst many scavenger receptors collaborate in the detection and ingestion of materials, not all the receptors engaged in the course of phagocytosis trigger engulfment alone. The engagement of certain phagocytosis and scavenger receptors can have dramatic impacts on the downstream immune response. For example, triggering the type A scavenger receptor MARCO with 500 nm negatively charge nanoparticles is associated with an anti-inflammatory tolerogenic immune response.
  • Non-opsonic receptors variably expressed naturally by professional phagocytes include lectin-like recognition molecules, such as CD169, CD33, and related receptors for sialylated residues.
  • phagocytes also express Dectin-1 (a receptor for fungal ⁇ -glucan with well- defined signaling capacity), related C-type lectins (e.g., MICL, Dectin-2, Mincle, and DNGR-1), and a group of scavenger receptors.
  • Dectin-1 a receptor for fungal ⁇ -glucan with well- defined signaling capacity
  • C-type lectins e.g., MICL, Dectin-2, Mincle, and DNGR-1
  • CD36-related family member revealed that apoprotein ligands bind to receptor helical bundles, whereas their exofacial domains form a channel through which lipids such as cholesterol are translocated to the membrane bilayer.
  • the recombinant nucleic acid encodes a chimeric antigenic receptor for phagocytosis (CAR-P). In some embodiments, the recombinant nucleic acid encodes a phagocytic receptor (PR) fusion protein.
  • CAR-P chimeric antigenic receptor for phagocytosis
  • PR phagocytic receptor
  • the ICD of a CFP encoded by the recombinant nucleic acid comprises a domain from a protein selected from the group consisting of TNFR1, CD40, MDA5, lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor.
  • a protein selected from the group consisting of TNFR1, CD40, MDA5, lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD
  • the ICD comprises the signaling domain derived from any one or more of: lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO (Macrophage Receptor with Collagenous Structure, aliases: SRA6, SCARA2), CD36 (Thrombospondin receptor, aliases: Scavenger Receptor class B, member 3), CD163 (Scavenger receptor, cysteine rich-type 1), MSR1, SCARA3, COLEC12 (aliases: Scavenger Receptor With C- Type Lectin, SCARA4, or Collectin 12), SCARA5, SCARB1, SCARB2, CD68 (SCARD, microsialin), OLR1 (Oxidized Low Density Lipoprotein Receptor 1, LOX1, or C-Type Lectin Domain Family 8 Member A), SCARF1, SCARF2, SRCRB4D, SSC5D, and CD169 (aliases,
  • the recombinant nucleic acid encodes, for example, an intracellular domain of human MARCO.
  • the PR subunit can comprise an intracellular domain having a 44 amino acid ICD of human MARCO having an amino acid sequence: MRNKKILKEDELLSETQQAAFHQIAMEPFEINVPKPKRRNGVNF.
  • the PR subunit comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to the intracellular domain of MARCO.
  • the PR comprises a transmembrane region of human MARCO.
  • the recombinant nucleic acid encodes an intracellular domain of human SRA1.
  • the CFP comprises an intracellular domain having a 50 amino acid ICD of human SRA1 having an amino acid sequence: MEQWDHFHNQQEDTDSCSESVKFDARSMTA LLPPNPKNSPSLQEKLKSFK.
  • the PR subunit comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to the intracellular domain of human SRA1.
  • the intracellular region of SRA has a phosphorylation site.
  • the CFP comprises a transmembrane region of human SRA1.
  • the recombinant nucleic acid comprises a sequence encoding an intracellular domain of CD36.
  • the recombinant nucleic acid comprises a sequence encoding TM domain of CD36.
  • Naturally occurring full length CD36 has two TM domains and two short intracellular domains, and an extracellular domain of CD36 binds to oxidized LDL. Both of the intracellular domains contain pairs of cysteines that are fatty acid acylated. It lacks known signaling domains (e.g. kinase, phosphatase, g-protein binding, or scaffolding domains).
  • N- terminal cytoplasmic domain is extremely short (5–7 amino acid residues) and is closely associated with the internal leaflet of the plasma membrane.
  • the carboxy-terminal domain contains 13 amino acids, containing a CXCX5K motif homologous to a region in the intracellular domain of CD4 and CD8 that is known to interact with signaling molecules.
  • the intracellular domain of CD36 is capable of assembling a signaling complex that activates lyn kinases, MAP kinases and Focal Adhesion Kinases (FAK), and inactivation of src homology 2-containing phosphotyrosine phosphatase (SHP- 2).
  • FAM Focal Adhesion Kinases
  • SHP-2 phosphotyrosine phosphatase
  • GEFs guanine nucleotide exchange factors
  • the recombinant nucleic acid encodes for example, an intracellular domain of human SCARA3.
  • the CFP may comprise an intracellular domain having a 56 amino acid ICD of human SCARA3 having an amino acid sequence: MKVRSAGGDGDALCVTEEDL AGDDEDMPTFPCTQKGRPGPRCSRCQKNLS LHTSVR.
  • the CFP comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to an intracellular domain of human SCARA3.
  • the CFP comprises a TM domain of SCARA3.
  • the TM domain of a PR is about 20-30 amino acids long.
  • the TM domain comprises multiple transmembrane spans.
  • the TM domain comprises about 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or more amino acids in length. In some embodiments, the TM domains of SRs are about 20-30 amino acids long.
  • Scavenger receptors may occur as homo or hetero dimers. MARCO, for example occurs as a homo trimer.
  • MARCO for example occurs as a homo trimer.
  • a scavenger receptor is a monomer.
  • the scavenger receptor is a homodimer.
  • the scavenger receptor is a heterodimer. In some embodiments, the scavenger receptor is a homotrimer.
  • the scavenger receptor is a heterotrimer. In some embodiments, the scavenger receptor is a homo tetramer. In some embodiments, the scavenger receptor is a hetero tetramer. In some embodiments, the scavenger receptor is multimer comprising two, three, four, five, six, seven, eight, night, ten or more subunits that are the same or different. [0304] In some embodiments, the TM domain or the ICD domain of the PSP is not derived from FcR, Megf10, Bai1 or MerTK. In some embodiments, the ICD of the PR does not comprise a CD3 zeta intracellular domain.
  • the intracellular domain and transmembrane domains are derived from FcR ⁇ .
  • the recombinant nucleic acid encodes a chimeric antigenic receptor for enhanced phagocytosis (CAR-P), which is a phagocytic scavenger receptor (PSR) fusion protein (PFP) comprising: (a) an extracellular domain comprising an extracellular antigen binding domain specific to an antigen of a target cell, (b) a transmembrane domain, and (c) a recombinant PSR intracellular signaling domain, wherein the recombinant PSR intracellular signaling domain comprises a first portion derived from a phagocytic receptor and a second portion derived from a non-phagocytic receptor.
  • CAR-P a chimeric antigenic receptor for enhanced phagocytosis
  • PSR phagocytic scavenger receptor
  • PFP fusion protein
  • the second portion is not a PI3K recruitment domain.
  • the second portion derived from a non-phagocytic receptor may comprise an intracellular signaling domain that enhances phagocytosis, and/or inflammatory potential of the engineered myeloid cells, such as phagocytic cells, expressing the recombinant nucleic acid.
  • the second portion derived from non-phagocytic receptor comprises more than one intracellular domains (ICD).
  • the second portion derived from non-phagocytic receptor comprises a second ICD.
  • the second portion derived from non- phagocytic receptor comprises a second and a third ICD.
  • the second portion derived from non-phagocytic receptor comprises a second, a third and a fourth ICD, wherein the second portion is encoded by the recombinant nucleic acid.
  • the intracellular portion comprises two, three, four, five, six, seven, or more ICDs.
  • the recombinant nucleic acid encodes a second intracellular domain in addition to the phagocytic ICD, which confers capability of potent pro-inflammatory immune activation, such as when myeloid cells, such as macrophages, engage in fighting infection.
  • the second intracellular domain (second ICD) is fused to the cytoplasmic terminus of the first phagocytic ICD.
  • the second intracellular domain provides a second signal is necessary to trigger inflammasomes and pro-inflammatory signals.
  • NLRs Nod-like receptors
  • TME tumor microenvironment
  • Influence of IL-10, glucocorticoid hormones, apoptotic cells, and immune complexes can interfere with innate immune cell function.
  • Immune cells including phagocytic cells settle into a tolerogenic phenotype.
  • myeloid cells such as macrophages
  • this phenotype commonly known as the M2 phenotype
  • M2 phenotype is distinct from the M1 phenotype, where the cells are potent and capable of killing pathogens.
  • Myeloid cells, such as macrophages, exposed to LPS or IFN ⁇ , for example, can polarize towards an Ml phenotype
  • myeloid cells, such as macrophages, exposed to IL-4 or IL-13 can polarize towards an M2 phenotype.
  • LPS or IFN ⁇ can interact with Toll-like receptor 4 (TLR4) on the surface of myeloid cells, such as macrophages, inducing the Trif and MyD88 pathways, inducing the activation of transcription factors IRF3, AP-1, and NFKB and thus activating TNFs genes, interferon genes, CXCL10, NOS2, IL-12, etc., for a pro-inflammatory Ml myeloid cell response.
  • TLR4 and IL-13 bind to IL-4R, activation the Jak/Stat6 pathway, which regulates the expression of CCL17, ARG1, IRF4, IL-10, SOCS3, etc., which are genes associated with an anti- inflammatory response (M2 response).
  • the recombinant nucleic acid encodes one or more additional intracellular domains, comprising a cytoplasmic domain for inflammatory response.
  • expression of the recombinant nucleic acid encoding the phagocytic receptor (PR) fusion protein (PFP) comprising the cytoplasmic domain for inflammatory response in the engineered myeloid cells, such as macrophages confers potent pro-inflammatory response similar to the M1 phenotype.
  • PR phagocytic receptor
  • PFP fusion protein
  • the cytoplasmic domain for inflammatory response comprises an intracellular signaling domain of TLR3, TLR4, TLR9, MYD88, TRIF, RIG-1, MDA5, CD40, IFN receptor, NLRP-1, NLRP-2, NLRP-3, NLRP-4, NLRP-5, NLRP-6, NLRP-7, NLRP-8, NLRP-9, NLRP-10, NLRP-11, NLRP-12, NLRP-13, NLRP-14, NOD1, NOD2, Pyrin, AIM2, NLRC4 and/or CD40.
  • TLR3, TLR4, TLR9, MYD88, TRIF RIG-1, MDA5, CD40, IFN receptor, NLRP-1, NLRP-2, NLRP-3, NLRP-4, NLRP-5, NLRP-6, NLRP-7, NLRP-8, NLRP-9, NLRP-10, NLRP-11, NLRP-12, NLRP-13, NLRP-14, NOD1, NOD2,
  • the phagocytic scavenger receptor (PR) fusion protein (PFP) comprises a pro-inflammatory cytoplasmic domain for activation of IL-1 signaling cascade.
  • the cytoplasmic portion of the chimeric receptor (for example, phagocytic receptor (PR) fusion protein (PFP)) comprises a cytoplasmic domain from a toll-like receptor, such as the intracellular signaling domains of toll-like receptor 3 (TLR3), toll-like receptor 4 (TLR4), toll-like receptor 7 (TLR7), toll-like receptor 8 (TLR8), toll-like receptor 9 (TLR9).
  • the cytoplasmic portion of the chimeric receptor comprises a suitable region from interleukin-1 receptor-associated kinase 1 (IRAK1). [0316] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from differentiation primary response protein (MYD88). [0317] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from myelin and lymphocyte protein (MAL). [0318] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from retinoic acid inducible gene (RIG-1).
  • RIG-1 retinoic acid inducible gene
  • the cytoplasmic portion of the CFP comprises a cytoplasmic domain of any one of MYD88, TLR3, TLR4, TLR7, TLR8, TLR9, MAL, or IRAK1.
  • the recombinant CFP intracellular signaling domain comprises a first portion derived from a phagocytic and a second portion derived from non-phagocytic receptor wherein the second portion derived from non-phagocytic receptor comprises a phosphorylation site.
  • the phosphorylation site comprises amino acid sequences suitable for an autophosphorylation site.
  • the phosphorylation site comprises amino acid sequences suitable phosphorylation by Src family kinases. In some embodiments, the phosphorylation site comprises amino acid sequences, which upon phosphorylation are capable of binding to SH2 domains in a kinase. In some embodiments, a receptor tyrosine kinase domain is fused at the cytoplasmic end of the PFP in addition to the first cytoplasmic portion. [0321] In some embodiments, the phosphorylation is a Tyrosine phosphorylation. [0322] In some embodiments the second intracellular domain is an Immune receptor Tyrosine Activation Motif (ITAM).
  • ITAM Immune receptor Tyrosine Activation Motif
  • the ITAM motif is present in mammalian ⁇ and ⁇ immunoglobulin proteins, TCR ⁇ receptors, FCR ⁇ receptors subunits, CD3 chains receptors and NFAT activation molecule.
  • the PFP intracellular domain comprises one ITAM motif. In some embodiments the PFP intracellular domain comprises more than one ITAM motifs. In some embodiments the PFP intracellular domain comprises two or more ITAM motifs. In some embodiments the PFP intracellular domain comprises three or more ITAM motifs. In some embodiments the PFP intracellular domain comprises four or more ITAM motifs. In some embodiments the PFP intracellular domain comprises five or more ITAM motifs. In some embodiments the PFP intracellular domain comprises six or more ITAM motifs.
  • the PFP intracellular domain comprises seven or more ITAM motifs. In some embodiments the PFP intracellular domain comprises eight or more ITAM motifs. In some embodiments the PFP intracellular domain comprises nine or more ITAM motifs. In some embodiments the PFP intracellular domain comprises ten or more ITAM motifs. [0324] In some embodiments one or more domains in the first phagocytic ICD comprises a mutation. [0325] In some embodiments one or more domains in the second ICD comprises a mutation to enhance a kinase binding domain, to generate a phosphorylation site, to generate an SH2 docking site or a combination thereof.
  • the recombinant nucleic acid comprises a coding sequence for a pro- inflammatory gene, which is co-expressed with the PFP in the engineered cell.
  • the pro-inflammatory gene is a cytokine. Examples include but not limited to TNF- ⁇ , IL-1 ⁇ , IL-1 ⁇ , IL-6, CSF, GMCSF, or IL-12 or interferons.
  • the recombinant nucleic acid encoding the proinflammatory gene can be monocistronic, wherein the two coding sequences for (a) the PSP and (b) the proinflammatory gene are post- transcriptionally or post-translationally cleaved for independent expression.
  • the two coding sequences comprise a self-cleavage domain, encoding a P2A sequence, for example.
  • the two coding regions are separated by a IRES site.
  • the two coding sequences are encoded by a bicistronic genetic element.
  • the coding regions for (a) the PSP and (b) the proinflammatory gene can be unidirectional, where each is under a separate regulatory control. In some embodiments the coding regions for both are bidirectional and drive in opposite directions. Each coding sequence is under a separate regulatory control.
  • Coexpression of the proinflammatory gene is designed to confer strong inflammatory stimulation of the myeloid cells, such as macrophages, and activate the surrounding tissue for inflammation.
  • Integrin activation domains [0332] Cell-cell and cell-substratum adhesion is mediated by the binding of integrin extracellular domains to diverse protein ligands; however, cellular control of these adhesive interactions and their translation into dynamic cellular responses, such as cell spreading or migration, requires the integrin cytoplasmic tails. These short tails bind to intracellular ligands that connect the receptors to signaling pathways and cytoskeletal networks (Calderwood DA, 2004, Integrin Activation, Journal of Cell Science 117, 657-666, incorporated herein in its entirety).
  • Integrins are heterodimeric adhesion receptors formed by the non-covalent association of ⁇ and ⁇ subunits. Each subunit is a type I transmembrane glycoprotein that has relatively large extracellular domains and, with the exception of the ⁇ 4 subunit, a short cytoplasmic tail. Individual integrin family members have the ability to recognize multiple ligands. Integrins can bind to a large number of extracellular matrix proteins (bone matrix proteins, collagens, fibronectins, fibrinogen, laminins, thrombospondins, vitronectin, and von Willebrand factor), reflecting the primary function of integrins in cell adhesion to extracellular matrices.
  • extracellular matrix proteins bone matrix proteins, collagens, fibronectins, fibrinogen, laminins, thrombospondins, vitronectin, and von Willebrand factor
  • Integrin ⁇ 2 subfamily consists of four different integrin receptors, ⁇ M ⁇ 2 (CD11b/CD18, Mac-1, CR3, Mo-1), ⁇ L ⁇ 2 (CD11a/CD18, LFA-1), ⁇ X ⁇ 2 (CD11c/CD18), and ⁇ D ⁇ 2 (CD11d/CD18).
  • leukocyte integrins are involved in virtually every aspect of leukocyte function, including the immune response, adhesion to and transmigration through the endothelium, phagocytosis of pathogens, and leukocyte activation.
  • the ⁇ subunits of all ⁇ 2 integrins contain an inserted region of ⁇ 200 amino acids, termed the I or A domain. Highly conserved I domains are found in several other integrin ⁇ subunits and other proteins, such as certain coagulation and complement proteins. I domains mediate protein- protein interactions, and in integrins, they are integrally involved in the binding of protein ligands.
  • I domains dominate the ligand binding functions of their integrins, other regions of the ⁇ subunits do influence ligand recognition.
  • a mAb (OKM1) recognizing an epitope outside the I domain but in the ⁇ M subunit inhibits ligand binding; and the EF-hand regions in ⁇ L ⁇ 2 and ⁇ 2 ⁇ 1, integrins with I domains in their ⁇ subunits, contribute to ligand recognition.
  • the ⁇ M subunit, and perhaps other ⁇ subunits contains a lectin-like domain, which is involved in engagement of non-protein ligands, and occupancy may modulate the function of the I domain.
  • integrins lack enzymatic activity
  • signaling is instead induced by the assembly of signaling complexes on the cytoplasmic face of the plasma membrane. Formation of these complexes is achieved in two ways; first, by receptor clustering, which increases the avidity of molecular interactions thereby increasing the on-rate of binding of effector molecules, and second, by induction of conformational changes in receptors that creates or exposes effector binding sites.
  • receptor clustering which increases the avidity of molecular interactions thereby increasing the on-rate of binding of effector molecules
  • induction of conformational changes in receptors that creates or exposes effector binding sites Within the ECM, integrins have the ability to bind fibronectin, laminins, collagens, tenascin, vitronectin and thrombospondin. Clusters of integrin/ECM interactions form focal adhesions, concentrating cytoskeletal components and signaling molecules within the cell.
  • integrins serve as a binding site for ⁇ -actinin and talin which then recruit vinculin, a protein involved in anchoring F-actin to the membrane.
  • Talin is activated by kinases such as protein kinase C (PKC ⁇ ).
  • PKC ⁇ protein kinase C
  • Integrins are activated by selectins. Leucocytes express L-selectin, activated platelets express P-selectin, and activated endothelial cells express E-and P-selectin. P-selectin-mediated adhesion enables chemokine-or platelet-activating factor-triggered activation of ⁇ 2 integrins, which stabilizes adhesion.
  • chemokines from adherent leucocytes.
  • the cytoplasmic domain of P-selectin glycoprotein ligand 1 formed a constitutive complex with Nef- associated factor 1.
  • Src kinases phosphorylated Nef-associated factor 1, which recruit the phosphoinositide-3-OH kinase p85-p110 ⁇ heterodimer and result in activation of leukocyte integrins.
  • E-selectin ligands transduce signals that also affect ⁇ 2 integrin function. Selectins trigger activation of Src family kinases.
  • SFKs activated by selectin engagement phosphorylate the immunoreceptor tyrosine-based activation motifs (ITAMs) in the cytoplasmic domains of DAP12 and FcR ⁇ .
  • ITAMs immunoreceptor tyrosine-based activation motifs
  • CD44 is sufficient to transduce signals from E- selectin.
  • CD44 triggers the inside-out signaling of integrins.
  • a final common step in integrin activation is binding of talin to the cytoplasmic tail of the ⁇ subunit.
  • Kindlins, another group of cytoplasmic adaptors bind to a different region of integrin ⁇ tails.
  • Kindlins increase the clustering of talin-activated integrins.
  • the intracellular domain of the recombinant CFP comprises an integrin activation domain.
  • the integrin activation domain comprises an intracellular domain of a selectin, for example, a P-selectin, L-selectin or E-selectin.
  • the intracellular domain of the recombinant CFP comprises an integrin activation domain of laminin.
  • the intracellular domain of the recombinant CFP comprises an integrin activation domain for activation of Talin.
  • the intracellular domain of the recombinant CFP comprises an integrin activation domain fused to the cytoplasmic end of the phagocytic receptor ICD domain.
  • Chimeric Receptor for Enhancing Antigen Cross presentation [0341] In some embodiments, the recombinant nucleic acid encodes a domain capable of enabling cross presentation of antigens.
  • MHC class I molecules present self-or pathogen-derived antigens that are synthesized within the cell, whereas exogenous antigens derived via endocytic uptake are loaded onto MHC class II molecules for presentation to CD4+ T cells.
  • DC can process exogenous antigens into the MHC-I pathway for presentation to CD8+ T cells. This is referred to as cross presentation of antigens. Soluble or exogenous antigenic components may get degraded by lysosomal proteases in the vacuoles and cross presented by DCs, instead of following the endocytic pathway.
  • chaperones such as heat shock protein 90 (Hsp90) have shown to help cross present antigens by certain APCs.
  • HSP-peptide complexes are known to be internalized by a distinct group of receptors compared to free polypeptides. These receptors were from the scavenger receptor families and included LOX-1, SREC-I/SCARF-I, and FEEL1/Stabilin-1. Both SREC-I and LOX-1 have been shown to mediate the cross presentation of molecular chaperone bound antigens and lead to activation of CD8 + T lymphocytes.
  • SREC-1 scavenger receptor expressed by endothelial cells
  • SREC-I scavenger receptor expressed by endothelial cells
  • SREC-I contains 10 repeats of EGF-like cysteine-rich motifs in the extracellular domain.
  • the structure of SREC-I was shown to be similar to that of a transmembrane protein with 16 EGF-like repeats encoded by the Caenorhabditis elegans gene ced-I, which functions as a cell surface phagocytic receptor that recognizes apoptotic cells.
  • Cross presentation of cancer antigens through the Class-I MHC pathway results in enhanced CD8+ T cell response, which is associated with cytotoxicity and therefore beneficial in tumor regression.
  • the intracellular domain of the PFP comprises a SREC1 intracellular domain. In some embodiments, the intracellular domain of the PFP comprises a SRECII intracellular domain. [0344] In some embodiments, the CFP comprises: an intracellular domain comprising a PSR intracellular signaling domain from SREC1 or SRECII. [0345] In some embodiments, the CFP comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising a CFP intracellular signaling domain from SREC1 or SRECII.
  • the CFP comprises: (i) a transmembrane domain, (ii) an intracellular domain comprising a intracellular signaling domain, and (iii) an extracellular domain from SREC1 or SRECII.
  • TRANSMEMBRANE DOMAIN OF CHIMERIC FUSION PROTEIN [0347]
  • the TM encoded by the recombinant nucleic acid comprises a sequence encoding a domain of a scavenger receptor (SR).
  • the TM can be the TM domain of or derived from any one or more of: lectin, dectin 1, mannose receptor (CD206), SRA1, MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, SRCRB4D, SSC5D, and CD169.
  • the TM domains are about 20-30 amino acids long.
  • TM domains of SRs are about 20-30 amino acids long.
  • the TM domain or the ICD domain of the CFP is not derived from Megf10, Bai1 or MerTK.
  • the ICD of the CFP does not comprise a CD3 ⁇ intracellular domain.
  • the TM is derived from the same phagocytic receptor as the ICD.
  • the TM region is derived from a plasma membrane protein.
  • the TM can be selected from an Fc receptor (FcR).
  • FcR Fc receptor
  • nucleic acid sequence encoding domains from specific FcRs are used for cell-specific expression of a recombinant construct.
  • An FCR-alpha region comprising the TM domain may be used for a myeloid cell, such as a macrophage, specific expression of the construct.
  • FcR ⁇ recombinant protein can be expressed in mast cells.
  • the PFP comprises the TM of FcR ⁇ .
  • the PFP comprises both the FcR ⁇ and ICD domains.
  • the PFP comprises both the FcR ⁇ and ICD domains.
  • the TM domain is derived from CD8.
  • the TM is derived from CD2.
  • the TM is derived from FcR ⁇ . EXTRACELLULAR DOMAIN OF CHIMERIC FUSION PROTEIN [0357]
  • the extracellular domain of a CFP provided herein comprises an antigen binding domain that binds to one or more targets.
  • the binding targets may be antigens or ligands.
  • a binding target may be an antigen on a target cell.
  • the target binding domain is specific for the target.
  • the extracellular domain can include an antibody or an antigen-binding domain selected from intrabodies, peptibodies, nanobodies, single domain antibodies. SMIPs, and multispecific antibodies.
  • an antibody fragment comprises a portion of an intact antibody, such as the antigen binding or variable region of the intact antibody.
  • an anti-HIV antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • Antibody fragments include, but are not limited to, Fab, Fab’, Fab’-SH, F(ab’) 2 , Fv, diabody, linear antibodies, multispecific formed from antibody fragments antibodies and scFv fragments, and other fragments described below.
  • the antibody is a full length antibody, e.g., an intact IgG1 antibody or other antibody class or isotype as described herein. (See, e.g., Hudson et al. Nat. Med.9:129-134 (2003); Pluckthiin, The Pharmacology of Monoclonal Antibodies, vol.113, pp.269-315 (1994); Hollinger et al., Proc. Natl. Acad. Sci.
  • a full length antibody, intact antibody, or whole antibody is an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g., E. coli or phage), as described herein.
  • An Fv is the minimum antibody fragment that contains a complete antigen-recognition and antigen-binding site.
  • a single-chain Fv is an antibody fragment that comprises the V H and V L antibody domains connected into a single polypeptide chain.
  • the sFv polypeptide can further comprise a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding.
  • a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding.
  • a diabody is a small antibody fragment prepared by constructing an sFv fragment with a short linker (about 5-10 residues) between the VH and VL domains such that inter-chain but not intra- chain pairing of the V domains is achieved, resulting in a bivalent fragment.
  • Bispecific diabodies are heterodimers of two crossover sFv fragments in which the V H and V L domains of the two antibodies are present on different polypeptide chains. (See, e.g., EP 404,097; WO 93/11161; and Hollinger et al, Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993)).
  • DAbs Domain antibodies
  • VH and VL immunoglobulins
  • DAbs are the robust variable regions of the heavy and light chains of immunoglobulins (VH and VL, respectively). They are highly expressed in microbial cell culture, show favorable biophysical properties including, for example, but not limited to, solubility and temperature stability, and are well suited to selection and affinity maturation by in vitro selection systems such as, for example, phage display. DAbs are bioactive as monomers and, owing to their small size and inherent stability can be formatted into larger molecules to create drugs with prolonged serum half-lives or other pharmacological activities.
  • Fv and sFv are the only species with intact combining sites that are devoid of constant regions. Thus, they are suitable for reduced nonspecific binding during in vivo use.
  • sFv fusion proteins can be constructed to yield fusion of an effector protein at either the amino or the carboxy terminus of an sFv.
  • the antibody fragment also can be a “linear antibody. (See, e.g., U.S. Pat. No. 5,641,870). Such linear antibody fragments can be monospecific or bispecific.
  • the extracellular domain includes a Fab binding domain. In yet other such embodiments, the extracellular domain includes a scFv.
  • the chimeric antigen receptor comprises an extracellular antigen binding domain is derived from the group consisting of an antigen-binding fragment (Fab), a single- chain variable fragment (scFv), a nanobody, a VH domain, a VL domain, a single domain antibody (sdAb), a VNAR domain, and a VHH domain, a bispecific antibody, a diabody, or a functional fragment of any thereof.
  • Fab antigen-binding fragment
  • scFv single-chain variable fragment
  • nanobody a VH domain, a VL domain, a single domain antibody (sdAb), a VNAR domain, and a VHH domain, a bispecific antibody, a diabody, or a functional fragment of any thereof.
  • the antigen-binding fragment Fab
  • a single-chain variable fragment scFv
  • a nanobody a V H domain, a V L domain, a single domain antibody (sdAb), a VNAR domain, and a V HH domain, a bispecific antibody, a diabody, or a functional fragment of any thereof specifically bind to one or more antigens.
  • the antigen is a cancer antigen
  • the target cell is a target cancer cell.
  • the antigen for a target cell is selected from the group consisting of CD3, CD4, CD5, CD7, CD19, CCR2, CCR4, CD30, CD37, TCRB1/2, TCR ⁇ , TCR ⁇ .
  • the target antigen is an autoimmune antigen.
  • the target cell is a B cell.
  • the target cell is a B cell that produces autoantibodies.
  • the target antigen is Dsg1 or Dsg3.
  • Various cancer antigen targets can be selected from cancer antigens known to one of skill in the art. Depending on the cancer and the cell type involved cancer antigens are mutated native proteins. The antigen binding domains are screened for specificity towards mutated/cancer antigens and not the native antigens.
  • the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: MUC16, CCAT2, CTAG1A, CTAG1B, MAGE A1, MAGEA2, MAGEA3, MAGEA4, MAGEA6, PRAME, PCA3, MAGEC1, MAGEC2, MAGED2, AFP, MAGEA8, MAGE9, MAGEA11, MAGEA12, IL13RA2, PLAC1, SDCCAG8, LSP1, CT45A1, CT45A2, CT45A3, CT45A5, CT45A6, CT45A8, CT45A10, CT47A1, CT47A2, CT47A3, CT47A4, CT47A5, CT47A6, CT47A8, CT47A9, CT47A10, CT47A11, CT47A12, CT47B1, SAGE1, and CT55.
  • the mutated/cancer antigens MUC16, CCAT2, CTAG1A, CTAG1B, MAGE A1, MAGEA2, MAGEA3, MAGEA4, MAGEA
  • the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: CD2, CD3, CD4, CD5, CD7, CD8, CD20, CD30, CXCR4, CD45, CD56, where the cancer is a T cell lymphoma.
  • the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: IDH1, ATRX, PRL3, or ETBR, where the cancer is a glioblastoma.
  • the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: CA125, ⁇ -hCG, urinary gonadotropin fragment, AFP, CEA, SCC, inhibin or extradiol, where the cancer is ovarian cancer.
  • the cancer antigen for a target cancer cell may be CD5.
  • the cancer antigen for a target cancer cell may be HER2.
  • the cancer antigen for a target cancer cell may be EGFR Variant III.
  • the cancer antigen for a target cancer cell may be CD19.
  • the SR subunit region comprises an extracellular domain (ECD) of the scavenger receptor.
  • ECD of the scavenger receptor comprises an ECD domain of the SR comprising the ICD and the TM domains.
  • the target antigen is the SR-ligand on the cancer cell, for example, any one of the ligand components from Table 2 or Table 3.
  • the SR-ECD contributes to the binding of the phagocyte to the target cell, and in turn is activated, and activates the phagocytosis of the target cell.
  • the CFP comprises an ECD or portion thereof of a scavenger receptor.
  • the CFP comprises an ICD or portion thereof of a scavenger receptor. In some embodiments, the CFP comprises a TM domain of a scavenger receptor. In some embodiments, the ECD encoded by the recombinant nucleic acid comprises a domain selected from the group consisting of lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169.
  • lectin dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1),
  • the extracellular domains of most scavenger receptors contain scavenger receptors with a broad binding specificity that may be used to discriminate between self and non-self in the nonspecific antibody- independent recognition of foreign substances.
  • the type I and II class A scavenger receptors (SR- AI1 and SR-AII) are trimeric membrane glycoproteins with a small NH2-terminal intracellular domain, and an extracellular portion containing a short spacer domain, an a-helical coiled-coil domain, and a triple-helical collagenous domain.
  • the type I receptor additionally contains a cysteine-rich COOH-terminal (SRCR) domain.
  • the SR ECD is designed to bind to pro-apoptotic cells.
  • the scavenger receptor ECD comprises a binding domain for a cell surface molecule of a cancer cell or an infected cell.
  • the extracellular domain of the PR subunit is linked by a linker to a target cell binding domain, such as an antibody or part thereof, specific for a cancer antigen.
  • the extracellular antigen binding domain comprises one antigen binding domain.
  • the extracellular antigen binding domain comprises more than one binding domain.
  • the binding domain are scFvs.
  • FIG. 2 shows a diagrammatic representation of an embodiment, where the PFP targets a single target on a cancer cell (left) or multiple targets (right). The one or more than one scFvs are fused to the recombinant PR at the extracellular domain.
  • the scFv fraction and the extracellular domain of the PR are linked via a linker.
  • the ECD antigen binding domain can bind to an intracellular antigen.
  • the intracellular antigen is a cancer antigen.
  • the extracellular antigen binding domain binds to the target ligand with an affinity of less than 1000 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 500 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 450 nM.
  • the extracellular antigen binding domain binds to the target ligand with an affinity of less than 400 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 350 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 250 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 200 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 100 nM.
  • the extracellular antigen binding domain binds to the target ligand with an affinity ranging between than 200 nM to 1000 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity ranging between than 300 nM to 1.5 mM. In some embodiments, the antigen binding domain binds to the target ligand with an affinity > 200 nM, > 300 nM or >500 nM. [0384] In some embodiments, the extracellular antigen binding domain binds to the target ligand, where the target ligand is a T cell, the binding characteristics are such that the target T cell is not triggered to activate T cell mediated lysis of the engineered cell.
  • Linkers may be used to link any of the polypeptides or peptide domains of the present disclosure.
  • the PFP fusion proteins described herein may comprises one or more linkers.
  • one or more of the domains and subunits of a PFP fusion protein can be directly fused to another domain or subunit, or can be connected to another domain or subunit via a linker.
  • the extracellular antigen binding domains comprising the antibody specific for the antigen on a target cell, parts of an antibody that can specifically bind to an antigen on a target cell, or scFvs specific for an antigen on a target cell are linked to the TM domain or other extracellular domains by a linker.
  • linkers are short peptide sequences.
  • the linker may be as simple as a covalent bond, or it may be a polymeric linker many atoms in length.
  • the linker is a polypeptide or based on amino acids. In other embodiments, the linker is not peptide-like. In certain embodiments, the linker is a covalent bond (e.g., a carbon-carbon bond, disulfide bond, carbon-heteroatom bond, etc.). [0388] In some embodiments, the linker is an amino acid or a plurality of amino acids (e.g., a peptide or protein). In some embodiments, the linker is a bond (e.g., a covalent bond), an organic molecule, group, polymer, or chemical moiety.
  • the linker is about 3 to about 104 (e.g., 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, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100) amino acids in length.
  • the linkers are stretches of Glycine and one or more Serine residues.
  • amino acids preferred for short peptide linkers include but are not limited to threonine (Thr), serine (Ser), proline (Pro), glycine (Gly), aspartic acid (Asp), lysine (Lys), glutamine (Gln), asparagine (Asn), and alanine (Ala) arginine (Arg), phenylalanine (Phe), glutamic acid (Glu).
  • Thr threonine
  • Ser serine
  • Pro proline
  • Gly aspartic acid
  • Als glutamine
  • Aln glutamine
  • Aln asparagine
  • Ala alanine
  • Arg arginine
  • Phe phenylalanine
  • glutamic acid Glu
  • Pro is a unique amino acid with a cyclic side chain which causes a very restricted conformation.
  • Pro-rich sequences are used as interdomain linkers, including the linker between the lipoyl and E3 binding domain in pyruvate dehydrogenase (GA2PA3PAKQEA3PAPA2KAEAPA3PA2KA).
  • the empirical linkers may be flexible linkers, rigid linkers, and cleavable linkers.
  • Sequences such as (G4S)x (where x is multiple copies of the moiety, designated as 1, 2, 3, 4, and so on) comprise a flexible linker sequence.
  • Other flexible sequences used herein include several repeats of glycine, e.g., (Gly)6 or (Gly)8.
  • a rigid linker may be used, for example, a linker (EAAAK)x, where x is an integer, 1, 2, 3, 4 etc. gives rise to a rigid linker.
  • Various linker lengths and flexibilities between domains or subunits of the fusion proteins provided herein can be employed, e.g., ranging from very flexible linkers of the form (GGGS)n, (GGGGS)n, and (G)n to more rigid linkers of the form (EAAAK)n, (SGGS)n, SGSETPGTSESATPES (see, e.g., Guilinger JP, Thompson DB, Liu DR.
  • n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15.
  • the linker comprises a (GGS)n motif, wherein n is 1, 3, or 7.
  • the linker comprises amino acid sequence SGGGGSG.
  • the linker comprises amino acid sequence GSGS. [0389]
  • the linkers are flexible. In some embodiments the linkers comprise a hinge region.
  • an extracellular spacer domain may position the binding domain away from the host cell surface to further enable proper cell/cell contact, binding, and activation.
  • the length of the extracellular spacer may be varied to optimize target molecule binding based on the selected target molecule, selected binding epitope, binding domain size and affinity.
  • an extracellular spacer domain is an immunoglobulin hinge region (e.g., IgGl, IgG2, IgG3, IgG4, IgA, IgD).
  • An immunoglobulin hinge region may be a wild type immunoglobulin hinge region or an altered wild type immunoglobulin hinge region.
  • a linker or a spacer used herein comprises an IgG4 hinge region, having a sequence: ESKYGPPCPPCP.
  • the hinge region comprises a hinge or a spacer comprising a sequence present in the extracellular regions of type 1 membrane proteins, such as CD8a, CD4, CD28 and CD7, which may be wild-type or variants thereof.
  • an extracellular spacer domain comprises all or a portion of an immunoglobulin Fc domain selected from: a CH1 domain, a CH2 domain, a CH3 domain, or combinations thereof.
  • the spacer or the linker may be further modified by post-translation modifications, such as glycosylation.
  • an extracellular spacer domain may comprise a stalk region of a type II C-lectin (the extracellular domain located between the C-type lectin domain and the transmembrane domain).
  • Type II C-lectins include CD23, CD69, CD72, CD94, NKG2A, and NKG2D.
  • an extracellular spacer domain may be derived from scavenger receptor MERTK.
  • the linker comprises at least 2, or at least 3 amino acids. In some embodiments, the linker comprises 4 amino acids. In some embodiments, the linker comprises 5 amino acids. In some embodiments, the linker comprises 6 amino acids. In some embodiments, the linker comprises 7 amino acids.
  • the linker comprises 8 amino acids. In some embodiments, the linker comprises 9 amino acids. In some embodiments, the linker comprises 8 amino acids. In some embodiments, the linker comprises 10 amino acids. In some embodiments the linker comprises greater than 10 amino acids. In some embodiments, the linker comprises 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids. In some embodiments there are 12 or more amino acids in the linker. In some embodiments, there are 14 or more amino acids in the linker. In some embodiments, there are 15 or more amino acids in the linker.
  • recombinant nucleic acids are prepared which encode one or more chimeric receptors that enhance phagocytosis in myeloid cells, such as macrophages, principally by blocking inhibitory signals.
  • Myeloid cells, such as macrophages especially in the tumor environment encounter phagocytosis dampening or inhibitory signals, such as CD47 mediated anti-phagocytic activity on target cells, e.g., cancer cells.
  • Chimeric receptors are generated which when expressed in a phagocytic cell blocks CD47 signaling.
  • CAR fusion protein may be designed for expression in a phagocytic cell that may enhance phagocytosis.
  • a composition comprising a recombinant nucleic acid encoding a chimeric antigen receptor (CAR) fusion protein (CFP) comprising: (a) a subunit comprising: (i) an extracellular domain; and (ii) a transmembrane domain; (b) an extracellular antigen binding domain specific to CD47 of a target cell; wherein: the extracellular domain of the subunit and the extracellular antigen binding domain are operatively linked; and the subunit does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase.
  • CAR chimeric antigen receptor
  • the extracellular antigen binding domain is derived from signal- regulatory protein alpha (SIRP ⁇ ). In some embodiments, the extracellular antigen binding domain is derived from signal-regulatory protein alpha (SIRP ⁇ ). In some embodiments, the transmembrane domain is derived from SIRP ⁇ . In some embodiments, the transmembrane domain is derived from SIRP ⁇ . [0394] In some embodiments, the additional CAR fusion protein (CFP) may be co-transfected with the recombinant PFP described above.
  • SIRP ⁇ signal- regulatory protein alpha
  • SIRP ⁇ signal-regulatory protein alpha
  • the transmembrane domain is derived from SIRP ⁇ . In some embodiments, the transmembrane domain is derived from SIRP ⁇ .
  • the additional CAR fusion protein (CFP) may be co-transfected with the recombinant PFP described above.
  • the scavenger receptor intracellular domain comprises a second intracellular domain comprising a signaling domain that activates phagocytosis; or a proinflammatory domain at the cytoplasmic terminus, which are operably linked.
  • the signaling domain that activates phagocytosis is derived from a receptor selected from the group consisting of the receptors listed in Table 2.
  • the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs.
  • ITAM Immunoreceptor Tyrosine-based Activation Motif
  • YXXL/I-X6-8-YXXL/I a negatively charged amino acid
  • D/E negatively charged amino acid
  • the phagocytic signaling domain in the intracellular region comprises a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain).
  • PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR.
  • PI3 kinase recruitment to the binding domain leads to the Akt mediated signaling cascade and activation of phagocytosis.
  • the PI3K-Akt signaling pathway is important in phagocytosis, regulation of the inflammatory response, and other activities, including vesicle trafficking and cytoskeletal reorganization.
  • the PI3kinase recruitment domain is an intracellular domain in a plasma membrane protein, which has tyrosine residues that can be phosphorylated, and which can in turn be recognized by the Src homology domain (SH2) domain of PI3Kp85.
  • SH2 domain of p85 recognizes the phosphorylated tyrosines on the cytosolic domain of the receptor.
  • PIP3 phosphatidylinositol-3,4,5-trisphosphate
  • Akt phosphatidylinositol-3,4,5-trisphosphate
  • PDK1 constitutively active 3′-phosphoinositide-dependent kinase 1
  • Phosphorylation of these two residues causes the activation of Akt which in turn phosphorylates, among other substrates, the enzyme glycogen synthase kinase- 3 (GSK-3).
  • GSK-3 has two isoforms, GSK-3 ⁇ and GSK-3 ⁇ both of which are constitutively active.
  • the isoforms are structurally related but functionally nonredundant. Inactivation of GSK-3 is observed when the residues Ser21 in GSK-3 ⁇ or Ser9 in GSK-3 ⁇ , located in their regulatory N- terminal domains, are phosphorylated by Akt and other kinases. Inhibition of GSK-3 by phosphorylation is important for the modulation of the inflammation and in phagocytosis processes.
  • a recombinant PFP comprises (a) an extracellular CD47 binding domain SIRP ⁇ , (b) a SIRP ⁇ transmembrane domain, and (c) an intracellular domain of SIRP ⁇ .
  • SIRP ⁇ signaling can activate pro-phagocytic signaling by engaging DAP12 activation.
  • Various members of the family transduce checkpoint signal upon contact with sialylated glycans on membrane proteins.
  • the intracellular domains of the Siglec proteins comprise multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs).
  • Phosphorylation of the Tyrosine residues at the ITIM motif recruit either of two SH2 domain– containing negative regulators: the inositol phosphatase SHIP (Src homology 2–containing inositol polyphosphate 5-phosphatase) or the tyrosine phosphatase SHP-1 (Src homology 2–containing protein tyrosine phosphatase-1).
  • SHIP inositol phosphatase SHIP
  • tyrosine phosphatase SHP-1 Src homology 2–containing protein tyrosine phosphatase-1
  • ITIMs can also bind to another tyrosine phosphatase, SHP-2, but evidence for SHP-2 playing a functional role in ITIM-mediated inhibition is less clear than for the other mediators. Therefore, activation of the Siglec membrane proteins at the extracellular ligand binding domain by binding with a sialic acid residue, (e.g. in sialylated membrane glycan proteins), the ITIMs receive the intracellular signals, which are phosphorylated, and initiate the SHP mediated signaling for immunomodulation, including reduction in phagocytic potential.
  • SHP-2 tyrosine phosphatase
  • the composition described herein comprises a recombinant nucleic acid construct encoding a chimeric Siglec receptor (SgR) fusion protein (SgFP), comprising: (a) a SgR subunit which comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (a) an extracellular domain comprising an antigen binding domain specific to a sialylated glycan of a cell surface protein of a target cell; (b) wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the SgFP does not comprise a functional intracellular domain of an endogenous receptor that binds a sialylated glycan, or (ii) the SgFP comprises an intracellular signaling domain that activates phagocytosis or an inflammatory pathway.
  • SgR chimeric Siglec receptor
  • SgFP chimeric Siglec receptor
  • the chimeric receptor is deficient in an intracellular domain, and therefore acts as a blocker for Siglec induced immunoregulatory intracellular signaling. Such is achieved by deletion of the nucleic acid region encoding the intracellular domain and cloning the remainder of the coding sequence of the Siglec receptor.
  • This construct can be designated as a siglec intracellular domain deletion construct [Siglec ⁇ ICD].
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a cancer antigen specific scFv fused with the extracellular domain (ECD) of a siglec receptor. This allows targetability of the construct to the cancer cell.
  • the chimeric receptor comprises the TM and the ICD of the siglec receptor, which can be the endogenous ICD, or the ICD fused with additional phagocytosis promoting domains, such as PI3K binding domain or the domains.
  • a chimeric receptor comprising an extracellular siglec domain is co- expressed with a sialidase.
  • the nucleic acid encoding a sialidase may be incorporated in the expression vector expressing the chimeric domain with a signal sequence for secretion.
  • the sialidase is expressed by the same cell that expressed the CAR-siglec receptor, expression of sialidase deprives the ECD of the siglec from binding to its natural ligand, but is activated by the scFv binding to its receptor, thereby ensuring the specificity of action of the chimeric receptor on a cancer-antigen expressing cell.
  • the chimeric receptors comprise one or more domains from TREM proteins, fused at the extracellular region with an antigen binding domain that can specifically bind to a cancer antigen, such as a cancer antigen-specific antibody or part or fragment thereof.
  • recombinant nucleic acids encoding a TREM chimeric antigen receptor encode a fusion proteins that comprises: (a) the at least a TREM transmembrane domain (TM) and a TREM intracellular domain (ICD); and (b) an extracellular domain (ECD) comprising an antigen binding domain that can specifically bind to a cancer antigen.
  • the fusion proteins are designed to target cancer cells and bind to the target cancer cells via the ECD comprising the antigen binding domain, and the binding triggers and enhance phagocytosis via signaling through the TREM TM and/or the intracellular domains.
  • the transmembrane domain of TREM trimerizes with DAP12 transmembrane domains and trigger intracellular pro-phagocytosis signaling cascade.
  • the TREM domains are contributed by TREM1, or by TREM2, or by TREM3 members.
  • the extracellular antigen binding domain is fused to the extracellular terminus of the TREM domains through a short spacer or linker.
  • the extracellular antigen binding domain comprises an antibody, specific to a cancer antigen.
  • the extracellular antigen binding domain comprises an antibody or an antigen binding part thereof that binds specifically to an antigen on the surface of a cancer cell.
  • the extracellular antigen binding domain is an antibody specific for a cancer antigen. In some embodiments, the extracellular antigen binding domain is a fraction of an antibody, wherein the fragment can bind specifically to the cancer antigen on a cancer cell. In some embodiments the antigen binding domain comprises a single chain variable fraction (scFv) specific for a cancer antigen binding domain.
  • the chimeric fusion protein comprises an extracellular domain (ECD) targeted to bind to CD5 (CD5 binding domain), for example, comprising a heavy chain variable region (VH) having an amino acid sequence as set forth in SEQ ID NO: 1.
  • the chimeric CFP comprises a CD5 binding heavy chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 1.
  • the extracellular domain (ECD) targeted to bind to CD5 comprises a light chain variable domain (V L ) having an amino acid sequence as set forth in SEQ ID NO: 2.
  • the chimeric CFP comprises a CD5 binding light chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 2.
  • the CFP comprises an extracellular domain targeted to bind to HER2 (HER2 binding domain) having for example a heavy chain variable domain amino acid sequence as set forth in SEQ ID NO: 8 and a light chain variable domain amino acid sequence as set forth in SEQ ID NO: 9.
  • the CFP comprises a HER2 binding heavy chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 8.
  • the CFP comprises a HER2 binding light chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 9.
  • the CFP comprises a hinge connecting the ECD to the transmembrane (TM).
  • the hinge comprises the amino acid sequence of the hinge region of a CD8 receptor.
  • the CFP may comprise a hinge having the amino acid sequence set forth in SEQ ID NO: 7 (CD8 ⁇ chain hinge domain).
  • the PFP hinge region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 7.
  • the CFP comprises a CD8 transmembrane region, for example having an amino acid sequence set forth in SEQ ID NO: 6.
  • the CFP TM region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 6.
  • the CFP comprises an intracellular domain having an FcR domain.
  • the CFP comprises an FcR domain intracellular domain comprises an amino acid sequence set forth in SEQ ID NO: 3, or at least a sequence having 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 3.
  • the CFP comprises an intracellular domain having a PI3K recruitment domain.
  • the PI3K recruitment domain comprises an amino sequence set forth in SEQ ID NO: 4.
  • the PI3K recruitment domain comprises an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 4.
  • the CFP comprises an intracellular domain having a CD40 intracellular domain.
  • the CD40 ICD comprises an amino sequence set forth in SEQ ID NO: 5.
  • the CD40 ICD comprises an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 5.
  • Table 4 - Sequences of chimeric antigen receptors and domains thereof [0410] Table 4 shows exemplary sequences of chimeric fusion protein domains and/or fragments thereof that are meant to be non-limiting for the disclosure. Underlines denote the CDR sequences in sequential order of CDR1, CDR2 and CDR3 for the respective heavy and light chains in accordance to the Kabat numbering system.
  • the instant disclosure therefore provides all the necessary building blocks for a construct design that contemplates using any one of the domains in combination with another to build a chimeric protein with the information provided herein. Any such domain can be used to generate a phagocytic receptor fusion protein, i..e., a CFP, or a CAR or a dual CFP-CAR construct. Characteristics of the PFP: [0412] The PFP structurally incorporates into the cell membrane of the cell in which it is expressed. Specific leader sequences in the nucleic acid construct, such as the signal peptide directs plasma membrane expression of the encoded protein.
  • the transmembrane domain encoded by the construct incorporates the expressed protein in the plasma membrane of the cell.
  • the transmembrane domain comprises a TM domain of an FcR- alpha receptor, which dimerizes with endogenous FcR ⁇ receptors in the myeloid cells, such as macrophages, ensuring myeloid cell specific expression.
  • the PFP renders the cell expressing it as potently phagocytic.
  • the cell When the recombinant nucleic acid encoding the PFP is expressed in a cell, the cell exhibits an increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid.
  • the recombinant nucleic acid When the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid.
  • the recombinant nucleic acid when expressed in a cell the cell exhibits at least 2-fold increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid.
  • the recombinant nucleic acid when expressed in a cell the cell exhibits at least 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold 30- fold or at least 5-fold increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid.
  • the composition comprises a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the PFP to the antigen of the target cell, the killing or phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP.
  • PR phagocytic or tethering receptor
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 11% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 12% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 13% compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 14% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 15% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 16% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 17% compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 18% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 19% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 30% compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 40% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 50% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 60% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 70% compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 80% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 90% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 100% compared to a cell not expressing the PFP. [0415] In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 4-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 6-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 8-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10-fold compared to a cell not expressing the PFP.
  • the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 50-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by about 50-fold compared to a cell not expressing the PFP. [0416] In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 30% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 40% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 50% compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 60% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 70% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 80% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 90% compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 100% compared to a cell not expressing the PFP. [0417] In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 4-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 6-fold compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 8-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 10-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 20-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 30-fold compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 40-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 50-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 100-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by about 100-fold compared to a cell not expressing the PFP.
  • the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP.
  • the cell when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased cytokine production.
  • the cytokine can comprise any one of: IL-1, IL-6, IL-12, IL-23, TNF, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27 and interferons.
  • the recombinant nucleic acid when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased cell migration.
  • Enhanced cell migration may be detected in cell culture by standard motility assays.
  • actin filament rearrangements may be detected and monitored using phalloidin staining and fluorescent microscopy.
  • time-lapsed microscopy is used for the purpose.
  • the recombinant nucleic acid when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased immune activity. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of MHC II. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of CD80.
  • the cell when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of CD86. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased iNOS production. [0422] In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits increased trogocytosis of a target cell expressing the antigen of a target cell compared to a cell not expressing the recombinant nucleic acid.
  • a design of a CAR is directed to enhancing a cytotoxic potential of a T cell in which the CAR is expressed.
  • the cytolytic activity of a T cell expressing a CAR may be at least 2-fold higher than a T cell that does not express a CAR in presence of a target cell.
  • a target cell can be a cell that comprises the target antigen, wherein the extracellular antigen binding domain of the CAR is designed to specifically bind to the target antigen.
  • the cytolytic activity of a T cell expressing a CAR may be at least 3- fold higher, 4-fold higher, 5-fold higher, 6-fold higher, 7-fold higher, 8-fold higher, 9-fold higher, 10-fold higher, 15-fold higher, 20-fold higher or 50 fold higher than a T cell that does not express a CAR in presence of a target cell.
  • a target cell is a cell that comprises the target antigen, wherein the extracellular antigen binding domain of the CAR is designed to specifically bind to the target antigen.
  • a target cell is a cancer cell, which expresses a cancer antigen to which the extracellular antigen binding domain of the CAR binds to, thereby activating the T cell and enhancing cytotoxicity.
  • an unmodified T cell for example can also bind to an potentially destroy the same target cell as a CAR-expressing T cell, provided that the unmodified T cell has a TCR that binds to the target antigen.
  • an unmodified T cell that binds to the same target as the CAR-expressing T cells of the invention, but at least shows a lower potential for target cell cytotoxicity compared to a CAR-expressing cell.
  • the chimeric receptors may be glycosylated, pegylated, and/or otherwise post-translationally modified.
  • glycosylation, pegylation, and/or other posttranslational modifications may occur in vivo or in vitro and/or may be performed using chemical techniques.
  • any glycosylation, pegylation and/or other posttranslational modifications may be N-linked or O-linked.
  • any one of the chimeric receptors may be enzymatically or functionally active such that, when the extracellular domain is bound by a ligand, a signal is transduced to polarize myeloid cells, such as macrophages.
  • the method for preparing CAR-Ps comprise the steps of (1) screening for PSR subunit framework; (2) screening for antigen binding specificity; (3) CAR-P recombinant nucleic acid constructs; (4) engineering cells and validation.
  • Screening for PSR subunit framework As described above, the design of the receptor comprises at least of one phagocytic receptor domain, which enables the enhanced signaling of phagocytosis. In essence a large body of plasma membrane proteins can be screened for novel phagocytic functions or enhancements domains. Methods for screening phagocytic receptor subunits are known to one of skill in the art. Additional information can be found in The Examples section.
  • primers and probes are constructed for identification, and or isolation of a protein, a polypeptide or a fragment thereof or a nucleic acid fragment encoding the same.
  • the primer or probe may be tagged for experimental identification.
  • tagging of a protein or a peptide may be useful in intracellular or extracellular localization.
  • antibodies and fragments thereof include, but are not limited to IgAs, IgDs, IgEs, IgGs, IgMs, Fab fragments, F(ab')2 fragments, monovalent antibodies, scFv fragments, scRv-Fc fragments, IgNARs, hcIgGs, V HH antibodies, nanobodies, and alphabodies.
  • Commercially available antibodies can be adapted to generate extracellular domains of a chimeric receptor.
  • antibodies examples include, but are not limited to: anti-HGPRT, clone 13H11.1 (EMD Millipore), anti-ROR1 (ab135669) (Abcam), anti-MUC1 [EP1024Y] (ab45167) (Abcam), anti-MUC16 [X75] (ab1107) (Abcam), anti-EGFRvIII [L8A4] (Absolute antibody), anti-Mesothelin [EPR2685 (2)] (ab134109) (Abcam), HER2 [3B5] (ab16901) (Abcam), anti-CEA (LS-C84299-1000) (LifeSpan BioSciences), anti-BCMA (ab5972) (Abcam), anti-Glypican 3 [9C2] (ab129381) (Abcam), anti-FAP (ab53066) (Abcam), anti-EphA2 [RM-0051- 8F21] (ab73254) (Abcam), anti-GD2 (
  • the recombinant nucleic acid can be generated following molecular biology techniques known to one of skill in the art. The methods include but are not limited to designing primers, generating PCR amplification products, restriction digestion, ligation, cloning, gel purification of cloned product, bacterial propagation of cloned DNA, isolation and purification of cloned plasmid or vector. General guidance can be found in: Molecular Cloning of PCR Products: by Michael Finney, Paul E.
  • DNA, mRNA and Circular RNA In some embodiments, naked DNA or messenger RNA (mRNA) may be used to introduce the nucleic acid inside the cell. In some embodiments, DNA or mRNA encoding the PFP is introduced into the phagocytic cell by lipid nanopaticle (LNP) encapsulation. mRNA is single stranded and may be codon optimized.
  • LNP lipid nanopaticle
  • the mRNA may comprise one or more modified or unnatural bases such as 5’-Methylcytosine, or Pseudouridine.
  • mRNA may be 50-10,000 bases long.
  • the transgene is delivered as an mRNA.
  • the mRNA may comprise greater than about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10,000 bases.
  • the mRNA may be more than 10,000 bases long.
  • the mRNA may be about 11,000 bases long.
  • the mRNA may be about 12,000 bases long.
  • the mRNA comprises a transgene sequence that encodes a fusion protein.
  • LNP encapsulated DNA or RNA can be used for transfecting myeloid cells, such as macrophages, or can be administered to a subject.
  • circular RNA (circRNAs) encoding the PFP is used.
  • circular RNAs (circRNAs) the 3′ and 5′ ends are covalently linked, constitute a class of RNA. CircRNA may be delivered inside a cell or a subject using LNPs.
  • Myeloid cells such as macrophages, especially in the tumor microenvironment encounter phagocytosis dampening or inhibitory signals, such as CD47 mediated anti-phagocytic activity by target cells, e.g., cancer cells.
  • Cluster of Differentiation 47 (CD47) is a receptor belonging to the immunoglobulin superfamily. It can bind to integrins, and thrombospondin 1 (TSP- 1), and is ubiquitously expressed in human cells.
  • Target cells including tumor cells express CD47 as the ‘don’t eat me’ signals to evade phagocytosis mediated killing and removal.
  • Phagocytic cells can express signal regulatory protein alpha receptors, (SIRP) which bind to CD47.
  • SIRP family members are receptor-type transmembrane glycoproteins involved in the negative regulation of receptor tyrosine kinase-coupled signaling processes.
  • SIRP- ⁇ can be phosphorylated by tyrosine kinases. The phospho-tyrosine residues of this PTP have been shown to recruit SH2 domain containing tyrosine phosphatases (PTP), and serve as substrates of PTPs.
  • PTP SH2 domain containing tyrosine phosphatases
  • SIRP- ⁇ is another member of the SIRP family, which is found to interact with TYROBP/DAP12, a protein bearing immunoreceptor tyrosine-based activation motifs. It can trigger activation of myeloid cells when associated with TYROBD. This protein was also reported to participate in the recruitment of tyrosine kinase SYK.
  • chimeric receptors generated to functionally block CD47 signaling when expressed in a phagocytic cell.
  • compositions and methods for phagocytic enhancement of the engineered myeloid cells, such as macrophages, by blocking CD47 signal are provided herein.
  • the recombinant nucleic acids encoding the CFP receptors described herein are transfected or transduced into myeloid cells, such as phagocytic cells, alone or in combination with other recombinant phagocytic receptors for creating engineered macrophages for use in immunotherapy.
  • the recombinant phagocytic receptors comprise an intracellular domain of a scavenger receptor.
  • an additional binding domain or an additional therapeutic agent comprises a SIRP ⁇ antagonist, a SIRP ⁇ blocker, an antibody, a chimeric SIRP ⁇ receptor, a cytokine, a proinflammatory gene, a procaspase, or an anti-cancer agent.
  • an additional binding domain or an additional therapeutic agent comprises a PD1 antagonist, a PD1 blocker, an antibody, a chimeric PD1 receptor, a cytokine, a proinflammatory gene, a procaspase, or an anti-cancer agent.
  • composition comprising a recombinant nucleic acid encoding a CFP comprising: (a) a subunit comprising: (i) an extracellular domain that can specifically bind to CD47 on a target cell; and (ii) a transmembrane domain; wherein the extracellular domain of the subunit and the extracellular antigen binding domain are operably linked; and the subunit does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase.
  • the extracellular antigen binding domain is derived from signal-regulatory protein alpha (SIRP ⁇ ).
  • the extracellular antigen binding domain is derived from signal-regulatory protein beta (SIRP ⁇ ).
  • SIRP ⁇ does not bind to CD47.
  • the transmembrane domain is derived from SIRP ⁇ .
  • the transmembrane domain is derived from SIRP ⁇ .
  • the recombinant nucleic acid of this category lacks any intracellular domain, and is therefore a non-signaling receptor and blocker of CD47 signaling. This construct is referred to as SIRP- ⁇ ICD.
  • the receptor binds to CD47 on the target cell, but renders it’s signaling inert by not having a functional SIRP ⁇ intracellular domain, thereby reducing CD47 signaling and inhibition of phagocytosis of the CD47+ cells by myeloid cells, such as macrophages, that express the CFP.
  • myeloid cells such as macrophages
  • Over-expression of this construct can largely reduce CD47 mediated anti-phagocytic activity of myeloid cells, such as macrophages, by cancer cells.
  • endogenous SIRP ⁇ may be further inhibited, in addition to overexpression of the SIRP- ⁇ ICD.
  • siRNA can be designed specifically targeting the portion of the mRNA encoding the ICD of SIRP ⁇ , such that the siRNA does not reduce or affect the expression of the SIRP- ⁇ ICD.
  • the SIRP- ⁇ ICD and or the inhibitor of endogenous SIRP ⁇ may be expressed in a cell expressing a fusion protein comprising a phagocytic receptor and an extracellular antigen binding domain specific for cancer antigen. Chimeric antigen receptor for blocking anti-phagocytic signal and activating phagocytosis A.
  • a recombinant nucleic acid comprising a recombinant nucleic acid encoding a chimeric antigen receptor (CAR) fusion protein (CFP) comprising: (a) a transmembrane domain; (b) an extracellular antigen binding domain specific to CD47 of a target cell; wherein: the transmembrane domain and extracellular antigen binding domain specific to CD47 are operatively linked; and the CFP does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase, and, (b) an intracellular domain from a phagocytic receptor, that is capable of activating intracellular signaling to enhance phagocytosis.
  • CAR chimeric antigen receptor
  • CFP chimeric antigen receptor
  • the recombinant nucleic acid construct comprises a nucleic acid sequence encoding an intracellular signaling domain of a scavenger receptor.
  • This class of receptors are termed herein the “switch receptor”, because they are designed to convert (or switch) a phagocytosis inhibitory signal to a phagocytosis promoting signal.
  • the intracellular domain of the chimeric receptor may comprise the intracellular domain of a scavenger receptor, selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor, which is fused at the extracellular terminus with the extracellular domain comprising the SIRP ⁇ CD47 binding domain.
  • a scavenger receptor selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163,
  • the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs.
  • the phagocytic signaling domain in the intracellular region can comprise a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain).
  • PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR.
  • PI3 kinase recruitment to the binding domain leads to the Akt mediated signaling cascade and activation of phagocytosis.
  • the PI3K-Akt signaling pathway is important in phagocytosis, regulation of the inflammatory response, and other activities, including vesicle trafficking and cytoskeletal reorganization.
  • the PI3kinase recruitment domain is an intracellular domain in a plasma membrane protein, which has tyrosine residues that can be phosphorylated, and which can in turn be recognized by the Src homology domain (SH2) domain of PI3Kp85.
  • SH2 domain of p85 recognizes the phosphorylated tyrosines on the cytosolic domain of the receptor.
  • PIP 3 phosphatidylinositol-3,4,5-trisphosphate
  • Akt phosphatidylinositol-3,4,5-trisphosphate
  • PDK1 constitutively active 3′-phosphoinositide-dependent kinase 1
  • Phosphorylation of these two residues causes the activation of Akt which in turn phosphorylates, among other substrates, the enzyme glycogen synthase kinase- 3 (GSK-3).
  • GSK-3 has two isoforms, GSK-3 ⁇ and GSK-3 ⁇ both of which are constitutively active.
  • the isoforms are structurally related but functionally nonredundant. Inactivation of GSK-3 is observed when the residues Ser21 in GSK-3 ⁇ or Ser9 in GSK-3 ⁇ , located in their regulatory N- terminal domains, are phosphorylated by Akt and other kinases. Inhibition of GSK-3 by phosphorylation is important for the modulation of the inflammation and in phagocytosis processes.
  • a recombinant PFP comprises (a) an extracellular CD47 binding domain SIRP ⁇ , (b) a SIRP ⁇ transmembrane domain, and (c) an intracellular domain of SIRP ⁇ .
  • SIRP ⁇ signaling can activate pro-phagocytic signaling by engaging DAP12 activation.
  • B. Alteration of Sialic Acid-binding Signal Transduction [0445]
  • Various members of the family transduce checkpoint signal upon contact with sialylated glycans on membrane proteins.
  • the intracellular domains of the Siglec proteins comprise multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs).
  • Phosphorylation of the Tyrosine residues at the ITIM motif recruit either of two SH2 domain–containing negative regulators: the inositol phosphatase SHIP (Src homology 2–containing inositol polyphosphate 5-phosphatase) or the tyrosine phosphatase SHP-1 (Src homology 2– containing protein tyrosine phosphatase-1).
  • a leucine in the (Y+2) position favors binding to SHIP, whereas an isoleucine in the (Y–2) position favors SHP-1 binding.
  • ITIMs can also bind to another tyrosine phosphatase, SHP-2, but evidence for SHP-2 playing a functional role in ITIM-mediated inhibition is less clear than for the other mediators. Therefore, activation of the Siglec membrane proteins at the extracellular ligand binding domain by binding with a sialic acid residue, (e.g. in sialylated membrane glycan proteins), the ITIMs receive the intracellular signals, which are phosphorylated, and initiate the SHP mediated signaling for immunomodulation, including reduction in phagocytic potential.
  • the composition described herein comprises a recombinant nucleic acid construct encoding a chimeric Siglec receptor (SgR) fusion protein (SgFP), comprising: (a) a SgR subunit which comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (a) an extracellular domain comprising an antigen binding domain specific to a sialylated glycan of a cell surface protein of a target cell; (b) wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the SgFP does not comprise a functional intracellular domain of an endogenous receptor that binds a sialylated glycan, or (ii) the SgFP comprises an intracellular signaling domain that activates phagocytosis or an inflammatory pathway.
  • SgR chimeric Siglec receptor
  • SgFP chimeric Siglec receptor
  • Siglec family receptors comprise the membrane proteins, siglec 1 (CD169), siglec 2 (CD22), siglec 3 (CD33), siglec 4 (MAG), siglec 5, siglec 6, siglec 7, siglec 8, siglec 9, siglec 10, siglec 11, siglec 12, siglec 13, siglec 14, siglec 15, siglec 16.
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising an extracellular domain (ECD) of a Siglec receptor that can bind to sialylated residues on membrane proteins of a target cell, which comprises any one of the siglec family members.
  • ECD extracellular domain
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a transmembrane protein (TM) domain of a Siglec receptor.
  • the chimeric receptor is deficient in an intracellular domain, and therefore acts as a blocker for Siglec induced immunoregulatory intracellular signaling. Such is achieved by deletion of the nucleic acid region encoding the intracellular domain and cloning the remainder of the coding sequence of the Siglec receptor.
  • This construct can be designated as a siglec intracellular domain deletion construct [Siglec ⁇ ICD].
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising an extracellular domain (ECD) of a Siglec receptor that can bind to sialylated residues on membrane proteins of a target cell.
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a transmembrane protein (TM) domain of a Siglec receptor.
  • the chimeric receptor comprises a TM domain of an unrelated membrane protein, for example CD8 TM or CD2 TM domains.
  • the chimeric antigenic receptor comprising the Siglec ECD and/or Siglec TM is deficient in endogenous Siglec intracellular domain (ICD) (e.g., achieved by a deletion of the intracellular domain [Siglec ⁇ ICD]), and wherein an intracellular domain of an unrelated protein is fused to the cytoplasmic end of the construct.
  • ICD Siglec intracellular domain
  • Siglec 2, 3, 5, 6, 7, 8, 9, 10, 11, and 12 family members comprise 2 or more intracellular ITIM motifs.
  • the intracellular domains of the siglec proteins comprising the ITIM motifs are deleted to generate Siglec ⁇ ICD, and fused with an ICD of a phagocytosis promoting protein, thereby altering the inhibitory signal generated by binding of the siglec to its ligand (sialylated glycan) on a cancer cell, into a pro-inflammatory and phagocytosis promoting signal.
  • the unrelated protein can comprise an intracellular domain that can generate phagocytosis activation signals or pro-inflammatory signals, such as the intracellular domains of the proteins: MRC1, ItgB5, MERTK, ELMO, BAIL Tyro3, Axl, Traf6, Syk, MyD88, Zap70, PI3K, FcyR1, FcyR2A, FcyR2B2, FcyR2C, FcyR3A, FcER1, FcaRl, BAFF-R, DAP12, NFAM1, and CD79b intracellular domains.
  • the intracellular domain of the chimeric receptor may comprise the intracellular domain of a scavenger receptor, selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor, which is fused at the extracellular terminus with the extracellular domain comprising the Siglec sialylated glycan binding domain.
  • a scavenger receptor selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO
  • the phagocytic signaling domain in the intracellular region can comprise a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain).
  • PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR.
  • the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs.
  • ITAM Immunoreceptor Tyrosine-based Activation Motif
  • the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a cancer antigen specific scFv fused with the extracellular domain (ECD) of a siglec receptor.
  • ECD extracellular domain
  • the chimeric receptor comprises the TM and the ICD of the siglec receptor, which can be the endogenous ICD, or the ICD fused with additional phagocytosis promoting domains, such as PI3K binding domain or the domains.
  • the siglec ECD region is prevented from activation by a concurrently expressed sialidase.
  • the sialidase may be encoded by the construct in a monocistronic design, where the entire construct is expressed as a single polypeptide and is readily cleaved by an endogenous T2A cleavage; or may be bicistronic, where the coding sequence of the sialidase enzyme is transcribed as a separate mRNA.
  • the siglec receptors bind to sialylated residues ubiquitously present on membrane proteins, and can activate the downstream signals.
  • the chimeric construct described herein encodes an extracellular domain, a transmembrane domain and an intracellular domain of the siglec protein, but is fused with a cancer targeting scFv at the extracellular terminus, and encodes for a sialidase at the intracellular terminus.
  • the sialidase coding sequence is fused with the coding sequence of an N-terminal signal peptide that signals for secretion of the protein sialidase upon translation.
  • the enzyme Upon expression of the sialidase and its release into the extracellular environment, the enzyme removes the digests sialic acid residues from the extracellular membrane proteins in the environment proximal to the membrane of the cell expressing the construct. Since the sialidase is expressed by the same cell that expressed the CAR- siglec receptor, expression of sialidase deprives the ECD of the siglec from binding to its natural ligand, but is activated by the scFv binding to its receptor, thereby ensuring the specificity of action of the chimeric receptor on a cancer-antigen expressing cell.
  • the chimeric antigenic receptor functions as an anti-inflammatory and phagocytosis regulatory signaling protein by activation of the endogenous ICD domains of the siglec receptor through activation of the ITIMs.
  • the chimeric antigenic receptor comprises endogenous ICD domains that have a pro-inflammatory signaling moiety, and/or a depletion of the endogenous siglec ITIM comprising ICDs, each of which are described in the previous section, and can be combined in modular ways with the scFv comprising CAR described herein.
  • TREM Chimeric Antigenic Receptor Domains for Enhancing Phagocytosis TREM Domains
  • TREM receptors are important regulators of the immune response, due to their ability to either amplify or decrease PRR-induced signals.
  • This family of proteins includes the members: TREM 1, 2, 3. TREMs share common structural properties, including the presence of a single extracellular immunoglobulin-like domain of the V-type, a trans- membrane domain and a short cytoplasmic tail.
  • the TREM trans-membrane domain possesses negatively charged residues that interact with the positively charged residues of the DNAX Activating Protein of 12 kDa (DAP12), a trans-membrane adaptor containing an immunoreceptor tyrosine-based activation motif (ITAM).
  • recombinant nucleic acids encoding a chimeric antigen receptor comprises sequences that encode at least the TREM TM domain, such that the chimeric receptor interacts with DAP12 and enhance phagocytosis via phosphorylation of residues within the ITAM in DAP12, which recruits several signaling molecules that activate phagocytosis.
  • the chimeric receptors comprise one or more domains from TREM proteins, fused at the extracellular region with an antigen binding domain that can specifically bind to a cancer antigen, such as a cancer antigen-specific antibody or part or fragment thereof.
  • recombinant nucleic acids encoding a TREM chimeric antigen receptor encode a fusion proteins that comprises: (a) the at least a TREM transmembrane domain (TM) and a TREM intracellular domain (ICD); and (b) an extracellular domain (ECD) comprising an antigen binding domain that can specifically bind to a cancer antigen.
  • the fusion proteins are designed to target cancer cells and bind to the target cancer cells via the ECD comprising the antigen binding domain, and the binding triggers and enhance phagocytosis via signaling through the TREM TM and/or the intracellular domains.
  • the transmembrane domain of TREM trimerizes with DAP12 transmembrane domains and trigger intracellular pro-phagocytosis signaling cascade.
  • the TREM domains are contributed by TREM1, or by TREM2, or by TREM3 members.
  • the extracellular antigen binding domain is fused to the extracellular terminus of the TREM domains through a short spacer or linker. [0465]
  • the extracellular antigen binding domain comprises an antibody, specific to a cancer antigen.
  • the extracellular antigen binding domain comprises an antibody or an antigen binding part thereof that binds specifically to an antigen on the surface of a cancer cell.
  • the extracellular antigen binding domain is an antibody specific for a cancer antigen.
  • the extracellular antigen binding domain is a fraction of an antibody, wherein the fragment can bind specifically to the cancer antigen on a cancer cell.
  • the antigen binding domain comprises a single chain variable fraction (scFv) specific for a cancer antigen binding domain.
  • scFv single chain variable fraction
  • the chimeric receptors described herein may comprise a FcR ⁇ (FcR ⁇ 1) domain.
  • the FcR ⁇ 1 transmembrane domain heterodimerizes with FcR ⁇ transmembrane domains in the myeloid cells, such as macrophages, and other phagocytic cells, such as mast cells, and the heterodimerization is required for expression of the protein on the cell surface.
  • expression of a recombinant protein that comprises the FcR ⁇ 1 TM domain is restricted to expression in cells that naturally express the Fc ⁇ R.
  • a recombinant chimeric protein having the FcR ⁇ 1 TM domain is precluded for expressing in any cells other than the phagocytic cells that express the FcR ⁇ .
  • FcR ⁇ expression is restricted to mast cells.
  • the chimeric receptors are designed with one or more domains comprising the FcR ⁇ 1 TM domain for myeloid cell specific expression of the chimeric protein.
  • the chimeric receptors are designed with one or more domains comprising the FcR ⁇ TM domain for mast-cell specific expression of the chimeric protein.
  • Pro-caspases domains [0468] In some aspects, described herein are recombinant nucleic acids encoding chimeric antigen receptors and a fusion protein comprising: an Src Homology 2 domain (SH2) l linked at the C terminus with a caspase cleavage sequence and a sequence encoding a Procaspase (SH2-ccs- Procaspase).
  • SH2 Src Homology 2 domain
  • the recombinant nucleic acid encodes chimeric antigen receptor which comprises a first polypeptide (a) comprising: an extracellular antigen binding domain that can specifically bind to a cancer antigen, a transmembrane domain and an intracellular signaling domain comprising an ITAM motif that can trigger or enhance phagocytosis, and a second polypeptide (b) an intracellular polypeptide comprising the Src Homology 2 domain (SH2) l linked at the C terminus with a caspase cleavage sequence and a sequence encoding a Procaspase (SH2-ccs-Procaspase).
  • the recombinant nucleic acid may be monocistronic or polycistronic.
  • the recombinant nucleic acid is monocistronic and the sequence encoding (a) and the sequence encoding (b) are separated by a T2A sequence that cleaves the two polypeptides endogenously after translation.
  • the Procaspase can be a Procaspase 1, Procaspase 2 or Procaspase 3.
  • the SH2 domain of the SH2-ccs-Procaspase directs the Procaspase to the phosphorylated intracellular domain of the ITAM motif of (a), and is activated.
  • Ubiquitylation Activation Domains [0469]
  • intracellular signaling domains may be added to the chimeric receptor to enhance phagocytotic signaling.
  • Ubiquitin signaling is involved in triggering of phagocytosis.
  • Monoubiquitylation of endocytic receptors can target them for lysosomal degradation.
  • IL-4 signaling mediated polyubiquitination of scavenger receptor MSR1 at the intracellular domain can lead to activation of the receptor, and trigger its interaction with MAP kinase pathway proteins that are involved in inducing inflammatory gene activation.
  • K63 polyubiquitylation of the MSR1 protein leads to its interaction with TAK1/MKK7/JNK in the phagosomes.
  • Ubiquitylation of K27 residue of MSR1 is also implicated in MSR signaling and pro-inflammatory gene activation, such as expression of pro-inflammatory cytokines.
  • Ubiquitin E3 ligase can promote ligation of ubiquitin to specific lysine residues on the intracellular signaling domain of MSR1, which is activated upon IL4 activation, and which in turn can activate intracellular signaling by binding of TAK1, MKK7 and JNK, and triggering expression of pro-inflammatory genes, such as TNF-alpha and IL-1 ⁇ .
  • the recombinant chimeric antigen receptors described herein may comprise an intracellular domain which comprises residues that can be ubiquitylated and activated to generate a proinflammatory signal.
  • the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain of MSR1 comprising the residues that can undergo E3 ligase mediated ubiquitylation.
  • the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to TAK1.
  • the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a MAP kinase protein, such as MKK7.
  • the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that comprises a MAP kinase protein, such as MKK7.
  • the intracellular domain of a chimeric antigen receptors described herein comprises an intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that comprises a JNK.
  • the intracellular domain of a chimeric antigen receptors described herein comprises an intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that can activate pro-inflammatory gene transcription.
  • a suitable ubiquitylation domain can be ligated at the intracellular portion of any one of the CARP receptors described in the disclosure.
  • one or more CFPs described herein may be co-expressed with a recombinant phagocytic receptor fusion protein (PFP) that has an extracellular domain comprising an antigen binding domain that can specifically bind to a cancer antigen on the surface of a cancer cell.
  • the fusion protein further comprises a transmembrane domain and an intracellular phagocytosis receptor domain, may comprise and further phagocytosis signaling enhancement domains, such as kinase binding domains.
  • the PFP is specifically designed to target a cancer cell and activate phagocytosis upon binding to the target.
  • the phagocytosis enhancement by PFP is augmented by co- expression of the CFP.
  • cells coexpressing the PFP with the SIRP-CFPs enhance the phagocytic potential of the cells, wherein additionally the PFPs provide specific cancer targeting to the cells.
  • cells coexpressing the PFP with SH2-ccs-Procaspase have specific targeting to cancer cells with the PFP, whereas the phagocytic activity is enhanced by the PFP intracellular domains, and cancer cell killing activity is enhanced due to clearance of apoptotic cells by SH2-ccs- Procaspase via Procaspase activation to caspase.
  • the scavenger receptor intracellular domain comprises a second intracellular domain comprising a signaling domain that activates phagocytosis; or a proinflammatory domain at the cytoplasmic terminus, which are operably linked.
  • the CD47 binding domain is operably linked with the one or more intracellular signaling domains of the phagocytic receptor, the signaling event originating from the engagement of the CD47 ligand at the extracellular end is altered upon passage through the intracellular domains to phagocytosis enhancing signal at the intracellular end of the recombinant receptor instead of the phagocytosis inhibition signal of a native CD47-binding SIRP ⁇ receptor.
  • the intracellular phagocytosis signaling domain may comprise domains selected from MRC1, ItgB5, MERTK, ELMO, BAIL Tyro3, Axl, Traf6, Syk, MyD88, Zap70, PI3K, FcyR1, FcyR2A, FcyR2B2, FcyR2C, FcyR3A, FcER1, FcaRl, BAFF-R, DAP12, NFAM1, and CD79b intracellular domains.
  • the function of the chimeric receptor may be further augmented by expressing an additional recombinant protein in the myeloid cell concurrently with or independent of the CFP expression.
  • the additional recombinant protein is co-expressed with the CFP described above, wherein the CFP can bind to an antigen that is expressed in the target cell, and the CFP enhances phagocytosis and killing of the target cell by the myeloid cell expressing the CFP.
  • the additional recombinant protein is designed to further enhance the functioning of the CFP expressing cell.
  • the additional recombinant protein is a second chimeric protein, such as a chimeric fusion protein, for example, a second chimeric protein.
  • the second chimeric protein is expressed in population of myeloid cells that expresses the cancer antigen targeting CFP.
  • the additional recombinant protein is a second chimeric fusion protein or phagocytosis receptor fusion protein (PFP).
  • the second chimeric protein is a truncated protein.
  • compositions comprising a recombinant nucleic acid encoding a chimeric CD47 receptor (CR) fusion protein comprising: (a) a CR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to CD47 of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the CR does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, (ii) the CR comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase, or (iii) the CR comprises an intracellular signaling domain derived from a phage
  • the CR does not comprise a functional intracellular domain of an endogenous receptor that binds CD47.
  • the CR comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase.
  • the CR comprises an intracellular signaling domain derived from a phagocytic or tethering receptor.
  • the antigen binding domain specific to CD47 comprises an antibody domain specific to CD47.
  • the antigen binding domain specific to CD47 comprises an extracellular domain derived from SIRP ⁇ or SIRP ⁇ .
  • the CR comprises an intracellular domain comprising an intracellular signaling domain.
  • a composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; wherein the intracellular signaling domain is derived from a phagocytic receptor; and wherein the recombinant nucleic acid encodes: (A) encodes a pro-inflammatory polypeptide, (B) comprises a binding motif for a molecule, such that upon binding of the molecule to the binding motif translation of the recombinant nucleic
  • composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; an wherein the PFP forms a functional complex with FcR ⁇ when expressed in a cell.
  • PR phagocytic receptor
  • composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the PFP forms a functional complex with DAP12 when expressed in a cell.
  • PR phagocytic receptor
  • the recombinant nucleic acid encodes: (A) encodes a pro- inflammatory polypeptide, (B) comprises a binding motif for a molecule, such that upon binding of the molecule to the binding motif translation of the recombinant nucleic acid is inhibited, (C) a procaspase domain, or (D) a procaspase binding domain.
  • the PFP forms a functional complex with FcR ⁇ when expressed in a cell.
  • the PFP forms a functional complex with DAP12 when expressed in a cell.
  • the PFP comprises an antigen binding domain that binds to a CD47 ligand, but does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, (i) the PFP comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase, or (ii) the PFP comprises an intracellular signaling domain derived from a phagocytic receptor.
  • the CFP or the PFP forms a functional complex with an FcR.
  • the CFP or the PFP forms a functional complex with FcR ⁇ .
  • the CFP or the PFP comprises a transmembrane domain of FcR- alpha or FcR ⁇ .
  • the CFP or the PFP forms a functional complex with a TREM.
  • the CFP or the PFP comprises an intracellular domain comprising an ITAM.
  • the CFP or the PFP comprises an intracellular domain comprising an ITIM.
  • a cell expressing the CFP or the PFP exhibits inhibits CD47 mediated anti-phagocytosis activity.
  • the truncated SIRP ⁇ binds to CD47 and inhibits.
  • the binding motif is in a UTR region of the recombinant nucleic acid. In some embodiments, the binding motif is an ARE sequence. [0500] In some embodiments, the transmembrane domain binds to a transmembrane domain of DAP12. [0501] In some embodiments, the transmembrane domain that binds to a transmembrane domain of DAP12 is derived from TREM. [0502] In some embodiments, the CFP or the PFP comprises an intracellular domain derived from a DAP12 monomer.
  • the recombinant polynucleotide further comprises a sequence encoding a first modified signal regulatory protein ⁇ (SIRP ⁇ ).
  • SIRP ⁇ first modified signal regulatory protein ⁇
  • the first modified SIRP ⁇ lacks an intracellular signaling domain of a wild type SIRP ⁇ .
  • the first modified SIRP ⁇ lacks an intracellular signaling domain of a wild type SIRP ⁇ .
  • the CFP or the PFP does not transmit a signal to block phagocytosis.
  • the composition further comprises a polynucleotide encoding a second modified signal regulatory protein ⁇ (SIRP ⁇ ), wherein the second modified SIRP ⁇ comprises a PI3K binding domain
  • SIRP ⁇ second modified signal regulatory protein ⁇
  • the PI3K binding domain is derived from CD19, CD28, CSFR or PDGFR.
  • the composition further comprises a polynucleotide encoding a third modified signal regulatory protein ⁇ (SIRP ⁇ ), wherein the third modified SIRP ⁇ comprises a pro-inflammatory domain.
  • the composition further comprises a polynucleotide encoding a recombinant SIRP, wherein the recombinant SIRP comprises an extracellular domain derived from SIRP ⁇ , a transmembrane domain derived from SIRP ⁇ , and an intracellular domain derived from SIRP ⁇ .
  • the composition further comprises when the recombinant SIRP binds to an antigen CD47, the third modified SIRP ⁇ does not transmit a signal to block phagocytosis.
  • the anti-CD47 binding domain is derived from signal-regulatory protein alpha (SIRP ⁇ ) or signal-regulatory protein beta (SIRP ⁇ ).
  • the intracellular signaling domain is derived from a phagocytic or tethering receptor.
  • the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from any one of the receptors listed in Table 2.
  • the intracellular signaling domain is derived from a receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169.
  • a receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain.
  • the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1.
  • the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP.
  • the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of any one of the proteins listed in Table 1.
  • the intracellular signaling domain of any one of the PFPs described herein is derived from a T cell receptor component or tethering receptor or wherein the intracellular signaling domain comprises a T cell activation domain.
  • the intracellular signaling domain comprises a PI3K recruitment domain.
  • the CFP or the PFP functionally incorporates into a cell membrane of a cell when the CFP or the PFP is expressed in the cell.
  • a cell expressing the CFP or the PFP exhibits an increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits at least a 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP.
  • the target cell expressing the antigen is a cancer cell.
  • the target cell expressing the antigen is at least 0.8 microns in diameter.
  • the intracellular signaling domain is derived from a scavenger receptor.
  • a cell expressing the CFP or the PFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP or the PFP.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof.
  • a cell expressing the CFP or the PFP exhibits an increase in effector activity compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in cross- presentation compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of TRAIL/TNF Family death receptors compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of B7-H2 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of LIGHT compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of HVEM compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD40 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of TL1A compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of 41BBL compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of OX40L compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of GITRL death receptors compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD30L compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of TIM4 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of TIM1 Ligand compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of SLAM compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD48 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD58 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD155 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of CD112 compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in expression of PDL1compared to a cell not expressing the CFP or the PFP.
  • provided herein is the composition of any of B7-DC compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in respiratory burst compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in ROS production compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in iNOS production compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in iNOS production compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in extra- cellular vesicle production compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in trogocytosis with a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in resistance to CD47 mediated inhibition of phagocytosis compared to a cell not expressing the CFP or the PFP.
  • a cell expressing the CFP or the PFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the CFP or the PFP.
  • the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain.
  • the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the CFP or the PFP.
  • the intracellular domain comprises an F-actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain.
  • a cell expressing the CFP or the PFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production.
  • the extracellular domain comprises an Ig binding domain.
  • the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, Fc ⁇ RI, Fc ⁇ RIIA, Fc ⁇ RIIB, Fc ⁇ RIIC, Fc ⁇ RIIIA, Fc ⁇ RIIIB, FcRn, TRIM21, FcRL5 binding domain.
  • the extracellular domain comprises an FcR extracellular domain.
  • the extracellular domain comprises an FcR ⁇ , FcR ⁇ , FcR ⁇ or FcR ⁇ extracellular domain.
  • the extracellular domain comprises FcR ⁇ (FCAR) extracellular domain.
  • the extracellular domain comprises an FcR ⁇ extracellular domain.
  • the extracellular domain comprises an FcR ⁇ (FCER1A) extracellular domain.
  • the extracellular domain comprises an FcR ⁇ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, FCGR3B) extracellular domain.
  • the extracellular domain comprises one more integrin ⁇ 1, ⁇ 2, ⁇ IIb, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, ⁇ 10, ⁇ 11, ⁇ IIb, ⁇ D, ⁇ E, ⁇ L, ⁇ M, ⁇ V, ⁇ X, ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8 domain.
  • the intracellular domain comprises a CD47 inhibition domain.
  • the PSR subunit further comprises an extracellular domain operatively linked to the transmembrane domain and the extracellular antigen binding domain.
  • the extracellular domain further comprises an extracellular domain of a receptor, a hinge, a spacer or a linker. In some embodiments, the extracellular domain comprises an extracellular portion of a PSR. In some embodiments, the extracellular portion of the PSR is derived from the same PSR as the PSR intracellular signaling domain. In some embodiments, the extracellular domain comprises an extracellular domain of a scavenger receptor or an immunoglobulin domain. [0580] In some embodiments, the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region. In some embodiments, the extracellular domain comprises a phagocytic engulfment marker.
  • the extracellular domain comprises a structure capable of multimeric assembly. In some embodiments, the extracellular domain comprises a scaffold for multimerization. [0582] In some embodiments, the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length. [0583] In some embodiments, the extracellular antigen binding domain specifically binds to the antigen of a target cell. In some embodiments, the extracellular antigen binding domain comprises an antibody domain.
  • the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof.
  • the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor.
  • the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen.
  • the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen.
  • the antigen is a cancer antigen or a pathogenic antigen or an autoimmune antigen.
  • the antigen comprises a viral antigen.
  • the antigen is a T-lymphocyte antigen.
  • the antigen is an extracellular antigen.
  • the antigen is an intracellular antigen.
  • the antigen is selected from the group consisting of Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), Mesothelin, EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B-Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2,
  • TK1 Thym
  • the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56. [0588] In some embodiments, the antigen is an ovarian cancer antigen or a T lymphoma antigen. In some embodiments, the antigen is an integrin receptor.
  • the antigen is an integrin receptor selected from the group consisting of ⁇ 1, ⁇ 2, ⁇ IIb, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, ⁇ 10, ⁇ 11, ⁇ D, ⁇ E, ⁇ L, ⁇ M, ⁇ V, ⁇ X, ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, and ⁇ 8.
  • the transmembrane domain and the extracellular antigen binding domain is operably linked through a linker.
  • the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker such as the hinge region of CD8 ⁇ , IgG1 or IgG4.
  • the extracellular domain comprises a multimerization scaffold.
  • the transmembrane domain comprises an FcR transmembrane domain.
  • the transmembrane domain comprises an FcR-alpha, FcR ⁇ or FcR ⁇ transmembrane domain.
  • the transmembrane domain comprises an Fc ⁇ R (FCAR) transmembrane domain.
  • the transmembrane domain comprises an FcR ⁇ (FCER1A) transmembrane domain. In some embodiments, the transmembrane domain comprises an FcR ⁇ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B) transmembrane domain.
  • the transmembrane domain comprises a T cell Receptor subunit, CD3 epsilon, CD3 gamma and CD3 delta, CD45, CD2 CD4, CD5, CD8, CD9, CD16, CD19, CD22, CD33, CD28, CD30, CD37, CD64, CD80, CD86, CD134, CD137 and CD 154, or a functional fragment thereof, or an amino acid sequence having at least one, two or three modifications but not more than 20, 10 or 5 modifications transmembrane domain.
  • the transmembrane domain comprises a transmembrane domain from a syntaxin such as syntaxin 3 or syntaxin 4 or syntaxin 5.
  • the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the CR or the PFP is expressed in a cell.
  • the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the CR or the PFP is expressed in a cell.
  • the transmembrane domain dimerizes with a transmembrane domain of an endogenous receptor when the CFP or the PFP is expressed in a cell.
  • the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the CFP or the PFP is expressed in a cell.
  • the transmembrane domain is derived from a protein that is different than the protein from which the intracellular signaling domain is derived.
  • the transmembrane domain is derived from a protein that is different than the protein from which the extracellular domain is derived.
  • the transmembrane domain comprises a transmembrane domain of a phagocytic receptor.
  • the transmembrane domain and the extracellular domain are derived from the same protein.
  • the transmembrane domain is derived from the same protein as the intracellular signaling domain.
  • the recombinant nucleic acid encodes a DAP12 recruitment domain.
  • the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12.
  • the transmembrane domain is at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length.
  • the transmembrane domain is at most 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length.
  • the intracellular domain comprises a phosphatase inhibition domain.
  • the intracellular domain comprises an ARP2/3 inhibition domain.
  • the intracellular domain comprises at least one ITAM domain.
  • the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 ITAM domains.
  • the intracellular domain further comprises at least one ITAM domain.
  • the intracellular domain further comprises at least one ITAM domain select from a group CD3 zeta TCR subunit, CD3 epsilon TCR subunit, CD3 gamma TCR subunit, CD3 delta TCR subunit, TCR zeta chain, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278, CD66d, functional fragments thereof, and amino acid sequences thereof having at least one but not more than 20 modifications thereto.
  • ITAM domain select from a group CD3 zet
  • the at least one ITAM domain comprises a Src-family kinase phosphorylation site. In some embodiments, the at least one ITAM domain comprises a Syk recruitment domain. In some embodiments, the intracellular domain comprises an F-actin depolymerization activation domain. [0615] In some embodiments, the intracellular domain comprises residues that can be ubiquitylated. In some embodiments, the intracellular domain can bind to an E3 ubiquitin ligase. [0616] In some embodiments, the intracellular domain can bind to a TAK1 kinase.
  • the intracellular domain can bind to a MAP kinase or a MAP kinase family member. [0618] In some embodiments, the intracellular domain can be activated upon ubiquitylation and can activate intracellular signaling resulting in pro-inflammatory gene transcription. [0619] In some embodiments, the intracellular domain lacks enzymatic activity. [0620] In some embodiments, the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. [0621] In some embodiments, the intracellular domain comprises a CD47 inhibition domain [0622] In some embodiments the intracellular signaling domain comprises a domain that activates integrin such as the intracellular region of PSGL-1.
  • the intracellular signaling domain comprises a domain that activates Rap1 GTPase, such as that from EPAC and C3G. [0624] In some embodiments, the intracellular signaling domain is from Paxillin. In some embodiments, the intracellular signaling domain activates focal adhesion kinase. In some embodiments, the intracellular signaling domain is derived from a single phagocytic receptor. In some embodiments, the intracellular signaling domain is derived from a single scavenger receptor. In some embodiments, the intracellular domain further comprises a phagocytosis enhancing domain. In some embodiments, the intracellular domain comprises a pro-inflammatory signaling domain.
  • the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain. In some embodiments, the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain. In some embodiments, the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, an NLRP family member, NLRP1- 14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40 or any combination thereof.
  • the CFP or the PFP does not comprise a full length intracellular signaling domain.
  • the pro-inflammatory signaling domain comprises an intracellular signaling domain from CD3-zeta, CD3-epsilon, CD3-delta or CD3-gamma.
  • the pro-inflammatory signaling domain comprises an intracellular signaling domain from TCR- alpha, TCR-beta, TCR-gamma or TCR-delta. [0625]
  • the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length.
  • the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length.
  • the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • the FcR ⁇ chain or the FcR ⁇ chain forms a complex with FcR ⁇ when expressed in a cell.
  • the FcR ⁇ chain or FcR ⁇ chain forms a complex with endogenous FcR ⁇ when expressed in a cell.
  • the FcR ⁇ chain or the FcR ⁇ chain does not incorporate into a cell membrane of a cell that does not express FcR ⁇ .
  • the CFP or the PFP does not comprise an FcR ⁇ chain intracellular signaling domain.
  • the CFP or the PFP does not comprise an FcR ⁇ chain intracellular signaling domain.
  • the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain.
  • the TREM is TREM1, TREM 2 or TREM 3.
  • the recombinant nucleic acid comprises a sequence encoding a pro-inflammatory polypeptide.
  • the composition further comprises a pro- inflammatory polypeptide.
  • the pro-inflammatory polypeptide is a chemokine, cytokine and nucleotides.
  • the chemokine is selected from the group consisting of CCL2, CXCL1, CXCL12, CXCL9, CXCL10, CXCL11.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon.
  • the recombinant nucleic acid comprises a homeostatic regulator of inflammation.
  • the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA.
  • the sequence in the UTR is a sequence that binds to an RNA binding protein.
  • the target cell is a mammalian cell. In some embodiments, the target cell is a human cell. In some embodiments, the target cell comprises a cell infected with a pathogen. In some embodiments, the target cell is a cancer cell. In some embodiments, the target cell is a cancer cell that is a lymphocyte.
  • the target cell is a cancer cell that is an ovarian cancer cell. In some embodiments, the target cell is a cancer cell that is an ovarian pancreatic cell. In some embodiments, the target cell is a cancer cell that is a glioblastoma cell.
  • the recombinant nucleic acid is DNA. In some embodiments, the recombinant nucleic acid is RNA. In some embodiments, the recombinant nucleic acid is mRNA. In some embodiments, the recombinant nucleic acid is a circRNA. In some embodiments, the recombinant nucleic acid is a tRNA.
  • the recombinant nucleic acid is a microRNA.
  • a vector comprising the composition described above.
  • vector is viral vector.
  • the viral vector is retroviral vector or a lentiviral vector.
  • the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides.
  • the vector is polycistronic.
  • each of the at least one nucleic acid sequence is operably linked to a separate promoter.
  • the vector further comprises one or more internal ribosome entry sites (IRESs).
  • the vector further comprises a 5’UTR and/or a 3’UTR flanking the at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector further comprises one or more regulatory regions.
  • a polypeptide encoded by the recombinant nucleic acid of the composition described above is provided herein.
  • a cell comprising a vector described above or the polypeptide described above. In some embodiments, the cell is a phagocytic cell.
  • the cell is a stem cell derived cell, myeloid cell, macrophage, a dendritic cell, lymphocyte, mast cell, monocyte, neutrophil, microglia, or an astrocyte.
  • the cell is an autologous cell.
  • the cell is an allogeneic cell.
  • the cell is an M1 myeloid cell, such as a macrophage.
  • the cell is an M2 myeloid cell, such as a macrophage.
  • the cell is a precursor cell of myeloid lineage.
  • the myeloid cell is a CD14+ cell.
  • the myeloid cell is a CD16- cell.
  • the myeloid cell is a CD14+ and CD16- cell.
  • the therapeutic agent comprises a T cell such as described herein.
  • the T cell is a T cell precursor.
  • the T cell is an undifferentiated and/or unpolarized.
  • the T cell is a CD4 T cells.
  • T cell is a CD8 T cell.
  • T cell is gamma delta T cell.
  • the cell is a NK T cell.
  • composition comprising (a) the nucleic acid composition or the vector or the polypeptide or the cell as described above; and (b) a pharmaceutically acceptable excipient.
  • the pharmaceutical composition further comprising an additional therapeutic agent.
  • the pharmaceutical composition comprises additional therapeutic agent which is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the CFP or the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • additional therapeutic agent which is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the CFP or the PFP, an agent that promotes
  • the pharmaceutically acceptable excipient comprises serum free media, a lipid, or a nanoparticle.
  • the disease is cancer.
  • the cancer is a solid cancer.
  • the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer.
  • the cancer is a liquid cancer.
  • the liquid cancer is a leukemia or a lymphoma. In some embodiments, the liquid cancer is a T cell lymphoma. In some embodiments, the disease is a T cell malignancy. In some embodiments the method further comprises administering an additional therapeutic agent to the subject.
  • the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F- actin disassembly, an agent that promotes PI3K recruitment to the CFP or the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5- trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • administering comprises infusing or injecting.
  • administering comprises administering directly to the solid cancer.
  • administering comprises a circRNA, mRNA, viral-, particle-, liposome-, or exosome-based delivery procedure.
  • a CD4+ T cell response or a CD8+ T cell response is elicited in the subject.
  • method comprising contacting a cell with the composition described above, the vector or the polypeptide described above.
  • contacting comprises transducing.
  • transducing comprises chemical transfection, electroporation, nucleofection, or viral infection.
  • a method of preparing a pharmaceutical composition comprising contacting a lipid to the composition described herein or the vector described herein.
  • contacting comprises forming a lipid nanoparticle.
  • a method of preparing a pharmaceutical composition comprising contacting an antibody to the composition as described herein or the vector described herein.
  • contacting comprises forming a lipid nanoparticle.
  • the recombinant nucleic comprises one or more regulatory elements within the noncoding regions that can be manipulated for desired expression profiles of the encoded proteins.
  • the noncoding region may comprise suitable enhancer.
  • the enhancer comprises a binding region for a regulator protein or peptide may be added to the cell or the system comprising the cell, for commencement of expression of the protein encoded under the influence of the enhancer.
  • a regulatory element may comprise a protein binding domain that remains bound with the cognate protein and continue to inhibit transcription and/or translation of recombinant protein until an extracellular signal is provided for the protein to decouple from the bound position to allow commencement of the protein synthesis. Examples include but are not limited to Tetracycline-inducible (Tet-Inducible or Tet-on) and Tetracycline repressible (Tet-off) systems known to one of skill in the art.
  • the 5’ and 3’ untranslated regions flanking the coding regions of the construct may be manipulated for regulation of expression of the recombinant protein encoded by the nucleic acid constructs described above.
  • the 3’UTR may comprise one or more elements that are inserted for stabilizing the mRNA.
  • AU-Rich Elements (ARE) sequences are inserted in the 3’ UTR that result in binding of RNA binding proteins that stabilize or destabilize the mRNA, allowing control of the mRNA half-life.
  • the 3’UTR may comprise a conserved region for RNA binding proteins (e.g.
  • GAPDH GAPDH binding to mature mRNA strand preventing translation.
  • glycolysis results in the uncoupling of the RNA binding proteins (e.g. GAPDH) allowing for mRNA strand translation.
  • the principle of the metabolic switch is to trigger expression of target genes when a cell enters a certain metabolic state.
  • GAPDH is a RNA binding protein (RBP). It binds to ARE sequences in the 3’UTR, preventing translation of mRNA.
  • RBP RNA binding protein
  • the environment in which the cell comprising the recombinant nucleic acid is present provides the metabolic switch to the gene expression.
  • hypoxic condition can trigger the metabolic switch inducing the disengaging of GAPDH from the mRNA.
  • the expression of the mRNA therefore can be induced when the myeloid cell, such as a macrophage, leaves the circulation and enters into a tumor environment, which is hypoxic. This allows for systemic administration of the nucleic acid or a cell comprising the nucleic acid, but ensures a local expression, specifically targeting the tumor environment.
  • the nucleic acid construct can be a split construct, for example, allowing a portion of the construct to be expressed under the control of a constitutive expression system whereas another portion of the nucleic acid is expressed under control of a metabolic switch, as described above.
  • the nucleic acid may be under bicistronic control.
  • the bicistronic vector comprises a first coding sequence under a first regulatory control, comprising the coding sequence of a target recognition moiety which may be under constitutive control; and a second coding sequence encoding an inflammatory gene expression which may be under the metabolic switch.
  • the bicistronic vector may be unidirectional. In some embodiments the bicistronic vector may be bidirectional.
  • the ARE sequences comprise protein binding motifs for binding ARE sequence that bind to ADK, ALDH18A1, ALDH6A1, ALDOA, ASS1, CCBL2, CS, DUT, ENO1, FASN, FDPS, GOT2,HADHB, HK2, HSD17B10, MDH2, NME1, NQ01, PKM2, PPP1CC, SUCLG1, TP11, GAPDH, or LDH.
  • a recombinant nucleic acid that comprises a sequence encoding a gene of interest, often referred to as a cargo, that is incorporated into a myeloid cell, e.g., a CD14+ cell ex vivo, in vitro or in vivo to produce an engineered myeloid cell (e.g., a myeloid cell expressing a chimeric receptor that can bind to and phagocytose a target cell in vivo, e.g., an ATAK cell).
  • the engineered cell is effective as an immunotherapeutic cell.
  • the recombinant nucleic acid may be a DNA or an RNA.
  • the recombinant nucleic acid is an mRNA.
  • a recombinant mRNA comprising a sequence encoding a gene of interest, e.g., a chimeric fusion protein that can be expressed in a myeloid cell.
  • the recombinant mRNA is variously modified to enhance stability, translatability, reduce any toxicity, reduce any unintended TLR activation or innate immune reaction to the foreign RNA.
  • optimization efforts are in progress as described herein.
  • an mRNA comprising about 3000 nucleotides or more (e.g., 4kb, 5 kb, 6 kb, 7 kb, 8 kb, 10kb etc.,) may be modified in one or more ways to confer stability in vivo.
  • the 5′ and 3′ UTR elements flanking the coding sequence profoundly influence the stability and translation of mRNA, both of which are critical concerns for ensuring stability of the mRNA.
  • the 5’-UTR comprises an anti-reverse cap analogues (ARCA) cap.
  • the 3’ UTR comprises a poly A tail, which may be 50-200 nucleotides long.
  • the mRNA coding region comprises one or more modified nucleotides, such as methylcytosine, or pseudouridine. Replacing rare codons with frequently used synonymous codons that have abundant cognate tRNA in the cytosol is a common practice to increase protein production from mRNA.
  • the G:C content of the coding region may be altered for stability optimization.
  • the immunostimulatory property of the mRNA cargo may be enhanced or complemented by co- administering the mRNA with a suitable adjuvant.
  • the mRNA is purified to obtain greater than 90%, greater than 95%, greater than 97%, 98% or 99% purity, with less than 5%, or less than 1% or less than 0.5% contaminants from the mRNA generation procedure.
  • the mRNA is prepared in vitro translation (IVT).
  • IVT capping of the 5’ end
  • mRNA is prepared by in vitro transcription (IVT). The transcription reaction is typically carried out by bacteriophage RNA polymerase (T7, T3, SP6) in the presence of a single (ss) or double stranded (ds) DNA template containing an appropriate promoter sequence and the mix of four NTPs.
  • the polymerase initiates the transcription by the attack of the 3 -OH of the first transcribed nucleotide defined by the promoter sequence on the -phosphate of the upcoming NTP, to yield 5 -triphosphate RNA.
  • the first transcribed nucleotide is usually a purine nucleotide; the most common is the use of T7 polymerase and 6.5 promoter, which imposes the initiation with GTP if an m7GpppG derived cap analog is added to the transcription mixture.
  • mRNA generated in vitro is 5’capped and a 3’-polyA tail is added.
  • the 5’Cap is modified from a native mRNA 5’Cap.
  • the 5’ Cap is Cap 0 ((m7GpppN1pG). [0664] IN some embodiments the 5’ Cap is Cap 1 (m7GpppN1 mpG). [0665] One aspect of this disclosure involves having an efficient Cap structure using suitable capping analogs. [0666] In some embodiments, the RNA products prepared by IVT mRNA are co-transcriptional capped with m7GpppN-derived dinucleotides. In some embodiments, the 5’-Cap is a trinucleotide cap analog. In some embodiments the first nucleotide base following a 5’-Cap plays a crucial role in mRNA stability. In some embodiments, the base is adenine (A).
  • the base is cytosine (C). In some embodiments, the base is guanine (G). In some embodiments, the base is uracil (U). In some embodiments, cap-proximal sequence is crucial for mRNA stability and function. [0667] In some embodiments, the Cap analog is m 7 GpppApG. [0668] In some embodiments, the Cap analog is m 7 GpppAmpG. [0669] In some embodiments, the Cap analog is m 7 Gpppm 6 ApG. [0670] In some embodiments, the Cap analog is m 7 Gpppm 6 A m pG. [0671] In some embodiments, the Cap analog is m 7 GpppCpG.
  • the Cap analog is m 7 GpppC m pG. [0673] In some embodiments, the Cap analog is m 7 GpppGpG. [0674] In some embodiments, the Cap analog is m 7 GpppGmpG. [0675] In some embodiments, the Cap analog is m 7 GpppUpG. [0676] In some embodiments, the Cap analog is m 7 GpppU m pG [0677] In some embodiments, the 5’-Cap is anti-reverse cap analogs (ARCAs) having chemically modified 3 -O or 2 -O positions of m7G (m2 7,3 -O GpppG).
  • ARCAs anti-reverse cap analogs
  • the 5’-Cap is N7-methylguanosine 5 -diphosphate (m7GDPIm).
  • the caping involves post-transcriptional enzymatic capping of 5 - triphosphates RNA using Vaccinia virus capping complex.
  • the 5’capping confers stability and translational efficiency to the mRNA.
  • the 5’ Cap analog is a trinucleotide analog, and is 2’-O- methyladenosine (Am).
  • the 5’ Cap analog is a trinucleotide analog, and N 6 ,2’-O-dimethyladenosine (m 6 Am) (FIG.3).
  • Dinucleotides pNpG and trinucleotide cap analogs are isolated from the reaction mixtures by ion-exchange chromatography on a DEAE Sephadex A-25 (HCO3 – form) column a described in Sikorski et al., Nucleic Acids Research, 2020, Vol.48, No.4.
  • the m 7 GpppAmpG -capped mRNA shows higher stability, as well as higher translational efficiency that a related Cap analog.
  • m 7 GpppAmpG capped mRNA shows at least 1.2-fold higher protein yield when transfected in suitable cells.
  • Transcription of the RNA may be performed in vitro using the linearized DNA template in the presence of the corresponding RNA polymerase and adenosine, guanosine, uridine and cytidine ribonucleoside triphosphates (rNTPs) under conditions that support polymerase activity while minimizing potential degradation of the resultant mRNA transcripts.
  • rNTPs adenosine, guanosine, uridine and cytidine ribonucleoside triphosphates
  • In vitro transcription can be performed using a variety of commercially available kits including, but not limited to RiboMax Large Scale RNA Production System (Promega), MegaScript Transcription kits (Life Technologies) as well as with commercially available reagents including RNA polymerases and rNTPs.
  • the methodology for in vitro transcription of mRNA is well known in the art. (see, e.g. Losick, R., 1972, In vitro transcription, Ann Rev Biochem v. 41409-46; Kamakaka, R. T. and Kraus, W. L. 2001. In Vitro Transcription. Current Protocols in Cell Biology. 2:11.6:11.6.1-11.6.17; Beckert, B.
  • RNA transcripts are well known in the art.
  • Well known procedures include phenol/chloroform extraction or precipitation with either alcohol (ethanol, isopropanol) in the presence of monovalent cations or lithium chloride.
  • Additional, non- limiting examples of purification procedures which can be used include size exclusion chromatography (Lukavsky, P. J. and Puglisi, J. D., 2004, Large-scale preparation and purification of polyacrylamide-free RNA oligonucleotides, RNA v.10,889-893), silica-based affinity chromatography and polyacrylamide gel electrophoresis (Bowman, J. C., Azizi, B., Lenz, T.
  • RNA glycosylation [0686]
  • the recombinant nucleic acid is an mRNA, wherein the mRNA is a glycosylated mRNA.
  • Glycans are the fourth fundamental biopolymer, next to proteins, polynucleotides, and polyalkanes (lipids).
  • glycans are composed of roughly 10 monomeric carbohydrate units, which are strung together and appended to proteins or lipids.
  • glycans are present in every cell studied to date, across the kingdoms of life. They perform a myriad of essential functions, especially in the context of cell surface events, where complex glycans facilitate the folding and purposeful trafficking of proteins and lipids for secretion or membrane presentation.
  • processes such as embryogenesis, host-pathogen recognition, and tumor-immune interactions rely on proper glycosylation.
  • RNAs in mammalian cells are modified with glycans (Flynn et al., bioRxiv 2019).
  • Chemical, genetic, and metabolic labeling coupled biorthogonal chemistry revealed sialylated glycans are conjugated to RNAs in mammalian cells. These conjugates are termed glycoRNAs.
  • GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. The glycoRNA conjugate is present in all cell types and tissues tested across human, mouse, and hamster.
  • RNA glycosylation revealed a distinct set of small RNAs including Y RNAs, small nuclear RNAs, and small nucleolar RNAs as modified putatively at guanosine.
  • Chemical, genetic, and enzymatic approaches revealated that sialylated N-glycans, produced by the oligosaccharyltransferase (OST) complex in the ER lumen, modify these RNA transcripts.
  • OST oligosaccharyltransferase
  • the mRNA comprising the gene of interest is subjected to glycosylation in a process that recapitulates an intracellular glycosylation process to ensure proper intracellular trafficking, translation, interaction with intracellular proteins, attachment to a cellular organelle structure or scaffolding.
  • Delivery of nucleic acids into a cell [0687] Nucleic acids encoding the CFP or PFP as described herein may be introduced to a cell, e.g. a myeloid cell, via different delivery approaches. A recombinant nucleic acid as described herein may be introduced to a cell in vitro, ex vivo or in vivo.
  • a nucleic acid is introduced into a myeloid cell in the form of a plasmid or a vector.
  • the vector is a viral vector.
  • the vector is an expression vector, for example, a vector comprising one or more promoters, and other regulatory components, including enhancer binding sequence, initiation and terminal codons, a 5’UTR, a 3’UTR comprising a transcript stabilization element, optional conserved regulatory protein binding sequences and others.
  • the vector is a phage, a cosmid, or an artificial chromosome.
  • a vector is introduced or incorporated in the cell by known methods of transfection, such as using lipofectamine, or calcium phosphate, or via physical means such as electroporation or nucleofection.
  • the vector is introduced or incorporated in the cell by infection, a process commonly known as viral transduction.
  • the vector for expression of the CFP is of a viral origin.
  • the recombinant nucleic acid is encoded by a viral vector capable of replicating in non-dividing cells.
  • the nucleic acid encoding the recombinant nucleic acid is encoded by a lentiviral vector, e.g. HIV and FIV-based vectors.
  • the lentiviral vector is prepared in-house and manufactured in large scale for the purpose. In some embodiments, commercially available lentiviral vectors are utilized, as is known to one of skill in the art.
  • the recombinant nucleic acid is encoded by a herpes simplex virus vector, a vaccinia virus vector, an adenovirus vector, or an adeno-associated virus (AAV) vector.
  • the myeloid cell may be modified by expressing a transgene via incorporation of the transgene in a transient expression vector. In some embodiments expression of the transgene may be temporally regulated by a regulator from outside the cell.
  • RNAs may be contemplated for retrotransposition of the transgene and stable integration into myeloid cells, such as macrophages or phagocytic cells.
  • Retrotransposon may be used for stable integration of a recombinant nucleic acid encoding a gene of interest, e.g. a fusion protein, such as a chimeric fusion protein or a chimeric antigen receptor for myeloid cells.
  • recombinant nucleic acid is an mRNA comprising a sequence encoding human ORF2 protein and/or a human ORF1 protein with the recombinant nucleic acid sequence as the payload.
  • the mRNA payload may be about 6kb in length, or about 10 kb in length.
  • the recombinant nucleic acid is a self-amplifying mRNA.
  • Self-amplifying or self-replicating RNA molecules e.g., described in US20110300205A1
  • which replicate in host cells leading to an amplification of the amount of RNA encoding the desired gene product can enhance efficiency of RNA delivery and expression of the encoded gene products. See, e.g., Johanning, F. W., et al., Nucleic Acids Res., 23(9):1495-1501 (1995); Khromykh, A.
  • saRNAs are genetically engineered replicons derived from self-replicating single- stranded RNA viruses.
  • the saRNA may be polycistronic. They can be delivered as viral replicon particles (VRPs) with the saRNA packaged into the viral particle, or as a completely synthetic saRNA produced after in vitro transcription.
  • VRPs viral replicon particles
  • VRPs To generate replication-defective VRPs, envelope proteins are provided in trans as defective helper constructs during production. Resulting VRPs therefore lack the ability to form infectious viral particles following a first infection, and only the RNA is capable of further amplification. VRPs may be derived from both positive- sense and negative-sense RNA viruses, however the latter are more complex and require reverse genetics to rescue the VRPs.
  • the pharmaceutical composition described herein comprises an saRNA that can be produced and delivered in a similar manner to conventional mRNA vaccines.
  • Positive-sense alphavirus genomes that have been commonly used for saRNA design include the Venezuelan equine encephalitis virus (VEE), Sindbis virus (SINV), and Semliki forest virus (SFV).
  • the saRNA may be used to engineer a myeloid cell in vitro, and the resultant engineered cell comprising the saRNA that comprises the gene of interest may be formulated in a pharmaceutical composition and administered to a subject in need thereof.
  • the saRNA may be delivered directly in vivo.
  • the saRNA may be delivered inside a suitable cell either in vitro or in vivo as a naked RNA.
  • the saRNA may be delivered inside a suitable cell either in vitro or in vivo as a nucleic acid and lipid complex (LN complex).
  • the saRNA may be delivered as a cationic nano-emulsion.
  • the saRNA may be delivered inside a suitable cell either in vitro or in vivo in liposomes. In some embodiments, saRNA may be delivered inside a suitable cell either in vitro or in vivo as a lipid nanoparticle (LNPs). Exemplary saRNA vaccines currently in development are discussed in Bloom et al., Gene Therapy (2021) 28:117–129. [0693] Various designs of LNP compositions are contemplated herein, as described in the later section. Circ-RNA [0694] In some embodiments, the recombinant nucleic acid described herein is a circular RNA (circRNA).
  • a circular RNA comprises a RNA molecule where the 5’ end and the 3’ end of the RNA molecule are joined together.
  • circRNAs have no free ends and may have longer half-life as compared to some other forms of RNAs or nucleic acid and may be resistant to digestion with RNase R exonuclease and turn over more slowly than its counterpart linear RNA in vivo.
  • the half-life of a circRNA is more than 20 hours. In some embodiments, the half-life of a circRNA is more than 30 hours. In some embodiments, the half-life of a circRNA is more than 40 hours. In some embodiments, the half-life of a circRNA is more than 48 hours.
  • a circRNA comprises an internal ribosome entry site (IRES) element that engages a eukaryotic ribosome and an RNA sequence element encoding a polypeptide operatively linked to the IRES for insertion into cells in order to produce a polypeptide of interest.
  • IRES internal ribosome entry site
  • circRNAs can be prepared by methods known to those skilled in the art. For example, circRNAs may be chemically synthesized or enzymatically synthesized. In some embodiments, a linear primary construct or linear mRNA may be cyclized, or concatemerized to create a circRNA.
  • the mechanism of cyclization or concatemerization may occur through methods such as, but not limited to, chemical, enzymatic, or ribozyme catalyzed methods.
  • the newly formed 5′-/3′-linkage may be an intramolecular linkage or an intermolecular linkage.
  • a linear primary construct or linear mRNA may be cyclized, or concatemerized using the chemical method to form a circRNA.
  • the 5′-end and the 3′-end of the nucleic acid e.g., linear primary construct or linear mRNA
  • the 5′-end may contain an NHS-ester reactive group and the 3′-end may contain a 3′-amino-terminated nucleotide such that in an organic solvent the 3′-amino-terminated nucleotide on the 3′-end of a linear RNA molecule will undergo a nucleophilic attack on the 5′-NHS-ester moiety forming a new 5′-/3′-amide bond.
  • a DNA or RNA ligase e.g.
  • a T4 ligase may be used to enzymatically link a 5′-phosphorylated nucleic acid molecule (e.g., a linear primary construct or linear mRNA) to the 3′-hydroxyl group of a nucleic acid forming a new phosphodiester linkage.
  • a linear primary construct or linear mRNA may be cyclized or concatemerized by using at least one non-nucleic acid moiety.
  • the at least one non-nucleic acid moiety may react with regions or features near the 5′ terminus and/or near the 3′ terminus of the linear primary construct or linear mRNA in order to cyclize or concatemerized the linear primary construct or linear mRNA.
  • a linear primary construct or linear mRNA may be cyclized or concatemerized due to a non-nucleic acid moiety that causes an attraction between atoms, molecules surfaces at, near or linked to the 5′ and 3′ ends of the linear primary construct or linear mRNA.
  • a linear primary construct or linear mRNA may be cyclized or concatemerized by intermolecular forces or intramolecular forces. Non-limiting examples of intermolecular forces.
  • a linear primary construct or linear mRNA may comprise a ribozyme RNA sequence near the 5′ terminus and near the 3′ terminus.
  • a circRNA may be synthesized by inserting DNA fragments into a plasmid containing sequences having the capability of spontaneous cleavage and self-circularization.
  • a circRNA is produced by making a DNA construct encoding an RNA cyclase ribozyme, expressing the DNA construct as an RNA, and then allowing the RNA to self-splice, which produces a circRNA free from intron in vitro.
  • a circRNA is produced by synthesizing a linear polynucleotide, combining the linear nucleotide with a complementary linking oligonucleotide under hybridization conditions, and ligating the linear polynucleotide.
  • the circRNA may be modified or unmodified.
  • the circRNA is chemically modified.
  • an A, G, U or C ribonucleotide of a circRNA may comprise chemical modifications.
  • any region of a circRNA, e.g. the coding region of the CFP or PFP, the flanking regions and/or the terminal regions may contain one, two, or more (optionally different) nucleoside or nucleotide modifications.
  • a modified circRNA introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified circRNA. Modifications such as to the sugar, the nucleobase, or the internucleoside linkage (e.g.
  • nucleobase e.g. a pyrimidine nucleobase
  • one or more atoms of nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro).
  • modifications e.g., one or more modifications
  • are present in each of the sugar and the internucleoside linkage. Additional modifications to circRNAs are described in US20170204422, the entire content of which is incorporated herein by reference.
  • the circRNA is conjugated to other polynucleotides, dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g.
  • intercalating agents e.g. acridines
  • cross-linkers e.g. psoralene, mitomycin C
  • porphyrins TPPC4, texaphyrin, Sapphyrin
  • polycyclic aromatic hydrocarbons e.g., phenazine, dihydrophenazine
  • artificial endonucleases e.g.
  • alkylating agents phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g.
  • the circRNA is administered directly to tissues of a subject. Additional description of circRNAs in U.S.
  • an additional safety layer may be designed to reduce or prevent expression of the polypeptide of interest in a non-intended cell type, e.g., in a non- myeloid cell.
  • a non-intended cell type e.g., in a non- myeloid cell.
  • the fusion protein may comprise (i) an extracellular domain comprising, e.g.
  • the expression construct may comprise a regulatory control unit for cell-specific expression, such as a cell-specific promoter or an enhancer that controls the expression of the polypeptide of interest.
  • a regulatory control unit for cell-specific expression such as a cell-specific promoter or an enhancer that controls the expression of the polypeptide of interest.
  • myeloid cell-specific promoters may be promoter elements of the lysM, csf1r, CD11c, CD68, macrophage SRA, and CD11b genes.
  • Such controls may additionally be under regulatable control, for example, by incorporating an inducible element.
  • the myeloid cell-specific promoter or enhancer may be human myeloid cell-specific gene promoters, enhancers or portions thereof.
  • cell specific expression of the construct may be incorporated in the design by incorporating one or more microRNA binding sites.
  • MicroRNAs miRNAs
  • MiRNAs are endogenous small non-coding single stranded RNAs that control diverse biological processes. MiRNAs are well suited to suppress previously active programs and, thereby, provide robustness to cell fate decisions (Babiarz, J. E. & Blelloch, R. Small RNAs—their biogenesis, regulation and function in embryonic stem cells. StemBook, ed.
  • MiRNAs identify their targets via base pairing of nucleotides 2-8 of the miRNA (the seed sequence) with complementary sequences within the target mRNA's open reading frame (ORF) and 3′ untranslated region (UTR) (The Stem Cell Research Community, StemBook, doi/10.3824/stembook. 1.47.1. (2009)).
  • MicroRNAs are single-stranded RNA molecules, typically between 21 and 23 nucleotides in length.
  • RNA binding protein DGCR8 which is required for the production of all canonical miRNAs, results in a cell cycle defect and an inability to silence the self-renewal program of ESCs when they are placed in differentiation-inducing conditions (Wang, Y. et al., Nat Genet 39:380-5 (2007).
  • one miRNA can target multiple mRNAs and one mRNA can be regulated by multiple miRNAs targeting different regions of the 3′ UTR.
  • miRNA can modulate gene expression and protein production by affecting, e.g., mRNA translation and stability (Baek et al., Nature 455(7209):64 (2008); Selbach et al., Nature 455(7209):58 (2008); Ambros, 2004, Nature, 431, 350-355; Bartel, 2004, Cell, 116, 281-297; Cullen, 2004, Virus Research., 102, 3-9; He et al., 2004, Nat. Rev. Genet., 5, 522-531; and Ying et al., 2004, Gene, 342, 25-28). [0703] Uchida et al.
  • WO2015066042A1 demonstrated use of oncolytic HSV in treating Glioblastoma multiforme, by inhibition of vector replication by a cellular microRNA that is highly expressed in normal brain but virtually absent in tumor cells.
  • Many of miRNAs are specific to or over-expressed in certain organs/tissues/cells (see, e.g., Hua et al., BMC Genomics, 2009, 10:214; Liang et al., BMC Genomics. 2007, 8:166; Landgraf et al., Cell.2007, 129:1401-1414; Lee et al., RNA.2008, 14:35-42).
  • binding site for particular microRNA(s) that are specifically expressed in non- myeloid cells. This method can be utilized herein to reduce off-target expression of the gene of interest encoded by the recombinant nucleic acid.
  • a binding site for miRNA for example is a short polynucleotide sequence within the recombinant mRNA that comprises a region that hybridizes with an miRNA present in a cell.
  • non-myeloid cells discussed in this concept include, but are not limited to blood, bone, brain, kidney, muscle, spinal cord, nerve, endocrine system, uterine, ear, foreskin, liver, intestine, bladder or skin.
  • the cells can include neural cells, lymphocytes, epidermal cells, islet cells, intestinal cells or fibroblasts.
  • Exemplary non-myeloid cells may include embryonic stem cells.
  • miRNA binding sites that may be incorporated in the recombinant mRNA of the invention may include for example, hsa-mir-302a, hsa-miR-302b, hsa-miR-302c, hsa-miR-302d, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373.
  • hsa-mir-302a hsa-miR-302b, hsa-miR-302c, hsa-miR-302d, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373.
  • Exemplary non-myeloid cells may include neuronal cells.
  • miRNA binding sites that may be incorporated in the recombinant mRNA of the invention may include those are enriched in certain cellular compartments, particularly in axons, dendrites and synapses (see, e.g., Schratt et al., Nature.439:283-289, 2006; Lugli et al., J Neurochem.106:650-661, 2008; Bicker and Schratt, J Cell Mol Med., 12:1466-1476, 2008; Smalheiser and Lugli, Neuromolecular Med. 11:133-140, 2009; Rajasethupathy, Neuron.
  • Exemplary non-myeloid cells may include endothelial cells.
  • Exemplary endothelial cell- specific miRNA may include miR-10a/b, miR-24/27, miR-125a, miR-126 and miR-221/222.
  • binding sites for more than one miRNAs are incorporated in the recombinant construct.
  • the miRNA binding sites are incorporated in more than one copy numbers, in tandem.
  • the recombinant nucleic acid may incorporate binding sites for two or more different miRNAs.
  • the recombinant nucleic acid may incorporate binding sites for 1, 2, 3, 4, 5, 6 or more different miRNAs.
  • the recombinant nucleic acid may incorporate multiple binding sites for each miRNA.
  • the recombinant nucleic acid may incorporate 1, 2, 3, 4, 5, 6 or binding sites for one or more miRNAs, in tandem.
  • MicroRNAs may upregulate the expression of a gene and the miRNA binding site may be utilized accordingly.
  • Nanoparticles for nucleic acid delivery [0713]
  • a nucleic acid is introduced into a myeloid cell with a nanoparticle (NP).
  • a nanoparticle may be of various shapes or sizes and may harbor the nucleic acid encoding the CFP or PFP.
  • the NP is a lipid nanoparticle (LNP).
  • the NP comprises poly(amino acids), polysaccharides and poly(alpha-hydroxy acids), gold, silver, carbon, iron, silica, or any combination thereof.
  • the NP comprises a polylactide-co-glycolide (PGLA) particle.
  • the nucleic acid is encapsulated in the NP, for example, via water/oil emulsion or water-oil-water emulsion.
  • the nucleic acid is conjugated to a component of or complexed with components of the NP.
  • a protein corona may form around NPs. A protein corona may form in a two-step process.
  • proteins of lower affinity bind either directly to the NP or to the proteins in the primary corona forming a secondary corona. Constituents of the protein corona may thus be impacted by the protein content of the serum and thus by the homeostatic or immune responses that regulate it.
  • proteins with high abundance such as albumin, comprise a significant proportion of the primary corona.
  • NPs with different charges bind significant amounts of less-abundant proteins in particular environments, e.g. in plasma with certain antigen or antibody.
  • NPs are engineered to reduce changes to NP charges or masking of functional groups, and/or increase the serum half-life of the NPs.
  • NP surface coating are designed to modulate opsonization events.
  • the NP’s surface may be coated with polymeric ethylene glycol (PEG) or its low molecular weight derivative polyethylene oxide (PEO).
  • PEG increases surface hydrophilicity, resulting in improved circulating NP half-life due to reduced serum protein binding.
  • the NP coated with PEG or PEO are engineered to result in reduced toxicity or increased biocompatibility of the NPs. Additional NP design and NP targeting for myeloid cells are described in Getts et al., Trends Immunol. 36(7): 419-427 (2015), the entirety of which is incorporated herein by reference. [0714] NPs described herein may be used to introduce the recombinant nucleic acid into a cell in in vitro/ex vivo cell culture or administered in vivo. In some embodiments, the NP is modified for in vivo administration.
  • the NP may comprise surface modification or attachment of binding moieties to bind specific toxins, proteins, ligands, or any combination thereof, before being taken up by liver or spleen phagocytes.
  • infused highly negatively charged ‘immune-modifying NPs’ IMPs
  • IMPs can absorb certain blood proteins, including S100 family and heat shock proteins, before finally being removed and destroyed by cells of the mononuclear phagocyte system.
  • this mechanism may also be used to capture and concentrate certain circulating proteins.
  • IMPs have been shown to bind Annexin 1. The accumulation of Annexin 1 and its presentation to particular leukocyte subsets can have broad immune outcomes.
  • Annexin 1-loaded NPs may reduce neutrophils via induction of apoptosis and/or promote T cell activation.
  • the NP is designed to target a cell surface receptor, e.g. a scavenger receptor.
  • an NP is a particle with highly negative surface charge.
  • the NP encapsulates the nucleic acid wherein the nucleic acid is a naked DNA molecule.
  • the NP encapsulates the nucleic acid wherein the nucleic acid is an mRNA molecule.
  • the NP encapsulates the nucleic acid wherein the nucleic acid is a circular RNA (circRNA) molecule.
  • the NP encapsulates the nucleic acid wherein the nucleic acid is a vector, a plasmid, or a portion or fragment thereof.
  • NPs may be delivered to a cell in vitro, ex vivo or in vivo.
  • a NP is delivered to a phagocytic cell ex vivo.
  • a NP is delivered to a phagocytic cell in vivo.
  • the NP is less than 100nm in diameter.
  • the NP is more than 100nm in diameter.
  • the NP is a rod-shaped NP.
  • the NP is a spherical NP.
  • the NP is a spherical NP for delivery to a phagocytic cell.
  • the NP is at least 100nm in diameter and does not trigger or triggers reduced toxicity when delivered to a cell.
  • the NP is positively charged.
  • the NP is negatively charged.
  • the NP is a cationic NP that is delivered and taken up by a myeloid cell ex vivo or in vivo.
  • Stiffness may affect the biological impact of NPs.
  • NPs made of rigid materials may be associated with increased potential for embolism, while flexible polymer-based NPs that can more easily deform may gain better access to tissues during the complex vascular changes associated with inflammation.
  • the fluidity of NPs affects the ability of antigen-loaded NP to stimulate immune responses.
  • intramuscular, solid-phase, antigen-containing liposome immunization may elicit a more robust Th1/Th17 response than similarly administered fluid-phase liposomes.
  • solid-phase particles may result from the formation of an immobilized antigen particle depot and may result in a prolonged supply of antigen for APCs also associated with upregulation of positive costimulatory molecules such as CD80, which support efficient T cell priming.
  • the lipid nanoparticle comprises a charged lipid, e.g., a cationic lipid.
  • Charged lipids may be synthetic or naturally derived. Examples of charged lipids include phosphatidylserines, phosphatidic acids, phosphatidylglycerols, phosphatidylinositols, sterol hemisuccinates, dialkyl trimethylammonium-propanes, (e.g. DOTAP, DOTMA), dialkyl dimethylaminopropanes, ethyl phosphocholines, dimethylaminoethane carbamoyl sterols (e.g. DC- Chol).
  • DOTAP phosphatidylglycerols
  • phosphatidylinositols sterol hemisuccinates
  • dialkyl trimethylammonium-propanes e.g. DOTAP, DOTMA
  • dialkyl dimethylaminopropanes
  • the lipid nanoparticle comprises a neutral lipid.
  • a neutral lipid is a lipid that exists in either an uncharged state or as a zwitterionic form at a selected pH.
  • such lipids include, but are not limited to, phosphotidylcholines such as 1,2- Distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-Dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine (POPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), phophatidylethanolamines such as 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE),
  • DOPE 1,2-Di
  • a neutral lipid is selected from the neutral lipid is selected from DSPC, DPPC, DMPC, DOPC, POPC, DOPE and SM. In some embodiments, the neutral lipid may be DSPC.
  • Neutral lipids may be synthetic or naturally derived.
  • a steroid or steroid analog may be incorporated into the LNP.
  • the molar ratio of the compound (e.g., the mRNA) to the neutral lipid ranges from about 2:1 to about 8:1.
  • the compositions further comprise a steroid or steroid analogue. In certain embodiments, the steroid or steroid analogue is cholesterol.
  • the polymer conjugated lipid is a pegylated lipid.
  • some embodiments include a pegylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy- polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a pegylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2′,3′- di(tetradecanoyloxy)propyl-1-O-(cw-methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), a pegylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as ⁇
  • PEG-DAG pegylated diacylglycerol
  • PEG-DMG 1-(monome
  • the molar ratio of the compound to the pegylated lipid ranges from about 100:1 to about 25:1.
  • the lipid components of the LNP described herein comprise components that have been previously described in US20170342442A1.
  • the lipid nanoparticle for the polynucleic acid delivery in myeloid cells ex vivo or in vivo comprises one or more ionizable cationic lipids as described in US patent 9,593,077.
  • the lipid nanoparticle for the polynucleic acid delivery in myeloid cells ex vivo or in vivo comprises one or more phospholipids selected from phospholipid is selected from the group consisting of phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, di stearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine, as described in US patent 9,504,651.
  • the subject is administered a pharmaceutical composition comprising a mRNA encoding the CFP or PFP as described herein.
  • the mRNA is co-formulated into nanoparticles (NPs), such as lipid nanoparticles (LNPs).
  • NPs nanoparticles
  • LNPs lipid nanoparticles
  • the LNP may comprise cationic lipids or ionizable lipids.
  • the mRNA is formulated into polymeric particles, for example, polyethyleneimine particles, poly(glycoamidoamine), ly( ⁇ -amino)esters (PBAEs), PEG particles, ceramide-PEGs, polyamindoamine particles, or polylactic-co-glycolic acid particles (PLGA).
  • the mRNA is administered by direct injection.
  • the mRNA is complexed with transfection agents, e.g. Lipofectamine 2000, jetPEI, RNAiMAX, or Invivofectamine.
  • a method of in vivo delivery of a composition comprising mRNA wherein the mRNA may be complexed with a cationic molecule wherein the delivery comprises systemic delivery, e.g., delivery via intravenous, intraperitoneal, intramuscular or subcutaneous, or any other suitable delivery course.
  • the delivery comprises local delivery, for example intratumoral or in a specific tissue or site of a disease, such as a site of an inflammation, e.g. arthritic joints.
  • the cationic molecule may be a cationic lipid, a cationic polymer, a cationic dendrimer, or a synthetic molecule.
  • the mRNA is at least 5 kb in length. In one embodiment, the mRNA is about 5.5 kb, about 6.0, about 6.5, about 7.0, about 7.5, about 8, about 8.5, about 9, about 9.5, about 10 kb in length. In some embodiments, the mRNA is at least 95% pure. In some embodiments, the mRNA is at least 98% pure. In some embodiments, the mRNA is at least99% pure. In some embodiments, the mRNA is sufficiently free of salts. In some embodiments, the mRNA is at least 99% pure. In some embodiments, the mRNA is effectively free of salts. In some embodiments, the mRNA is about 0.1 microgram/microliter in concentration.
  • the mRNA is complexed with a cationic molecule, e.g., a cationic polymer and is formulated in a microsome or a nanoparticle.
  • the nanoparticle is a lipid nanoparticle, comprising one or more cationic lipids and one or more neutral lipids.
  • the mRNA of a length greater than 5kb is complexed with a cationic molecule, and is administered systemically.
  • the mRNA may be a naked mRNA.
  • the mRNA may be modified or unmodified. For example, the mRNA may be chemically modified.
  • nucleobases and/or sequences of the mRNA are modified to increase stability and half-life of the mRNA.
  • the mRNA is glycosylated. Additional mRNA modification and delivery approaches as described in Flynn et al., BioRxiv 787614 (2019) and Kowalski et al. Mol. Ther. 27(4): 710-728 (2019) are each incorporated herein by reference in its entirety.
  • the mRNA is a self-amplifying mRNA.
  • the mRNA is a genome-integrating mRNA.
  • the mRNA is a circular RNA.
  • the mRNA comprises a sequence comprising a binding site for an miRNA that is expressed in a non-myeloid cell, but is not expressed in a myeloid cell.
  • the NP is a Lipid nanoparticle (LNP).
  • LNPs may comprise a polar and or a nonpolar lipid.
  • cholesterol is present in the LNPs for efficient delivery.
  • LNPs are 100-300 nm in diameter provide efficient means of mRNA delivery to various cell types, including myeloid cells, such as macrophages.
  • LNP may be used to introduce the recombinant nucleic acids into a cell in in vitro cell culture.
  • the LNP encapsulates the nucleic acid wherein the nucleic acid is a naked DNA molecule. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is an mRNA molecule. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is inserted in a vector, such as a plasmid vector. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is a circRNA molecule. [0732] In some embodiments, the LNP is used to deliver the nucleic acid into the subject. LNP can be used to deliver nucleic acid systemically in a subject. It can be delivered by injection.
  • the LNP comprising the nucleic acid is injected by intravenous route. In some embodiments the LNP is injected subcutaneously.
  • Pharmaceutical Composition [0733] Provided herein is a pharmaceutical composition, comprising engineered myeloid cells, such as macrophages, comprising a recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient. [0734] Also provided herein is a pharmaceutical composition, comprising a recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition may comprise DNA, mRNA, self-amplifying mRNA, self-integrating mRNA (e.g., retrotransposons), or circRNA or a liposomal composition of any one of these.
  • the liposome is an LNP.
  • a pharmaceutical composition comprising a vector comprising the recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition may comprise DNA, mRNA or circRNA inserted in a plasmid vector or a viral vector.
  • the engineered myeloid cells are grown in cell culture sufficient for a therapeutic administration dose, and washed, and resuspended into a pharmaceutical composition comprising a pharmaceutically acceptable solution.
  • “Pharmaceutically acceptable solution” may refer to those solutions or comprising an acid or aqueous salt of the acid, which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic
  • the pharmaceutically acceptable solution may comprise a pharmaceutically acceptable base or a salt thereof.
  • “Pharmaceutically acceptable base” refers to those basic solutions or salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • basic ion exchange resins such as
  • Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine.
  • solvate may refer to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • the excipient comprises a sterile buffer, (e.g. HEPES or PBS) at neutral pH.
  • the pH of the pharmaceutical composition is at 7.5.
  • the pH may vary within an acceptable range.
  • the engineered cells may be comprised in sterile enriched cell suspension medium comprising complement deactivated or synthetic serum.
  • the pharmaceutic composition further comprises cytokines, chemokines or growth factors for cell preservation and function.
  • the pharmaceutical composition may comprise additional therapeutic agents, co-administered with the engineered cells.
  • a pharmaceutical composition comprising one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: a CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembran
  • CAR chimeric antigen receptor
  • the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence. In some embodiments, the first sequence and the second sequence are operatively linked by a linker sequence. In some embodiments, the linker sequence comprises a protease cleavage site. In some embodiments, the protease cleavage site is a T2A cleavage site or a P2A cleavage site [0743] In some embodiments, the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle. In some embodiments, the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle.
  • the one or more polynucleic acids comprises a first polynucleic acid molecule comprising the first sequence and a second polynucleic acid molecule comprising the second sequence.
  • the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle.
  • the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle.
  • after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject.
  • the CAR after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject.
  • the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject.
  • the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon.
  • at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit.
  • the CAR intracellular domain further comprises a costimulatory domain.
  • the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137).
  • the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain.
  • the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89.
  • the CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain.
  • the CFP after administration of the pharmaceutical composition to a human subject the CFP is specifically expressed in myeloid cells, monocytes or macrophages of the human subject.
  • the CAR is specifically expressed in T cells of the human subject.
  • the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89.
  • the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain.
  • the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from Fc ⁇ R, Fc ⁇ R, Fc ⁇ R, CD40 or CD3zeta.
  • the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain.
  • PI3K phosphoinositide 3-kinase
  • the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4.
  • the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89.
  • the one or more polynucleic acids is an mRNA.
  • the nanoparticle delivery vehicle comprises a lipid nanoparticle.
  • the lipid nanoparticle comprises a polar lipid.
  • the polar lipid is a cationic lipid.
  • the lipid nanoparticle comprises a non-polar lipid.
  • the nanoparticle comprises polyethylene glycol (PEG).
  • the nanoparticle comprises cholesterol as one of the lipid components. In some embodiments, the lipid nanoparticle is from 100 to 300 nm in diameter. [0755] In some embodiments, the first antigen binding domain and the second antigen binding domain bind to the same antigen. In some embodiments, the first antigen binding domain and the second antigen binding domain bind to different antigens. In some embodiments, the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75.
  • an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75.
  • the pharmaceutical composition is a lipid nanoparticle (LNP) encapsulated mRNA encoding a CAR, (alternatively, or interchangeably, a monocyte CAR, a chimeric fusion protein (CFP) or a phagocytic fusion protein (PFP)), where the CAR comprises an antigen binding domain that can bind to an antigen on a cell surface, a transmembrane domain and an intracellular signaling domain, and the mRNA comprises a 5’UTR, a 3’UTR, further comprising a 5’CAP at the terminal, where the 5’ CAP has a general formula 5’-m7GpppAmpG –, described as CC1 cap throughout the specification provided herein.
  • LNP lipid nanoparticle
  • the second nucleotide is methylated.
  • the second nucleotide is Adenine. This is a Cap 1 structure, and is identical to most eukaryotic mRNA capping structure.
  • the CAR of the pharmaceutical composition comprises a TROP2 binding domain.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula m 7 GpppAmpG; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a) and an intracellular cytoplasmic sequence from the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a) respectively from which the TMD is derived.
  • an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol
  • the TMD has a sequence that is identical to the sequence of the TMD of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a).
  • the second nucleotide of the 5’Cap is further modified.
  • the adenosine at the second position is modified at the N6 position.
  • the mRNA further comprises a poly A tail.
  • the poly A tail is at least 80 nucleotides long.
  • the poly A tail is about 100, about 110, about 120 or about 150 nucleotides long.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine comprising a further modification in the N6 position; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a) and an intracellular cytoplasmic sequence from the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a) respectively from which the TMD is derived.
  • the CFP comprises a transmembrane domain from
  • the TMD has a sequence that is identical to the sequence of the TMD of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a).
  • the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., TROP2.
  • the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., GPC3.
  • the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., HER2.
  • the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., CD5.
  • the antigen binding domain of the CAR is an scFv.
  • the antigen binding domain of the CAR is a VHH domain.
  • the mRNA further comprises a poly A tail.
  • the poly A tail is at least 80 nucleotides long.
  • the poly A tail is about 100, about 110, about 120 or about 150 nucleotides long.
  • Myeloid cell-specific CFP constructs are designed to comprise a transmembrane domain that dimerizes with an endogenous Fc-gamma receptor that is specifically expressed in a myeloid cell, thereby becoming functionally active as a result of dimerization.
  • myeloid cell-specific CFP constructs comprises an Fc ⁇ R1 (CD89) transmembrane domain.
  • myeloid cell-specific CFP constructs comprises a human Fc ⁇ R1 (CD89) transmembrane domain.
  • myeloid cell-specific CFP constructs comprises a mouse Fc ⁇ R1 (CD89) transmembrane domain.
  • myeloid cell-specific CFP constructs comprises a rodent Fc ⁇ R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a equine Fc ⁇ R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a porcine Fc ⁇ R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a ovine Fc ⁇ R1 (CD89) transmembrane domain. In some embodiments, the CD89 TMD is at least 80% homologous to a human or a mouse CD89 TMD.
  • myeloid cell-specific CFP constructs comprises a mouse Fc ⁇ RIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a rodent Fc ⁇ RIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a equine Fc ⁇ RIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a porcine Fc ⁇ RIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a ovine Fc ⁇ RIII (CD16) transmembrane domain.
  • the CD16 TMD is at least 80% homologous to a human or a mouse CD16 TMD. In some embodiments, the CD16TMD is at least 85% homologous to a human or a mouse CD16 TMD. In some embodiments, CD16 TMD is at least 90% homologous to a human or a mouse CD16 TMD. In some embodiments, CD16 TMD is at least 95% homologous to a human or a mouse CD16 TMD. In one embodiment, the Fc ⁇ RIII is Fc ⁇ RIIIA (CD16a).
  • any myeloid cell specific CFP can be designed or generated using a TMD from a CD89 molecule, a CD16 molecule, a CD64 molecule or a CD32a molecule, and a myeloid cell specific CFP having the desired antigen binding domain can be created based on the compositions and methods described in the instant disclosure.
  • the CFP exhibits myeloid cell-specific expression.
  • the myeloid cell-specific expression is an Fc-gamma dependent expression, that is, if the cell that comprises the nucleic acid comprising the CFP construct expresses an Fc-gamma endogenously, the CFP will co-express in the cell.
  • the CFP even though it expresses in a cell other than a myeloid cell, does so only transiently, and may not be detectable.
  • only a myeloid cell expresses a functional CFP in an Fc-gamma dependent manner, that is, the CFP is not functional in a cell that does not express Fc-gamma endogenously.
  • the myeloid cell specific CFP comprises an antigen binding domain that binds to a cancer cell-specific target antigen, and an intracellular region comprising 1, 2, 3 or more domain, e.g., intracellular cell signaling domain, for example, a PI3 kinase recruitment domain.
  • the myeloid cell specific CFP comprises an antigen binding domain that binds to a CD5 target antigen.
  • the respective CFP is designed herein comprising : an extracellular domain comprising an anti-CD5 antibody or a part thereof, e.g., an anti-CD5 ScFv, a hinge domain, e.g. a CD8 hinge domain, a transmembrane domain capable of dimerizing with an Fc-gamma receptor endogenously expressed by a myeloid cell and an intracellular region comprising 1, 2, 3 or more domain, e.g., intracellular cell signaling domain including, for example, a CD40 intracellular signaling domain, and/or a PI3kinase recruitment domain.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position and the encoded CFP is an anti-CD5 CFP that comprises sequence that is at least 80% identical to the sequence of G S NQ QGSGS G Q S GQ G N S N . (SEQ ID NO.
  • the anti-CD5 heavy chain variable domain comprises an amino acid sequence that is at least 80% identical to the sequence
  • the anti-CD5 light chain variable domain comprises an amino acid sequence that is at least 80% identical to the sequence DIQMTQSPSSLSASVGDRVTITCRASQDINSYLSWFQQKPGKAPKTLIYRANRLESGVPSRFS GSGSGTDYTLTISSLQYEDFGIYYCQQYDESPWTFGGGTKLEIK.
  • the anti-CD5 binding domain comprises a heavy chain CDR1 sequence, a CDR2 sequence and a CDR3 sequence, wherein the CDR1 may be denoted by a sequence of NYGMN, a CDR2 may be denoted by a sequence of WINTHTGEPTYADSFKG and a CDR3 may be denoted by a sequence of RGYDWYFDV; or the anti-CD5 binding domain may comprise a sequence of wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to Kabat scheme.
  • the anti-CD5 binding domain comprises a light chain CDR1 sequence, a CDR2 sequence and a CDR3 sequence, wherein the CDR1 sequence may be RASQDINSYLS; the CDR2 sequence may be RANRLES, and the CDR3 sequence may be QQYDESPWT; or wherein the anti-CD5 binding domain may comprise a sequence of DIQMTQSPSSLSASVGDRVTITCRASQDINSYLSWFQQKPGKAPKTLIYRANRLESGVPSRFS GSGSGTDYTLTISSLQYEDFGIYYCQQYDESPWTFGGGTKLEIK wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to Kabat scheme.
  • an exemplary anti-CD5 CAR sequence comprises a signal sequence of MWLQSLLLLGTVACSIS, or a sequence that is at least 90% identical to the above sequence.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and the encoded CFP is an anti- HER2 CFP that comprises a sequence that is at least 80% identical to : SDGVYTGLSTRNQETYETLKHEKPPQGSYEDMRGILYAAPQLRSIRGQPGPNHEEDADSYE (wherein
  • the anti-HER2 CFP antigen binding domain comprises a heavy chain variable sequence that is at least 80% identical to : Q Q [0771] In some embodiments, the anti-HER2 CFP antigen binding domain comprises a light chain variable sequence that is at least 80% identical to : [0772] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc
  • the antigen binding domain binds to TROP2 antigen, and comprises a heavy chain variable domain having a sequence that has at least 80%, or at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence QVQLQQSGSELKKPGASVKVSCKASGYTFTNYGMNWVKQAPGQGLKWMGWINTYTGEP TYTDDFKGRFAFSLDTSVSTAYLQISSLKADDTAVYFCARGGFGSSYWYFDVWGQGSLVT VSS.
  • the antigen binding domain binds to TROP2 antigen, and comprises a light chain variable domain having a sequence that has at least 80%, or at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence DIQLTQSPSSLSASVGDRVSITCKASQDVSIAVAWYQQKPGKAPKLLIYSASYRYTGVPDRF SGSGSGTDFTLTISSLQPEDFAVYYCQQHYITPLTFGAGTKVEIKR.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the CFP comprises a TROP2 binding domain that comprises a heavy chain CDR3 sequence, GGFGSSYWYFDV.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a); and wherein the antigen binding domain binds to TROP2 antigen, and wherein the binding domain comprises a heavy chain CDR3 sequence, GGFGSSYWYFDV.
  • the mRNA encodes a TROP2-binding CFP that comprises an antigen binding domain having a heavy chain (VH) CDR1 sequence of NYGMN and a VH CDR2 sequence of WINTYTGEPTYTDDFKG.
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA
  • the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc ⁇ R1 (CD64), Fc ⁇ RIIIA (CD16), or Fc ⁇ RIIA (CD32a); wherein, the encoded chimeric antigen receptor comprises GPC3 binding variable heavy chain (VH), and a variable light chain (VL), wherein the variable heavy chain comprises a HCDR1, a HCDR2 and
  • composition comprising an polynucleotide, having a sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to
  • the polynucleotide is an mRNA, further comprising a 5’ UTR, a 3’ UTR, a poly A tail, and a 5’- cap, wherein the cap is a cap-1 analog.
  • the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position.
  • the polynucleotide e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non-cationic lipid, and PEG.
  • the mRNA encodes a chimeric fusion protein having an anti- CD70 binding domain, comprising a sequence that is at least 80% identical to, or a sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence : QQ Q Q Q Q ; wherein, in some embodiments, the mRNA comprises a 5’ UTR, a 3’ UTR, a poly A tail, and a 5’-cap, wherein the cap is a cap-1 analog.
  • the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position.
  • the polynucleotide e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non- cationic lipid, and PEG.
  • the mRNA encodes a chimeric fusion protein having an anti- CD137 binding domain, comprising a sequence that is at least 80% identical to, or a sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence : ; wherein, in some embodiments, the mRNA comprises a 5’ UTR, a 3’ UTR, a poly A tail, and a 5’-cap, wherein the cap is a cap-1 analog.
  • the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position.
  • the polynucleotide e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non- cationic lipid, and PEG.
  • the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer.
  • Treatment Methods Provided herein are methods for treating cancer in a subject using a pharmaceutical composition comprising engineered myeloid cells, such as phagocytic cells (e.g., macrophages), expressing a recombinant nucleic acid encoding a CFP, such as a phagocytic receptor (PR) fusion protein (PFP), to target, attack and kill cancer cells directly or indirectly.
  • engineered myeloid cells such as phagocytic cells
  • phagocytic cells are also designated as CAR-P cells in the descriptions herein.
  • Cancers include, but are not limited to T cell lymphoma, cutaneous lymphoma, B cell cancer (e.g., multiple myeloma, Waldenstrom's macroglobulinemia), the heavy chain diseases (such as, for example, alpha chain disease, gamma chain disease, and mu chain disease), benign monoclonal gammopathy, and immunocytic amyloidosis, melanomas, breast cancer, lung cancer, bronchus cancer, colorectal cancer, prostate cancer (e.g., metastatic, hormone refractory prostate cancer), pancreatic cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma
  • cancers include human sarcomas and carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma
  • human sarcomas and carcinomas e.g.,
  • the cancer is an epithelial cancer such as, but not limited to, bladder cancer, breast cancer, cervical cancer, colon cancer, gynecologic cancers, renal cancer, laryngeal cancer, lung cancer, oral cancer, head and neck cancer, ovarian cancer, pancreatic cancer, prostate cancer, or skin cancer.
  • the cancer is breast cancer, prostate cancer, lung cancer, or colon cancer.
  • the epithelial cancer is non-small-cell lung cancer, nonpapillary renal cell carcinoma, cervical carcinoma, ovarian carcinoma (e.g., serous ovarian carcinoma), or breast carcinoma.
  • the epithelial cancers can be characterized in various other ways including, but not limited to, serous, endometrioid, mucinous, clear cell, or undifferentiated.
  • the present disclosure is used in the treatment, diagnosis, and/or prognosis of lymphoma or its subtypes, including, but not limited to, mantle cell lymphoma. Lymphoproliferative disorders are also considered to be proliferative diseases.
  • cellular immunotherapy comprises providing the patient a medicament comprising live cells.
  • a patient or a subject having cancer is treated with autologous cells, the method comprising, isolation of PBMC-derived myeloid cells, such as macrophages, modifying the cells ex vivo to generate phagocytic myeloid cells capable of tumor lysis by introducing into the cells a recombinant nucleic acid encoding a CFP, and administering the modified myeloid cells into the subject.
  • a subject is administered one or more doses of a pharmaceutical composition comprising therapeutic myeloid cells, such as phagocytic cells, wherein the cells are allogeneic.
  • HLA may be matched for compatibility with the subject, and such that the cells do not lead to graft versus Host Disease, GVHD.
  • a subject arriving at the clinic is HLA typed for determining the HLA antigens expressed by the subject.
  • HLA ⁇ typing is conventionally carried out by either serological methods using antibodies or by PCR ⁇ based methods such as Sequence Specific Oligonucleotide Probe Hybridization (SSOP), or Sequence Based Typing (SBT).
  • SSOP Sequence Specific Oligonucleotide Probe Hybridization
  • SBT Sequence Based Typing
  • the sequence information may be identified by either sequencing methods or methods employing mass spectrometry, such as liquid chromatography—mass spectrometry (LC-MS or LC- MS/MS, or alternatively HPLC-MS or HPLC-MS/MS).
  • the phagocytic cell is derived from the subject, transfected or transduced with the recombinant nucleic acid in vitro, expanded in cell culture in vitro for achieving a number suitable for administration, and then administered to the subject.
  • the steps of transfected or transduced with the recombinant nucleic acid in vitro, expanded in cell culture in vitro for achieving a number suitable for administration takes 2 days, or 3 days, or 4 days or 5 days or 6 days or 7 days or 8 days or 9 days or 10 days.
  • sufficient quantities of transfected or transduced myeloid cells, such as macrophages, comprising the recombinant nucleic acid are preserved aseptically, which are administered to the subject as “off the shelf” products after HLA typing and matching the product with the recipients HLA subtypes.
  • the engineered phagocytes are cryopreserved.
  • the engineered phagocytes are cryopreserved in suitable media to withstand thawing without considerable loss in cell viability.
  • the subject is administered a pharmaceutical composition comprising the DNA, or the mRNA or the circRNA in a vector, or in a pharmaceutically acceptable excipient described above.
  • the administration of the off the shelf cellular products may be instantaneous, or may require 1 day, 2 days or 3 days or 4 days or 5 days or 6 days or 7 days or more prior to administration.
  • the pharmaceutical composition comprising cell, or nucleic acid may be preserved over time from preparation until use in frozen condition. In some embodiments, the pharmaceutical composition may be thawed once.
  • the pharmaceutical composition may be thawed more than once. In some embodiments, the pharmaceutical composition is stabilized after a freeze-thaw cycle prior administering to the subject. In some embodiments the pharmaceutical composition is tested for final quality control after thawing prior administration.
  • a composition comprising 10 ⁇ 6 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10 ⁇ 7 engineered cells are administered per administration dose. In some embodiments, a composition comprising 5X 10 ⁇ 7 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10 ⁇ 8 engineered cells are administered per administration dose. In some embodiments, a composition comprising 2x10 ⁇ 8 engineered cells are administered per administration dose.
  • a composition comprising 5x10 ⁇ 8 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10 ⁇ 9 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10 ⁇ 10 engineered cells are administered per administration dose. [0794] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once. [0795] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered more than once.
  • the engineered myeloid cells such as phagocytic cells
  • the engineered myeloid cells such as phagocytic cells
  • the engineered myeloid cells such as phagocytic cells
  • the engineered myeloid cells, such as phagocytic cells are administered once every two weeks.
  • the engineered myeloid cells are administered once every three weeks. [0801] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once monthly. [0802] In some embodiments, the engineered phagocytic cells are administered once in every 2 months, once in every 3 months, once in every 4 months, once in every 5 months or once in every 6 months. [0803] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by injection. [0804] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by infusion.
  • the engineered myeloid cells are administered by intravenous infusion.
  • the engineered myeloid cells such as phagocytic cells, are administered by subcutaneous infusion.
  • the pharmaceutical composition comprising the recombinant nucleic acid or the engineered cells may be administered by any route which results in a therapeutically effective outcome.
  • enteral into the intestine
  • gastroenteral epidural
  • epidural into the dura mater
  • oral by way of the mouth
  • transdermal peridural
  • intracerebral into the cerebrum
  • intracerebroventricular into the cerebral ventricles
  • epicutaneous application onto the skin
  • intradermal into the skin itself
  • subcutaneous under the skin
  • nasal administration through the nose
  • intravenous into a vein
  • intravenous bolus intravenous drip
  • intraarterial into an artery
  • intramuscular into a muscle
  • intracardiac into the heart
  • intraosseous infusion into the bone marrow
  • intrathecal into the spinal canal
  • intraperitoneal infusion or injection into the peritoneum
  • intravesical infusion intravitreal, (through the eye), intracavernous injection (into a pathologic cavity), intracavitary (into the base of the penis), intravaginal administration
  • compositions may be administered in a way which allows them to cross the blood-brain barrier, vascular barrier, or other epithelial barrier.
  • the subject is administered a pharmaceutical composition comprising the nucleic acid encoding the CFP or PFP as described herein.
  • the subject is administered a pharmaceutical composition comprising DNA, mRNA, or circRNA.
  • the subject is administered a vector harboring the nucleic acid, e.g., DNA, mRNA, or circRNA.
  • the nucleic acid is administered or in a pharmaceutically acceptable excipient described above.
  • the subject is administered a nanoparticle (NP) associated with the nucleic acid, e.g. a DNA, an mRNA, or a circRNA encoding the CFP or PFP as described herein.
  • NP nanoparticle
  • the nucleic acid is encapsulated in the nanoparticle.
  • the nucleic acid is conjugated to the nanoparticle.
  • the NP is a polylactide-co- glycolide (PGLA) particle.
  • the NP is administered subcutaneously.
  • the NP is administered intravenously.
  • the NP is engineered in relation to the administration route.
  • the size, shape, or charges of the NP maybe engineered according to the administration route.
  • subcutaneously administered NPs are less than 200nm in size. In some embodiments, subcutaneously administered NPs are more than 200nm in size. In some embodiments, subcutaneously administered NPs are at least 30nm in size. In some embodiments, the NPs are intravenously infused. In some embodiments, intravenously infused NPs are at least 5nm in diameter. In some embodiments, intravenously infused NPs are at least 30nm in diameter. In some embodiments, intravenously infused NPs are at least 100nm in diameter. In certain embodiments, the administered NPs, e.g.
  • the subject is administered a pharmaceutical composition comprising a circRNA encoding the CFP or PFP as described herein.
  • the circRNA may be administered in any route as described herein.
  • the circRNA may be directly infused.
  • the circRNA may be in a formulation or solution comprising one or more of sodium chloride, calcium chloride, phosphate and/or EDTA.
  • the circRNA solution may include one or more of saline, saline with 2 mM calcium, 5% sucrose, 5% sucrose with 2 mM calcium, 5% Mannitol, 5% Mannitol with 2 mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2 mM calcium and mannose.
  • the circRNA solution is lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of circRNA in the buffer solution over a period of time and/or under a variety of conditions.
  • the circRNA is formulated in a lyophilized gel- phase liposomal composition.
  • the circRNA formulation comprises a bulking agent, e.g. sucrose, trehalose, mannitol, glycine, lactose and/or raffinose, to impart a desired consistency to the formulation and/or stabilization of formulation components. Additional formulation and administration approaches for circRNA as described in US Publications No. US2012060293, and US20170204422 are herein incorporated by reference in entirety. EMBODIMENTS 1.
  • a composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the PFP to the antigen of the target cell, the killing or phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP.
  • the composition of embodiment 1 or 2 wherein the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1.
  • composition of embodiment 5, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain.
  • a composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1.
  • the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I,
  • compositions comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1,
  • composition of embodiment 11 wherein upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 55% compared to a cell not expressing the PFP.
  • the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1.
  • the intracellular signaling domain comprises a pro-inflammatory signaling domain.
  • the composition of embodiment 14, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. 16.
  • a composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. 17.
  • PR phagocytic or tethering receptor
  • PFP fusion protein
  • composition of embodiment 16 wherein upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. 18.
  • a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206,
  • the target cell expressing the antigen is a cancer cell.
  • the target cell expressing the antigen is at least 0.8 microns in diameter. 28.
  • composition according to embodiment 29, wherein the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof.
  • the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof.
  • a cell expressing the PFP exhibits an increase in expression of TIM1 ligand compared to a cell not expressing the PFP.
  • a cell expressing the PFP exhibits an increase in respiratory burst compared to a cell not expressing the PFP.
  • 57. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in ROS production compared to a cell not expressing the PFP.
  • a cell expressing the PFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the PFP.
  • the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain.
  • composition of embodiment 64 wherein the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the PFP.
  • the intracellular domain comprises an F-actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain.
  • a cell expressing the PFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production. 68.
  • composition of any one of the preceding embodiments, wherein the extracellular domain comprises an Ig binding domain.
  • the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, FcR ⁇ I, FcR ⁇ IIA, FcR ⁇ IIB, FcR ⁇ IIC, FcR ⁇ IIIA, FcR ⁇ IIIB, FcRn, TRIM21, FcRL5 binding domain.
  • the composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcR extracellular domain. 71.
  • composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcR-alpha, FcR ⁇ , FcR ⁇ or FcR ⁇ extracellular domain.
  • FCAR FcR ⁇
  • FcR ⁇ extracellular domain
  • FcR ⁇ extracellular domain
  • FCER1A FcR ⁇
  • composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcR ⁇ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, FCGR3B) extracellular domain.
  • FcR ⁇ FcR ⁇
  • FCGR2A FCGR2A
  • FCGR2B FCGR2C
  • FCGR3A FCGR3B
  • composition of any one of the preceding embodiments, wherein the extracellular domain comprises one or more integrin ⁇ 1, ⁇ 2, ⁇ IIb, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, ⁇ 8, ⁇ 9, ⁇ 10, ⁇ 11, ⁇ D, ⁇ E, ⁇ L, ⁇ M, ⁇ V, ⁇ X, ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, or ⁇ 8 domains.
  • the intracellular domain comprises a CD47 inhibition domain.
  • the composition of embodiment 81, wherein the extracellular portion of the PSR is derived from the same PSR as the PSR intracellular signaling domain. 83.
  • composition of any one of the embodiments 79-82, wherein the extracellular domain comprises an extracellular domain of a scavenger receptor or an immunoglobulin domain.
  • the composition of embodiment 83, wherein the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region.
  • the composition of any one of the embodiments 79-84, wherein the extracellular domain comprises a phagocytic engulfment marker.
  • the composition of any one of the embodiments 79-85, wherein the extracellular domain comprises a structure capable of multimeric assembly.
  • the composition of any one of the embodiments 79-86, wherein the extracellular domain comprises a scaffold for multimerization. 88.
  • composition of any one of the preceding embodiments, wherein the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. 89.
  • the composition of any one of the preceding embodiments, wherein the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length.
  • the composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain specifically binds to the antigen of a target cell.
  • the composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises an antibody domain. 92.
  • the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single- domain antibody (sdAb), a nanobody, a V H domain, a V L domain, a VNAR domain, a V HH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof.
  • the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor.
  • composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen.
  • the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen.
  • the antigen is a cancer antigen or a pathogenic antigen or an autoimmune antigen.
  • the antigen comprises a viral antigen.
  • composition of any one of the preceding embodiments, wherein the antigen is a T- lymphocyte antigen. 99. The composition of any one of the preceding embodiments, wherein the antigen is an extracellular antigen. 100. The composition of any one of the preceding embodiments, wherein the antigen is an intracellular antigen. 101.
  • composition of any one of the preceding embodiments, wherein the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56.
  • the antigen is an ovarian cancer antigen or a T lymphoma antigen.
  • the antigen is an integrin receptor. 105.
  • the antigen comprises 2 or more antigens. 107.
  • composition of any one of the preceding embodiments, wherein the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker.
  • the composition of any one of the preceding embodiments, wherein the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker such as the hinge region of CD8 ⁇ , IgG1 or IgG4.
  • the composition of any one of the preceding embodiments, wherein the extracellular domain comprises a multimerization scaffold.
  • the composition of any one of the preceding embodiments, wherein the transmembrane domain comprises an FcR transmembrane domain.
  • composition of any one of the preceding embodiments, wherein the transmembrane domain comprises an FcR- ⁇ with no more than 20, 10 or 5 modifications transmembrane domain. 112.
  • the composition of any one of the preceding embodiments, wherein the transmembrane domain comprises a transmembrane domain from a syntaxin such as syntaxin 3 or syntaxin 4 or syntaxin 5.
  • a syntaxin such as syntaxin 3 or syntaxin 4 or syntaxin 5.
  • the composition of any one of the preceding embodiments, wherein the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the PFP is expressed in a cell. 114.
  • composition of any one of the preceding embodiments, wherein the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the PFP is expressed in a cell.
  • the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the PFP is expressed in a cell.
  • composition of any one of the preceding embodiments, wherein the transmembrane domain is derived from the same protein as the intracellular signaling domain. 122. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes a DAP12 recruitment domain. 123. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12. 124. The composition of any one of the preceding embodiments, wherein the transmembrane domain is at least 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. 125.
  • composition of any one of the preceding embodiments, wherein the transmembrane domain is at most 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length.
  • the intracellular domain comprises a phosphatase inhibition domain.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain comprises an ARP2/3 inhibition domain. 128.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain comprises at least one ITAM domain.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 ITAM domains. 130.
  • composition of any one of the preceding embodiments, wherein the intracellular domain further comprises at least one ITAM domain.
  • the intracellular domain further comprises at least one ITAM domain select from a group CD3 zeta TCR subunit, CD3 epsilon TCR subunit, CD3 gamma TCR subunit, CD3 delta TCR subunit, TCR zeta chain, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278,
  • composition of embodiment 129, wherein the at least one ITAM domain comprises a Src- family kinase phosphorylation site.
  • the at least one ITAM domain comprises a Syk recruitment domain.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain comprises a F-actin depolymerization activation domain. 135.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain lacks enzymatic activity.
  • the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. 137.
  • composition of any one of the preceding embodiments, wherein the intracellular domain comprises a CD47 inhibition domain. 138. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain comprises a domain that activate integrin such as the intracellular region of PSGL-1. 139. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain comprises a domain that activate Rap1 GTPase, such as that from EPAC and C3G. 140. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain are from paxillin. 141. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain activates focal adhesion kinase. 142.
  • composition of any one of the preceding embodiments, wherein the intracellular signaling domain is derived from a single phagocytic receptor. 143. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain is derived from a single scavenger receptor. 144. The composition of any one of the preceding embodiments, wherein the intracellular domain further comprises a phagocytosis enhancing domain. 145. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a pro-inflammatory signaling domain. 146. The composition of embodiment 145, wherein the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain. 147.
  • composition of embodiment 145 or 146, wherein the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain.
  • the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, an NLRP family member, NLRP1-14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40, a caspase domain or a procaspase binding domain or any combination thereof.
  • TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5 an IFN-receptor
  • an NLRP family member NLRP1-14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40, a caspase domain or a
  • composition of any one of the preceding embodiments, wherein the PFP does not comprise a full length intracellular signaling domain.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length.
  • the composition of any one of the preceding embodiments, wherein the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. 152.
  • composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • the composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes an FcR ⁇ chain extracellular domain, an FcR ⁇ chain transmembrane domain and/or an FcR ⁇ chain intracellular domain.
  • 154. The composition of embodiment 152 or 153, wherein the FcR ⁇ chain or the FcR ⁇ chain forms a complex with FcR ⁇ when expressed in a cell. 155.
  • composition of embodiment 154 wherein the FcR ⁇ chain or FcR ⁇ chain forms a complex with endogenous FcR ⁇ when expressed in a cell.
  • composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain.
  • TREM is TREM1, TREM 2 or TREM 3. 161.
  • the composition of any one of the preceding embodiments, wherein the recombinant nucleic acid comprises a sequence encoding a pro-inflammatory polypeptide. 162.
  • the composition of any one of the preceding embodiments, wherein the composition further comprises a pro-inflammatory polypeptide. 163.
  • the pro-inflammatory polypeptide is a chemokine, cytokine and nucleotides. 164.
  • composition of embodiment 163, wherein the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon.
  • the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon.
  • the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL-12
  • composition of embodiment 163, wherein the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon.
  • the nucleotide is selected from ATP, ADP, UTP, UDP, and/or UDP-glucose. 167.
  • composition of any one of the preceding embodiments, wherein the recombinant nucleic acid comprises a sequence encoding a homeostatic regulator of inflammation.
  • the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA.
  • UTR untranslated region
  • the composition of embodiment 168, wherein the sequence in the UTR is a sequence that binds to an RNA binding protein.
  • composition of embodiment 169 or 170, wherein the sequence in the UTR comprises a consensus sequence of WWWU(AUUUA)UUUW, wherein W is A or U. 172.
  • the composition of any one of the preceding embodiments, wherein the target cell is a mammalian cell.
  • the target cell is a human cell.
  • the composition of any one of the preceding embodiments, wherein the target cell comprises a cell infected with a pathogen. 176.
  • composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is RNA. 183. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is mRNA. 184. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a circRNA. 185. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a tRNA. 186. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a microRNA. 187. A vector comprising the composition of any one of embodiments 1-186. 188.
  • the vector of embodiment 187, wherein the vector is viral vector. 189.
  • the vector of embodiment 188, wherein the viral vector is retroviral vector or a lentiviral vector.
  • the vector of any one of embodiments 187-189, wherein the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides. 191.
  • the vector of embodiment 190 or 191, wherein each of the at least one nucleic acid sequence is operably linked to a separate promoter. 193.
  • the vector of any one of embodiments 187-192, wherein the vector further comprises one or more internal ribosome entry sites (IRESs).
  • IRSs internal ribosome entry sites
  • 196. A polypeptide encoded by the recombinant nucleic acid of the composition of any one of embodiments 1-186.
  • 197. A cell comprising the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195 or the polypeptide of embodiment 196.
  • the cell of any one of embodiments 197-201, wherein the cell is an M1 cell.
  • a pharmaceutical composition comprising (a) the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195, the polypeptide of embodiment 196 or the cell of any one of embodiments 197-203; and (b) a pharmaceutically acceptable excipient.
  • composition of embodiment 204 or 205 wherein the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3
  • a method of treating a disease in a subject in need thereof comprising administering to the subject the pharmaceutical composition of any one of embodiments 204-207. 209.
  • the method of embodiment 208, wherein the disease is cancer.
  • the method of embodiment 209, wherein the cancer is a solid cancer.
  • the method of embodiment 210, wherein the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer.
  • the method of embodiment 209, wherein the cancer is a liquid cancer. 213.
  • the method of embodiment 212, wherein the liquid cancer is leukemia or a lymphoma.
  • the method of embodiment 212, wherein the liquid cancer is a T cell lymphoma. 215.
  • the method of embodiment 208, wherein the disease is a T cell malignancy. 216.
  • the method of any one of embodiments 208-215, wherein the method further comprises administering an additional therapeutic agent to the subject. 217.
  • the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof.
  • administering comprises infusing or injecting.
  • administering comprises administering directly to the solid cancer.
  • the method of any one of embodiments 208-219, wherein administering comprises a circRNA, mRNA, viral-, particle-, liposome-, or exosome-based delivery procedure.
  • the method of any one of embodiments 208-220, wherein a CD4+ T cell response or a CD8+ T cell response is elicited in the subject.
  • a method of preparing a cell the method comprising contacting a cell with the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195 or the polypeptide of embodiment 196. 223.
  • contacting comprises transducing.
  • 224 The method of embodiment 223, where transducing comprises chemical transfection, electroporation, nucleofection, or viral infection.
  • 225 A method of preparing a pharmaceutical composition comprising contacting a lipid to the composition of any one of embodiments 1-186 or the vector of any one of embodiments 187-195. 226. The method of embodiment 225, where contacting comprises forming a lipid nanoparticle. 227.
  • a method of preparing a pharmaceutical composition comprising contacting an antibody to the composition of any one of embodiments 1-186 or the vector of any one of embodiments 187-195. 228. The method of embodiment 225, where contacting comprises forming a lipid nanoparticle.
  • CFP chimeric receptors fusion proteins
  • J774 macrophage cells are transiently transfected with the library of 5800 plasma proteins.
  • High- throughput multiplex assays (ranging from 6-well plate assay set up to up to 384-well plate assay with robotic handling) are set up to evaluate various potential functions of the plasma membranes.
  • Exemplary assays include, but are not limited to phagocytosis assay, cytokine production assay, inflammasome activation assay, and iNOS activation assay. Exemplary simplified methods are described in the following paragraphs. Variations of each method are also used and are understood by a skilled artisan. Variations of each method are also used and are understood by a skilled artisan.
  • Exemplary intracellular signaling domains tested for include but are not limited to CD40-FcR ⁇ ; FcR ⁇ -CD40; NLRP3; FcR ⁇ -SH2-Procaspase; FcR ⁇ -Myd88; FcR ⁇ -IFN receptor; FcR-TNFR1; FcR ⁇ - TNFR2; FcR-AIM2; FcR ⁇ -TRIFN; FcR ⁇ -Procaspase; TRIFC; RIG1; MDA5; TBK; CD64; CD16A; CD89; FcR ⁇ ; SIRP ⁇ ; (two consecutive intracellular domains are represented as hyphenated terms, for example, FcR ⁇ -Myd88 refers to an intracellular domain comprising an FcR ⁇ intracellular signaling domain as signaling domain 1; and an Myd88 intracellular signaling domain as signaling domain 2).
  • the extracellular linker domains screened include but are not limited to CD64, CD16A, CD89, SIRP ⁇ , FcR ⁇ , CD8 hinge.
  • the transmembrane domains tested include but are not limited to CD8, CD64, CD16A, CD89, FcR ⁇ , SIRP ⁇ , TNFR1 and CD40. MDA5 domains were also screened.
  • Phagocytosis assay [0813] Antigen-linked silica or polysterene beads ranging in diameters 1 nm, 5 nm or 10 nm were used for a screen of macrophages. Inert beads are coated in a supported lipid bilayer and the antigens are ligated to the lipid bilayer.
  • J774 macrophage cell lines are prepared, each cell line expressing a cloned recombinant plasma membrane protein.
  • the recombinant plasma membrane protein may also express a fluorescent tag.
  • the cell lines are maintained and propagated in complete RPMI media with heat inactivated serum and antibiotics (Penicillin/Streptomycin).
  • antibiotics Penicillin/Streptomycin.
  • cells are plated at a density of 1x10 ⁇ 6 cells/ml per well in 6 well plates or in a relative proportion in 12 or 24 well plates, and incubated for 2-6 hours. The cells are then washed once in Phosphate Buffer Saline, and the beads are added in serum depleted or complement depleted nutrient media.
  • Cells are visualized by light microscopy at 30 minutes and 2 hours after addition of the beads. Immunofluorescence reaction may be performed using tagged antibody, and fluorescent confocal microscopy is used to detect the interaction and co-localization of cellular proteins at engulfment. Confidence levels are determined by Kruskal-Wallis test with Dunn’s multiple comparison correction. [0814] In some examples, dye loaded tumor cells are fed to macrophage cell lines and phagocytosis is assessed by microscopy. Cytokine production: [0815] Macrophage cell lines are cultured as above.
  • each J774 cell line expressing a plasma membrane protein is plated in multi-wells and challenged with antigen-linked beads and cytokine production was assayed by collecting the supernatants at 4 hours and 24 hours. Cytokines are assayed from the supernatant by ELISA. In another fraction, cells are collected at 4 and 24 hours after incubation with the beads and flow cytometry is performed for detection of cytokines.
  • cytokines are assayed in a multiplex format, which can be selected from: IL-1 ⁇ , IL-1 ⁇ , IL-6, IL-12, IL-23, TNF- ⁇ , GMCSF, CXCL1, CXCL3, CXCL9, CXCL-10, MIP1- ⁇ and MIP-2.
  • Macrophage inflammatory cytokine array kit (R&D Systems) is used.
  • Intracellular signaling pathway for inflammatory gene and cytokine activation can be identified by western blot analysis for phosphorylation of MAP kinases, JNK, Akt signaling pathway, Interferon activation pathway including phosphorylation and activation of STAT-1.
  • Inflammasome activation assay [0817] Activation of NLRP3 inflammasome is assayed by ELISA detection of increased IL-1 production and detection caspase-1 activation by western blot, detecting cleavage of procaspase to generate the shorter caspase. In a microwell plate multiplex setting, Caspase-Glo (Promega Corporation) is used for faster readout of Caspase 1 activation.
  • iNOS activation assay [0818] Activation of the oxidative burst potential is measured by iNOS activation and NO production using a fluorimetric assay NOS activity assay kit (AbCAM).
  • Cancer cell killing assay [0819] Raji B cells are used as cancer antigen presenting cells.
  • Raji cells are incubated with whole cell crude extract of cancer cells, and co-incubated with J774 macrophage cell lines.
  • the macrophages can destroy the cells after 1 hour of infection, which can be detected by microscopy or detected by cell death assay.
  • Screening for high affinity antigen binding domains [0820] Cancer ligands are subjected to screening for antibody light chain and heavy chain variable domains to generate extracellular binding domains for the CFPs. Human full length antibodies or scFv libraries are screened. Also potential ligands are used for immunizing llama for development of novel immunoglobulin binding domains in llama, and preparation of single domain antibodies.
  • Specific useful domains identified from the screens are then reverse transcribed, and cloned into lentiviral expression vectors to generate the CFP constructs.
  • a recombinant nucleic acid encoding a CFP can generated using one or more domains from the extracellular, TM and cytoplasmic regions of the highly phagocytic receptors generated from the screen. Briefly plasma membrane receptors showing high activators of pro-inflammatory cytokine production and inflammasome activation are identified.
  • Bioinformatics studies are performed to identify functional domains including extracellular activation domains, transmembrane domains and intracellular signaling domains, for example, specific kinase activation sites, SH2 recruitment sites.
  • screened functional domains are then cloned in modular constructions for generating novel CFPs.
  • candidate CFPs are candidate CFPs, and each of these chimeric construct is tested for phagocytic enhancement, production of cytokines and chemokines, and/or tumor cell killing in vitro and/or in vivo.
  • a microparticle based phagocytosis assay was used to examine changes in phagocytosis. Briefly, streptavidin coupled fluorescent polystyrene microparticles (6 ⁇ m diameter) are conjugated with biotinylated recombinantly expressed and purified cancer ligand.
  • Myeloid cells expressing the novel CFP were incubated with the ligand coated microparticles for 1-4 h and the amount of phagocytosis was analyzed and quantified using flow cytometry. Plasmid or lentiviral constructions of the designer CFPs are then prepared and tested in macrophage cells for cancer cell lysis. [0822] Exemplary functional domain containing CFPs are described in the following sections.
  • CD14+ monocytes are isolated from the mononuclear cell fraction using CliniMACS GMP grade CD14 microbeads and LS separation magnetic columns (Miltenyi Biotec). Briefly, cells are resuspended to appropriate concentration in PEA buffer (phosphate-buffered saline [PBS] plus 2.5 mmol/L ethylenediaminetetraacetic acid [EDTA] and human serum albumin [0.5% final volume of Alburex 20%, Octopharma]), incubated with CliniMACS CD14 beads per manufacturer’s instructions, then washed and passed through a magnetized LS column.
  • PEA buffer phosphate-buffered saline [PBS] plus 2.5 mmol/L ethylenediaminetetraacetic acid [EDTA] and human serum albumin [0.5% final volume of Alburex 20%, Octopharma]
  • PBMCs are collected by leukapheresis from cirrhotic donors who gave informed consent to participate in the study.
  • Leukapheresis of peripheral blood for mononuclear cells (MNCs) is carried out using an Optia apheresis system by sterile collection. A standard collection program for MNC is used, processing 2.5 blood volumes.
  • Isolation of CD14 cells is carried out using a GMP-compliant functionally closed system (CliniMACS Prodigy system, Miltenyi Biotec).
  • the leukapheresis product is sampled for cell count and an aliquot taken for pre-separation flow cytometry.
  • the percentage of monocytes (CD14+) and absolute cell number are determined, and, if required, the volume is adjusted to meet the required criteria for selection ( ⁇ 20 x 10 9 total white blood cells; ⁇ 400 x 10 6 white blood cells/mL; ⁇ 3.5 x 10 9 CD14 cells, volume 50–300 mL).
  • CD14 cell isolation and separation is carried out using the CliniMACS Prodigy with CliniMACS CD14 microbeads (medical device class III), TS510 tubing set and LP-14 program.
  • the selected CD14+ positive monocytes are washed in PBS/EDTA buffer (CliniMACS buffer, Miltenyi) containing pharmaceutical grade 0.5% human albumin (Alburex), then re-suspended in TexMACS (or comparator) medium for culture.
  • Cell count and purity [0824] Cell counts of total MNCs and isolated monocyte fractions are performed using a Sysmex XP-300 automated analyzer (Sysmex). Assessment of macrophage numbers is carried out by flow cytometry with TruCount tubes (Becton Dickinson) to determine absolute cell number, as the Sysmex consistently underestimated the number of monocytes.
  • the purity of the separation is assessed using flow cytometry (FACSCanto II, BD Biosciences) with a panel of antibodies against human leukocytes (CD45-VioBlue, CD15-FITC, CD14-PE, CD16-APC), and product quality is assessed by determining the amount of neutrophil contamination (CD45int, CD15pos).
  • FACSCanto II flow cytometry
  • CD45-VioBlue human leukocytes
  • CD14-PE CD16-APC
  • product quality is assessed by determining the amount of neutrophil contamination (CD45int, CD15pos).
  • Cell culture development of cultures with healthy donor samples [0825]
  • Optimal culture medium for macrophage differentiation is investigated, and three candidates are tested using for the cell product.
  • the effect of monocyte cryopreservation on deriving myeloid cells and macrophages for therapeutic use is examined.
  • Functional assays are conducted to quantify the phagocytic capacity of myeloid cells and macrophages and their capacity for further polarization, and phagocytic potential as described elsewhere in the disclosure.
  • Full-scale process validation with subject samples [0826] Monocytes cultured from leukapheresis from Prodigy isolation are cultured at 2 x 10 6 monocytes per cm 2 and per mL in culture bags (MACS GMP differentiation bags, Miltenyi) with GMP-grade TexMACS (Miltenyi) and 100 ng/mL M-CSF. Monocytes are cultured with 100 ng/mL GMP-compliant recombinant human M-CSF (R&D Systems).
  • Cells are cultured in a humidified atmosphere at 37°C, with 5% CO2 for 7 days. A 50% volume media replenishment is carried out twice during culture (days 2 and 4) with 50% of the culture medium removed, then fed with fresh medium supplemented with 200 ng/mL M-CSF (to restore a final concentration of 100 ng/mL).
  • Cell Dissociation Buffer Gibco, Thermo Fisher
  • Leukapheresis- derived macrophages are removed from the culture bags at day 7 using PBS/EDTA buffer (CliniMACS buffer, Miltenyi) containing pharmaceutical grade 0.5% human albumin from serum (HAS; Alburex).
  • Harvested cells are resuspended in excipient composed of two licensed products: 0.9% saline for infusion (Baxter) with 0.5% human albumin (Alburex).
  • Flow cytometry characterization [0828] Monocyte and macrophage cell surface marker expression is analyzed using either a FACSCanto II (BD Biosciences) or MACSQuant 10 (Miltenyi) flow cytometer. Approximately 20,000 events are acquired for each sample.
  • Cell surface expression of leukocyte markers in freshly isolated and day 7 matured cells is carried out by incubating cells with specific antibodies (final dilution 1:100). Cells are incubated for 5 min with FcR block (Miltenyi) then incubated at 4°C for 20 min with antibody cocktails. Cells are washed in PEA, and dead cell exclusion dye DRAQ7 (BioLegend) is added at 1:100.
  • Cells are stained for a range of surface markers as follows: CD45- VioBlue, CD14-PE or CD14-PerCP-Vio700, CD163-FITC, CD169-PE and CD16-APC (all Miltenyi), CCR2-BV421, CD206-FITC, CXCR4-PE and CD115-APC (all BioLegend), and 25F9- APC and CD115-APC (eBioscience). Both monocytes and macrophages are gated to exclude debris, doublets and dead cells using forward and side scatter and DRAQ7 dead cell discriminator (BioLegend) and analyzed using FlowJo software (Tree Star).
  • CD45- VioBlue CD14-PE or CD14-PerCP-Vio700
  • CD163-FITC CD169-PE and CD16-APC (all Miltenyi)
  • CCR2-BV421, CD206-FITC CXCR4-PE and CD115-APC
  • 25F9- APC and CD115-APC
  • a panel is developed as Release Criteria (CD45-VB/CD206-FITC/CD14-PE/25F9 APC/DRAQ7) that defined the development of a functional macrophage from monocytes. Macrophages are determined as having mean fluorescence intensity (MFI) five times higher than the level on day 0 monocytes for both 25F9 and CD206.
  • MFI mean fluorescence intensity
  • a second panel is developed which assessed other markers as part of an Extended Panel, composed of CCR2-BV421/CD163-FITC/CD169-PE/CD14-PerCP- Vio700/CD16-APC/DRAQ7), but is not used as part of the Release Criteria for the cell product.
  • monocytes and macrophages from buffy coat CD14 cells are tested for phagocytic uptake using pHRodo beads, which fluoresce only when taken into acidic endosomes. Briefly, monocytes or macrophages are cultured with 1–2 uL of pHRodo Escherichia coli bioparticles (LifeTechnologies, Thermo Fisher) for 1 h, then the medium is taken off and cells washed to remove non-phagocytosed particles. Phagocytosis is assessed using an EVOS microscope (Thermo Fisher), images captured and cellular uptake of beads quantified using ImageJ software (NIH freeware).
  • pHRodo beads which fluoresce only when taken into acidic endosomes. Briefly, monocytes or macrophages are cultured with 1–2 uL of pHRodo Escherichia coli bioparticles (LifeTechnologies, Thermo Fisher) for 1 h, then the medium is taken off and cells washed
  • the capacity to polarize toward defined differentiated macrophages is examined by treating day 7 macrophages with IFN ⁇ (50 ng/mL) or IL-4 (20 ng/mL) for 48 h to induce polarization to M1 or M2 phenotype (or M[IFN ⁇ ] versus M[IL-4], respectively). After 48 h, the cells are visualized by EVOS bright-field microscopy, then harvested and phenotyped as before. Further analysis is performed on the cytokine and growth factor secretion profile of macrophages after generation and in response to inflammatory stimuli.
  • Macrophages are generated from healthy donor buffy coats as before, and either left untreated or stimulated with TNF ⁇ (50 ng/mL, Peprotech) and polyinosinic:polycytidylic acid (poly I:C, a viral homolog which binds TLR3, 1 g/mL, Sigma) to mimic the conditions present in the inflamed liver, or lipopolysaccharide (LPS, 100 ng/mL, Sigma) plus IFN ⁇ (50 IU/mL, Peprotech) to produce a maximal macrophage activation. Day 7 macrophages are incubated overnight and supernatants collected and spun down to remove debris, then stored at ⁇ 80°C until testing.
  • TNF ⁇ 50 ng/mL, Peprotech
  • poly I:C polyinosinic:polycytidylic acid
  • LPS lipopolysaccharide
  • IFN ⁇ 50 IU/mL, Peprotech
  • Secretome analysis is performed using a 27-plex human cytokine kit and a 9-plex matrix metalloprotease kit run on a Magpix multiplex enzyme linked immunoassay plate reader (BioRad).
  • Product stability [0830] Various excipients are tested during process development including PBS/EDTA buffer; PBS/EDTA buffer with 0.5% HAS (Alburex), 0.9% saline alone or saline with 0.5% HAS. The 0.9% saline (Baxter) with 0.5% HAS excipient is found to maintain optimal cell viability and phenotype (data not shown).
  • CD5-FcR-PI3K CFP construct [0832]
  • a CD5-targeted CFP was constructed using known molecular biology techniques.
  • the CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8 ⁇ chain hinge and CD8 ⁇ chain TM domain via a short linker.
  • the TM domain is fused at the cytosolic end with an FcR ⁇ cytosolic portion, and a PI3K recruitment domain.
  • the construct was prepared in a vector having a fluorescent marker and a drug (ampicillin) resistance and amplified by transfecting a bacterial host.
  • the sequence is provided below: CD5-FcR-PI3K MWLQSLLLLGTVACSISEIQLVQSGGGLVKPGGSVRISCAASGYTFTNYGMNWVRQAPGK GLEWMGWINTHTGEPTYADSFKGRFTFSLDDSKNTAYLQINSLRAEDTAVYFCTRRGYDW YFDVWGQGTTVTVSSGGGGSGGGGSGGSDIQMTQSPSSLSASVGDRVTITCRASQDINSY LSWFQQKPGKAPKTLIYRANRLESGVPSRFSGSGSGTDYTLTISSLQYEDFGIYYCQQYDESP WTFGGGTKLEIKSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRP EACRPAAGGAVHTRGLDIYIWAPLAGTCGVLLLSLVITLYCRRLKIQ
  • mRNA was generated by in vitro reverse transcription of the purified plasmids using suitable primers. The purified mRNA was transduced into a cell line for expression analysis.
  • Example 4. HER2-FcR-PI3K CFP construct
  • a HER2-targeted CFP was constructed using known molecular biology techniques. The CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to HER2 on a target cell, attached to a CD8 ⁇ chain hinge and CD8 ⁇ chain TM domain via a short linker.
  • the TM domain is fused at the cytosolic end with an FcR ⁇ cytosolic portion, and a PI3K recruitment domain as in the previous example.
  • the sequence is provided below: HER2-FcR-PI3K MWLQSLLLLGTVACSISDIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAP KLLIYSASFLYSGVPSRFSGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKVEIKR TGSTSGSGKPGSGEGSEVQLVESGGGLVQPGGSLRLSCAASGFNIKDTYIHWVRQAPGKGL EWVARIYPTNGYTRYADSVKGRFTISADTSKNTAYLQMNSLRAEDTAVYYCSRWGGDGFY AMDVWGQGTLVTVSSSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPL SLRPEACRPAAGGAVHTRGLDIYI
  • CD5-FcR-CD40 CFP construct [0835]
  • a CD5-targeted CFP was constructed using known molecular biology techniques having an intracellular domain comprising CD40 sequence.
  • the CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8 ⁇ chain hinge and CD8 ⁇ chain TM domain via a short linker.
  • the TM domain is fused at the cytosolic end with an FcR ⁇ cytosolic portion, followed by a CD40 cytosolic portion.
  • CD5-FcR-CD40 MWLQSLLLLGTVACSISEIQLVQSGGGLVKPGGSVRISCAASGYTFTNYGMNWVRQAPGK GLEWMGWINTHTGEPTYADSFKGRFTFSLDDSKNTAYLQINSLRAEDTAVYFCTRRGYDW YFDVWGQGTTVTVSSGGGGSGGGGSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQDINSY LSWFQQKPGKAPKTLIYRANRLESGVPSRFSGSGSGTDYTLTISSLQYEDFGIYYCQQYDESP WTFGGGTKLEIKSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRP EACRPAAGGAVHTRGLDIYIWAPLAGTCGVLLLSLVITLYCRLKIQVRKAAITSYEKSDGVY TGLSTRNQETYETLKHEKPPQKKVAKKPTNKAPHPKQEPQEINFPDDLPG
  • CD5-FcR-MDA5 CFP construct [0837]
  • a CD5-targeted CFP was constructed using known molecular biology techniques having an intracellular domain comprising two caspase activation (CARD) domains of MDA-5 intracellular domain sequence (Tandem CARD ICD sequence as in SEQ ID NO: 23).
  • the CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8 ⁇ chain hinge and CD8 ⁇ chain TM domain via a short linker.
  • CD5-FcR-TNFR1 or TNFR2 CFP construct [0839] In this example, a CD5-targeted CFP was constructed using known molecular biology techniques having a TNFR1 or TNFR2 intracellular domain. As shown graphically in FIG.
  • the CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8 ⁇ chain hinge and CD8 ⁇ chain TM domain via a short linker.
  • the TM domain is fused at the cytosolic end with an FcR ⁇ cytosolic portion, followed by an intracellular signaling domain (ICD) sequence TNFR1 (SEQ ID NO: 21) or an ICD of TNFR2 (SEQ ID NO: 22).
  • ICD intracellular signaling domain
  • the full length sequence of the CFP having a TNFR1 intracellular signaling domain is depicted below: (SEQ ID NO: 25) .
  • the full length sequence of the CFP having a TNFR2 intracellular signaling domain is depicted below:
  • Example 8 Bicistronic constructs for in vivo delivery
  • design and expression of bicistronic construct is demonstrated in which a single mRNA comprises sequence encoding a CAR and a CFP for expression in a T cell and a myeloid cell respectively, each of which can comprise an extracellular binding domain that binds to the same (or two different) cancer cell surface antigen.
  • the design is directed towards in vivo delivery and cell specific expression of either the CAR or the CFP, depending on whether the cell is a T cell lineage cell or a myeloid lineage cell.
  • the single mRNA can be delivered via an LNP.
  • the molecular design of a bicistronic CFP-CAR dual construct comprises (a) a sequence encoding a CAR-T and a sequence encoding a CFP, separated by an autocleavage sequence, such as a T2A, or a P2A sequence.
  • a CAR-T sequence comprises a CD3 epsilon transmembrane region and cytoplasmic region that facilitate TCR complex assembly and is expressed in a T cell.
  • This design promotes a T cell-specific expression of a CAR with a binder (the extracellular antigen binding domain) that specifically binds to a desired antigen, e.g., CD19 cell surface antigen on a cancer cell.
  • a binder the extracellular antigen binding domain
  • a desired antigen e.g., CD19 cell surface antigen on a cancer cell.
  • CD19 binder CAR multimerizes with members of the TCR endogenously expressed by T cells.
  • the design not only facilitates T cell specific expression, but also the CAR activates a T cell upon binding of the extracellular antigen binding domain to the antigen that it specifically binds to. Therefore a CAR, that is functional specifically in a T cell is designed.
  • a CFP sequence, as designed here for specific expression on a myeloid cell comprises a transmembrane and intracellular domain of Fc-alpha R, which assembles in a myeloid cell with endogenously expressed Fc-gamma receptors and expresses on a myeloid cell surface.
  • the CFP comprises an extracellular binding domain specific for a cancer cell antigen, and can be a binder that binds the antigen that the CAR binds.
  • Each of the binders can be an scFv or can be any other form of binder as described elsewhere in the specification.
  • the design can include a CAR with a binder that binds to a different antigen in a CFP but is directed to the same tumor tissue.
  • a CFP as described here will exhibit myeloid cell specific expression and function as the Fc-alpha R domain promotes expression and function of the CFP in a cell that naturally expresses Fcgamma receptor, and myeloid cells specifically express Fcgamma receptor.
  • Sequences Sequence for ⁇ MYL222 QQ Q Sequence for MYL223
  • Example 9.5’-Cap structures for stability and efficient translation.
  • several Cap structures are tested for efficiency in mRNA translation.
  • mRNA constructs comprising a sequence encoding a CFP as described herein is transcribed in vitro and 5’capping is performed using a different Cap analog for each test set.
  • the IVT mRNAs are purified by HPLC and electroporated into a myeloid or suitable cell line.
  • the Cap analogs are listed in Table 7. [0846] Table 7.
  • Cap analogues G, nucleotide base Guanine; C, nucleotide base Cytosine; A, nucleotide base Adenine; U, nucleotide base Uracil; m, methylation; p-phosphate.
  • Gppp m6 AmpG analog shows the highest protein expression in the test cell line. This test is repeated for different constructs of varying mRNA length and starting nucleotide – thereby generating a better understanding and confidence in the use of the Cap analog.
  • the Cap analogs are designed for suitable expression of the construct comprise an anti-CD5 scFv, a CD8 hinge, a CD8 transmembrane domain, an FcR intracellular domain and an intracellular PI3kinase recruitment domain (anti-CD5-CD8hinge- CD8TM-FcR-PI3K) having a translated amino acid sequence (SEQ ID NO: 50): [0849]
  • the Cap analogs are designed for suitable expression of the construct comprise an anti-HER2 scFv, a CD8 hinge, a CD8 transmembrane domain, a CD40 intracellular domain, an FcR intracellular domain and an intracellular PI3kinase recruitment domain (anti-HER2-CD8hinge-CD8TM-CD40-FcR-PI3K) having a translated amino acid sequence (SEQ ID NO: 51): [0850] Q Q Q Q Q QQ [0851]
  • Example 10 Example 10
  • FIG. 4 illustrates graphically a simplified generalized work plan for evaluating the functional aspects.
  • RNA/1x10 ⁇ 6 cells 100ug/ml was used for electroporation of the cells.
  • the mRNA sets were: Uncapped AG-anti-HER2-CAR-polyA; CC1-Capped-anti-HER2-CAR- polyA; m6A-Capped-anti-HER2-CAR-polyA; EC1-Capped-anti-HER2-CAR-polyA RNA was prepared by in vitro transcription (IVT) in IVT buffer at pH 6.5, as described elsewhere in the examples. Transfection using lipofectamine was also used in one exemplary set.
  • FIGs. 6 and 7 demonstrate representative results of these experiments. As shown in FIG.
  • FIGs. 14A and its continuing figure FIG. 14B show flow cytometry sorting of electroporated cells from a single donor (Donor 23B), using 0.1 microM RNA.
  • FIG. 15 shows HER2 binder (CAR) expression of monocytes for the same subject for the experiment at 4h, 24h and 48h respectively.
  • FIG. 16 shows a summary graph plot, indicating the increase and the duration of expression of the anti-HER2 CAR in these cells over the indicated time. Similar results were observed in case of cells from donor PD-004-04, as shown in FIGs.
  • FIGs. 20-22 demonstrate data showing results from using cells from the donor PD-004-04 (same donor’s cells used in results shown in FIGs 17-19), where the cells were transfected with lipofectamine instead of electroporation. Additional data from cells from Donor MYE-101-0024A (Donor 24A) electroporated with 0.1 microM RNA – Mock RNA, uncapped AG-anti-HER2 CAR-polyA; CC1- cap-anti-HER2 CAR-polyA; EC1-cap-anti-HER2 CAR-polyA or ARCA-cap-anti-HER2 CAR- polyA are shown in FIGs. 23-25.
  • the anti-HER2 CAR comprises an antigen binding domain having an scFV, having a heavy chain variable domain sequence of and a light chain variable domain having a sequence of:
  • the sequence additionally has a CD8 hinge domain having an amino acid sequence of ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLD; a CD8 TM domain having a sequence of: IYIWAPLAGTCGVLLLSLVITLYC; an FcR intracellular domain having a sequence of: and intracellular PI3 K recruitment domain having a sequence of: YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM.
  • Table 8 A summary of data from a representative example from three monocyte donor’s cells is presented in the Table below.
  • Example 11 Functional evaluation of 5’-Cap structures: immune activation by capped RNA
  • the protocol workflow is outlined in FIG. 8. Following transfection (e.g., lipofection) of the respective mRNA constructs with the different Cap analogues the Type 1 interferons, Interferon alpha and beta (IFN-a and IFN-b) were measured at 4 and 20h.
  • mRNA constructs, with or without HPLC purification were used for the study. Monocytes from a single human donor were used per assay.
  • 0.6 x 10 ⁇ 6 cells were plated in 24 well plate in 0.5ml recovery media/0.5 ug RNA/ul transfection reagent (TransIT reagent) and 1 ul Boost reagent in 50 ul final Opti-Mem. Cells were also collected for assessing IFN recognition pathways via qPCR.
  • the mRNA encoded by the constructs was a TROP2 binder CAR, having anti-TROP2 antigen binding domain, a CD89 TM domain, with short CD89 extracellular and intracellular regions.
  • FIG. 9A shows that IFN-a and IFN-b were induced upon transfection with an in house ARCA construct and commercial ARCA construct, both at 4h and 20h. ARCA in house construct was HPLC purified.
  • in-house ARCA RNA was comparable to the commercial ARCA.
  • in-house generated and HPLC purified CC1 and EC1 caps did not result in IFN response.
  • interferon type I response As is well understood by one skilled in the art that the activation of interferons can lead to an inflammatory sequel which is harmful. Additionally, presence of double stranded RNA in the RNA preparation, even in small amounts, is likely to activate the antiviral innate immune response, marked by interferon type I response.
  • IFN alpha and beta are exemplary type I interferons. Release of these interferons in turn activate a number of inflammatory cytokines and chemokines.
  • FIG. 9B shows induction of CXCL10 and IFITM, which followed similar pattern as the interferons.
  • FIGs 26-28 demonstrate the flow cytometry data of an experiment with monocyte cells from donor PD-004-04, and comparing the different mRNA preparations with the caps with or without HPLC purification, as shown in the figures.
  • Example 12 Large scale ARCA- versus CC1-capped GP75-FcR-PI3K RNA production for comparison studies in vivo
  • ARCA and CC1 cap structures were evaluated for their efficacy in an in vivo tumor model. Both ARCA capped and CC1-capped RNA were successfully generated in large scale for the studies.
  • An exemplary GP75-FcR-PI3K mRNA sequence including 5’-UTR (underlined) and 3’- UTR (underlined) is shown below: [0859] Pre-capped in vitro transcribed mRNA and CC1-capped mRNA before and after HPLC purification were resolved in gel electrophoresis, a representative is shown in FIG.
  • Example 13 Comparison of effectiveness of anti-gp75 myeloid cell CAR mRNA with ARCA and CC1 Cap in in vivo tumor model [0861] In this example, a comparison of the activities of two mRNAs encoding gp75-targeting CAR for myeloid cells, one with ARCA 5’-Cap and one with CC1-5’Cap in a mouse murine model was evaluated.
  • tumor cells were inoculated in the mice and allowed to grow to a certain size, and the treatment was initiated with intravenous (i.v.) administration of anti-gp75-CAR monocytes (2 x 10 6 ), generated from the bone marrow of C57BL/6 mice from day 10 at intervals for 6 doses, spread across 8 days till day 18.
  • Anti-gp75 CAR expressing murine monocytes were prepared as follows: Fresh murine monocytes were isolated at different days corresponding to different doses and mRNA (gp75-FLAG-FcR-PI3K) capped with ARCA or CC1 cap was nucleofected.
  • FIG. 11 shows comparable cell viability after nucleofection of anti-gp75-CAR, which indicates that the same cell number take up the mRNA at different doses.
  • FIG. 11 lower panel shows consistent high CAR expression across all injection. In each case, the CC1-capped mRNA had a higher expression compared to ARCA-capped mRNA. However, measurement of tumor volume over time shows interesting differences.
  • mice injected with CC1-capped anti-gp75 CAR mRNA showed the least increase in tumor volume, and the tumor volume was about half the size of the tumor size of untreated mice.
  • ARCA-capped mRNA encoding the same anti-gp75 CAR led to a lesser suppression of tumor, compared to the CC1-Capped anti-gp75 CAR.
  • FIGs. 5-7 showing efficiency of the CCI-Capped mRNA in expression of the mRNA, and also FIGs. 9A and 9B showing reduced immune activation.
  • the results indicate that the beneficial effect of using CC1-Cap for the mRNA is also evident in vivo, and that CC1 is a superior design over the ARCA.
  • Example 14 Capping Efficiency
  • capping efficiency is determined and compared across CC1, EC1 and m6A Cap structures.
  • An improved IVT buffer was used, composed of 40mM Bis-Tris pH 6.5, 25 mM MgCl2, 10mM DTT and 2 mM spermidine.
  • FIG. 13 shows gel electrophoresis data showing post capping samples. The indicated ratios on the figure represents a ratio of Cap: ATP if an AG initiator; or the ratio of Cap: GTP if an GG initiator.
  • CC1 and m6A showed a higher capping efficiency compared to EC1 for Cap:ATP ratio: 1:1.25.
  • a summary of the capping efficiencies of ARCA and CC1 and EC1 capping is shown in the table below. From the data it is understood that 2.5-fold more EC1 cap is needed to obtain 100% capping efficiency compared to CC1.
  • Table 9 A comparison of benefits of CC1/EC1 vs ARCA. [0866] Thus, from the above examples, it is shown that compared to ARCA, CC1 and EC1 mRNA express 3-fold higher.
  • EC1 mRNA has the lowest dsRNA content.
  • ARCA-capped RNA produced in-house requires HPLC purification to remove double stranded DNA (dsRNA) to a level compared to the commercial ARCA-GMP lot. It was further observed that even after removal of dsRNA, ARCA mRNA triggers higher levels of IFN type I upregulation compared to CC1 and EC1 cap, suggesting remaining innate immune recognition by IFIT family of proteins, which recognize Cap 0.

Abstract

Compositions and methods for making and using engineered myeloid cells that express a chimeric fusion protein for immunotherapy and compositions and methods for making and using engineered T cells that express a chimeric antigen receptor for immunotherapy.

Description

ENGINEERED CHIMERIC FUSION PROTEIN COMPOSITIONS AND METHODS OF USE THEREOF CROSS REFERENCE [0001] This application claims the benefit of U.S. Provisional Application No.63/347,183 filed on May 31, 2022, which is incorporated herein by reference in its entirety. BACKGROUND [0002] Cellular immunotherapy is a promising new technology for fighting difficult to treat diseases, such as cancer, and persistent infections and also certain diseases that are refractory to other forms of treatment. A major breakthrough has come across with the discovery of CAR-T cell and their potential use in immunotherapy. However, newer avenues are sought for using other cell types towards development of improved therapeutics. Myeloid cells are a major cellular compartment of the immune system comprising monocytes, dendritic cells, tissue macrophages, and granulocytes. Their enormous plasticity and heterogeneity, during both homeostasis and disease can be harnessed to develop safe and efficacious therapeutics. Myeloid cells can be engineered to specifically target and destroy a range of pathogenic agents or pathogenic cell types, mount a strong immunological response that can further induce a T cell response in vivo. [0003] Engineered myeloid cells can also be short-lived in vivo, phenotypically diverse, sensitive, plastic, and are often found to be difficult to manipulate in vitro. For example, exogenous gene expression in monocytes has been difficult compared to exogenous gene expression in non- hematopoietic cells. There are significant technical difficulties associated with transfecting myeloid cells (e.g., monocytes/macrophages). As professional phagocytes, myeloid cells, such as monocytes/macrophages, comprise many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process. This is especially true of activated macrophages which undergo a dramatic change in their physiology following exposure to immune or inflammatory stimuli. Viral transduction of these cells has been hampered because macrophages are end-stage cells that generally do not divide; therefore, some of the vectors that depend on integration into a replicative genome have met with limited success. Furthermore, macrophages are quite responsive to “danger signals,” and therefore several of the original viral vectors that were used for gene transfer induced potent anti-viral responses in these cells making these vectors inappropriate for gene delivery. [0004] Thus, engineered myeloid cells have recently captured interest for development as a cell therapy tool. Major hindrances remain for successful generation of myeloid cells into therapeutic tools, which may include low exogenous protein expression in myeloid cells. Methods for increasing expression of recombinant constructs delivered to myeloid cells for engineering are sought. SUMMARY [0005] The diverse functionality of myeloid cells makes them an ideal cell therapy candidate that can be engineered to have numerous therapeutic effects. The present disclosure is related to immunotherapy using myeloid cells (e.g., CD14+ cells) of the immune system, particularly phagocytic cells. A number of therapeutic indications could be contemplated using myeloid cells. For example, myeloid cell immunotherapy could be exceedingly important in cancer, autoimmunity, fibrotic diseases and infections. The present disclosure is related to immunotherapy using myeloid cells, including phagocytic cells of the immune system, particularly macrophages. It is an object of the invention disclosed herein to harness one or more of these functions of myeloid cells for therapeutic uses. For example, it is an object of the invention disclosed herein to harness the phagocytic activity of myeloid cells, including engineered myeloid cells, for therapeutic uses. For example, it is an object of the invention disclosed herein to harness the ability of myeloid cells, including engineered myeloid cells, to promote T cell activation. For example, it is an object of the invention disclosed herein to harness the ability of myeloid cells, including engineered myeloid cells, to promote secretion of tumoricidal molecules. For example, it is an object of the invention disclosed herein to harness the ability of myeloid cells, including engineered myeloid cells, to promote recruitment and trafficking of immune cells and molecules. The present disclosure provides innovative methods and compositions that can successfully transfect or transduce a myeloid cell, or otherwise induce a genetic modification in a myeloid cell, with the purpose of augmenting a functional aspect of a myeloid cell, additionally, without compromising the cell’s differentiation capability, maturation potential, and/or its plasticity. [0006] The present disclosure involves making and using engineered myeloid cells (e.g., CD14+ cells, such as macrophages or other phagocytic cells, which can attack and kill (ATAK) diseased cells directly and/or indirectly, such as cancer cells and infected cells. Engineered myeloid cells, such as macrophages and other phagocytic cells, can be prepared by incorporating nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric fusion protein (CFP), that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology. It has been found that myeloid cells can be engineered to have a broad and diverse range of activities. For example, it has been found that myeloid cells can be engineered to express a chimeric fusion protein (CFP) containing an antigen binding domain to have a broad and diverse range of activities. For example, it has been found that myeloid cells can be engineered to have enhanced phagocytic activity such that upon binding of the CFP to an antigen on a target cell, the cell exhibits increased phagocytosis of the target cell. It has also been found that myeloid cells can be engineered to promote T cell activation such that upon binding of the CFP to an antigen on a target cell, the cell promotes activation of T cells, such as T cells in the tumor microenvironment. The engineered myeloid cells can be engineered to promote secretion of tumoricidal molecules such that upon binding of the CFP to an antigen on a target cell, the cell promotes secretion of tumoricidal molecules from nearby cells. The engineered myeloid cells can be engineered to promote recruitment and trafficking of immune cells and molecules such that upon binding of the CFP to an antigen on a target cell, the cell promotes recruitment and trafficking of immune cells and molecules to the target cell or a tumor microenvironment. [0007] In some embodiments, the one or more polynucleic acids is an mRNA. [0008] In some embodiments, the recombinant polynucleic acid comprises a sequence encoding the CFP. [0009] In some embodiments, the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements, e.g., a LINE1 element, e.g. sequences encoding L1 -ORF1 and L1-ORF2. In some embodiments, the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements and a sequence for a guide RNA. In some embodiments, the recombinant polynucleic acid comprises a sequence encoding the CFP and one or more retrotransposable elements in which the retrotransposable elements is fused with a heterologous protein or functional portion thereof. [0010] In some embodiments, the recombinant polynucleic acid comprises a sequence encodes for two CFPs (e.g., a CFP and a CAR) in tandem, separated by an autocleavable moiety (e.g., T2A, P2A etc). [0011] In some embodiments, the mRNA is 2kb - 20,000kb long. [0012] In some embodiments, the recombinant polynucleic acid comprises one or more elements for stabilizing structures. Particularly important for long mRNA is to have 5’ Cap protection. In some embodiments, the RNA products required for generating engineered myeloid cells of therapeutic interest are generated in vitro, 5’capped and 3’-polyA tail. [0013] In some embodiments, the 5’Cap is modified from a native mRNA 5’Cap. [0014] One aspect of this disclosure involves having an efficient Cap structure using suitable capping analogs. [0015] In some embodiments, the RNA products prepared by IVT mRNA are co-transcriptional capped with m7GpppN-derived dinucleotides. In some embodiments, the 5’-Cap is a trinucleotide cap analog. In some embodiments the first nucleotide base following a 5’-Cap plays a crucial role in mRNA stability. In some embodiments, the base is adenine (A). In some embodiments, the base is cytosine (C). In some embodiments, the base is guanine (G). In some embodiments, the base is uracil (U). [0016] In some embodiments, the Cap analog is m7GpppApG. [0017] In some embodiments, the Cap analog is m7GpppAmpG. [0018] In some embodiments, the Cap analog is m7Gpppm6ApG. [0019] In some embodiments, the Cap analog is m7Gpppm6AmpG. [0020] In some embodiments, the Cap analog is m7GpppCpG. [0021] In some embodiments, the Cap analog is m7GpppCmpG. [0022] In some embodiments, the Cap analog is m7GpppGpG. [0023] In some embodiments, the Cap analog is m7GpppGmpG. [0024] In some embodiments, the Cap analog is m7GpppUpG. [0025] In some embodiments, the Cap analog is m7GpppUmpG. [0026] In some embodiments, the 5’-Cap is anti-reverse cap analogs (ARCAs) having chemically modified 3 -O or 2 -O positions of m7G (m27,3 -OGpppG). [0027] In some embodiments, the 5’-Cap is N7-methylguanosine 5 -diphosphate (m7GDPIm). [0028] In some embodiments, the caping involves post-transcriptional enzymatic capping of 5 - triphosphates RNA using Vaccinia virus capping complex. In some embodiments, the 5’capping confers stability and translational efficiency to the mRNA. [0029] In some embodiments, cap-proximal sequence is crucial for mRNA stability and function. [0030] In some embodiments, the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer. [0031] Also provided herein is a method for treating a disease in a subject in need thereof comprising administering a therapeutically effective amount of a pharmaceutical composition described herein to the subject. In some embodiments, the disease is cancer. In some embodiments, the disease is an infection. In some embodiments, the subject is human. [0032] In one aspect, provided herein is a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a CD5 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least three intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (A) a first intracellular signaling domain from Fc epsilon receptor Ig (FCER1G), and (B) a second intracellular signaling domain comprising a PI3K recruitment domain. [0033] In some embodiments, the CFP has the sequence according to SEQ ID NO:50. In some embodiments, the Cap structure is a trinucleotide cap analog. [0034] In one aspect, provided herein is a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a HER2 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least three intracellular signaling domains comprise: (A) a first intracellular signaling domain from CD40, (B) a second intracellular signaling domain from Fc epsilon receptor Ig (FCER1G), (C) a third intracellular signaling domain comprising a PI3K recruitment domain. In some embodiments, the CFP has the sequence according to SEQ ID NO:51. [0035] In some embodiments, the Cap structure is a trinucleotide cap analog. In some embodiments, the Cap comprises a 2’-O-methyl modification. In some embodiments, the Cap comprises an N6 methyl modification. In some embodiments, the Cap comprises both a 2’-O-methyl modification and an N6 methyl modification. [0036] In some embodiments, the mRNA is in vitro transcribed. In some embodiments, the mRNA is HPLC purified. [0037] In one aspect, provided herein is a composition comprising an mRNA comprising a sequence encoding a CFP; a 5’UTR, a 3’UTR, the 5’UTR terminating in a 5’cap analog represented by the formula 5’-m7GpppAmpG, wherein, G represents Guanine, A represents Adenine, p represents phosphate group, and m represents methylation; and wherein, the CFP comprises an antigen binding domain having a variable heavy chain sequence with at least 95% sequence identity to
Figure imgf000006_0002
[0038] In some embodiments, the CFP comprises an antigen binding domain having a variable light chain sequence with at least 95% sequence identity to
Figure imgf000006_0003
[0039] In some embodiments, the CFP comprises an scFv having a sequence that is at least 95% identical to the sequence
Figure imgf000006_0001
[0040] In some embodiments, the CFP further comprises a transmembrane domain comprising a CD8 transmembrane domain, the sequence comprising at least 95% identity to IYIWAPLAGTCGVLLLSLVITLYC. [0041] In some embodiments, the CFP further comprises a CD8 hinge domain having a sequence that has at least 95% sequence identity to the sequence
Figure imgf000006_0004
In some embodiments, the CFP further comprises an intracellular signaling domain encoding an FCER1G intracellular signaling domain having a sequence: RRLKIQVRKAAITSYEKSDGVYTGLSTRNQETYETLKHEKPPQ. In some embodiments, the CFP further comprises an intracellular signaling domain encoding an intracellular signaling domain from PI3kinase recruitment domain, having the sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM. In some embodiments, the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS. [0042] In some embodiments, the CFP specifically binds to CD5 antigen. In some embodiments, the cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG. In some embodiments, a nucleotide in the second position of the 5’cap analog is methylated. In some embodiments, the nucleotide in the second position of the 5’cap is further modified at the N6 position. In some embodiments, expression of the sequence encoded by the mRNA in a cell is enhanced compared to an mRNA that does not comprise the 5’cap specified herein. [0043] In some embodiments, the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to
Figure imgf000007_0004
[0044] In some embodiments, the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYSASFLYSGVPSRF SGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKVEIK. [0045] In some embodiments, the CFP further comprises an antigen binding domain that is an scFv having a sequence that is at least 95% identical to the sequence
Figure imgf000007_0001
In some embodiments, the CFP further comprises a CD8 hinge domain having a sequence ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLD. [0046] In some embodiments, the CFP further comprises a CD8 transmembrane domain having a sequence, IYIWAPLAGTCGVLLLSLVITLYC. [0047] In some embodiments, the intracellular signaling domain from CD40 comprises a sequence
Figure imgf000007_0002
Q. In some embodiments, the intracellular signaling domain from FCER1G comprises a sequence . In some embodiments, the
Figure imgf000007_0003
intracellular signaling domain from PI3K recruitment domain comprises a sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM. [0048] In one aspect, provided herein is a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; and wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a GPC3 binding domain and (ii) a CD89 transmembrane domain. In some embodiments, the CD89 transmembrane domain is operably linked to the extracellular domain. In some embodiments, the CFP further comprises (iii) a CD89 intracellular domain. In some embodiments, the CFP further comprises an extracellular domain from CD89. In some embodiments, the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG. [0049] In some embodiments, the GPC3 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is FYSYTY. In some embodiments, the HCDR1 sequence is DYEMH; and the HCDR2 sequence is ALDPKTGDTAYSQKFKG. In some embodiments, the GPC3 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is SQNTHVPPT. In some embodiments, the LCDR1 sequence is RSSQSLVHSNRNTYLH; and the LCDR2 sequence is KVSNRFS. In some embodiments, the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000008_0003
Figure imgf000008_0004
In some embodiments, the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000008_0001
Q Q Q Q Q
Figure imgf000008_0002
In some embodiments, wherein the CD89 transmembrane domain is LIRMAVAGLVLVALLAILV. In some embodiments, CFP comprises a CD89 sequence that is at least 95 % identical to the sequence
Figure imgf000008_0005
Figure imgf000008_0006
SVC . In some embodiments, CFP further comprises a signal sequence MWLQSLLLLGTVACSIS. [0050] In one aspect, provided herein is a composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a TROP2 binding domain and (ii) a CD89 transmembrane domain. [0051] In some embodiments, the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV. In some embodiments, the the HCDR1 sequence is TNYGM; and the HCDR2 sequence is WINTYTGEPTYTDDFKG. In some embodiments, the the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLTF. In some embodiments, the the LCDR1 sequence is KASQDVSIA; and the LCDR2 sequence is SASYRYT. In some embodiments, the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000009_0001
VSS. In some embodiments, the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000009_0002
Figure imgf000009_0003
In some embodiments, the CD89 transmembrane domain is LIRMAVAGLVLVALLAILV. In some embodiments, the CFP comprises a CD89 extracellular sequence DSIHQDYTTQN. In some embodiments, the CFP comprises a CD89 intracellular sequence
Figure imgf000009_0004
In some embodiments, the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS. [0052] In one aspect, provided herein is a composition comprising: (a) a lipid nanoparticle comprising (i) a cationic lipid (ii) a non-cationic lipid (iii) a neutral lipid; the lipid nanoparticle encapsulating: (b) an mRNA encoding a chimeric fusion protein (CFP), wherein the mRNA comprises a 5’cap analog represented by the formula 5’-m7GpppAmpG; the CFP comprise an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain; wherein the transmembrane domain of the protein CD89, Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a). In some embodiments, the 5’cap is a cap 1 structure. In some embodiments, a nucleotide in the second position of the 5’cap is methylated. In some embodiments, the nucleotide in the second position of the 5’cap is further modified. In some embodiments, the nucleotide in the second position of the 5’cap is modified at the N6 position. In some embodiments, the lipid nanoparticle further comprises cholesterol. In some embodiments, the lipid nanoparticle is less than 200 nM in diameter. In some embodiments, the chimeric antigen receptor comprises an antigen binding domain that binds to a cancer antigen. In some embodiments, the chimeric antigen receptor comprises an antigen binding domain that is an scFv, and that specifically binds to a cancer antigen. In some embodiments, the chimeric antigen receptor comprises an antigen binding domain that binds to TROP2. In some embodiments, the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV. In some embodiments, the HCDR1 sequence is TNYGM; and the HCDR2 sequence is WINTYTGEPTYTDDFKG. In some embodiments, the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLTF. In some embodiments, the LCDR1 sequence is KASQDVSIA; and the LCDR2 sequence is SASYRYT. In some embodiments, the chimeric antigen receptor comprises GPC3 binding variable heavy chain (VH), and a variable light chain (VL), wherein the variable heavy chain comprises a HCDR1, a HCDR2 and a HCDR3, the HCDR3 having a sequence of ATACADTTQYAYDY, ATACADTTLYEYDY, ATACVDTTQYEYDY, ATACADATQHEYDY, ATACADTTQYDYDY, ATACADTTQYEYDY, ATACADTTHYEYDY, ATACVITTLYEYDY, ATACAETTLYEYDY, ATACADTTQHEYDY, ATACVDTTHYEYDY, ATACASTTLYEYDY, ATACVVTTLYEYDY, ATACGGATGPYDY, ATACAGAIGPYDY, ATACVVVGDQNDY, ATACVVVGDRNDY, ATDCAGGTSTPYDY, ATDCAGGTATPYDY, ATACVVADRNEYDY, ATSCVVVTKNEYDY, ATACSGLTHEYDY, ATTCSGLTHEYDY, ATACANWSSLGPYDY, ATACANWSTLGPYDY, ATACSDPRVYEYDY, ATTCASPEKYEYDY, ATHCGGTSWGTSYDY, ATHCGGSSWSNEYDY, YARYSGRTY, ASSAWPAGPKHQVEYDY, ATACGSLVGMYDY, ATACGSAVHEYDY, ATDCVGFGSNWFDY, ATACASPVIYEYDY, ATDCAGGVGHEYDY, ATDCSLHGSDYPYDY and AVRIYSGSFDNTLAYDY. In some embodiments, the chimeric antigen receptor comprises an antigen binding domain that is an variable heavy chain (VHH), and that specifically binds to a cancer antigen. [0053] Provided herein is a pharmaceutical composition comprising the composition described above, and a pharmaceutically acceptable excipient. Provided herein is a cell comprising the recombinant mRNA. The cell is a live cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a CD14+ cell. In some embodiments, the cell is a CD14+ CD16- cell. In some embodiments the cell is a CD14+ CD16+ cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a monocyte. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a monocyte precursor. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a macrophage cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a dendritic cell. In some embodiments, the cell is a myeloid cell. In some embodiments, the cell is a precursor of a dendritic cell. [0054] Provided herein is a method of expressing a chimeric fusion protein in a cell, the method comprising in vitro transcribing a recombinant mRNA encoding the chimeric fusion protein, 5’capping the mRNA with a Cap structure selected from the Cap structures of Table 7, thereby generating a 5’-capped recombinant mRNA; contacting the cell with a composition comprising the 5’-capped recombinant mRNA encoding the chimeric fusion protein, wherein the mRNA is incorporated into the cell. In some embodiments, the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG. In some embodiments, a nucleotide in the second position of the 5’cap is methylated, and is further modified at the N6 position. In some embodiments, the mRNA expression is enhanced compared to a recombinant mRNA that does not comprise the 5’cap structure specified herein. In some embodiments, the composition further comprises a lipid nanoparticle encapsulating the 5’-capped recombinant mRNA. [0055] Provided herein is a method of treating cancer in a subject, the method comprising A method of treating a cancer in a subject, the method comprising administering to the subject a composition comprising the cell that comprises the recombinant mRNA described herein. In some embodiments, the cell is a CD14+ cell. In some embodiments, the cell does not express CD16. [0056] In some embodiments, the cell is CD14+ and CD16+. [0057] Provided herein is a method of treating cancer in a subject, the method comprising administering to the subject in need thereof any one of the compositions described above, an mRNA, a naked mRNA, an mRNA encapsulated in a lipid nanoparticle, or a cell comprising the mRNA, wherein the mRNA comprises a 5’cap as described herein at least at the time of uptake of the mRNA by a cell. In some embodiments, the cancer is ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer. In some embodiments, the cancer is breast cancer. INCORPORATION BY REFERENCE [0058] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. BRIEF DESCRIPTION OF THE DRAWINGS [0059] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings. [0060] FIG. 1 outlines the ARCA versus Clean Cap processes of mRNA capping in vitro. ARCA (left), Clean Cap analog (right). [0061] FIG. 2 depicts a diagram showing naturally occurring m6Am Cap (top left), and the structure of bases (top right); and four trinucleotide Cap analog structures. [0062] FIG.3 depicts structures of an adenine Cap analogue. [0063] FIG. 4 depicts graphical structures of different Cap analogs, ARCA (Cap0), and two other (Cap1) structures, termed herein as CC1 (Cap1) and EC1 (Cap 1), whose structural design are shown. The polynucleotide chain in blue is mRNA terminating in the cap at it’s 5’ end. [0064] FIG. 5 depicts a generalized workflow for evaluating the effect of the Cap analogs described in FIG.4. EP, electroporation. [0065] FIG. 6 depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct on the expression level at 24 hours after introduction to primary monocytes derived from human donors. The mRNA encodes a chimeric fusion protein (CFP) or CAR, chimeric antigenic receptor, that binds HER2 (HER2 binding CAR). The expression was determined by binding to HER2. EP, electroporation. Left, data from individual donor sets. Right, summary data from the results shown on the left graph. The data shown is an average of six experimental runs. [0066] FIG. 7 depicts data from experiment conducted as described for FIG. 6, showing the expression level of HER2 binding CAR over the time period indicated in the X axis after introduction of the mRNA in primary monocytes derived from human donors. [0067] FIG.8 depicts a graphical representation of a workflow for evaluating the effect of the cap analogs for immunogenic response. Immunogenic response is evaluated by measurement of Type 1 interferon response 4h and 20h post transfection. [0068] FIG. 9A depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct, interferon-alpha (IFN- ^) and interferon-beta (IFN- ^) release at 4h and 20h after introduction to primary monocytes derived from human donors. Data on commercial ARCA is boxed. [0069] FIG. 9B depicts data from an experiment conducted to determine the effect of three Cap structures in the mRNA construct, CXCL-10 and IFITM1 generation at 4h and 20h after introduction to primary monocytes derived from human donors. Data on commercial ARCA is boxed. [0070] FIG. 10 depicts visualization of RNA products from a large scale production of capped mRNA, involving comparison studies of ARCA -vs – CC1 capped mRNA encoding a GP-75 binding CAR, gp-75-FcR-PI3K. Left figure, mRNA resolved in an electrophoresis, against a molecular size marker ladder (left-most well). Right figure, dot blot depiction of the prepared mRNA samples for detection of double-stranded RNA produced which are contaminant components in the preparation. [0071] FIG. 11 shows results from examination of murine monocytes after nucleofection with a recombinant mRNA encoding anti-gp75 CAR (gp75-FLAG-FcR-PI3K) capped with ARCA or CC1 cap, before administering the anti-gp75 CAR-expressing monocytes into mice. GP75-FcR-PI3K is a structural depiction of the domains of the CAR, having an anti-GP75 extracellular antigen binding domain comprising an anti-GP75 scFv; an FcR transmembrane domain, and a PI3Kinase intracellular domain. An aliquot of the cells were examined in vitro for cell survival after nucleofection (upper panel) assessed with 7-AAD staining, which showed that cell survival was not affected by the nucleofection. Lower panel shows high level of expression of the gp75 CAR constructs in the cells for each of the doses. Doses 1, 2, 3, and 4 correspond to the cell preparations for the monocyte doses administered to the mice in FIG.12. [0072] FIG. 12 depicts experiment results, showing the effect of monocytes expressing the indicated anti-gp75 CAR mRNA (gp75-FLAG-FcR-PI3K) capped with ARCA or CC1 cap on tumor size in a syngeneic mouse tumor model. The GP75 tumor is induced in the mouse, followed by injecting the monocytes of FIG. 11, by intravenous administration at the times indicated at the bottom of the graph in black triangles. The monocytes express the mRNA composition, wherein the mRNA is a ARCA-capped, or CC1-capped, wherein the mRNA encodes the anti-GP75 CAR as described in FIG.11 above. [0073] FIG. 13 depicts a gel electrophoresis showing electrophoretic mobility of capped and uncapped mRNA of the indicated preparations. The bands for capped and uncapped mRNA are shown by arrows. The ratio of the amount of cap to the ATP or GTP amount in the preparation are indicated at the top. [0074] FIG. 14A-14B represents data from an independent experiment, involving an assay using monocytes from a single donor (MYE101-0023B) using the protocol depicted in FIG. 5; mRNA encoding anti-HER2 CARs were tested for the effect of different cap structures in the mRNA constructs as depicted at the top of the figures. The data agrees with the data shown in FIG.6. [0075] FIG. 15 depicts data showing binding of HER2, and an assay that determines the expression level of the anti-HER2 CAR in the cells from the experiment of FIGs.14A-14B. 0.1 microgram RNA was electroporated in 2x 10γ6 cells in 20 microliters reaction volume, final RNA concentration 0.1 uM. [0076] FIG. 16 depicts data from the same experiment of FIGs 14A, 14B and 15, the data showing duration of expression of the anti-HER2 CAR based on the cap structures. [0077] FIG. 17 depicts data from experiment performed with cells from a donor PD-004-04 electroporated with 0.1 microM final concentration mRNA of any one of uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti-HER2 CAR-polyA, m6A-capped -anti-HER2 CAR-polyA, EC1- capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR-polyA mRNA constructs. Data showing flow cytometry of cells electroporated with the constructs. [0078] FIG. 18 depicts data from the experiment described in FIG. 17, showing expression of anti-HER2 as determined by binding to HER2. [0079] FIG. 19 depicts data from the experiment described in FIG. 17 and FIG. 18, showing expression of anti-HER2 over time. [0080] FIG. 20 depicts data from a parallel experiment similar to that described in FIG. 17, performed with cells from a donor PD-004-04 but transfected (lipofected) with the mRNA, the different mRNA sets are: uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti-HER2 CAR- polyA, m6A-capped -anti-HER2 CAR-polyA, EC1-capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR-polyA mRNA constructs. Data showing flow cytometry of cells transfected with the constructs. Transfection was done with 6x10^5 cells/24 well/0.5 microgram RNA. [0081] FIG. 21 depicts data showing anti-HER2 binder expression in the different experimental sets described in FIG.20. [0082] FIG. 22 depicts data from the experiment described in FIG. 20 and FIG. 21, showing expression of anti-HER2 over time following the lipofection. [0083] FIG. 23 depicts data from an experiment performed with cells from a donor MYE-101- 0024A electroporated with 0.1 microM of uncapped AG-anti-HER2 CAR-polyA; CC1-capped-anti- HER2 CAR-polyA, EC1-capped-anti-HER2 capped CAR-polyA or ARCA-capped anti-HER2 CAR- polyA mRNA constructs. Data showing flow cytometry of cells electroporated with the constructs. RNA was made with IVT buffer at pH 6.5 or ARCA kit. [0084] FIG. 24 depicts data showing expression of anti-HER2 binder (CAR) in the experiment described in FIG.23. [0085] FIG. 25 depicts data from the experiment described in FIG. 23 and FIG. 24, showing expression of anti-HER2 CAR over time following the lipofection. [0086] FIG. 26 depicts data from an experiment where the monocyte donor is PD004-04. Monocytes were electroporated with mRNA that were not purified or purified by HPLC. 2x10^6 cells were electroporated with RNA, final concentration 0.5 uM. [0087] FIG.27A depicts data indicating expression of anti-HER2 CAR in the different sets, in the experiment described for FIG.26 at 4h and 24hs. [0088] FIG. 27B depicts data indicating expression of anti-HER2 CAR in the different sets, at 48h after electroporation. [0089] FIG.28 depicts data showing the duration of the HER2 binder expression. DETAILED DESCRIPTION [0090] All terms are intended to be understood as they would be understood by a person skilled in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the disclosure pertains. [0091] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. [0092] Although various features of the present disclosure can be described in the context of a single embodiment, the features can also be provided separately or in any suitable combination. Conversely, although the present disclosure can be described herein in the context of separate embodiments for clarity, the disclosure can also be implemented in a single embodiment. [0093] Reference in the specification to “some embodiments,” “an embodiment,” “one embodiment” or “other embodiments” means that a feature, structure, or characteristic described in connection with the embodiments is included in at least some embodiments, but not necessarily all embodiments, of the present disclosure. [0094] As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open- ended and do not exclude additional, unrecited elements or method steps. It is contemplated that any embodiment discussed in this specification can be implemented with respect to any method or composition of the disclosure, and vice versa. Furthermore, compositions of the disclosure can be used to achieve methods of the disclosure. [0095] The term “about” or “approximately” as used herein when referring to a measurable value such as a parameter, an amount, a temporal duration, and the like, is meant to encompass variations of +/-30% or less, +/-20% or less, +/-10% or less, +/-5% or less, or +/-1% or less of and from the specified value, insofar such variations are appropriate to perform in the present disclosure. It is to be understood that the value to which the modifier “about” or “approximately” refers is itself also specifically disclosed. [0096] Provided herein are engineered myeloid cells (including, but not limited to, neutrophils, monocytes, myeloid dendritic cells (mDCs), mast cells and macrophages), designed to specifically bind a target cell. The engineered myeloid cells can attack and kill target cells directly (e.g., by phagocytosis) and/or indirectly (e.g., by activating T cells). In some embodiments, the target cell is a cancer cell. [0097] While cancer is one exemplary embodiment described in detail in the instant disclosure, the methods and technologies described herein are contemplated to be useful in targeting an infected or otherwise diseased cell inside the body. Similarly, therapeutic and vaccine compositions using the engineered cells are described herein. [0098] Provided herein are compositions and methods for treating diseases or conditions, such as cancer. The compositions and methods provided herein utilize human myeloid cells, including, but not limited to, neutrophils, monocytes, myeloid dendritic cells (mDCs), mast cells and macrophages, to target diseased cells, such as cancer cells. The compositions and methods provided herein can be used to eliminate diseased cells, such as cancer cells and or diseased tissue, by a variety of mechanisms, including T cell activation and recruitment, effector immune cell activation (e.g., CD8 T cell and NK cell activation), antigen cross presentation, enhanced inflammatory responses, reduction of regulatory T cells and phagocytosis. For example, the myeloid cells can be used to sustain immunological responses against cancer cells. [0099] Provided herein is a composition comprising a recombinant polynucleic acids, wherein the one or more polynucleic acids encodes one or more chimeric fusion proteins, and wherein the recombinant polynucleic acid when introduced systemically, is expressed in specific cell types in vivo. [0100] In one aspect, the recombinant polynucleic acid is an mRNA. In some embodiments the recombinant polynucleic acid is an mRNA comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for myeloid cell-specific expression. [0101] In one aspect, the recombinant polynucleic acid is an mRNA. In some embodiments the recombinant polynucleic acid is an mRNA comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for non-myeloid cell-specific expression. In some embodiments, the non-myeloid cell is a lymphocyte, e.g., B cell, a T cell, an NK cell etc. [0102] In some embodiments, the recombinant polynucleic acid comprises a first sequence comprising a sequence encoding a chimeric fusion protein (CFP) that is designed for myeloid cell- specific expression, and a second sequence encoding a chimeric receptor that is designed for non- myeloid cell specific expression. In some embodiments, non-myeloid cell is a T cell. In some embodiments, a single polynucleotide is designed that comprises a first sequence that encodes a CFP, which predominantly expresses in a myeloid cell; and a second sequence that encodes a chimeric antigen receptor (CAR) that is predominantly expressed in a T cell. In some embodiments, the first and the second sequences are intervened by a self-cleavage sequence such as a T2A or P2A sequence, thereby post-translationally generating two peptides, one comprising the CFP, the other comprising the CAR. Also provided is a recombinant polypeptide translation product of the recombinant polynucleic acid described above, comprising (1) a single polypeptide comprising a CFP that is expressed in a myeloid cell, and (2) a single polypeptide comprising a CAR that is expressed in a T cell. In some embodiments, a polynucleic acid construct comprising sequence encoding the CFP arm and the sequence encoding the CAR arm as described herein can be delivered in vivo in a suitable vehicle such that the CFP is expressed in a myeloid cell in vivo, and the CAR is expressed in a lymphoid cell in vivo. [0103] In some embodiments, the recombinant polynucleic acid is an mRNA comprising: (a) a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) a CFP extracellular domain comprising a first antigen binding domain, and (ii) a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and (b) a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: (i) an CAR extracellular domain comprising a second antigen binding domain, (ii) a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR) complex and/or at least one endogenous TCR polypeptide, and (iii) a CAR intracellular domain operatively linked to the transmembrane domain. Also provided herein is a pharmaceutical composition comprising: (A). one or more lipid nanoparticles, (B) one or more polynucleic acids, the one or more polynucleic acids comprising: (a) a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) a CFP extracellular domain comprising a first antigen binding domain, and (ii) a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and (b) a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: (i) a CAR extracellular domain comprising a second antigen binding domain, (ii) a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR) complex and/or at least one endogenous TCR polypeptide, and (iii) a CAR intracellular domain operatively linked to the transmembrane domain; wherein the one or more lipid nanoparticles of (A) encapsulate the one or more polynucleic acids of (B). In some embodiments, the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence, wherein the first sequence and the second sequence are operatively linked by a linker sequence. [0104] In some embodiments, the linker sequence comprises a protease cleavage site such as a T2A cleavage site or a P2A cleavage site. The P2A or the T2A sequence can cleave the first arm and the second arm, releasing the CFP and the CAR arms in vivo for independent expression. [0105] In some embodiments, the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle. In some embodiments, the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle. [0106] In some embodiments, the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle. In some embodiments, the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle. In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject. In some embodiments, the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject. In some embodiments, the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR- beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR- delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. In some embodiments, each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. In some embodiments, the CAR intracellular domain further comprises a costimulatory domain. In some embodiments, the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). In some embodiments, the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain. In some embodiments, the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89. In some embodiments, CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain. In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is expressed or specifically expressed in myeloid cells, monocytes or macrophages of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is expressed or specifically expressed in T cells of the human subject. [0107] In some embodiments, the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89. In some embodiments, the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain. In some embodiments, the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from FcγR, FcαR, FcεR, CD40 or CD3zeta. In some embodiments, the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain. In some embodiments, the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4. In some embodiments, the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89. [0108] In some embodiments, the one or more polynucleic acids is an mRNA. In some embodiments, the nanoparticle delivery vehicle comprises a lipid nanoparticle. In some embodiments, the lipid nanoparticle comprises a polar lipid. In some embodiments, the lipid nanoparticle comprises a non-polar lipid. In some embodiments the lipid nanoparticle is from 100 to 300 nm in diameter. In some embodiments the first antigen binding domain and the second antigen binding domain bind to the same antigen. In some embodiments, the first antigen binding domain and the second antigen binding domain bind to different antigens. [0109] In some embodiments, the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75. In some embodiments, the T cell is a CD3+ T cell, a CD4+ T cell, a CD8+ T cell, an NK T cell, an αβ T cell or a δγ T cell. [0110] In some embodiments, the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer. [0111] The present disclosure is based on the important finding that engineered myeloid cells overcome at least some of the limitations of CAR-T cells, including being readily recruited to solid tumors; having an engineerable duration of survival, therefore lowering the risk of prolonged persistence resulting in aplasia and immunodeficiency; myeloid cells cannot be contaminated with T cells; myeloid cells can avoidance of fratricide, for example because they do not express the same antigens as malignant T cells; and myeloid cells have a plethora of anti-tumor functions that can be deployed. In some respects, engineered myeloid derived cells can be safer immunotherapy tools to target and destroy diseased cells. [0112] Moreover, myeloid cells, such as macrophages, have been ubiquitously found in the tumor environment (TME) and are notably the most abundant cells in some tumor types. As part of their role in the immune system, myeloid cells, such as macrophages, are naturally engaged in clearing diseased cells. The present invention relates too harnessing myeloid cell function and specifically for targeting, killing and directly and/or indirectly clearing diseased cells as well as the delivery payloads such as antigens and cytokines. [0113] The present disclosure is also based on the important finding that engineered myeloid cells can promote endogenous T cell activity, therefore can amplify the activity of existing therapies focused on activating T cells. Engineered T cells and myeloid cells can be achieved through the in vivo incorporation of nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric fusion protein (CFP), that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, ex vivo using recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology. [0114] The present disclosure involves, in addition to the programming of myeloid cells in vivo, programming T cells in vivo. It has been found that the combination of T cell and myeloid cell engineering can have robust anti-tumor cell activity. For example, it has been found that T cells and myeloid cells can be engineered to express a chimeric antigen receptor (CAR) and chimeric fusion protein (CFP), respectively, each containing an antigen binding domain to have a broad and diverse range of activities. For example, it has been found that T cells can be engineered to have enhanced cytolytic activity such that upon binding of the CAR to an antigen on a target cell, the cell exhibits increased lysis of the target cell. It has also been found that co-engineering myeloid cells with T cells can promote broad immune activation in tumor microenvironment, resulting in robust and durable anti-tumor responses. Engineered T cells can be achieved through the in vivo incorporation of nucleic acid sequences (e.g., mRNA, plasmids, viral constructs) encoding a chimeric antigen receptor (CAR) that has an extracellular binding domain specific to disease associated antigens (e.g., cancer antigens), into the cells using, for example, ex vivo using recombinant nucleic acid technology, synthetic nucleic acids, gene editing techniques (e.g., CRISPR), transduction (e.g., using viral constructs), electroporation, or nucleofection, or in vivo using mRNA delivery technology including but not limited to LNP technology. [0115] Engineered myeloid cells can also be short-lived in vivo, phenotypically diverse, sensitive, plastic, and are often found to be difficult to manipulate in vitro. For example, exogenous gene expression in monocytes has been difficult compared to exogenous gene expression in non- hematopoietic cells. There are significant technical difficulties associated with transfecting myeloid cells (e.g., monocytes/macrophages). As professional phagocytes, myeloid cells, such as monocytes/macrophages, comprise many potent degradative enzymes that can disrupt nucleic acid integrity and make gene transfer into these cells an inefficient process. This is especially true of activated macrophages which undergo a dramatic change in their physiology following exposure to immune or inflammatory stimuli. Viral transduction of these cells has been hampered because macrophages are end-stage cells that generally do not divide; therefore, some of the vectors that depend on integration into a replicative genome have met with limited success. Furthermore, macrophages are quite responsive to “danger signals,” and therefore several of the original viral vectors that were used for gene transfer induced potent anti-viral responses in these cells making these vectors inappropriate for gene delivery. The present disclosure provides innovative methods and compositions that can successfully transfect or transduce a myeloid cell, or otherwise induce a genetic modification in a myeloid cell, with the purpose of augmenting a functional aspect of a myeloid cell, additionally, without compromising the cell’s differentiation capability, maturation potential, and/or its plasticity. [0116] Provided herein is a pharmaceutical composition comprising: (A) a recombinant nucleic acid comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient; wherein the recombinant nucleic acid is a recombinant self-amplifying nucleic acid. [0117] In some embodiments, the recombinant self-amplifying nucleic acid is a recombinant self- amplifying RNA. [0118] In some embodiments, the recombinant self-amplifying nucleic acid is a recombinant self- amplifying mRNA. [0119] In some embodiments, the recombinant self-amplifying nucleic acid comprises a 5' Cap. In some embodiments, the recombinant self-amplifying nucleic acid comprises a 3' polyadenylated (polyA) tail sequence. [0120] In some embodiments, the recombinant self-amplifying nucleic acid comprises a subgenomic promoter upstream of the sequence encoding a CFP. In some embodiments, the recombinant self-amplifying nucleic acid comprises a sequence encoding one or more replication genes from a virus. In some embodiments, the sequence encoding one or more replication genes from a virus is upstream of the sequence encoding a CFP. In some embodiments, the sequence encoding one or more replication genes from the virus is upstream of the subgenomic promoter. In some embodiments, the recombinant self-amplifying nucleic acid comprises a promoter upstream of the sequence encoding one or more replication genes from a virus. In some embodiments, the one or more replication genes comprise one or more non-structural proteins (NSPs). In some embodiments, the one or more NSPs comprises one or more of NSP1, NSP2, NSP3 and/or NSP4. In some embodiments, the recombinant self-amplifying nucleic acid lacks one or more viral genes encoding viral structural proteins required to make an infectious viral particle. In some embodiments, the virus is an RNA virus. In some embodiments, the virus is a single stranded virus. In some embodiments, the virus is an alphavirus or a flavivirus, selected from Venezuelan equine encephalitis virus (VEE), Sindbis virus (SINV), vaccinia virus and Semliki forest virus (SFV). In some embodiments, the recombinant self-amplifying nucleic acid is encapsulated within a lipid nanoparticle (LNP). [0121] Provided herein is a pharmaceutical composition comprising: (A) a recombinant RNA comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient, wherein the recombinant RNA is an in vitro glycosylated RNA comprising at least one residue that comprises a glycan. [0122] In some embodiments, the glycan is an N-linked glycan. In some embodiments, the at least one residue that comprises a glycan is a guanosine (G). In some embodiments, the glycan is sialylated glycan. In some embodiments, the glycan is fucosylated. In some embodiments, the glycan is wherein the recombinant RNA is a recombinant mRNA. In some embodiments, the recombinant RNA is encapsulated within a lipid nanoparticle (LNP). In some embodiments, the in vitro glycosylated RNA is glycosylated at specific residues. In some embodiments, the recombinant mRNA comprises 1,2 ,3 ,4, 5, 6, 7, 8, 9, 10 or more glycans. In some embodiments, the recombinant mRNA is a self-amplifying mRNA. Provided herein is a pharmaceutical composition comprising: (A) a recombinant nucleic acid comprising a sequence encoding a chimeric fusion protein (CFP) comprising: a phagocytic or tethering receptor (PR) subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain from a phagocytic or tethering receptor, wherein the transmembrane domain and the intracellular domain are operatively linked; and (B) a pharmaceutically acceptable excipient wherein the recombinant nucleic acid comprises a sequence complementary to an miRNA, wherein the miRNA: (i) is expressed in a myeloid cell and binding of the miRNA to the sequence complementary to the miRNA promotes expression of the CFP in the myeloid cell, or (ii) is expressed in a non-myeloid cell and not substantially expressed in a myeloid cell, wherein binding of the miRNA to the sequence complementary to the miRNA promotes degradation of the recombinant nucleic acid in a non-myeloid cell. [0123] In some embodiments, the non-myeloid cell is a neuronal cell. In some embodiments, the non-myeloid cell is an epithelial cell. In some embodiments, the non-myeloid cell is a hepatocyte. In some embodiments, the non-myeloid cell is a cardiomyocyte. In some embodiments, the non- myeloid cell is an embryonic stem cell. In some embodiments, the non-myeloid cell is an endothelial cell. In some embodiments, the miRNA is hsa-mir-302a, hsa-miR-302b, hsa-miR-302c, hsa-miR- 302d, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373, miR-10a/b, miR-24/27, miR-125a, miR-126 or miR-221/222. [0124] In some embodiments, the myeloid cell is a CD14+ cell. In some embodiments, the CD14+ cell is CD16- or CD16low. In some embodiments, the composition further comprises one or more lipid and/or carbohydrate or one or more polymer components. In some embodiments, the recombinant mRNA that is at least 5 kb in length. In some embodiments, the pharmaceutical composition is formulated for systemic delivery. In some embodiments, the recombinant mRNA is encapsulated in a lipid nanoparticle (LNP). In some embodiments, the lipid nanoparticle is about 100 nm in diameter. In some embodiments, the lipid nanoparticle comprises at least a cationic lipid, and/or a non-cationic lipid. In some embodiments, the cationic lipid is selected from a group consisting of: phosphatidylserines, phosphatidic acids, phosphatidylglycerols, phosphatidylinositols, sterol hemisuccinates, dialkyl trimethylammonium-propanes, (e.g. DOTAP, DOTMA), dialkyl dimethylaminopropanes, ethyl phosphocholines, dimethylaminoethane carbamoyl sterols (e.g. DC- Chol). In some embodiments, the non-cationic lipid is a neutral lipid. In some embodiments, the non-cationic lipid is selected from the group consisting of: DSPC, DPPC, DMPC, DOPC, POPC, DOPE and SM. In some embodiments, the molar ratio of the mRNA to the neutral lipid ranges from about 2:1 to about 8:1. In some embodiments, the lipid composition further comprises a pegylated lipid. In some embodiments, the PEGylated lipid comprises a 50-200 carbon linear chain. [0125] In some embodiments, the pegylated lipid comprises one or more of a pegylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy-polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a pegylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2′,3′-di(tetradecanoyloxy)propyl-1-O-(cw- methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), or, a pegylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as ω-methoxy(polyethoxy)ethyl-N-(2,3- di(tetradecanoxy)propyl)carbamate, or 2,3-di(tetradecanoxy)propyl-N-(ω- methoxy(polyethoxy)ethyl)carbamate. In some embodiments, the molar ratio of the mRNA to the pegylated lipid ranges from about 100:1 to about 25:1. In some embodiments, the lipid nanoparticle further comprises a steroid or steroid analog. In some embodiments, the steroid or steroid analogue is cholesterol. In some embodiments, the molar ratio of the mRNA to cholesterol ranges from about 2:1 to 1:1. In some embodiments, the pharmaceutical composition further comprising one or more phospholipids. In some embodiments, the one or more phospholipids selected from the group consisting of phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, di stearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine. In some embodiments, the CFP further comprises an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell. In some embodiments, the intracellular signaling domain comprises a phagocytosis activation domain. In some embodiments, the intracellular signaling domain is derived from a receptor selected from the group consisting of the receptors listed in Table 2. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. In some embodiments, the antigen is an antigen or a surface molecule that is expressed by a diseased cell or a pathogenic cell or a pathogen. In some embodiments, the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof. [0126] In some embodiments, the antigen is selected from the group consisting of Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), Mesothelin, EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B-Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2, a Natural Killer Group 2D (NKG2D) ligand, Disialoganglioside 2 (GD2), CD2, CD3, CD4, CD5, CD7, CD8, CD19, CD20, CD22, CD24, CD30, CD33, CD38, CD44v6, CD45, CD56CD79b, CD97, CD117, CD123, CD133, CD138, CD171, CD179a, CD213A2, CD248, CD276, PSCA, CS-1, CLECL1, GD3, PSMA, FLT3, TAG72, EPCAM, IL-1, an integrin receptor, PRSS21, VEGFR2, PDGFR-β, SSEA-4, EGFR, NCAM, prostase, PAP, ELF2M, GM3, TEM7R, CLDN6, TSHR, GPRC5D, ALK, IGLL1 and combinations thereof. In some embodiments, the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56. [0127] In some embodiments, the antigen is human CD5. In some embodiments, the antigen is human HER2. [0128] In some embodiments, the antigen is human CD19. In some embodiments, the antigen is a viral antigen expressed on a mammalian cell. In some embodiments, the antigen is a bacterial antigen expressed on a mammalian cell. In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least greater than 20% compared to a cell not expressing the CFP, wherein killing or phagocytosis activity of a cell expressing the CFP is increased by at least 20% compared to a cell not expressing the CFP, wherein killing or phagocytosis activity is measured by flow cytometry. In some embodiments, the cell expressing the CFP exhibits at least a 1.2-fold, 1.3-fold, 1.4-fold, 1.5-fold, 1.6-fold, 1.7-fold, 1.8-fold, 1.9-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, or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP, wherein phagocytosis is measured by flow cytometry. In some embodiments, the target cell expressing the antigen is a diseased cell. [0129] In some embodiments, the cell expressing the CFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP. In some embodiments, the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof. [0130] In some embodiments, a cell expressing the CFP exhibits an increase in effector activity compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in cross-presentation compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP. In some embodiments, the cell expressing the CFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP. In some embodiments, the pharmaceutical composition is aqueous. [0131] In some embodiments, the pharmaceutical composition comprises one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: an CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR) complex and/or at least one endogenous TCR polypeptide, and a CAR intracellular domain operatively linked to the transmembrane domain. [0132] Also provided herein is a pharmaceutical composition comprising one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: a CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR) complex and/or at least one endogenous TCR polypeptide, and a CAR intracellular domain operatively linked to the transmembrane domain. [0133] In some embodiments, the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence. In some embodiments, the first sequence and the second sequence are operatively linked by a linker sequence. In some embodiments, the linker sequence comprises a protease cleavage site In some embodiments, the protease cleavage site is a T2A cleavage site or a P2A cleavage site. [0134] In some embodiments, the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle. In some embodiments, the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle. [0135] In some embodiments, the one or more polynucleic acids comprises a first polynucleic acid molecule comprising the first sequence and a second polynucleic acid molecule comprising the second sequence. [0136] In some embodiments, the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle. In some embodiments, the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle. [0137] In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject. [0138] In some embodiments, the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject. In some embodiments, the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. In some embodiments, each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. [0139] In some embodiments, the CAR intracellular domain further comprises a costimulatory domain. In some embodiments, the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). [0140] In some embodiments, the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain. [0141] In some embodiments, the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89. In some embodiments, the CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain. [0142] In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is specifically expressed in myeloid cells, monocytes or macrophages of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is specifically expressed in T cells of the human subject. [0143] In some embodiments, the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89. In some embodiments, the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain. In some embodiments, the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from FcγR, FcαR, FcεR, CD40 or CD3zeta. In some embodiments, the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain. In some embodiments, the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4. In some embodiments, the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89. [0144] Provided herein is a method for treating a disease in a subject in need thereof comprising administering a therapeutically effective amount of the pharmaceutical composition described above to the subject. [0145] In some embodiments, the disease is cancer. In some embodiments, the disease is an infection. In some embodiments, the subject is human. [0146] Provided herein are compositions comprising a recombinant nucleic acid encoding a chimeric fusion protein (CFP), such as a phagocytic receptor (PR) fusion protein (PFP), a scavenger receptor (SR) fusion protein (SFP), an integrin receptor (IR) fusion protein (IFP) or a caspase- recruiting receptor (caspase-CAR) fusion protein. A CFP encoded by the recombinant nucleic acid can comprise an extracellular domain (ECD) comprising an antigen binding domain that binds to an antigen of a target cell. The extracellular domain can be fused to a hinge domain or an extracellular domain derived from a receptor, such as CD2, CD8, CD28, CD64 or CD68, a phagocytic receptor, a scavenger receptor or an integrin receptor. The CFP encoded by the recombinant nucleic acid can further comprise a transmembrane domain, such as a transmembrane domain derived from CD2, CD8, CD28, CD64 or CD68, a phagocytic receptor, a scavenger receptor or an integrin receptor. In some embodiments, a CFP encoded by the recombinant nucleic acid further comprises an intracellular domain comprising an intracellular signaling domain, such as an intracellular signaling domain derived from a phagocytic receptor, a scavenger receptor or an integrin receptor. For example, the intracellular domain can comprise one or more intracellular signaling domains derived from a phagocytic receptor, a scavenger receptor or an integrin receptor. For example, the intracellular domain can comprise one or more intracellular signaling domains that promote phagocytic activity, inflammatory response, nitric oxide production, integrin activation, enhanced effector cell migration (e.g., via chemokine receptor expression), antigen presentation, and/or enhanced cross presentation. In some embodiments, the CFP is a phagocytic receptor fusion protein (PFP). In some embodiments, the CFP is a phagocytic scavenger receptor fusion protein (PFP). In some embodiments, the CFP is an integrin receptor fusion protein (IFP). In some embodiments, the CFP is an inflammatory receptor fusion protein. In some embodiments, a CFP encoded by the recombinant nucleic acid further comprises an intracellular domain comprising a recruitment domain. For example, the intracellular domain can comprise one or more PI3K recruitment domains, caspase recruitment domains or caspase activation and recruitment domains (CARDs). [0147] Provided herein is a composition comprising a recombinant nucleic acid encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising a phagocytic receptor intracellular signaling domain; and an extracellular antigen binding domain specific to an antigen, e.g., an antigen of or presented on a target cell; wherein the transmembrane domain and the extracellular antigen binding domain are operatively linked such that antigen binding to the target by the extracellular antigen binding domain of the fused receptor activated in the intracellular signaling domain of the phagocytic receptor. [0148] Provided herein is a composition comprising a recombinant nucleic acid sequence encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the CFP to the antigen of the target cell, the killing or phagocytosis activity of a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP. [0149] Provided herein is a composition comprising a recombinant nucleic acid sequence encoding a CFP comprising a phagocytic or tethering receptor (PR) subunit (e.g., a phagocytic receptor fusion protein (PFP)) comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the CFP to the antigen of the target cell, the killing or phagocytosis activity of a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4- fold, 4.5-fold, 5-fold, 5.5-fold, 6-fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, 10- fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold,-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25- fold, 30-fold, 40-fold, 50-fold, 75-fold, or 100-fold compared to a cell not expressing the CFP. [0150] In one aspect, provided herein is a pharmaceutical composition comprising: (a) a myeloid cell, such as a neutrophil, monocyte, myeloid dendritic cell (mDC), mast cell or macrophage cell comprising a recombinant polynucleic acid, wherein the recombinant polynucleic acid comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising an anti-CD5 binding domain, and (ii) a transmembrane domain operatively linked to the extracellular domain; and (b) a pharmaceutically acceptable carrier; wherein the myeloid cell expresses the CFP and exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing CD5 compared to a myeloid cell not expressing the CFP. In some embodiments, the CD5 binding domain is a CD5 binding protein that comprises an antigen binding fragment of an antibody, an Fab fragment, an scFv domain or an sdAb domain. In some embodiments, the CD5 binding domain comprises an scFv comprising: (i) a variable heavy chain (VH) sequence of SEQ ID NO: 1 (see, Table 4) or with at least 90% sequence identity to SEQ ID NO: 1; and (ii) a variable light chain (VL) sequence of SEQ ID NO: 2 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 2. In some embodiments, the CD5 binding domain comprises an scFv comprising SEQ ID NO: 33 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 33. In some embodiments, the HER2 binding domain comprises an scFv comprising: (i) a variable heavy chain (VH) sequence of SEQ ID NO: 8 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 8; and (ii) a variable light chain (VL) sequence of SEQ ID NO: 9 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 9. In some embodiments, the CD5 binding domain comprises an scFv comprising SEQ ID NO: 32 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 32. In some embodiments, the CFP further comprises an intracellular domain, wherein the intracellular domain comprises one or more intracellular signaling domains, and wherein a wild-type protein comprising the intracellular domain does not comprise the extracellular domain. [0151] In some embodiments, the extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the transmembrane domain and the anti-CD5 binding domain. In some embodiments, the extracellular hinge domain comprises a sequence of SEQ ID NO: 7 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 7. [0152] In some embodiments, the CFP comprises an extracellular domain fused to a transmembrane domain of SEQ ID NO: 30 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 30. In some embodiments, the CFP comprises an extracellular domain fused to a transmembrane domain of SEQ ID NO: 31 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 31. [0153] In some embodiments, the transmembrane domain comprises a CD8 transmembrane domain. In some embodiments, the transmembrane domain comprises a sequence of SEQ ID NO: 6 or 29 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 6 or 29. In some embodiments, the transmembrane domain comprises a sequence of SEQ ID NO: 18 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 18. In some embodiments, the transmembrane domain comprises a sequence of SEQ ID NO: 34 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 34. In some embodiments, the transmembrane domain comprises a sequence of SEQ ID NO: 19 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 19. [0154] In some embodiments, the CFP comprises one or more intracellular signaling domains that comprise a phagocytic signaling domain. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, FcRα, and Bai1. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, an FcR, and Bai1. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than CD3ζ. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from FcRγ, FcRα or FcRε. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from CD3ζ. In some embodiments, the CFP comprises an intracellular signaling domain of any one of SEQ ID NOs: 3, 20, 27 and 28 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any one of SEQ ID NOs: 3, 20, 27 and 28. In some embodiments, the one or more intracellular signaling domains further comprises a proinflammatory signaling domain. In some embodiments, the proinflammatory signaling domain comprises a PI3-kinase (PI3K) recruitment domain. In some embodiments, the proinflammatory signaling domain comprises a sequence of SEQ ID NO: 4 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 4. In some embodiments, the proinflammatory signaling domain is derived from an intracellular signaling domain of CD40. In some embodiments, the proinflammatory signaling domain comprises a sequence of SEQ ID NO: 5 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 5. In some embodiments, the CFP comprises an intracellular signaling domain of SEQ ID NO: 21 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 21. In some embodiments, the CFP comprises an intracellular signaling domain of SEQ ID NO: 23 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 23. [0155] In some embodiments, the CFP comprises a sequence of SEQ ID NO: 14 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 14. In some embodiments, the CFP comprises a sequence of SEQ ID NO: 15 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 15. In some embodiments, the CFP comprises a sequence of SEQ ID NO: 16 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 16. In some embodiments, the CFP comprises a sequence of SEQ ID NO: 24 or with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 24. In some embodiments, the CFP comprises a sequence of SEQ ID NO:25 or with at least 70%, 75%, 80%, 85%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 25. [0156] In some embodiments, the CFP comprises: (a) an extracellular domain comprising: (i) a scFv that specifically binds CD5, and (ii) a hinge domain derived from CD8; a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68; (b) a CD8 transmembrane domain, a CD28 transmembrane domain, a CD2 transmembrane domain or a CD68 transmembrane domain; and (c) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from FcRα, FcRγ or FcRε, and (ii) a second intracellular signaling domain: (A) comprising a PI3K recruitment domain, or (B) derived from CD40. In some embodiments, the CFP comprises as an alternative (c) to the above: an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from a phagocytic receptor intracellular domain, and (ii) a second intracellular signaling domain derived from a scavenger receptor phagocytic receptor intracellular domain comprising: (A) comprising a PI3K recruitment domain, or (B) derived from CD40. Exemplary scavenger receptors from which an intracellular signaling domain may be derived may be found in Table 2. In some embodiments, the CFP comprises and intracellular signaling domain derived from an intracellular signaling domain of an innate immune receptor. [0157] In some embodiments, the recombinant polynucleic acid is an mRNA. In some embodiments, the recombinant polynucleic acid is a circRNA. In some embodiments, the recombinant polynucleic acid is a viral vector. In some embodiments, the recombinant polynucleic acid is delivered via a viral vector. [0158] In some embodiments, the nanoparticle delivery vehicle comprises a lipid nanoparticle. In some embodiments, the lipid nanoparticle comprises a polar lipid. In some embodiments, the lipid nanoparticle comprises a non-polar lipid. In some embodiments, the lipid nanoparticle is from 100 to 300 nm in diameter. [0159] In some embodiments, the first antigen binding domain and the second antigen binding domain bind to the same antigen. In some embodiments, the first antigen binding domain and the second antigen binding domain bind to different antigens. In some embodiments, the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75. [0160] In some embodiments, the myeloid cell is a CD14+ cell, a CD14+/CD16- cell, a CD14+/CD16+ cell, a CD14-/CD16+ cell, CD14-/CD16- cell, a dendritic cell, an M0 macrophage, an M2 macrophage, an M1 macrophage or a mosaic myeloid cell/macrophage/dendritic cell. [0161] In some embodiments, the T cell is a T cell precursor. In some embodiments, the T cell is an undifferentiated and/or unpolarized. In some embodiments, the T cell is a CD4 T cells. In some embodiments, T cell is a CD8 T cell. In some embodiments, T cell is gamma delta T cell. In some embodiments, the cell is a NK T cell. [0162] In one aspect, provided herein is a method of treating cancer in a human subject in need thereof comprising administering a pharmaceutical composition to the human subject, the pharmaceutical composition comprising: (a) a myeloid cell comprising a recombinant polynucleic acid sequence, wherein the polynucleic acid sequence comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising an anti-CD5 binding domain, and (ii) a transmembrane domain operatively linked to the extracellular domain; and (b) a pharmaceutically acceptable carrier; wherein the myeloid cell expresses the CFP. [0163] In some embodiments, upon binding of the CFP to CD5 expressed by a target cancer cell of the subject killing or phagocytosis activity of the myeloid cell is increased by greater than 20% compared to a myeloid cell not expressing the CFP. In some embodiments, growth of a tumor is inhibited in the human subject. [0164] In some embodiments, the cancer is a CD5+ cancer. In some embodiments, the cancer is leukemia, T cell lymphoma, or B cell lymphoma. [0165] In some embodiments, the anti-CD5 binding domain is a CD5 binding protein that comprises an antigen binding fragment of an antibody, an scFv domain, an Fab fragment, or an sdAb domain. In some embodiments, the anti-CD5 binding domain is a protein or fragment thereof that binds to CD5, such as a ligand of CD5 (e.g., a natural ligand of CD5). [0166] In some embodiments, the CFP further comprises an intracellular domain, wherein the intracellular domain comprises one or more intracellular signaling domains, wherein the one or more intracellular signaling domains comprises a phagocytosis signaling domain and wherein a wild-type protein comprising the intracellular domain does not comprise the extracellular domain. [0167] In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from a receptor other than Megf10, MerTk, FcRα and Bai1. In some embodiments, the phagocytosis signaling domain comprises an intracellular signaling domain derived from FcRγ, FcRα or FcRε. [0168] In some embodiments, the one or more intracellular signaling domains further comprises a proinflammatory signaling domain. In some embodiments, the proinflammatory signaling domain comprises a PI3-kinase (PI3K) recruitment domain. In some embodiments, the transmembrane domain comprises a CD8 transmembrane domain. In some embodiments, the extracellular domain comprises a hinge domain derived from CD8, a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68. [0169] In some embodiments, the CFP comprises: (a) an extracellular domain comprising: (i) a scFv that specifically binds CD5, and (ii) a hinge domain derived from CD8, a hinge domain derived from CD28 or at least a portion of an extracellular domain from CD68; (b) a CD8 transmembrane domain, a CD28 transmembrane domain, a CD2 transmembrane domain or a CD68 transmembrane domain; and (c) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (i) a first intracellular signaling domain derived from FcRγ or FcRε, and (ii) a second intracellular signaling domain that: (A) comprises a PI3K recruitment domain, or (B) is derived from CD40. In some embodiments, the recombinant nucleic acid is mRNA or circRNA. In some embodiments, the myeloid cell is a CD14+ cell, a CD14+/CD16- cell, a CD14+/CD16+ cell, a CD14-/CD16+ cell, CD14-/CD16- cell, a dendritic cell, an M0 macrophage, an M2 macrophage, an M1 macrophage or a mosaic myeloid cell/macrophage/dendritic cell. [0170] In some embodiments, the method further comprises administering an additional therapeutic agent selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity, an agent that promotes sequestration of lymphocytes in primary and/or secondary lymphoid organs, an agent that increases concentration of naïve T cells and central memory T cells in secondary lymphoid organs, and any combination thereof. [0171] In some embodiments, the myeloid cell further comprises: (a) an endogenous peptide or protein that dimerizes with the CFP, (b) a non-endogenous peptide or protein that dimerizes with the CFP; and/or (c) a second recombinant polynucleic acid sequence, wherein the second recombinant polynucleic acid sequence comprises a sequence encoding a peptide or protein that interacts with the CFP; wherein the dimerization or the interaction potentiates phagocytosis by the myeloid cell expressing the CFP as compared to a myeloid cell that does not express the CFP. [0172] In some embodiments, the myeloid cell exhibits (i) an increase in effector activity, cross- presentation, respiratory burst, ROS production, iNOS production, inflammatory mediators, extra- cellular vesicle production, phosphatidylinositol 3,4,5-trisphosphate production, trogocytosis with the target cell expressing the antigen, resistance to CD47 mediated inhibition of phagocytosis, resistance to LILRB1 mediated inhibition of phagocytosis, or any combination thereof; and/or (ii) an increase in expression of a IL-1, IL3, IL-6, IL-10, IL-12, IL-13, IL-23, TNFα, a TNF family of cytokines, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL-17, IP-10, RANTES, an interferon, MHC class I protein, MHC class II protein, CD40, CD48, CD58, CD80, CD86, CD112, CD155, a TRAIL/TNF Family death receptor, TGFβ, B7-DC, B7-H2, LIGHT, HVEM, TL1A, 41BBL, OX40L, GITRL, CD30L, TIM1, TIM4, SLAM, PDL1, an MMP (e.g., MMP2, MMP7 and MMP9) or any combination thereof. [0173] In some embodiments, the intracellular signaling domain is derived from a phagocytic or tethering receptor or wherein the intracellular signaling domain comprises a phagocytosis activation domain. In some embodiments, the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. In some embodiments, the intracellular signaling domain is derived from a protein, such as receptor (e.g., a phagocytic receptor), selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fcα receptor I, CR1, CD35, CD3ζ, a complement receptor, CR3, CR4, Tim-1, Tim-4 and CD169. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. [0174] In some embodiments, the intracellular signaling domain is derived from an ITAM domain containing receptor. [0175] Provided herein is a composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcRα, or Bai1. [0176] In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP. In some embodiments, the CFP functionally incorporates into a cell membrane of a cell when the CFP is expressed in the cell. In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 5.5-fold, 6- fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold,-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30-fold, 40-fold, 50-fold, 75-fold, or 100-fold compared to a cell not expressing the CFP. [0177] In some embodiments, the intracellular signaling domain is derived from a receptor, such as a phagocytic receptor, selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fcα receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim-1, Tim-4 and CD169. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain. [0178] Provided herein is a composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a receptor, such as a phagocytic receptor, selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fcα receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim-1, Tim-4 and CD169. [0179] In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP. In some embodiments, the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcRα, or Bai1. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain. In some embodiments, the intracellular signaling domain comprises a PI3K recruitment domain, such as a PI3K recruitment domain derived from CD19. In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. [0180] Provided herein is a composition comprising a recombinant nucleic acid encoding a CFP, such as a phagocytic or tethering receptor (PR) fusion protein (PFP), comprising: a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; and an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. [0181] Provided herein is a composition of an engineered CFP, such as a phagocytic receptor fusion protein, that may be expressed in a cell, such as a myeloid cell, such as to generate an engineered myeloid cell that can target a target cell, such as a diseased cell. [0182] A target cell is, for example, a cancer cell. In some embodiments, the engineered myeloid cell, after engulfment of a cancer cell may present a cancer antigen on its cell surface to activate a T cell. An “antigen” is a molecule capable of stimulating an immune response. Antigens recognized by T cells, whether helper T lymphocytes (T helper (TH) cells) or cytotoxic T lymphocytes (CTLs), are not recognized as intact proteins, but rather as small peptides that associate with MHC proteins (such as class I or class II MHC proteins) on the surface of cells. During the course of a naturally occurring immune response, antigens that are recognized in association with class II MHC molecules on antigen presenting cells (APCs) are acquired from outside the cell, internalized, and processed into small peptides that associate with the class II MHC molecules. [0183] In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least greater than 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, or 1000% compared to a cell not expressing the CFP. In some embodiments, the CFP functionally incorporates into a cell membrane of a cell when the CFP is expressed in the cell. In some embodiments, upon binding of the CFP to the antigen of the target cell, the killing activity of a cell expressing the CFP is increased by at least 1.1-fold, 1.5-fold, 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 5.5-fold, 6- fold, 6.5-fold, 7-fold, 7.5-fold, 8-fold, 8.5-fold, 9-fold, 9.5-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold,-fold, 17-fold, 18-fold, 19-fold, 20-fold, 25-fold, 30-fold, 40-fold, 50-fold, 75-fold, or 100-fold compared to a cell not expressing the CFP. [0184] In some embodiments, the target cell expressing the antigen is a cancer cell. In some embodiments, the target cell expressing the antigen is at least 0.8 microns in diameter. [0185] In some embodiments, a cell expressing the CFP exhibits an increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits at least a 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10- fold, 20-fold, 30-fold or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP. In some embodiments, the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL- 23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP- 10, RANTES, an interferon and combinations thereof. In some embodiments, a cell expressing the CFP exhibits an increase in effector activity compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in cross-presentation compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TRAIL/TNF Family death receptors compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of B7-H2 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of LIGHT compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of HVEM compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD40 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TL1A compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of 41BBL compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of OX40L compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of GITRL death receptors compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD30L compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TIM4 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of TIM1 ligand compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of SLAM compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD48 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD58 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD155 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of CD112 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of PDL1 compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in expression of B7-DC compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in respiratory burst compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in ROS production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in iNOS production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in iNOS production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in extra-cellular vesicle production compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in trogocytosis with a target cell expressing the antigen compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in resistance to CD47 mediated inhibition of phagocytosis compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the CFP. In some embodiments, a cell expressing the CFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production. [0186] In some embodiments, the extracellular domain of a CFP comprises an Ig binding domain. In some embodiments, the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, FcRγI, FcRγIIA, FcRγIIB, FcRγIIC, FcRγIIIA, FcRγIIIB, FcRn, TRIM21, FcRL5 binding domain. In some embodiments, the extracellular domain of a CFP comprises an FcR extracellular domain. In some embodiments, the extracellular domain of a CFP comprises an FcRα, FcRβ, FcRε or FcRγ extracellular domain. In some embodiments, the extracellular domain comprises an FcRα (FCAR) extracellular domain. In some embodiments, the extracellular domain comprises an FcRβ extracellular domain. In some embodiments, the extracellular domain comprises an FCER1A extracellular domain. In some embodiments, the extracellular domain comprises an FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, or FCGR3B extracellular domain. In some embodiments, the extracellular domain comprises an integrin domain or an integrin receptor domain. In some embodiments, the extracellular domain comprises one or more integrin α1, α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αD, αE, αL, αM, αV, αX, β1, β2, β3, β4, β5, β6, β7, or β8 domains. [0187] In some embodiments, the CFP further comprises an extracellular domain operatively linked to the transmembrane domain and the extracellular antigen binding domain. In some embodiments, the extracellular domain further comprises an extracellular domain of a receptor, a hinge, a spacer and/or a linker. In some embodiments, the extracellular domain comprises an extracellular portion of a phagocytic receptor. In some embodiments, the extracellular portion of the CFP is derived from the same receptor as the receptor from which the intracellular signaling domain is derived. In some embodiments, the extracellular domain comprises an extracellular domain of a scavenger receptor. In some embodiments, the extracellular domain comprises an immunoglobulin domain. In some embodiments, the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region. In some embodiments, the extracellular domain comprises a phagocytic engulfment domain. In some embodiments, the extracellular domain comprises a structure capable of multimeric assembly. In some embodiments, the extracellular domain comprises a scaffold for multimerization. In some embodiments, the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length. In some embodiments, the extracellular antigen binding domain specifically binds to the antigen of a target cell. In some embodiments, the extracellular antigen binding domain comprises an antibody domain. In some embodiments, the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof. In some embodiments, the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor. In some embodiments, the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen. In some embodiments, the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen. [0188] In some embodiments, the antigen is a cancer associated antigen, a lineage associated antigen, a pathogenic antigen or an autoimmune antigen. In some embodiments, the antigen comprises a viral antigen. In some embodiments, the antigen is a T lymphocyte antigen. In some embodiments, the antigen is an extracellular antigen. In some embodiments, the antigen is an intracellular antigen. In some embodiments, the antigen is selected from the group consisting of an antigen from Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B- Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2, a Natural Killer Group 2D (NKG2D) ligand, Disialoganglioside 2 (GD2), CD2, CD3, CD4, CD5, CD7, CD8, CD19, CD20, CD22, CD24, CD30, CD33, CD38, CD44v6, CD45, CD56CD79b, CD97, CD117, CD123, CD133, CD138, CD171, CD179a, CD213A2, CD248, CD276, PSCA, CS-1, CLECL1, GD3, PSMA, FLT3, TAG72, EPCAM, IL-1, an integrin receptor, PRSS21, VEGFR2, PDGFRβ, SSEA-4, EGFR, NCAM, prostase, PAP, ELF2M, GM3, TEM7R, CLDN6, TSHR, GPRC5D, ALK, Dsg1, Dsg3, IGLL1 and combinations thereof. In some embodiments, the antigen is an antigen of a protein selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, and CD56. In some embodiments, the antigen is an ovarian cancer antigen or a T lymphoma antigen. In some embodiments, the antigen is an antigen of an integrin receptor. In some embodiments, the antigen is an antigen of an integrin receptor or integrin selected from the group consisting of α1, α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αD, αE, αL, αM, αV, αX, β 1, β 2, β 3, β 4, β 5, β 6, β 7, and β8. In some embodiment, the antigen is an antigen of an integrin receptor ligand. In some embodiments, the antigen is an antigen of fibronectin, vitronectin, collagen, or laminin. In some embodiments, the antigen binding domain can bind to two or more different antigens. [0189] In some embodiments, the antigen binding domain comprises an autoantigen or fragment thereof, such as Dsg1 or Dsg3. In some embodiments, the extracellular antigen binding domain comprises a receptor domain or an antibody domain wherein the antibody domain binds to an auto antigen, such as Dsg1 or Dsg3. [0190] In some embodiments, the transmembrane domain and the extracellular antigen binding domain are operatively linked through a linker. In some embodiments, the transmembrane domain and the extracellular antigen binding domain are operatively linked through a linker such as a hinge region of CD8α, IgG1 or IgG4. [0191] In some embodiments, the extracellular domain comprises a multimerization scaffold. [0192] In some embodiments, the transmembrane domain comprises a CD8 transmembrane domain. In some embodiments, the transmembrane domain comprises a CD28 transmembrane domain. In some embodiments, the transmembrane domain comprises a CD68 transmembrane domain. In some embodiments, the transmembrane domain comprises a CD2 transmembrane domain. In some embodiments, the transmembrane domain comprises an FcR transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRγ transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRα transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRβ transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRε transmembrane domain. In some embodiments, the transmembrane domain comprises a transmembrane domain from a syntaxin, such as syntaxin 3 or syntaxin 4 or syntaxin 5. In some embodiments, the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an endogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the CFP is expressed in a cell. In some embodiments, the transmembrane domain is derived from a protein that is different than the protein from which the intracellular signaling domain is derived. In some embodiments, the transmembrane domain is derived from a protein that is different than the protein from which the extracellular domain is derived. In some embodiments, the transmembrane domain comprises a transmembrane domain of a phagocytic receptor. In some embodiments, the transmembrane domain and the extracellular domain are derived from the same protein. In some embodiments, the transmembrane domain is derived from the same protein as the intracellular signaling domain. In some embodiments, the recombinant nucleic acid encodes a DAP12 recruitment domain. In some embodiments, the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12. [0193] In some embodiments, the transmembrane domain is at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. In some embodiments, the transmembrane domain is at most 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. [0194] In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor. In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcRα, or Bai1. In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a phagocytic receptor selected from the group consisting of TNFR1, MDA5, CD40, lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim-1, Tim-4 and CD169. In some embodiments, the intracellular signaling domain comprises a PI3K recruitment domain. In some embodiments, the intracellular signaling domain comprises an intracellular signaling domain derived from a scavenger receptor. In some embodiments, the intracellular domain comprises a CD47 inhibition domain. In some embodiments, the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain. In some embodiments, the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the PFP. In some embodiments, the intracellular domain comprises an F- actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain. In some embodiments, the intracellular domain comprises a phosphatase inhibition domain. In some embodiments, the intracellular domain comprises an ARP2/3 inhibition domain. In some embodiments, the intracellular domain comprises at least one ITAM domain. In some embodiments, the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more ITAM domains. In some embodiments, the intracellular domain comprises at least one ITAM domain select from an ITAM domain of CD3 zeta, CD3 epsilon, CD3 gamma, CD3 delta, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278, CD66d, functional fragments thereof, and amino acid sequences thereof having at least one but not more than 20 modifications thereto. In some embodiments, the at least one ITAM domain comprises a Src-family kinase phosphorylation site. In some embodiments, the at least one ITAM domain comprises a Syk recruitment domain. In some embodiments, the intracellular domain comprises an F-actin depolymerization activation domain. In some embodiments, the intracellular domain lacks enzymatic activity. [0195] In some embodiments, the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. In some embodiments, the intracellular domain does not comprise a domain derived from a MerTK intracellular domain. In some embodiments, the intracellular domain does not comprise a domain derived from a TLR4 intracellular domain. In some embodiments, the intracellular domain comprises a CD47 inhibition domain. In some embodiments, the intracellular signaling domain comprises a domain that activates integrin, such as the intracellular region of PSGL-1. In some embodiments, the intracellular signaling domain comprises a domain that activates Rap1 GTPase, such as that from EPAC and C3G. In some embodiments, the intracellular signaling domain is derived from paxillin. In some embodiments, the intracellular signaling domain activates focal adhesion kinase. In some embodiments, the intracellular signaling domain is derived from a single phagocytic receptor. In some embodiments, the intracellular signaling domain is derived from a single scavenger receptor. In some embodiments, the intracellular domain comprises a phagocytosis enhancing domain. [0196] In some embodiments, the intracellular domain comprises a pro-inflammatory signaling domain. In some embodiments, the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain. In some embodiments, the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain. In some embodiments, the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, STING, an NLRP family member, NLRP1-14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40, Tank1-binding kinase (TBK), a caspase domain, a procaspase binding domain or any combination thereof. [0197] In some embodiments, the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a connexin (Cx) protein. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Cx43, Cx46, Cx37, Cx40, Cx33, Cx50, Cx59, Cx62, Cx32, Cx26, Cx31, Cx30.3, Cx31.1, Cx30, Cx25, Cx45, Cx47, Cx31.3, Cx36, Cx31.9, Cx39, Cx40.1 or Cx23. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Cx43. [0198] In some embodiments, the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a SIGLEC protein. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from Siglec-1 (Sialoadhesin), Siglec-2 (CD22), Siglec-3 (CD33), Siglec-4 (MAG), Siglec-5, Siglec-6, Siglec-7, Siglec-8, Siglec-9, Siglec-10, Siglec-11, Siglec-12, Siglec-13, Siglec-14, Siglec-15, Siglec- 16 or Siglec-17. [0199] In some embodiments, the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a C-type lectin protein. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from a mannose receptor protein. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an asialoglycoprotein receptor protein. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from macrophage galactose-type lectin (MGL), DC-SIGN (CLEC4L), Langerin (CLEC4K), Myeloid DAP12 associating lectin (MDL)-1 (CLEC5A), a DC associated C type lectin 1 (Dectin1) subfamily protein, dectin 1/CLEC7A, DNGR1/CLEC9A, Myeloid C type lectin like receptor (MICL) (CLEC12A), CLEC2 (CLEC1B), CLEC12B, a DC immunoreceptor (DCIR) subfamily protein, DCIR/CLEC4A, Dectin 2/CLEC6A, Blood DC antigen 2 (BDCA2) ( CLEC4C), Mincle (macrophage inducible C type lectin) (CLEC4E), a NOD-like receptor protein, NOD-like receptor, MHC Class II transactivator (CIITA), IPAF, BIRC1, a RIG-I-like receptor (RLR) protein, RIG-I, MDA5, LGP2, NAIP5/Birc1e, an NLRP protein, NLRP1, NLRP2, NLRP3, NLRP4, NLRP5, NLRP6, NLRP7, NLRP89, NLRP9, NLRP10, NLRP11, NLRP12, NLRP13, NLRP14, an NLR protein, NOD1 or NOD2, or any combination thereof. [0200] In some embodiments, the intracellular domain comprises a signaling domain, such as an intracellular signaling domain, derived from a cell adhesion molecule. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an IgCAMs, a cadherin, an integrin, a C-type of lectin-like domains protein (CTLD) and/or a proteoglycan molecule. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from an E-cadherin, a P-cadherin, an N-cadherin, an R- cadherin, a B-cadherin, a T-cadherin, or an M-cadherin. For example, the intracellular domain can comprise a signaling domain, such as an intracellular signaling domain, derived from a selectin, such as an E-selectin, an L-selectin or a P-selectin. [0201] In some embodiments, the CFP does not comprise a full length intracellular signaling domain. In some embodiments, the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. [0202] In some embodiments, the recombinant nucleic acid encodes an FcRα chain extracellular domain, an FcRα chain transmembrane domain and/or an FcRα chain intracellular domain. In some embodiments, the recombinant nucleic acid encodes an FcRβ chain extracellular domain, an FcRβ chain transmembrane domain and/or an FcRβ chain intracellular domain. In some embodiments, the FcRα chain or the FcRβ chain forms a complex with FcRγ when expressed in a cell. In some embodiments, the FcRα chain or FcRβ chain forms a complex with endogenous FcRγ when expressed in a cell. In some embodiments, the FcRα chain or the FcRβ chain does not incorporate into a cell membrane of a cell that does not express FcRγ. In some embodiments, the CFP does not comprise an FcRα chain intracellular signaling domain. In some embodiments, the CFP does not comprise an FcRβ chain intracellular signaling domain. In some embodiments, the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain. In some embodiments, the TREM is TREM1, TREM 2 or TREM 3. [0203] In some embodiments, the recombinant nucleic acid comprises a sequence encoding a pro- inflammatory polypeptide. In some embodiments, the composition further comprises a proinflammatory nucleotide or a nucleotide in the recombinant nucleic acid, for example, an ATP, ADP, UTP, UDP, and/or UDP-glucose. [0204] In some embodiments, the composition further comprises a pro-inflammatory polypeptide. In some embodiments, the pro-inflammatory polypeptide is a chemokine, cytokine. In some embodiments, the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL- 12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, and interferon. In some embodiments, the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, and interferon. [0205] In some embodiments, the myeloid cells are specifically targeted for delivery. Myeloid cells can be targeted using specialized biodegradable polymers, such as PLGA (poly(lactic-co- glycolic) acid and/or polyvinyl alcohol (PVA). In some embodiments, one or more compounds can be selectively incorporated in such polymeric structures to affect the myeloid cell function. In some embodiments, the targeting structures are multilayered, e.g., of one or more PLGA and one or more PVA layers. In some embodiments, the targeting structures are assembled in an order for a layered activity. In some embodiments, the targeted polymeric structures are organized in specific shaped components, such as labile structures that can adhere to a myeloid cell surface and deliver one or more components such as growth factors and cytokines, such as to maintain the myeloid cell in a microenvironment that endows a specific polarization. In some embodiments, the polymeric structures are such that they are not phagocytosed by the myeloid cell, but they can remain adhered on the surface. In some embodiments the one or more growth factors may be M1 polarization factors, such as a cytokine. In some embodiments the one or more growth factors may be an M2 polarization factor, such as a cytokine. In some embodiments, the one or more growth factors may be a macrophage activating cytokine, such as IFNγ. In some embodiments the polymeric structures are capable of sustained release of the one or more growth factors in an in vivo environment, such as in a solid tumor. [0206] In some embodiments, the recombinant nucleic acid comprises a sequence encoding a homeostatic regulator of inflammation. In some embodiments, the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA. In some embodiments, the sequence in the UTR is a sequence that binds to an RNA binding protein. In some embodiments, translation is inhibited or prevented upon binding of the RNA binding protein to the sequence in an untranslated region (UTR). In some embodiments, the sequence in the UTR comprises a consensus sequence of WWWU(AUUUA)UUUW, wherein W is A or U. In some embodiments, the recombinant nucleic acid is expressed on a bicistronic vector. [0207] In some embodiments, the target cell is a mammalian cell. In some embodiments, the target cell is a human cell. In some embodiments, the target cell comprises a cell infected with a pathogen. In some embodiments, the target cell is a cancer cell. In some embodiments, the target cell is a cancer cell that is a lymphocyte. In some embodiments, the target cell is a cancer cell that is an ovarian cancer cell. In some embodiments, the target cell is a cancer cell that is a breast cell. In some embodiments, the target cell is a cancer cell that is a pancreatic cell. In some embodiments, the target cell is a cancer cell that is a glioblastoma cell. [0208] In some embodiments, the recombinant nucleic acid is DNA. In some embodiments, the recombinant nucleic acid is RNA. In some embodiments, the recombinant nucleic acid is mRNA. In some embodiments, the recombinant nucleic acid is an unmodified mRNA. In some embodiments, the recombinant nucleic acid is a modified mRNA. In some embodiments, the recombinant nucleic acid is a circRNA. In some embodiments, the recombinant nucleic acid is a tRNA. In some embodiments, the recombinant nucleic acid is a microRNA. [0209] Also provided herein is a vector comprising a recombinant nucleic acid sequence encoding a CFP described herein. In some embodiments, the vector is viral vector. In some embodiments, the viral vector is retroviral vector or a lentiviral vector. In some embodiments, the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector is polycistronic. In some embodiments, each of the at least one nucleic acid sequence is operably linked to a separate promoter. In some embodiments, the vector further comprises one or more internal ribosome entry sites (IRESs). In some embodiments, the vector further comprises a 5’UTR and/or a 3’UTR flanking the at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector further comprises one or more regulatory regions. [0210] Also provided herein is a polypeptide encoded by the recombinant nucleic acid of a composition described herein. [0211] Also provided herein is a cell comprising a composition described herein, a vector described herein or a polypeptide described herein. In some embodiments, the cell is a phagocytic cell. In some embodiments, the cell is a stem cell derived cell, a myeloid cell, a macrophage, a dendritic cell, a lymphocyte, a mast cell, a monocyte, a neutrophil, a microglia, or an astrocyte. In some embodiments, the cell is an autologous cell. In some embodiments, the cell is an allogeneic cell. In some embodiments, the cell is an M1 cell. In some embodiments, the cell is an M2 cell. In some embodiments, the cell is an M1 macrophage cell. In some embodiments, the cell is an M2 macrophage cell. In some embodiments, the cell is an M1 myeloid cell. In some embodiments, the cell is an M2 myeloid cell. [0212] Also provided herein is a pharmaceutical composition comprising a composition described herein, such as a recombinant nucleic acid described herein, a vector described herein, a polypeptide described herein or a cell described herein; and a pharmaceutically acceptable excipient. [0213] In some embodiments, the pharmaceutical composition further comprises an additional therapeutic agent. In some embodiments, the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. In some embodiments, the pharmaceutically acceptable excipient comprises serum free media, a lipid, or a nanoparticle. [0214] Also provided herein is a method of treating a disease in a subject in need thereof comprising administering to the subject a pharmaceutical composition described herein. In some embodiments, the disease is cancer. In some embodiments, the cancer is a solid cancer. In some embodiments, the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer. In some embodiments, the cancer is a liquid cancer. In some embodiments, the liquid cancer is leukemia or a lymphoma. In some embodiments, the liquid cancer is a T cell lymphoma. In some embodiments, the disease is a T cell malignancy. [0215] In some embodiments, the method further comprises administering an additional therapeutic agent to the subject. In some embodiments, the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. [0216] In some embodiments, administering comprises infusing or injecting. In some embodiments, administering comprises administering directly to the solid cancer. In some embodiments, administering comprises a circRNA-based delivery procedure, anon-particle encapsulated mRNA-based delivery procedure, an mRNA-based delivery procedure, viral-based delivery procedure, particle-based delivery procedure, liposome-based delivery procedure, or an exosome-based delivery procedure. In some embodiments, a CD4+ T cell response or a CD8+ T cell response is elicited in the subject. [0217] Also provided herein is a method of preparing a cell, the method comprising contacting a cell with a composition described herein, a vector described herein or a polypeptide described herein. In some embodiments, contacting comprises transducing. In some embodiments, contacting comprises chemical transfection, electroporation, nucleofection, or viral infection or transduction. [0218] Also provided herein is a method of preparing a pharmaceutical composition comprising contacting a lipid to a composition described herein or a vector described herein. In some embodiments, contacting comprises forming a lipid nanoparticle. [0219] Also provided herein is a method of preparing a pharmaceutical composition comprising contacting an antibody to a composition described herein or the vector described herein. In some embodiments, contacting comprises forming a lipid nanoparticle. Definitions [0220] An “agent” can refer to any cell, small molecule chemical compound, antibody or fragment thereof, nucleic acid molecule, or polypeptide. [0221] An “alteration” or “change” can refer to an increase or decrease. For example, an alteration can be an increase or decrease of 1%, 2%, 3%, 4%, 5%, 10%, 20%, 30%, or by 40%, 50%, 60%, or even by as much as 70%, 75%, 80%, 90%, or 100%. For example, an alteration can be an increase or decrease of 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 30-fold, or by 40-fold, 50-fold, 60- fold, or even by as much as 70-fold, 75-fold, 80-fold, 90-fold, or 100-fold. [0222] An “antigen presenting cell” or “APC” as used herein includes professional antigen presenting cells (e.g., B lymphocytes, macrophages, monocytes, dendritic cells, Langerhans cells), as well as other antigen presenting cells (e.g., keratinocytes, endothelial cells, astrocytes, fibroblasts, oligodendrocytes, thymic epithelial cells, thyroid epithelial cells, glial cells (brain), pancreatic beta cells, and vascular endothelial cells). An APC can express Major Histocompatibility complex (MHC) molecules and can display antigens complexed with MHC on its surface which can be recognized by T cells and trigger T cell activation and an immune response. Professional antigen- presenting cells, notably dendritic cells, play a key role in stimulating naive T cells. Nonprofessional antigen-presenting cells, such as fibroblasts, may also contribute to this process. APCs can also cross-present peptide antigens by processing exogenous antigens and presenting the processed antigens on class I MHC molecules. Antigens that give rise to proteins that are recognized in association with class I MHC molecules are generally proteins that are produced within the cells, and these antigens are processed and associate with class I MHC molecules. [0223] A “biological sample” can refer to any tissue, cell, fluid, or other material derived from an organism. [0224] The term “epitope” can refer to any protein determinant, such as a sequence or structure or amino acid residues, capable of binding to an antibody or binding fragment thereof, a T cell receptor, and/or an antibody-like molecule. Epitopic determinants typically consist of chemically active surface groups of molecules such as amino acids or sugar side chains and generally have specific three dimensional structural characteristics as well as specific charge characteristics. A “T cell epitope” can refer to peptide or peptide-MHC complex recognized by a T cell receptor. [0225] Chimeric fusion proteins (CFPs) may be used to designate a chimeric receptor made via recombinant DNA technology, and is exclusively detailed in the specification. In some cases a CFP may be termed a phagocytic receptor fusion protein (PFP) and the terms can be used interchangeably. For the purpose of describing constructs that have one arm of the recombinant polynucleic acid a comprising a sequence encoding chimeric receptor for expression in a myeloid cell and another arm comprising a sequence encoding chimeric receptor for expression in a T cell, e.g., the bicistronic constructs, the chimeric receptor designed for expression in a T cell is termed CAR to differentiate from the chimeric protein designed for expression in a myeloid cell, which is termed CFP. For the purpose of discussing these dual constructs from bicistronic recombinant nucleic acids as described herein, a CAR construct may comprise a CD3z domain and a CFP construct may comprise a CD89 transmembrane domain. [0226] An engineered cell, such as an engineered myeloid cell, can refer to a cell that has at least one exogenous nucleic acid sequence in the cell, even if transiently expressed. Expressing an exogenous nucleic acid may be performed by various methods described elsewhere, and encompasses methods known in the art. The present disclosure relates to preparing and using engineered cells, for example, engineered myeloid cells, such as engineered phagocytic cells. The present disclosure relates to, inter alia, an engineered cell comprising an exogenous nucleic acid encoding, for example, a chimeric fusion protein (CFP). [0227] The term “immune response” includes, but is not limited to, T cell mediated, NK cell mediated and/or B cell mediated immune responses. These responses may be influenced by modulation of T cell costimulation and NK cell costimulation. Exemplary immune responses include T cell responses, e.g., cytokine production, and cellular cytotoxicity. In addition, immune responses include immune responses that are indirectly affected by NK cell activation, B cell activation and/or T cell activation, e.g., antibody production (humoral responses) and activation of cytokine responsive cells, e.g., macrophages. Immune responses include adaptive immune responses. The adaptive immune system can react to foreign molecular structures, such as antigens of an intruding organism. Unlike the innate immune system, the adaptive immune system is highly specific to a pathogen. Adaptive immunity can also provide long-lasting protection. Adaptive immune reactions include humoral immune reactions and cell-mediated immune reactions. In humoral immune reactions, antibodies secreted by B cells into bodily fluids bind to pathogen-derived antigens leading to elimination of the pathogen through a variety of mechanisms, e.g. complement-mediated lysis. In cell-mediated immune reactions, T cells capable of destroying other cells are activated. For example, if proteins associated with a disease are present in a cell, they can be fragmented proteolytically to peptides within the cell. Specific cell proteins can then attach themselves to the antigen or a peptide formed in this manner, and transport them to the surface of the cell, where they can be presented to molecular defense mechanisms, such as T cells. Cytotoxic T cells can recognize these antigens and kill cells that harbor these antigens. [0228] A “ligand” can refer to a molecule which is capable of binding or forming a complex with another molecule, such as a receptor. A ligand can include, but is not limited to, a protein, a glycoprotein, a carbohydrate, a lipoprotein, a hormone, a fatty acid, a phospholipid, or any component that binds to a receptor. In some embodiments, a receptor has a specific ligand. In some embodiments, a receptor may have promiscuous binding to a ligand, in which case it can bind to several ligands that share at least a similarity in structural configuration, charge distribution or any other physicochemical characteristic. A ligand may be a biomolecule. A ligand may be an abiotic material. For example, a ligand may be a negative charged particle that is a ligand for scavenger receptor MARCO. For example, a ligand may be TiO2, which is a ligand for the scavenger receptor SRA1. [0229] The term “major histocompatibility complex (MHC)”, “MHC molecule”, or “MHC protein” refers to a protein capable of binding an antigenic peptide and present the antigenic peptide to T lymphocytes. Such antigenic peptides can represent T cell epitopes. The human MHC is also called the HLA complex. Thus, the terms “human leukocyte antigen (HLA)”, “HLA molecule” or “HLA protein” are used interchangeably with the terms “major histocompatibility complex (MHC)”, “MHC molecule”, and “MHC protein”. HLA proteins can be classified as HLA class I or HLA class II. The structures of the proteins of the two HLA classes are very similar; however, they have very different functions. Class I HLA proteins are present on the surface of almost all cells of the body, including most tumor cells. Class I HLA proteins are loaded with antigens that usually originate from endogenous proteins or from pathogens present inside cells, and are then presented to naïve or cytotoxic T-lymphocytes (CTLs). HLA class II proteins are present on antigen presenting cells (APCs), including but not limited to dendritic cells, B cells, and macrophages. They mainly present peptides which are processed from external antigen sources, e.g. outside of cells, to helper T cells. [0230] In the HLA class II system, phagocytes such as macrophages and immature dendritic cells can take up entities by phagocytosis into phagosomes – though B cells exhibit the more general endocytosis into endosomes – which fuse with lysosomes whose acidic enzymes cleave the uptaken protein into many different peptides. Autophagy is another source of HLA class II peptides. The most studied subclass II HLA genes are: HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1, HLA-DRA, and HLA-DRB1. [0231] Presentation of peptides by HLA class II molecules to CD4+ helper T cells can lead to immune responses to foreign antigens. Once activated, CD4+ T cells can promote B cell differentiation and antibody production, as well as CD8+ T cell (CTL) responses. CD4+ T cells can also secrete cytokines and chemokines that activate and induce differentiation of other immune cells. HLA class II molecules are typically heterodimers of α-and β-chains that interact to form a peptide- binding groove that is more open than class I peptide-binding grooves. [0232] HLA alleles are typically expressed in codominant fashion. For example, each person carries 2 alleles of each of the 3 class I genes, (HLA-A, HLA-B and HLA-C) and so can express six different types of class II HLA. In the class II HLA locus, each person inherits a pair of HLA-DP genes (DPA1 and DPB1, which encode α and β chains), HLA-DQ (DQA1 and DQB1, for α and β chains), one gene HLA-DRα (DRA1), and one or more genes HLA-DRβ (DRB1 and DRB3, -4 or- 5). HLA‐DRB1, for example, has more than nearly 400 known alleles. That means that one heterozygous individual can inherit six or eight functioning class II HLA alleles: three or more from each parent. Thus, the HLA genes are highly polymorphic; many different alleles exist in the different individuals inside a population. Genes encoding HLA proteins have many possible variations, allowing each person’s immune system to react to a wide range of foreign invaders. Some HLA genes have hundreds of identified versions (alleles), each of which is given a particular number. In some embodiments, the class I HLA alleles are HLA-A*02:01, HLA-B*14:02, HLA- A*23:01, HLA-E*01:01 (non-classical). In some embodiments, class II HLA alleles are HLA- DRB*01:01, HLA-DRB*01:02, HLA-DRB*11:01, HLA-DRB*15:01, and HLA-DRB*07:01. [0233] A “myeloid cell” can refer broadly to cells of the myeloid lineage of the hematopoietic cell system, and can exclude, for example, the lymphocytic lineage. Myeloid cells comprise, for example, cells of the granulocyte lineage and monocyte lineages. Myeloid cells are differentiated from common progenitors derived from the hematopoietic stem cells in the bone marrow. Commitment to myeloid cell lineages may be governed by activation of distinct transcription factors, and accordingly myeloid cells may be characterized as cells having a level of plasticity, which may be described as the ability to further differentiate into terminal cell types based on extracellular and intracellular stimuli. Myeloid cells can be rapidly recruited into local tissues via various chemokine receptors on their surface. Myeloid cells are responsive to various cytokines and chemokines. [0234] A myeloid cell, for example, may be a cell that originates in the bone marrow from a hematopoietic stem cell under the influence of one or more cytokines and chemokines, such as G- CSF, GM-CSF, Flt3L, CCL2, VEGF and S100A8/9. In some embodiments, the myeloid cell is a precursor cell. In some embodiments, the myeloid cell may be a cell having characteristics of a common myeloid progenitor, or a granulocyte progenitor, a myeloblast cell, or a monocyte-dendritic cell progenitor or a combination thereof. A myeloid can include a granulocyte or a monocyte or a precursor cell thereof. A myeloid can include an immature granulocyte, an immature monocyte, an immature macrophage, an immature neutrophil, and an immature dendritic cell. A myeloid can include a monocyte or a pre-monocytic cell or a monocyte precursor. In some cases, a myeloid cell as used herein may refer to a monocyte having an M0 phenotype, an M1 phenotype or an M2 phenotype. A myeloid can include a dendritic cell (DC), a mature DC, a monocyte derived DC, a plasmacytoid DC, a pre-dendritic cell, or a precursor of a DC. A myeloid can include a neutrophil, which may be a mature neutrophil, a neutrophil precursor, or a polymorphonucleocyte (PMN). A myeloid can include a macrophage, a monocyte-derived macrophage, a tissue macrophage, a macrophage of an M0, an M1 or an M2 phenotype. A myeloid can include a tumor infiltrating monocyte (TIM). A myeloid can include a tumor associated monocyte (TAM). A myeloid can include a myeloid derived suppressor cell (MDSC). A myeloid can include a tissue resident macrophage. A myeloid can include a tumor associated DC (TADC). Accordingly, a myeloid cell may express one or more cell surface markers, for example, CD11b, CD14, CD15, CD16, CD38, CCR5, CD66, Lox-1, CD11c, CD64, CD68, CD163, CCR2, CCR5, HLA-DR, CD1c, CD83, CD141, CD209, MHC-II, CD123, CD303, CD304, a SIGLEC family protein and a CLEC family protein. In some cases, a myeloid cell may be characterized by a high or a low expression of one or more of cell surface markers, for example, CD11b, CD14, CD15, CD16, CD66, Lox-1, CD11c, CD64, CD68, CD163, CCR2, CCR5, HLA-DR, CD1c, CD83, CD141, CD209, MHC-II, CD123, CD303, CD304 or a combination thereof. [0235] “Phagocytosis” is used interchangeably with “engulfment” and can refer to a process by which a cell engulfs a particle, such as a cancer cell or an infected cell. This process can give rise to an internal compartment (phagosome) containing the particle. This process can be used to ingest and or remove a particle, such as a cancer cell or an infected cell from the body. A phagocytic receptor may be involved in the process of phagocytosis. The process of phagocytosis can be closely coupled with an immune response and antigen presentation. The processing of exogenous antigens follows their uptake into professional antigen presenting cells by some type of endocytic event. Phagocytosis can also facilitate antigen presentation. For example, antigens from phagocytosed cells or pathogens, including cancer antigens, can be processed and presented on the cell surface of APCs. [0236] A “polypeptide” can refer to a molecule containing amino acids linked together via a peptide bond, such as a glycoprotein, a lipoprotein, a cellular protein or a membrane protein. A polypeptide may comprise one or more subunits of a protein. A polypeptide may be encoded by a recombinant nucleic acid. In some embodiments, polypeptide may comprise more than one peptide sequence in a single amino acid chain, which may be separated by a spacer, a linker or peptide cleavage sequence. A polypeptide may be a fused polypeptide. A polypeptide may comprise one or more domains, modules or moieties. [0237] A “receptor” can refer to a chemical structure composed of a polypeptide, which transduces a signal, such as a polypeptide that transduces an extracellular signal to a cell. A receptor can serve to transmit information in a cell, a cell formation or an organism. A receptor comprises at least one receptor unit and can contain two or more receptor units, where each receptor unit comprises a protein molecule, e.g., a glycoprotein molecule. A receptor can contain a structure that binds to a ligand and can form a complex with the ligand. Signaling information can be transmitted by a conformational change of the receptor following binding with the ligand on the surface of a cell. [0238] The term “antibody” refers to a class of proteins that are generally known as immunoglobulins, including, but not limited to IgGl, IgG2, IgG3, and IgG4), IgA (including IgAl and IgA2), IgD, IgE, IgM, and IgY, The term “antibody” includes, but is not limited to, full length antibodies, single-chain antibodies, single domain antibodies (sdAb) and antigen-binding fragments thereof. Antigen-binding antibody fragments include, but are not limited to, Fab, Fab’ and F(ab’)2, Fd (consisting of VH and CH1), single-chain variable fragment (scFv), single-chain antibodies, disulfide-linked variable fragment (dsFv) and fragments comprising a VL and/or a VH domain. Antibodies can be from any animal origin. Antigen-binding antibody fragments, including single- chain antibodies, can comprise variable region(s) alone or in combination with tone or more of a hinge region, a CH1 domain, a CH2 domain, and a CH3 domain. Also included are any combinations of variable region(s) and hinge region, CH1, CH2, and CH3 domains. Antibodies can be monoclonal, polyclonal, chimeric, humanized, and human monoclonal and polyclonal antibodies which, e.g., specifically bind an HLA-associated polypeptide or an HLA-peptide complex. [0239] The term “recombinant nucleic acid” refers a nucleic acid prepared, expressed, created or isolated by recombinant means. A recombinant nucleic acid can contain a nucleotide sequence that is not naturally occurring. A recombinant nucleic acid may be synthesized in the laboratory. A recombinant nucleic acid may be prepared by using recombinant DNA technology, for example, enzymatic modification of DNA, such as enzymatic restriction digestion, ligation, and DNA cloning. A recombinant nucleic acid can be DNA, RNA, analogues thereof, or a combination thereof. A recombinant DNA may be transcribed ex vivo or in vitro, such as to generate a messenger RNA (mRNA). A recombinant mRNA may be isolated, purified and used to transfect a cell. A recombinant nucleic acid may encode a protein or a polypeptide. [0240] The process of introducing or incorporating a nucleic acid into a cell can be via transformation, transfection or transduction. Transformation is the process of uptake of foreign nucleic acid by a bacterial cell. This process is adapted for propagation of plasmid DNA, protein production, and other applications. Transformation introduces recombinant plasmid DNA into competent bacterial cells that take up extracellular DNA from the environment. Some bacterial species are naturally competent under certain environmental conditions, but competence is artificially induced in a laboratory setting. Transfection is the introduction of small molecules such as DNA, RNA, or antibodies into eukaryotic cells. Transfection may also refer to the introduction of bacteriophage into bacterial cells. ‘Transduction’ is mostly used to describe the introduction of recombinant viral vector particles into target cells, while ‘infection’ refers to natural infections of humans or animals with wild-type viruses. [0241] The term “vector”, can refer to a nucleic acid molecule capable of autonomous replication in a host cell, and which allow for cloning of nucleic acid molecules. As known to those skilled in the art, a vector includes, but is not limited to, a plasmid, cosmid, phagemid, viral vectors, phage vectors, yeast vectors, mammalian vectors and the like. For example, a vector for exogenous gene transformation may be a plasmid. In certain embodiments, a vector comprises a nucleic acid sequence containing an origin of replication and other elements necessary for replication and/or maintenance of the nucleic acid sequence in a host cell. In some embodiments, a vector or a plasmid provided herein is an expression vector. Expression vectors are capable of directing the expression of genes and/or nucleic acid sequence to which they are operatively linked. In some embodiments, an expression vector or plasmid is in the form of circular double stranded DNA molecules. A vector or plasmid may or may not be integrated into the genome of a host cell. In some embodiments, nucleic acid sequences of a plasmid are not integrated in a genome or chromosome of the host cell after introduction. For example, the plasmid may comprise elements for transient expression or stable expression of the nucleic acid sequences, e.g. genes or open reading frames harbored by the plasmid, in a host cell. In some embodiments, a vector is a transient expression vector. In some embodiments, a vector is a stably expressed vector that replicates autonomously in a host cell. In some embodiments, nucleic acid sequences of a plasmid are integrated into a genome or chromosome of a host cell upon introduction into the host cell. Expression vectors that can be used in the methods as disclosed herein include, but are not limited to, plasmids, episomes, bacterial artificial chromosomes, yeast artificial chromosomes, bacteriophages or viral vectors. A vector can be a DNA or RNA vector. In some embodiments, a vector provide herein is a RNA vector that is capable of integrating into a host cell’s genome upon introduction into the host cell (e.g., via reverse transcription), for example, a retroviral vector or a lentiviral vector. Other forms of expression vectors known by those skilled in the art which serve the equivalent functions can also be used, for example, self-replicating extrachromosomal vectors or vectors capable of integrating into a host genome. Exemplary vectors are those capable of autonomous replication and/or expression of nucleic acids to which they are linked. [0242] The terms “spacer” or “linker” as used in reference to a fusion protein refers to a peptide sequence that joins two other peptide sequences of the fusion protein. In some embodiments, a linker or spacer has no specific biological activity other than to join or to preserve some minimum distance or other spatial relationship between the proteins or RNA sequences. In some embodiments, the constituent amino acids of a spacer can be selected to influence some property of the molecule such as the folding, flexibility, net charge, or hydrophobicity of the molecule. Suitable linkers for use in an embodiment of the present disclosure are well known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers. In some embodiments, a linker is used to separate two or more polypeptides, e.g. two antigenic peptides by a distance sufficient to ensure that each antigenic peptide properly folds. Exemplary peptide linker sequences adopt a flexible extended conformation and do not exhibit a propensity for developing an ordered secondary structure. Amino acids in flexible linker protein region may include Gly, Asn and Ser, or any permutation of amino acid sequences containing Gly, Asn and Ser. Other near neutral amino acids, such as Thr and Ala, also can be used in the linker sequence. [0243] The terms “treat,” “treated,” “treating,” “treatment,” and the like are meant to refer to reducing, preventing, or ameliorating a disorder and/or symptoms associated therewith (e.g., a neoplasia or tumor or infectious agent or an autoimmune disease). “Treating” can refer to administration of the therapy to a subject after the onset, or suspected onset, of a disease (e.g., cancer or infection by an infectious agent or an autoimmune disease). “Treating” includes the concepts of “alleviating”, which can refer to lessening the frequency of occurrence or recurrence, or the severity, of any symptoms or other ill effects related to the disease and/or the side effects associated with therapy. The term “treating” also encompasses the concept of “managing” which refers to reducing the severity of a disease or disorder in a patient, e.g., extending the life or prolonging the survivability of a patient with the disease, or delaying its recurrence, e.g., lengthening the period of remission in a patient who had suffered from the disease. It is appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition, or symptoms associated therewith be completely eliminated. The term “prevent”, “preventing”, “prevention” and their grammatical equivalents as used herein, can refer to avoiding or delaying the onset of symptoms associated with a disease or condition in a subject that has not developed such symptoms at the time the administering of an agent or compound commences. In certain embodiments, treating a subject or a patient as described herein comprises administering a therapeutic composition, such as a drug, a metabolite, a preventive component, a nucleic acid, a peptide, or a protein that encodes or otherwise forms a drug, a metabolite or a preventive component. In some embodiments, treating comprises administering a cell or a population of cells to a subject in need thereof. In some embodiments, treating comprises administering to the subject one or more of engineered cells described herein, e.g. one or more engineered myeloid cells, such as phagocytic cells. Treating comprises treating a disease or a condition or a syndrome, which may be a pathological disease, condition or syndrome, or a latent disease, condition or syndrome. In some cases, treating, as used herein may comprise administering a therapeutic vaccine. In some embodiments, the engineered phagocytic cell is administered to a patient or a subject. In some embodiments, a cell administered to a human subject results in reduced immunogenicity. For example, an engineered phagocytic cell may lead to no or reduced graft versus host disease (GVHD) or fratricide effect. In some embodiments, an engineered cell administered to a human subject is immunocompatible to the subject (i.e. having a matching HLA subtype that is naturally expressed in the subject). Subject specific HLA alleles or HLA genotype of a subject can be determined by any method known in the art. In exemplary embodiments, the methods include determining polymorphic gene types that can comprise generating an alignment of reads extracted from a sequencing data set to a gene reference set comprising allele variants of the polymorphic gene, determining a first posterior probability or a posterior probability derived score for each allele variant in the alignment, identifying the allele variant with a maximum first posterior probability or posterior probability derived score as a first allele variant, identifying one or more overlapping reads that aligned with the first allele variant and one or more other allele variants, determining a second posterior probability or posterior probability derived score for the one or more other allele variants using a weighting factor, identifying a second allele variant by selecting the allele variant with a maximum second posterior probability or posterior probability derived score, the first and second allele variant defining the gene type for the polymorphic gene, and providing an output of the first and second allele variant. [0244] A “fragment” can refer to a portion of a protein or nucleic acid. In some embodiments, a fragment retains at least 50%, 75%, or 80%, or 90%, 95%, or even 99% of the biological activity of a reference protein or nucleic acid. [0245] The terms “isolated,” “purified”, “biologically pure” and their grammatical equivalents refer to material that is free to varying degrees from components which normally accompany it as found in its native state. “Isolate” denotes a degree of separation from original source or surroundings. “Purify” denotes a degree of separation that is higher than isolation. A “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide of the present disclosure is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized. Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography. The term “purified” can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. For a protein that can be subjected to modifications, for example, phosphorylation or glycosylation, different modifications can give rise to different isolated proteins, which can be separately purified. [0246] The terms “neoplasia” or “cancer” refers to any disease that is caused by or results in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both. Glioblastoma is one non-limiting example of a neoplasia or cancer. The terms “cancer” or “tumor” or “hyperproliferative disorder” refer to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Cancer cells are often in the form of a tumor, but such cells can exist alone within an animal, or can be a non- tumorigenic cancer cell, such as a leukemia cell. [0247] The term “vaccine” is to be understood as meaning a composition for generating immunity for the prophylaxis and/or treatment of diseases (e.g., neoplasia/tumor/infectious agents/autoimmune diseases). Accordingly, vaccines as used herein are medicaments which comprise recombinant nucleic acids, or cells comprising and expressing a recombinant nucleic acid and are intended to be used in humans or animals for generating specific defense and protective substance by vaccination. A “vaccine composition” can include a pharmaceutically acceptable excipient, carrier or diluent. Aspects of the present disclosure relate to use of the technology in preparing a phagocytic cell-based vaccine. [0248] The term “pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans. A “pharmaceutically acceptable excipient, carrier or diluent” refers to an excipient, carrier or diluent that can be administered to a subject, together with an agent, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent. [0249] Nucleic acid molecules useful in the methods of the disclosure include, but are not limited to, any nucleic acid molecule with activity or that encodes a polypeptide. Polynucleotides having substantial identity to an endogenous sequence are typically capable of hybridizing with at least one strand of a double-stranded nucleic acid molecule. “Hybridize” refers to when nucleic acid molecules pair to form a double-stranded molecule between complementary polynucleotide sequences, or portions thereof, under various conditions of stringency. (See, e.g., Wahl, G. M. and S. L. Berger (1987) Methods Enzymol. 152:399; Kimmel, A. R. (1987) Methods Enzymol. 152:507). For example, stringent salt concentration can ordinarily be less than about 750 mM NaCl and 75 mM trisodium citrate, less than about 500 mM NaCl and 50 mM trisodium citrate, or less than about 250 mM NaCl and 25 mM trisodium citrate. Low stringency hybridization can be obtained in the absence of organic solvent, e.g., formamide, while high stringency hybridization can be obtained in the presence of at least about 35% formamide, or at least about 50% formamide. Stringent temperature conditions can ordinarily include temperatures of at least about 30° C, at least about 37°C, or at least about 42°C. Varying additional parameters, such as hybridization time, the concentration of detergent, e.g., sodium dodecyl sulfate (SDS), and the inclusion or exclusion of carrier DNA, are well known to those skilled in the art. Various levels of stringency are accomplished by combining these various conditions as needed. In an exemplary embodiment, hybridization can occur at 30° C in 750 mM NaCl, 75 mM trisodium citrate, and 1% SDS. In another exemplary embodiment, hybridization can occur at 37° C in 500 mM NaCl, 50 mM trisodium citrate, 1% SDS, 35% formamide, and 100 µg/ml denatured salmon sperm DNA (ssDNA). In another exemplary embodiment, hybridization can occur at 42° C in 250 mM NaCl, 25 mM trisodium citrate, 1% SDS, 50% formamide, and 200 µg/ml ssDNA. Useful variations on these conditions will be readily apparent to those skilled in the art. For most applications, washing steps that follow hybridization can also vary in stringency. Wash stringency conditions can be defined by salt concentration and by temperature. As above, wash stringency can be increased by decreasing salt concentration or by increasing temperature. For example, stringent salt concentration for the wash steps can be less than about 30 mM NaCl and 3 mM trisodium citrate, or less than about 15 mM NaCl and 1.5 mM trisodium citrate. Stringent temperature conditions for the wash steps can include a temperature of at least about 25°C, of at least about 42°C, or at least about 68°C. In exemplary embodiments, wash steps can occur at 25° C in 30 mM NaCl, 3 mM trisodium citrate, and 0.1% SDS. In other exemplary embodiments, wash steps can occur at 42° C in 15 mM NaCl, 1.5 mM trisodium citrate, and 0.1% SDS. In another exemplary embodiment, wash steps can occur at 68° C in 15 mM NaC1, 1.5 mM trisodium citrate, and 0.1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art. Hybridization techniques are well known to those skilled in the art and are described, for example, in Benton and Davis (Science 196:180, 1977); Grunstein and Hogness (Proc. Natl. Acad. Sci., USA 72:3961, 1975); Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, New York, 2001); Berger and Kimmel (Guide to Molecular Cloning Techniques, 1987, Academic Press, New York); and Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York. [0250] “Substantially identical” refers to a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein). Such a sequence can be at least 60%, 80% or 85%, 90%, 95%, 96%, 97%, 98%, or even 99% or more identical at the amino acid level or nucleic acid to the sequence used for comparison. Sequence identity is typically measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. In an exemplary approach to determining the degree of identity, a BLAST program can be used, with a probability score between e-3 and e-m° indicating a closely related sequence. A “reference” is a standard of comparison. It will be understood that the numbering of the specific positions or residues in the respective sequences depends on the particular protein and numbering scheme used. Numbering might be different, e.g., in precursors of a mature protein and the mature protein itself, and differences in sequences from species to species may affect numbering. One of skill in the art will be able to identify the respective residue in any homologous protein and in the respective encoding nucleic acid by methods well known in the art, e.g., by sequence alignment to a reference sequence and determination of homologous residues. [0251] The term “subject” or “patient” refers to an organism, such as an animal (e.g., a human) which is the object of treatment, observation, or experiment. By way of example only, a subject includes, but is not limited to, a mammal, including, but not limited to, a human or a non-human mammal, such as a non-human primate, murine, bovine, equine, canine, ovine, or feline. [0252] The term “therapeutic effect” refers to some extent of relief of one or more of the symptoms of a disorder (e.g., a neoplasia, tumor, or infection by an infectious agent or an autoimmune disease) or its associated pathology. “Therapeutically effective amount” as used herein refers to an amount of an agent which is effective, upon single or multiple dose administration to the cell or subject, in prolonging the survivability of the patient with such a disorder, reducing one or more signs or symptoms of the disorder, preventing or delaying, and the like beyond that expected in the absence of such treatment. “Therapeutically effective amount” is intended to qualify the amount required to achieve a therapeutic effect. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the “therapeutically effective amount” (e.g., ED50) of the pharmaceutical composition required. ENGINEERED MYELOD CELLS “TARGETED” TO ATTACK DISEASED CELLS [0253] The present disclosure involves compositions and methods for preparing targeted killer myeloid cells; by leveraging the innate functional role in immune defense, ranging from properties related to detecting foreign bodies, particles, diseased cells, cellular debris, inflammatory signal, chemoattract; activating endogenous DAMP and PAMP signaling pathways; trigger myelopoiesis, extravasation; chemotaxis; phagocytes; pinocytosis; recruitment; engulfment; scavenging; activating intracellular oxidative burst and lysis or killing of pathogens, detecting, engulfing and killing diseased or damaged cells; removing unwanted cellular, tissue or acellular debris in vivo; antigen presentation and role in activating innate immunity; activating and modulating an immune response cascade; activating T cell repertoire; autophagy; inflammatory and non-inflammatory apoptosis; pyroptosis, immune editing to response to stress and restoration of tissue homeostasis. In one aspect, provided herein are methods and compositions to augment one or more functions of a myeloid cell for use in a therapeutic application, the one or more functions may be one or more of: detecting foreign bodies, particles, diseased cells, cellular debris, inflammatory signal, chemoattract; activating endogenous DAMP and PAMP signaling pathways; trigger myelopoiesis, extravasation; chemotaxis; phagocytosis; pinocytosis; recruitment; trogocytosis; engulfment; scavenging; activating intracellular oxidative burst and intracellular lysis or killing of pathogens, detecting, engulfing and killing diseased or damaged cells; removing unwanted cellular, tissue or acellular debris in vivo; antigen presentation and role in activating innate immunity; activating and modulating an immune response cascade; activating T cell repertoire; autophagy; inflammatory and non-inflammatory apoptosis; pyroptosis, immune editing to response to stress and restoration of tissue homeostasis. In one embodiment, the compositions and methods are also directed to augmenting the targeting, and killing function of certain myeloid cells, by genetic modification of these cells. The compositions and methods described herein are directed to creating engineered myeloid cells, wherein the engineered myeloid cells comprise at least one genetic modification, and can be directed to recognize and induce effector functions against a pathogen, a diseased cell, such as a tumor or cancer cell, such that the engineered myeloid cell is capable of recognizing, targeting, phagocytosing, killing and/or eliminating the pathogen or the diseased cell or the cancer cell, and additionally, may activate a specific immune response cascade following the phagocytosis, killing and/or eliminating the pathogen or the diseased cell. [0254] Myeloid cells appear to be the most abundant cells in a tumor (FIG. 1B). Myeloid cells are also capable of recognizing a tumor cell over a healthy normal cell of the body and mount an immune response to a tumor cell of the body. As sentinels of innate immune response, myeloid cells are able to sense non-self or aberrant cell types and clear them via a process called phagocytosis. This can be directed to a therapeutic advantage in driving myeloid cell mediated phagocytosis and lysis of tumor cells. However, these naturally occurring tumor-infiltrating myeloid cells (TIMs) may be subjected to influence of the tumor microenvironment (TME). TIMs constitute a heterogeneous population of cells. Many TIMs originate from circulating monocytes and granulocytes, which in turn stem from bone marrow-derived hematopoietic stem cells. However, in the presence of persistent stimulation by tumor-derived factors the monocyte and granulocyte progenitors divert from their intrinsic pathway of terminal differentiation into mature macrophages, DCs or granulocytes, and may become tumor promoting myeloid cell types. Differentiation into pathological, alternatively activated immature myeloid cells is favored. These immature myeloid cells include tumor-associated DCs (TADCs), tumor-associated neutrophils (TANs), myeloid- derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs). Alternative to this emergency myelopoiesis, TAMs may also originate from tissue-resident macrophages, which in turn can be of embryonic or monocytic origin. These tissue-resident macrophages undergo changes in phenotype and function during carcinogenesis, and proliferation may help to maintain TAMs derived from tissue-resident macrophages. A tumor microenvironment may drive a tumor infiltrating myeloid cell to become myeloid derived suppressor cells and acquire the ability to suppress T cells. As a result, innovative methods are necessary to create therapeutically effective TIMs that can infiltrate a tumor, and can target tumor cells for phagocytic uptake and killing. [0255] In one aspect, provided herein are engineered myeloid cells that are capable of targeting specific target cells, for example, tumor cells or pathogenic cells. In some embodiments, engineered myeloid cells provided herein are potent in infiltrating, targeting, and killing tumor cells. An engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which encodes one or more proteins that help target the phagocytic cell to a target cell, for example a tumor cell or a cancer cell. In one embodiment, the engineered myeloid cell is capable of readily infiltrating a tumor. In one embodiment, the engineered myeloid cell has high specificity for the target cell, with none or negligible cross-reactivity to a non-tumor, non-diseased cell of the subject while in circulation. In one embodiment, the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which will help the cell to overcome/bypass the TME influence and mount a potent anti-tumor response. In one embodiment, the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which augments phagocytosis of the target cell. In another embodiment, the engineered myeloid/phagocytic cell described herein is designed to comprise a recombinant nucleic acid, which augments reduce or eliminate trogocytosis and/or enhance phagocytic lysis or of the target cell. [0256] Accordingly, in some embodiments, the compositions disclosed herein comprise a myeloid cell, comprising a recombinant nucleic acid encoding a chimeric receptor fusion protein (CFP), for example, a phagocytic receptor (PR) fusion protein (PFP). The recombinant nucleic acid can comprise a sequence encoding a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising a PR intracellular signaling domain; and an extracellular antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular antigen binding domain are operatively linked; wherein the PR intracellular signaling domain is derived from a receptor with a signal transduction domain. The recombinant nucleic acid further encodes for one or more polypeptides that constitute one or more plasma membrane receptors that helps engage the phagocytic cell to the target cell, and enhance its phagocytic activity. [0257] In some embodiments, the myeloid cell described herein comprises one or more recombinant proteins comprising a chimeric receptor, wherein the chimeric receptor is capable of responding to a first phagocytic signal directed to a target cell, which may be a diseased cell, a tumor cell or a pathogen, and a second signal, which is an inflammatory signal, that augments the phagocytic and killing response to target initiated by the first signal. PHAGOCYTIC CELLS, MYELOID CELLS, ENGINEERED MYELOID CELLS [0258] Provided herein are methods and compositions for immunotherapy, comprising ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a specific target, which can be a specific cell type or class of cells in a patient or a subject. In some embodiments, the subject is a patient having a disease. The terms subject and patient may often be used interchangeably in this section. In some embodiments, the phagocytic cell is derived from the subject having a disease, wherein the disease is, for example, cancer. The autologous cells from the subject may be modified in vitro and administered into the cell, where the modified phagocytic cell is redesigned to specifically attack and kill the cancer cells in the subject. [0259] In some embodiments, the subject has a disease that is not a cancer. [0260] In some embodiments, the subject has a disease that is an infection. In some embodiments, the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards an infection, for example an infected cell within the subject. [0261] In some embodiments, the subject has a disease that is a viral, a bacterial, a fungal or a protozoal infection. In some embodiments, the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a virus infected cell, a bacteria infected cell, a fungus infected cell or a protozoa infected cell inside the infected subject. In some embodiments the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a virus, a bacteria, a fungus or any pathogen in a subject, such that the virus, the bacteria, the fungus or the pathogen in a subject is phagocytosed, and/or killed. In some embodiments the methods and compositions for immunotherapy provided herein are for ‘improving’ or ‘modifying’ or ‘engineering’ a phagocytic cell and targeting it towards a viral antigen, a bacterial antigen, a fungal antigen or an antigen of a pathogen in a subject, such that there is at least one improved immune response within the subject to the virus, the bacteria, the fungus or the pathogen in the subject. [0262] In some embodiments, the myeloid cells, such as phagocytic cells, are allogeneic In some embodiments, the methods and compositions for immunotherapy provided herein comprises obtaining myeloid cells, such as phagocytic cells, derived from an allogeneic source. The myeloid cells, such as phagocytic cells, can thereafter be modified or engineered and introduced into a diseased subject, such that the modified or engineered cells from the allogeneic source are capable of attacking a diseased cell of the subject, phagocytose the diseased cell and/or kill the diseased cell directly or indirectly, or improve at least one immune response of the subject to the disease. In some embodiments, the allogeneic source is a human. In some embodiments, the allogeneic source is a healthy human. [0263] Phagocytes are the natural sentinels of the immune system and form the first line of defense in the body. They engulf a pathogen, a pathogen infected cell a foreign body or a cancerous cell and remove it from the body. Most potential pathogens are rapidly neutralized by this system before they can cause, for example, a noticeable infection. This can involve receptor-mediated uptake through the clathrin coated pit system, pinocytosis, particularly macropinocytosis as a consequence of membrane ruffling and phagocytosis. The phagocytes therefore can be activated by a variety of non-self (and self) elements and exhibit a level of plasticity in recognition of their “targets”. [0264] Mononuclear phagocytic system (MPS), comprised of monocytes, macrophages, and dendritic cells, is essential in tissue homeostasis and in determining the balance of an immune response through its role in antigen presentation. The MPS is a cell lineage which originates from bone marrow progenitor cells and gives rise to blood monocytes, tissue macrophages and dendritic cells. Thus, the process of generating a macrophage from the MPS begins with a promonocyte in the BM which undergoes a differentiation process into a monocyte that is ready to enter the systemic circulation. After a short period (<48h) in the circulation, these newly formed monocytes rapidly infiltrate into peripheral tissues where a majority of them differentiate into macrophages or dendritic cells (DC). Anti-microbe phagocytosis clears and degrades disease-causing microbes, induces pro- inflammatory signaling through cytokine and chemokine secretion, and recruits immune cells to mount an effective inflammatory response. This type of phagocytosis is often referred to as "inflammatory phagocytosis" (or "immunogenic phagocytosis"). However, in some instances, such as with certain persistent infections, anti-inflammatory responses may follow microbial uptake. Anti- microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells. Phagocytosis of damaged, apoptotic cells or cell is typically a non-inflammatory (also referred to as a "nonimmunogenic") process. Transformed or malignant cells (self-cells), and cells are phagocytosed and apoptotic cells are removed promptly without causing damage to the surrounding tissues or inducing a pro-inflammatory immune response. This type of apoptotic cell clearance is non-inflammatory and include release of "find me" signals from apoptotic cells to recruit phagocytes to the location of apoptotic cells; accompanied by "eat me" signals exposed on the surface of apoptotic cells are bound by phagocytes via specific receptors; cytoskeletal rearrangement to engulf the apoptotic cell; followed by the ingested apoptotic cell is digested and specific phagocytic responses are elicited (e.g., secretion of anti-inflammatory cytokines). [0265] Phagocytosis, the cellular uptake of particulates, e.g. particles >0.5 µm within a plasma- membrane envelope, is closely related to and partly overlaps the endocytosis of soluble ligands by fluid-phase macropinocytic and receptor pathways. Variants associated with the uptake of apoptotic cells, also known as efferocytosis, and that of necrotic cells arising from infection and inflammation (necroptosis and pyroptosis). The uptake of exogenous particles (heterophagy) has features in common with autophagy, an endogenous process of sequestration and lysosomal disposal of damaged intracellular organelles There is a spectrum of uptake mechanisms depending on the particle size, multiplicity of receptor-ligand interactions, and involvement of the cytoskeleton. Once internalized, the phagosome vacuole can fuse selectively with primary lysosomes, or the product of the endoplasmic reticulum (ER) and Golgi complex, to form a secondary phagolysosome (Russell, D.G. (2011). Immunol. Rev. 240, 252–268). This pathway is dynamic in that it undergoes fusion and fission with endocytic and secretory vesicles macrophages, DCs, osteoclasts, and eosinophils. Anti- microbe phagocytosis clears and degrades disease-causing microbes, induces pro-inflammatory signaling through cytokine and chemokine secretion, and recruits immune cells to mount an effective inflammatory response. This type of phagocytosis is often referred to as "inflammatory phagocytosis" (or "immunogenic phagocytosis"). However, in some instances, such as with certain persistent infections, anti-inflammatory responses may follow microbial uptake. Anti-microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells. Phagocytosis of damaged, self-derived apoptotic cells or cell debris (e.g., efferocytosis), in contrast, is typically a non-inflammatory (also referred to as a "nonimmunogenic") process. Billions of damaged, dying, and unwanted cells undergo apoptosis each day. Unwanted cells include, for example, excess cells generated during development, senescent cells, infected cells (intracellular bacteria or viruses), transformed or malignant cells, and cells irreversibly damaged by cytotoxic agents. [0266] The bone marrow is the source of circulating neutrophils and monocytes that will replace selected tissue-resident macrophages and amplify tissue myeloid populations during inflammation and infection. After phagocytosis, newly recruited monocytes and tissue macrophages secrete their products by generating them from pre-existing phospholipids and arachidonates in the plasma membrane and by releasing radicals generated by activation of a respiratory burst or induction of inducible nitric oxide synthesis; apart from being achieved by synthesis of the low-molecular-weight products (arachidonate metabolites, superoxide anions, and nitric oxide) generated as above, secretion induced by phagocytosis in macrophages is mainly achieved by new synthesis of RNA and changes in pH, resulting in progressive acidification. [0267] In some embodiments, phagocytes provided herein are monocytes or cells of the monocyte lineage. [0268] In some embodiments, myeloid cells are phagocytic macrophages are MARCO+ SignR1+ and are found in the outer marginal zone rapidly clear capsulated bacteria. Similar CD169+ F4/80_ macrophages line the subcapsular sinus in lymph nodes and have been implicated in virus infection. It was noted that endothelial macrophages, including Kupffer cells in the liver, clear microbial and antigenic ligands from blood and lymph nodes to provide a sinusoidal immune function comparable to but distinct from mucosal immunity. Not all tissue macrophages are constitutively phagocytic, even though they still express typical macrophage markers. In the marginal zone of the rodent spleen, metallophilic macrophages, which lack F4/80, strongly express CD169, sialic acid-binding immunoglobulin (Ig)-like lectin 1 (SIGLEC1 [sialoadhesin]), but are poorly phagocytic. Non- professional phagocytes include epithelial cells, and fibroblasts. Fibroblasts are ‘‘working-class phagocytes” that clear apoptotic debris by using integrins other than CD11b-CD18 through adhesion molecules ICAM and vitronectin receptors. Astrocytes have also been reported to engulf, even if not efficiently degrade, apoptotic corpses. Plasma-membrane receptors relevant to phagocytosis can be opsonic, FcRs (activating or inhibitory) for mainly the conserved domain of IgG antibodies, and complement receptors, such as CR3 for iC3b deposited by classical (IgM or IgG) or alternative lectin pathways of complement activation. CR3 can also mediate recognition in the absence of opsonins, perhaps by depositing macrophage-derived complement. Anti-microbe phagocytosis is commonly performed by professional phagocytes of the myeloid lineage, such as immature dendritic cells (DCs) and macrophages and by tissue-resident immune cells. [0269] In some embodiments, for the purpose of the instant cellular engineering program disclosed herein, cells that are used for engineering for use in immunotherapy are potently phagocytic. [0270] In some embodiments, for the purpose of the instant cellular engineering program disclosed herein, cells that are used for engineering for use in immunotherapy are obtained from whole blood, peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue. [0271] In some embodiments, cells that are used for engineering for use in immunotherapy are obtained from peripheral blood. [0272] Among the liver MPS, a variety of structural and functional distinctions have been characterized, both stimulatory and inhibitory with respect to the purpose of generation of cells for cancer immunotherapy. Table 1 - Exemplary phenotypic characteristics of liver monocytes, macrophages and DCs
Figure imgf000065_0001
[0273] In some embodiments the myeloid cells that are engineered for use in immunotherapy in the instant application comprise myeloid cells selected from the group consisting of macrophages, dendritic cells, mast cells, monocytes, neutrophils, microglia, and astrocytes. [0274] In some embodiments the myeloid cells that are engineered for use in immunotherapy are phagocytic cells. In some embodiments, the phagocytic cells are monocytes. [0275] In some embodiments, the myeloid cells that are engineered for use in immunotherapy in the instant application are monocytes, monocyte derived macrophages, and/or dendritic cells. [0276] In some embodiments the myeloid cells that are engineered for use in immunotherapy in the instant application are monocytes or macrophages. [0277] In some embodiments the cells that myeloid cells obtained from the peripheral blood. [0278] In some embodiments, the myeloid cells are selected by selection marker CD14+CD16low. In some embodiments the myeloid cells are selected via elutriation. [0279] In some embodiments, the myeloid cells are isolated from leukapheresis column of the subject. In some embodiments the subject is the same subject who is administered the pharmaceutical composition comprising engineered cells. [0280] In some embodiments the subject is not the same subject who is administered the pharmaceutical composition comprising engineered cells. [0281] In some embodiments, the leukapheresis is performed on the same subject once a week to collect more myeloid cells. In some embodiments, the leukapheresis is performed on the same subject more than once in a span of 8-10 days to collect more myeloid cells. In some embodiments, the leukapheresis is performed on the same subject more than twice in a span of one month to collect more myeloid cells. [0282] In some embodiments, myeloid cells are isolated from a leukapheresis sample or a peripheral blood sample. In some embodiments, the myeloid cell is a progenitor cell. In some embodiments, the myeloid cell is a monocyte precursor cell. In some embodiments, a myeloid cell described herein is not differentiated into a terminal cell and do not exhibit a terminal cell phenotype, such as tissue macrophages. In some embodiments, the myeloid cells comprise CD14+ cells. In some embodiments, the myeloid cells do not express CD16. In some embodiments the myeloid cells express low amounts of CD16. In some embodiments, the myeloid cells are pre- selected for the purpose of engineering from a biological sample, such as peripheral blood or an apheresis sample by selection of CD14+ cells. In some embodiments, the selection is performed without contacting with or engaging with the myeloid cell to be selected. In some embodiments, the myeloid cells are selected prior to engineering from a biological sample by sorting, for example a flow cytometry based cell sorter (FACS). In some embodiments, the myeloid cells expressing CD16 are captured by an antibody and the remaining myeloid cells were collected and used for engineering. In some embodiments, one or more other cell surface molecules are targeted for capturing in the negative selection process in addition to CD16, in order to obtain the myeloid cells, for example CD3, CD8, CD11c, CD40, or CD206. CHIMERIC FUSION PROTEINS FOR EXPRESSION IN ENGINEERED MYELOID CELLS [0283] In one aspect, provided herein are myeloid cells comprising at least one exogenous recombinant nucleic acid that encodes for a fusion protein. The fusion protein may be a chimeric protein comprising at least a transmembrane domain and an extracellular domain that comprises a region that can bind to a target cell. For example, the chimeric protein may bind to a target, e.g. a target antigen, an antigenic peptide, or a ligand on the target cell. In some embodiments, the target cell is a cancer cell. In some embodiments, a target is a cancer antigen. In some embodiments, the chimeric protein is expressed in the myeloid cell and activates the myeloid cell to overcome TME induced suppressive signal and act as an activated pro-inflammatory myeloid cell. In one embodiment, the chimeric protein that is expressed in the myeloid cell is capable of being responsive to a second signal other than the target (the first signal), wherein the second signal is a pro- inflammatory signal and an activating signal. In some embodiments, the chimeric protein that is expressed in the myeloid cell is capable of being responsive to multiple signals other than the target or the first signal. The chimeric protein may be able to respond to one, two, three, four, five, or more signals besides the target or the first signal. [0284] In another embodiment, the chimeric protein that is expressed in the myeloid cell is specific for binding to a target. In some embodiments, the target is a cancer antigen. Expression of the chimeric protein endows target specificity to the myeloid cell. [0285] In one embodiment, the chimeric protein that is expressed in the myeloid cell is capable of multiplexing, for example, has multiple domains for activation and processing of more than one signal or signal types. In some embodiments, activation of the multiple domains simultaneously leads to an augmented effector response to the myeloid cell. An effector response for the myeloid cell encompasses, for example, enhanced phagocytosis, pro-inflammatory activation, and killing of target cell. In some embodiments, the chimeric protein that is expressed in the myeloid cell, capable of multiplexing is capable of binding to more than one ligands, such as a target antigen and a helper molecule. In some embodiments, the chimeric protein is capable of binding to multiple target antigens on a cancer cell. In some embodiments, the chimeric protein is capable of multiplexing is capable of binding to multiple target antigens on multiple cells. In some embodiments, the chimeric protein may bind to a macrophage-monocyte inhibitory target on a cancer cell, and create a stimulatory signal upon contact using the pro-inflammatory domain fused to the intracellular end, a process termed as “signal switch”. For example, an extracellular domain of the chimeric protein may comprise a CD47-binding domain, whereas, the chimeric fusion protein lacks the transmembrane and/or intracellular domain of the native CD47 receptor, but comprises a PI3K recruiter domain at the intracellular region, thereby converting the macrophage-monocyte inhibitory signal from contact with the tumor cell to a pro-inflammatory phagocytosis enhancing signal. [0286] In some embodiments, the chimeric protein is capable of binding to multiple units of the expressed chimeric protein, for example, multimerizing. Multimerizing comprises dimer, trimer, tetramer, pentamer, hexamer, heptamer, octamer, nonamer, or decamer formations. In some embodiments, multimerizing can occur via association of the transmembrane region, the extracellular region or the intracellular region or combinations thereof. For example, a chimeric protein comprising a region of the collagenous domain of the phagocytic receptor MARCO may form a trimer for its effective function. In some embodiments, the chimeric protein is capable of associating with other molecules for example, another receptor. For example, the chimeric protein comprises an Fc-alpha transmembrane domain that dimerizes with Fcγ TM domain, wherein the Fcγ may be an endogenous receptor. [0287] In some embodiments, the chimeric protein capable of multiplexing comprises multiple intracellular domains that can be activated by more than one signal and can in turn activate multiple intracellular signaling molecules. For example, the chimeric protein may comprise, a phagocytosis receptor domain and a pro-inflammatory domain. For example, the chimeric protein comprises a FcR signaling domain and an additional phosphorylation domain that recruits procaspases. [0288] Provided herein is recombinant nucleic acid encoding a CFP that is phagocytic receptor (PR) fusion protein (PFP). The PFP can comprise a PR subunit comprising: a transmembrane (TM) domain, and an intracellular domain (ICD) comprising a PR intracellular signaling domain. In some embodiments, the recombinant nucleic acid encoding the PFP when expressed in a cell, the PFP functionally incorporates into the cell membrane of the cell. In some embodiments, the recombinant nucleic acid encodes for a transmembrane domain that specifically incorporates in the membrane of a myeloid cell, such as a phagocytic cell, e.g., a macrophage. [0289] In some embodiments, the suitable PR is selected after screening a library of membrane spanning proteins. The PR subunit is fused at the extracellular domain with a cancer cell binding antibody. In some embodiments, the PR may be fused with one or more additional domains at the intracellular end. INTRACELLULAR DOMAIN OF CFP FUSION PROTEIN [0290] In some embodiments the CFP subunit comprises a TM domain of a phagocytic receptor. [0291] In some embodiments the CFP subunit comprises an ICD domain of a phagocytic receptor. [0292] In some embodiments, the phagocytic receptor is a scavenger receptor. Whilst many scavenger receptors collaborate in the detection and ingestion of materials, not all the receptors engaged in the course of phagocytosis trigger engulfment alone. The engagement of certain phagocytosis and scavenger receptors can have dramatic impacts on the downstream immune response. For example, triggering the type A scavenger receptor MARCO with 500 nm negatively charge nanoparticles is associated with an anti-inflammatory tolerogenic immune response. Whereas, particles with positive charge are engulfed by a subset of phagocytosis receptors that activate proinflammatory pathways such as NLRP3 and/or fibrotic responses. Furthermore, certain scavenger receptor pathways such as the scavenger receptor expressed by endothelial cells (SREC- I), have been shown to play a role in antigen cross presentation. Therefore, identifying and understanding potential receptors that can be harnessed to enhance macrophage activity and clinical efficacy is important step in the CFP development platform. [0293] Non-opsonic receptors variably expressed naturally by professional phagocytes include lectin-like recognition molecules, such as CD169, CD33, and related receptors for sialylated residues. In addition, phagocytes also express Dectin-1 (a receptor for fungal β-glucan with well- defined signaling capacity), related C-type lectins (e.g., MICL, Dectin-2, Mincle, and DNGR-1), and a group of scavenger receptors. SR-A, MARCO, and CD36 vary in domain structure and have distinct though overlapping recognition of apoptotic and microbial ligands. CD36-related family member revealed that apoprotein ligands bind to receptor helical bundles, whereas their exofacial domains form a channel through which lipids such as cholesterol are translocated to the membrane bilayer. Table 2 - Scavenger receptors in human
Figure imgf000069_0001
Figure imgf000070_0001
Table 3 - Selected ligands of SR family members
Figure imgf000070_0002
Figure imgf000071_0001
(ND, not defined) [0294] In some embodiments, the recombinant nucleic acid encodes a chimeric antigenic receptor for phagocytosis (CAR-P). In some embodiments, the recombinant nucleic acid encodes a phagocytic receptor (PR) fusion protein. [0295] In some embodiments, the ICD of a CFP encoded by the recombinant nucleic acid comprises a domain from a protein selected from the group consisting of TNFR1, CD40, MDA5, lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor. [0296] In some embodiments, the ICD comprises the signaling domain derived from any one or more of: lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO (Macrophage Receptor with Collagenous Structure, aliases: SRA6, SCARA2), CD36 (Thrombospondin receptor, aliases: Scavenger Receptor class B, member 3), CD163 (Scavenger receptor, cysteine rich-type 1), MSR1, SCARA3, COLEC12 (aliases: Scavenger Receptor With C- Type Lectin, SCARA4, or Collectin 12), SCARA5, SCARB1, SCARB2, CD68 (SCARD, microsialin), OLR1 (Oxidized Low Density Lipoprotein Receptor 1, LOX1, or C-Type Lectin Domain Family 8 Member A), SCARF1, SCARF2, SRCRB4D, SSC5D, and CD169 (aliases, Sialoadhesin receptor, SIGLEC1). [0297] In some embodiments, the recombinant nucleic acid encodes, for example, an intracellular domain of human MARCO. The PR subunit can comprise an intracellular domain having a 44 amino acid ICD of human MARCO having an amino acid sequence: MRNKKILKEDELLSETQQAAFHQIAMEPFEINVPKPKRRNGVNF. In some embodiments the PR subunit comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to the intracellular domain of MARCO. In some embodiments, the PR comprises a transmembrane region of human MARCO. [0298] In some embodiments, the recombinant nucleic acid encodes an intracellular domain of human SRA1. The CFP comprises an intracellular domain having a 50 amino acid ICD of human SRA1 having an amino acid sequence: MEQWDHFHNQQEDTDSCSESVKFDARSMTA LLPPNPKNSPSLQEKLKSFK. In some embodiments the PR subunit comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to the intracellular domain of human SRA1. The intracellular region of SRA has a phosphorylation site. [0299] In some embodiments, the CFP comprises a transmembrane region of human SRA1. [0300] In some embodiments, the recombinant nucleic acid comprises a sequence encoding an intracellular domain of CD36. In some embodiments, the recombinant nucleic acid comprises a sequence encoding TM domain of CD36. Naturally occurring full length CD36 has two TM domains and two short intracellular domains, and an extracellular domain of CD36 binds to oxidized LDL. Both of the intracellular domains contain pairs of cysteines that are fatty acid acylated. It lacks known signaling domains (e.g. kinase, phosphatase, g-protein binding, or scaffolding domains). N- terminal cytoplasmic domain is extremely short (5–7 amino acid residues) and is closely associated with the internal leaflet of the plasma membrane. The carboxy-terminal domain contains 13 amino acids, containing a CXCX5K motif homologous to a region in the intracellular domain of CD4 and CD8 that is known to interact with signaling molecules. The intracellular domain of CD36 is capable of assembling a signaling complex that activates lyn kinases, MAP kinases and Focal Adhesion Kinases (FAK), and inactivation of src homology 2-containing phosphotyrosine phosphatase (SHP- 2). Members of the guanine nucleotide exchange factors (GEFs) have been identified as potential key signaling intermediates. [0301] In some embodiments, the recombinant nucleic acid encodes for example, an intracellular domain of human SCARA3. The CFP may comprise an intracellular domain having a 56 amino acid ICD of human SCARA3 having an amino acid sequence: MKVRSAGGDGDALCVTEEDL AGDDEDMPTFPCTQKGRPGPRCSRCQKNLS LHTSVR. In some embodiments, the CFP comprises a variant which is at least 70%, 75%, 80%, 85%, 90% or 95% identical to an intracellular domain of human SCARA3. In some embodiments the CFP comprises a TM domain of SCARA3. [0302] In some embodiments, the TM domain of a PR is about 20-30 amino acids long. In some embodiments, the TM domain comprises multiple transmembrane spans. In some embodiment, the TM domain comprises about 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or more amino acids in length. In some embodiments, the TM domains of SRs are about 20-30 amino acids long. [0303] Scavenger receptors may occur as homo or hetero dimers. MARCO, for example occurs as a homo trimer. In some embodiments, a scavenger receptor is a monomer. In some embodiments, the scavenger receptor is a homodimer. In some embodiments, the scavenger receptor is a heterodimer. In some embodiments, the scavenger receptor is a homotrimer. In some embodiments, the scavenger receptor is a heterotrimer. In some embodiments, the scavenger receptor is a homo tetramer. In some embodiments, the scavenger receptor is a hetero tetramer. In some embodiments, the scavenger receptor is multimer comprising two, three, four, five, six, seven, eight, night, ten or more subunits that are the same or different. [0304] In some embodiments, the TM domain or the ICD domain of the PSP is not derived from FcR, Megf10, Bai1 or MerTK. In some embodiments, the ICD of the PR does not comprise a CD3 zeta intracellular domain. [0305] In some embodiments, the intracellular domain and transmembrane domains are derived from FcRβ. [0306] In one aspect the recombinant nucleic acid encodes a chimeric antigenic receptor for enhanced phagocytosis (CAR-P), which is a phagocytic scavenger receptor (PSR) fusion protein (PFP) comprising: (a) an extracellular domain comprising an extracellular antigen binding domain specific to an antigen of a target cell, (b) a transmembrane domain, and (c) a recombinant PSR intracellular signaling domain, wherein the recombinant PSR intracellular signaling domain comprises a first portion derived from a phagocytic receptor and a second portion derived from a non-phagocytic receptor. [0307] In some embodiments, the second portion is not a PI3K recruitment domain. [0308] The second portion derived from a non-phagocytic receptor may comprise an intracellular signaling domain that enhances phagocytosis, and/or inflammatory potential of the engineered myeloid cells, such as phagocytic cells, expressing the recombinant nucleic acid. In some embodiment, the second portion derived from non-phagocytic receptor comprises more than one intracellular domains (ICD). In some embodiments, the second portion derived from non-phagocytic receptor comprises a second ICD. In some embodiments, the second portion derived from non- phagocytic receptor comprises a second and a third ICD. In some embodiments, the second portion derived from non-phagocytic receptor comprises a second, a third and a fourth ICD, wherein the second portion is encoded by the recombinant nucleic acid. In some embodiments, the intracellular portion comprises two, three, four, five, six, seven, or more ICDs. The respective second portions comprising a second, or third or fourth ICD derived from non-phagocytic receptor are described as follows. Chimeric antigen receptors for enhancing intracellular signaling and inflammation activation [0309] In one aspect, the recombinant nucleic acid encodes a second intracellular domain in addition to the phagocytic ICD, which confers capability of potent pro-inflammatory immune activation, such as when myeloid cells, such as macrophages, engage in fighting infection. The second intracellular domain (second ICD) is fused to the cytoplasmic terminus of the first phagocytic ICD. The second intracellular domain provides a second signal is necessary to trigger inflammasomes and pro-inflammatory signals. Nod-like receptors (NLRs) are a subset of receptors that are activated in innate immune response, and oligomerize to form multi-protein complexes that serve as platforms to recruit proinflammatory caspases and induce their cleavage and activation. This leads to direct activation of ROS, and often result in a violent cell death known as pyroptosis. There are four inflammasome complexes, NLRP1m, NLRP3, IPAF and AIM2. [0310] The tumor microenvironment (TME) constitutes an immunosuppressive environment. Influence of IL-10, glucocorticoid hormones, apoptotic cells, and immune complexes can interfere with innate immune cell function. Immune cells, including phagocytic cells settle into a tolerogenic phenotype. In myeloid cells such as macrophages, this phenotype, commonly known as the M2 phenotype, is distinct from the M1 phenotype, where the cells are potent and capable of killing pathogens. Myeloid cells, such as macrophages, exposed to LPS or IFNγ, for example, can polarize towards an Ml phenotype, whereas myeloid cells, such as macrophages, exposed to IL-4 or IL-13 can polarize towards an M2 phenotype. LPS or IFNγ can interact with Toll-like receptor 4 (TLR4) on the surface of myeloid cells, such as macrophages, inducing the Trif and MyD88 pathways, inducing the activation of transcription factors IRF3, AP-1, and NFKB and thus activating TNFs genes, interferon genes, CXCL10, NOS2, IL-12, etc., for a pro-inflammatory Ml myeloid cell response. Similarly, IL-4 and IL-13 bind to IL-4R, activation the Jak/Stat6 pathway, which regulates the expression of CCL17, ARG1, IRF4, IL-10, SOCS3, etc., which are genes associated with an anti- inflammatory response (M2 response). Expression of CD14, CD80, D206 and low expression of CD163 are indicators of myeloid cells, such as macrophages, polarization towards the M1 phenotype. [0311] In some embodiments, the recombinant nucleic acid encodes one or more additional intracellular domains, comprising a cytoplasmic domain for inflammatory response. In some embodiments, expression of the recombinant nucleic acid encoding the phagocytic receptor (PR) fusion protein (PFP) comprising the cytoplasmic domain for inflammatory response in the engineered myeloid cells, such as macrophages, confers potent pro-inflammatory response similar to the M1 phenotype. [0312] In some embodiments, the cytoplasmic domain for inflammatory response comprises an intracellular signaling domain of TLR3, TLR4, TLR9, MYD88, TRIF, RIG-1, MDA5, CD40, IFN receptor, NLRP-1, NLRP-2, NLRP-3, NLRP-4, NLRP-5, NLRP-6, NLRP-7, NLRP-8, NLRP-9, NLRP-10, NLRP-11, NLRP-12, NLRP-13, NLRP-14, NOD1, NOD2, Pyrin, AIM2, NLRC4 and/or CD40. [0313] In some embodiments, the phagocytic scavenger receptor (PR) fusion protein (PFP) comprises a pro-inflammatory cytoplasmic domain for activation of IL-1 signaling cascade. [0314] In some embodiments, the cytoplasmic portion of the chimeric receptor (for example, phagocytic receptor (PR) fusion protein (PFP)) comprises a cytoplasmic domain from a toll-like receptor, such as the intracellular signaling domains of toll-like receptor 3 (TLR3), toll-like receptor 4 (TLR4), toll-like receptor 7 (TLR7), toll-like receptor 8 (TLR8), toll-like receptor 9 (TLR9). [0315] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from interleukin-1 receptor-associated kinase 1 (IRAK1). [0316] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from differentiation primary response protein (MYD88). [0317] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from myelin and lymphocyte protein (MAL). [0318] In some embodiments, the cytoplasmic portion of the chimeric receptor comprises a suitable region from retinoic acid inducible gene (RIG-1). [0319] In some embodiments the cytoplasmic portion of the CFP comprises a cytoplasmic domain of any one of MYD88, TLR3, TLR4, TLR7, TLR8, TLR9, MAL, or IRAK1. [0320] In some embodiments, the recombinant CFP intracellular signaling domain comprises a first portion derived from a phagocytic and a second portion derived from non-phagocytic receptor wherein the second portion derived from non-phagocytic receptor comprises a phosphorylation site. In some embodiments, the phosphorylation site comprises amino acid sequences suitable for an autophosphorylation site. In some embodiments, the phosphorylation site comprises amino acid sequences suitable phosphorylation by Src family kinases. In some embodiments, the phosphorylation site comprises amino acid sequences, which upon phosphorylation are capable of binding to SH2 domains in a kinase. In some embodiments, a receptor tyrosine kinase domain is fused at the cytoplasmic end of the PFP in addition to the first cytoplasmic portion. [0321] In some embodiments, the phosphorylation is a Tyrosine phosphorylation. [0322] In some embodiments the second intracellular domain is an Immune receptor Tyrosine Activation Motif (ITAM). The ITAM motif is present in mammalian α and β immunoglobulin proteins, TCR γ receptors, FCR γ receptors subunits, CD3 chains receptors and NFAT activation molecule. [0323] In some embodiments the PFP intracellular domain comprises one ITAM motif. In some embodiments the PFP intracellular domain comprises more than one ITAM motifs. In some embodiments the PFP intracellular domain comprises two or more ITAM motifs. In some embodiments the PFP intracellular domain comprises three or more ITAM motifs. In some embodiments the PFP intracellular domain comprises four or more ITAM motifs. In some embodiments the PFP intracellular domain comprises five or more ITAM motifs. In some embodiments the PFP intracellular domain comprises six or more ITAM motifs. In some embodiments the PFP intracellular domain comprises seven or more ITAM motifs. In some embodiments the PFP intracellular domain comprises eight or more ITAM motifs. In some embodiments the PFP intracellular domain comprises nine or more ITAM motifs. In some embodiments the PFP intracellular domain comprises ten or more ITAM motifs. [0324] In some embodiments one or more domains in the first phagocytic ICD comprises a mutation. [0325] In some embodiments one or more domains in the second ICD comprises a mutation to enhance a kinase binding domain, to generate a phosphorylation site, to generate an SH2 docking site or a combination thereof. Co-expression of an Inflammatory Gene [0326] In one aspect, the recombinant nucleic acid comprises a coding sequence for a pro- inflammatory gene, which is co-expressed with the PFP in the engineered cell. In some embodiments, the pro-inflammatory gene is a cytokine. Examples include but not limited to TNF-α, IL-1α, IL-1 ^, IL-6, CSF, GMCSF, or IL-12 or interferons. [0327] The recombinant nucleic acid encoding the proinflammatory gene can be monocistronic, wherein the two coding sequences for (a) the PSP and (b) the proinflammatory gene are post- transcriptionally or post-translationally cleaved for independent expression. [0328] In some embodiments, the two coding sequences comprise a self-cleavage domain, encoding a P2A sequence, for example. [0329] In some embodiments the two coding regions are separated by a IRES site. [0330] In some embodiments the two coding sequences are encoded by a bicistronic genetic element. The coding regions for (a) the PSP and (b) the proinflammatory gene can be unidirectional, where each is under a separate regulatory control. In some embodiments the coding regions for both are bidirectional and drive in opposite directions. Each coding sequence is under a separate regulatory control. [0331] Coexpression of the proinflammatory gene is designed to confer strong inflammatory stimulation of the myeloid cells, such as macrophages, and activate the surrounding tissue for inflammation. Integrin activation domains [0332] Cell-cell and cell-substratum adhesion is mediated by the binding of integrin extracellular domains to diverse protein ligands; however, cellular control of these adhesive interactions and their translation into dynamic cellular responses, such as cell spreading or migration, requires the integrin cytoplasmic tails. These short tails bind to intracellular ligands that connect the receptors to signaling pathways and cytoskeletal networks (Calderwood DA, 2004, Integrin Activation, Journal of Cell Science 117, 657-666, incorporated herein in its entirety). Integrins are heterodimeric adhesion receptors formed by the non-covalent association of α and β subunits. Each subunit is a type I transmembrane glycoprotein that has relatively large extracellular domains and, with the exception of the β4 subunit, a short cytoplasmic tail. Individual integrin family members have the ability to recognize multiple ligands. Integrins can bind to a large number of extracellular matrix proteins (bone matrix proteins, collagens, fibronectins, fibrinogen, laminins, thrombospondins, vitronectin, and von Willebrand factor), reflecting the primary function of integrins in cell adhesion to extracellular matrices. Many “counter-receptors” are ligands, reflecting the role of integrins in mediating cell-cell interactions. Integrins undergo conformational changes to increase ligand affinity. [0333] The Integrin β2 subfamily consists of four different integrin receptors, αMβ2 (CD11b/CD18, Mac-1, CR3, Mo-1), αLβ2 (CD11a/CD18, LFA-1), αXβ2 (CD11c/CD18), and αDβ2 (CD11d/CD18). These leukocyte integrins are involved in virtually every aspect of leukocyte function, including the immune response, adhesion to and transmigration through the endothelium, phagocytosis of pathogens, and leukocyte activation. [0334] The α subunits of all β2 integrins contain an inserted region of ∼200 amino acids, termed the I or A domain. Highly conserved I domains are found in several other integrin α subunits and other proteins, such as certain coagulation and complement proteins. I domains mediate protein- protein interactions, and in integrins, they are integrally involved in the binding of protein ligands. Although the I domains dominate the ligand binding functions of their integrins, other regions of the α subunits do influence ligand recognition. As examples, in αMβ2 a mAb (OKM1) recognizing an epitope outside the I domain but in the αM subunit inhibits ligand binding; and the EF-hand regions in αLβ2 and α2β1, integrins with I domains in their α subunits, contribute to ligand recognition. The αM subunit, and perhaps other α subunits, contains a lectin-like domain, which is involved in engagement of non-protein ligands, and occupancy may modulate the function of the I domain. [0335] As integrins lack enzymatic activity, signaling is instead induced by the assembly of signaling complexes on the cytoplasmic face of the plasma membrane. Formation of these complexes is achieved in two ways; first, by receptor clustering, which increases the avidity of molecular interactions thereby increasing the on-rate of binding of effector molecules, and second, by induction of conformational changes in receptors that creates or exposes effector binding sites. Within the ECM, integrins have the ability to bind fibronectin, laminins, collagens, tenascin, vitronectin and thrombospondin. Clusters of integrin/ECM interactions form focal adhesions, concentrating cytoskeletal components and signaling molecules within the cell. The cytoplasmic tail of integrins serve as a binding site for α-actinin and talin which then recruit vinculin, a protein involved in anchoring F-actin to the membrane. Talin is activated by kinases such as protein kinase C (PKCα). [0336] Integrins are activated by selectins. Leucocytes express L-selectin, activated platelets express P-selectin, and activated endothelial cells express E-and P-selectin. P-selectin-mediated adhesion enables chemokine-or platelet-activating factor-triggered activation of β2 integrins, which stabilizes adhesion. It also facilitates release of chemokines from adherent leucocytes. The cytoplasmic domain of P-selectin glycoprotein ligand 1 formed a constitutive complex with Nef- associated factor 1. After binding of P-selectin, Src kinases phosphorylated Nef-associated factor 1, which recruit the phosphoinositide-3-OH kinase p85-p110δ heterodimer and result in activation of leukocyte integrins. E-selectin ligands transduce signals that also affect β2 integrin function. Selectins trigger activation of Src family kinases. SFKs activated by selectin engagement phosphorylate the immunoreceptor tyrosine-based activation motifs (ITAMs) in the cytoplasmic domains of DAP12 and FcRγ. In some respects, CD44 is sufficient to transduce signals from E- selectin. CD44 triggers the inside-out signaling of integrins. A final common step in integrin activation is binding of talin to the cytoplasmic tail of the β subunit. Kindlins, another group of cytoplasmic adaptors, bind to a different region of integrin β tails. Kindlins increase the clustering of talin-activated integrins. Kindlins are responsive to selectin signaling, however, kindlins are found mostly in hematopoietic cells, such as neutrophils. Selectin signaling as well as signaling upon integrin activation by chemokines components have shared components, including SFKs, Syk, and SLP-76. [0337] In some embodiments, the intracellular domain of the recombinant CFP comprises an integrin activation domain. The integrin activation domain comprises an intracellular domain of a selectin, for example, a P-selectin, L-selectin or E-selectin. [0338] In some embodiments, the intracellular domain of the recombinant CFP comprises an integrin activation domain of laminin. [0339] In some embodiments, the intracellular domain of the recombinant CFP comprises an integrin activation domain for activation of Talin. [0340] In some embodiments, the intracellular domain of the recombinant CFP comprises an integrin activation domain fused to the cytoplasmic end of the phagocytic receptor ICD domain. Chimeric Receptor for Enhancing Antigen Cross presentation [0341] In some embodiments, the recombinant nucleic acid encodes a domain capable of enabling cross presentation of antigens. In general, MHC class I molecules present self-or pathogen-derived antigens that are synthesized within the cell, whereas exogenous antigens derived via endocytic uptake are loaded onto MHC class II molecules for presentation to CD4+ T cells. MHC I-restricted presentation of endogenous antigens, in which peptides are generated by the proteasome. However, in some cases, DC can process exogenous antigens into the MHC-I pathway for presentation to CD8+ T cells. This is referred to as cross presentation of antigens. Soluble or exogenous antigenic components may get degraded by lysosomal proteases in the vacuoles and cross presented by DCs, instead of following the endocytic pathway. In some instances, chaperones, such as heat shock protein 90 (Hsp90) have shown to help cross present antigens by certain APCs. HSP-peptide complexes are known to be internalized by a distinct group of receptors compared to free polypeptides. These receptors were from the scavenger receptor families and included LOX-1, SREC-I/SCARF-I, and FEEL1/Stabilin-1. Both SREC-I and LOX-1 have been shown to mediate the cross presentation of molecular chaperone bound antigens and lead to activation of CD8+ T lymphocytes. [0342] SREC-1 (scavenger receptor expressed by endothelial cells) has no significant homology to other types of scavenger receptors but has unique domain structures. It contains 10 repeats of EGF-like cysteine-rich motifs in the extracellular domain. Recently, the structure of SREC-I was shown to be similar to that of a transmembrane protein with 16 EGF-like repeats encoded by the Caenorhabditis elegans gene ced-I, which functions as a cell surface phagocytic receptor that recognizes apoptotic cells. [0343] Cross presentation of cancer antigens through the Class-I MHC pathway results in enhanced CD8+ T cell response, which is associated with cytotoxicity and therefore beneficial in tumor regression. In some embodiments, the intracellular domain of the PFP comprises a SREC1 intracellular domain. In some embodiments, the intracellular domain of the PFP comprises a SRECII intracellular domain. [0344] In some embodiments, the CFP comprises: an intracellular domain comprising a PSR intracellular signaling domain from SREC1 or SRECII. [0345] In some embodiments, the CFP comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising a CFP intracellular signaling domain from SREC1 or SRECII. [0346] In some embodiments, the CFP comprises: (i) a transmembrane domain, (ii) an intracellular domain comprising a intracellular signaling domain, and (iii) an extracellular domain from SREC1 or SRECII. TRANSMEMBRANE DOMAIN OF CHIMERIC FUSION PROTEIN [0347] In some embodiments, the TM encoded by the recombinant nucleic acid comprises a sequence encoding a domain of a scavenger receptor (SR). In some embodiments, the TM can be the TM domain of or derived from any one or more of: lectin, dectin 1, mannose receptor (CD206), SRA1, MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, SRCRB4D, SSC5D, and CD169. [0348] In some embodiments, the TM domains are about 20-30 amino acids long. TM domains of SRs are about 20-30 amino acids long. [0349] In some embodiments, the TM domain or the ICD domain of the CFP is not derived from Megf10, Bai1 or MerTK. In some embodiments, the ICD of the CFP does not comprise a CD3ζ intracellular domain. [0350] In some embodiments the TM is derived from the same phagocytic receptor as the ICD. [0351] In some embodiments, the TM region is derived from a plasma membrane protein. The TM can be selected from an Fc receptor (FcR). In some embodiments, nucleic acid sequence encoding domains from specific FcRs are used for cell-specific expression of a recombinant construct. An FCR-alpha region comprising the TM domain may be used for a myeloid cell, such as a macrophage, specific expression of the construct. FcRα recombinant protein can be expressed in mast cells. [0352] In some embodiments, the PFP comprises the TM of FcRβ. [0353] In some embodiments, the PFP comprises both the FcRβ and ICD domains. In some embodiments, the PFP comprises both the FcRα and ICD domains. [0354] In some embodiments, the TM domain is derived from CD8. [0355] In some embodiments, the TM is derived from CD2. [0356] In some embodiments the TM is derived from FcRα. EXTRACELLULAR DOMAIN OF CHIMERIC FUSION PROTEIN [0357] In some embodiments, the extracellular domain of a CFP provided herein comprises an antigen binding domain that binds to one or more targets. The binding targets may be antigens or ligands. For example, a binding target may be an antigen on a target cell. In some embodiments, the target binding domain is specific for the target. In some embodiments, the extracellular domain can include an antibody or an antigen-binding domain selected from intrabodies, peptibodies, nanobodies, single domain antibodies. SMIPs, and multispecific antibodies. [0358] In some embodiments, an antibody fragment comprises a portion of an intact antibody, such as the antigen binding or variable region of the intact antibody. In a further aspect of the invention, an anti-HIV antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. Antibody fragments include, but are not limited to, Fab, Fab’, Fab’-SH, F(ab’)2, Fv, diabody, linear antibodies, multispecific formed from antibody fragments antibodies and scFv fragments, and other fragments described below. In another embodiment, the antibody is a full length antibody, e.g., an intact IgG1 antibody or other antibody class or isotype as described herein. (See, e.g., Hudson et al. Nat. Med.9:129-134 (2003); Pluckthiin, The Pharmacology of Monoclonal Antibodies, vol.113, pp.269-315 (1994); Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993); WO93/01161; and U.S. Pat. Nos. 5,571,894, 5,869,046, 6,248,516, and 5,587,458). A full length antibody, intact antibody, or whole antibody is an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein. Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g., E. coli or phage), as described herein. [0359] An Fv is the minimum antibody fragment that contains a complete antigen-recognition and antigen-binding site. This fragment contains a dimer of one heavy-and one light-chain variable region domain in tight, non-covalent association. From the folding of these two domains emanate six hypervariable loops (three loops each from the H and L chain) that contribute the amino acid residues for antigen binding and confer antigen binding specificity to the antibody. However, even a single variable region (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. [0360] A single-chain Fv (sFv or scFv) is an antibody fragment that comprises the VH and VL antibody domains connected into a single polypeptide chain. The sFv polypeptide can further comprise a polypeptide linker between the VH and VL domains that enables the sFv to form the desired structure for antigen binding. (See, e.g., Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994); Borrebaeck 1995, infra. The sFv can be used in a chimeric antigen receptor (CAR). [0361] A diabody is a small antibody fragment prepared by constructing an sFv fragment with a short linker (about 5-10 residues) between the VH and VL domains such that inter-chain but not intra- chain pairing of the V domains is achieved, resulting in a bivalent fragment. Bispecific diabodies are heterodimers of two crossover sFv fragments in which the VH and VL domains of the two antibodies are present on different polypeptide chains. (See, e.g., EP 404,097; WO 93/11161; and Hollinger et al, Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993)). [0362] Domain antibodies (dAbs), which can be produced in fully human form, are the smallest known antigen-binding fragments of antibodies, ranging from about 11 kDa to about 15 kDa. DAbs are the robust variable regions of the heavy and light chains of immunoglobulins (VH and VL, respectively). They are highly expressed in microbial cell culture, show favorable biophysical properties including, for example, but not limited to, solubility and temperature stability, and are well suited to selection and affinity maturation by in vitro selection systems such as, for example, phage display. DAbs are bioactive as monomers and, owing to their small size and inherent stability can be formatted into larger molecules to create drugs with prolonged serum half-lives or other pharmacological activities. (See, e.g., W09425591 and US20030130496). [0363] Fv and sFv are the only species with intact combining sites that are devoid of constant regions. Thus, they are suitable for reduced nonspecific binding during in vivo use. sFv fusion proteins can be constructed to yield fusion of an effector protein at either the amino or the carboxy terminus of an sFv. The antibody fragment also can be a “linear antibody. (See, e.g., U.S. Pat. No. 5,641,870). Such linear antibody fragments can be monospecific or bispecific. [0364] In some embodiments, the extracellular domain includes a Fab binding domain. In yet other such embodiments, the extracellular domain includes a scFv. [0365] In some embodiments the chimeric antigen receptor comprises an extracellular antigen binding domain is derived from the group consisting of an antigen-binding fragment (Fab), a single- chain variable fragment (scFv), a nanobody, a VH domain, a VL domain, a single domain antibody (sdAb), a VNAR domain, and a VHH domain, a bispecific antibody, a diabody, or a functional fragment of any thereof. In some embodiments, the antigen-binding fragment (Fab), a single-chain variable fragment (scFv), a nanobody, a VH domain, a VL domain, a single domain antibody (sdAb), a VNAR domain, and a VHH domain, a bispecific antibody, a diabody, or a functional fragment of any thereof specifically bind to one or more antigens. [0366] In some embodiments, the antigen is a cancer antigen, and the target cell is a target cancer cell. In some embodiments, the antigen for a target cell is selected from the group consisting of CD3, CD4, CD5, CD7, CD19, CCR2, CCR4, CD30, CD37, TCRB1/2, TCR αβ, TCRαδ. CD22, HER2 (ERBB2/neu), Mesothelin, PSCA, CD123, CD30, CD171, CD138, CS-1, CLECL1, CD33, CD79b, EGFRvIII, GD2, GD3, BCMA, PSMA, ROR1, FLT3, TAG72, CD38, CD44v6, CEA, EPCAM, B7H3 (CD276), KIT (CD 117), CD213A2, IL-1 IRa, PRSS21, VEGFR2, CD24, MUC-16, PDGFR- β, SSEA-4, CD20, MUC1, EGFR, NCAM, Prostase, PAP, ELF2M, Ephrin B2, FAP, EphA2, GM3, TEM1/CD248, TEM7R, CLDN6, TSHR, GPRC5D, CD97, CD179a, ALK, and IGLL1. [0367] In some embodiments, the target antigen is an autoimmune antigen. In some embodiments, the target cell is a B cell. In some embodiments, the target cell is a B cell that produces autoantibodies. In some embodiments, the target antigen is Dsg1 or Dsg3. [0368] Various cancer antigen targets can be selected from cancer antigens known to one of skill in the art. Depending on the cancer and the cell type involved cancer antigens are mutated native proteins. The antigen binding domains are screened for specificity towards mutated/cancer antigens and not the native antigens. [0369] In some embodiments, for example, the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: MUC16, CCAT2, CTAG1A, CTAG1B, MAGE A1, MAGEA2, MAGEA3, MAGEA4, MAGEA6, PRAME, PCA3, MAGEC1, MAGEC2, MAGED2, AFP, MAGEA8, MAGE9, MAGEA11, MAGEA12, IL13RA2, PLAC1, SDCCAG8, LSP1, CT45A1, CT45A2, CT45A3, CT45A5, CT45A6, CT45A8, CT45A10, CT47A1, CT47A2, CT47A3, CT47A4, CT47A5, CT47A6, CT47A8, CT47A9, CT47A10, CT47A11, CT47A12, CT47B1, SAGE1, and CT55. [0370] In some embodiments, for example, the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: CD2, CD3, CD4, CD5, CD7, CD8, CD20, CD30, CXCR4, CD45, CD56, where the cancer is a T cell lymphoma. [0371] In some embodiments, for example, the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: IDH1, ATRX, PRL3, or ETBR, where the cancer is a glioblastoma. [0372] In some embodiments, for example, the cancer antigen for a target cancer cell can be one or more of the mutated/cancer antigens: CA125, β-hCG, urinary gonadotropin fragment, AFP, CEA, SCC, inhibin or extradiol, where the cancer is ovarian cancer. [0373] In some embodiments the cancer antigen for a target cancer cell may be CD5. [0374] In some embodiments the cancer antigen for a target cancer cell may be HER2. [0375] In some embodiments the cancer antigen for a target cancer cell may be EGFR Variant III. [0376] In some embodiments the cancer antigen for a target cancer cell may be CD19. [0377] In some embodiments, the SR subunit region comprises an extracellular domain (ECD) of the scavenger receptor. In some embodiments, the ECD of the scavenger receptor comprises an ECD domain of the SR comprising the ICD and the TM domains. In some embodiments the target antigen is the SR-ligand on the cancer cell, for example, any one of the ligand components from Table 2 or Table 3. In some embodiments, the SR-ECD contributes to the binding of the phagocyte to the target cell, and in turn is activated, and activates the phagocytosis of the target cell. [0378] In some embodiments, the CFP comprises an ECD or portion thereof of a scavenger receptor. In some embodiments, the CFP comprises an ICD or portion thereof of a scavenger receptor. In some embodiments, the CFP comprises a TM domain of a scavenger receptor. In some embodiments, the ECD encoded by the recombinant nucleic acid comprises a domain selected from the group consisting of lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169. The extracellular domains of most scavenger receptors contain scavenger receptors with a broad binding specificity that may be used to discriminate between self and non-self in the nonspecific antibody- independent recognition of foreign substances. The type I and II class A scavenger receptors (SR- AI1 and SR-AII) are trimeric membrane glycoproteins with a small NH2-terminal intracellular domain, and an extracellular portion containing a short spacer domain, an a-helical coiled-coil domain, and a triple-helical collagenous domain. The type I receptor additionally contains a cysteine-rich COOH-terminal (SRCR) domain. These receptors are present in myeloid cells, such as macrophages, in diverse tissues throughout the body and exhibit an unusually broad ligand binding specificity. They bind a wide variety of polyanions, including chemically modified proteins, such as modified LDL, and they have been implicated in cholesterol deposition during atherogenesis. They may also play a role in cell adhesion processes in macrophage-associated host defense and inflammatory conditions. [0379] In some embodiments, the SR ECD is designed to bind to pro-apoptotic cells. In some embodiments, the scavenger receptor ECD comprises a binding domain for a cell surface molecule of a cancer cell or an infected cell. [0380] In some embodiments, the extracellular domain of the PR subunit is linked by a linker to a target cell binding domain, such as an antibody or part thereof, specific for a cancer antigen. [0381] In some embodiments, the extracellular antigen binding domain comprises one antigen binding domain. In some embodiments, the extracellular antigen binding domain comprises more than one binding domain. In some embodiments the binding domain are scFvs. FIG. 2 shows a diagrammatic representation of an embodiment, where the PFP targets a single target on a cancer cell (left) or multiple targets (right). The one or more than one scFvs are fused to the recombinant PR at the extracellular domain. In some embodiments the scFv fraction and the extracellular domain of the PR are linked via a linker. [0382] In some embodiments, the ECD antigen binding domain can bind to an intracellular antigen. In some embodiments, the intracellular antigen is a cancer antigen. [0383] In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 1000 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 500 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 450 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 400 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 350 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 250 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 200 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity of less than 100 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity ranging between than 200 nM to 1000 nM. In some embodiments, the extracellular antigen binding domain binds to the target ligand with an affinity ranging between than 300 nM to 1.5 mM. In some embodiments, the antigen binding domain binds to the target ligand with an affinity > 200 nM, > 300 nM or >500 nM. [0384] In some embodiments, the extracellular antigen binding domain binds to the target ligand, where the target ligand is a T cell, the binding characteristics are such that the target T cell is not triggered to activate T cell mediated lysis of the engineered cell. In some embodiments, binding of the TCR to a ligand on the engineered cell is avoided, bypassed or inhibited. LINKERS [0385] Linkers may be used to link any of the polypeptides or peptide domains of the present disclosure. The PFP fusion proteins described herein may comprises one or more linkers. For example, one or more of the domains and subunits of a PFP fusion protein can be directly fused to another domain or subunit, or can be connected to another domain or subunit via a linker. In some embodiments, the extracellular antigen binding domains comprising the antibody specific for the antigen on a target cell, parts of an antibody that can specifically bind to an antigen on a target cell, or scFvs specific for an antigen on a target cell are linked to the TM domain or other extracellular domains by a linker. In some embodiments, where there are more than one scFv at the extracellular antigen binding domain, the more than scFvs are linked with each other by linkers. [0386] In some embodiments, linkers are short peptide sequences. [0387] The linker may be as simple as a covalent bond, or it may be a polymeric linker many atoms in length. In certain embodiments, the linker is a polypeptide or based on amino acids. In other embodiments, the linker is not peptide-like. In certain embodiments, the linker is a covalent bond (e.g., a carbon-carbon bond, disulfide bond, carbon-heteroatom bond, etc.). [0388] In some embodiments, the linker is an amino acid or a plurality of amino acids (e.g., a peptide or protein). In some embodiments, the linker is a bond (e.g., a covalent bond), an organic molecule, group, polymer, or chemical moiety. In some embodiments, the linker is about 3 to about 104 (e.g., 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, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100) amino acids in length. In some embodiments the linkers are stretches of Glycine and one or more Serine residues. Other amino acids preferred for short peptide linkers include but are not limited to threonine (Thr), serine (Ser), proline (Pro), glycine (Gly), aspartic acid (Asp), lysine (Lys), glutamine (Gln), asparagine (Asn), and alanine (Ala) arginine (Arg), phenylalanine (Phe), glutamic acid (Glu). Of these Pro, Thr, and Gln are frequently used amino acids for natural linkers. Pro is a unique amino acid with a cyclic side chain which causes a very restricted conformation. Pro-rich sequences are used as interdomain linkers, including the linker between the lipoyl and E3 binding domain in pyruvate dehydrogenase (GA2PA3PAKQEA3PAPA2KAEAPA3PA2KA). For the purpose of the disclosure, the empirical linkers may be flexible linkers, rigid linkers, and cleavable linkers. Sequences such as (G4S)x (where x is multiple copies of the moiety, designated as 1, 2, 3, 4, and so on) comprise a flexible linker sequence. Other flexible sequences used herein include several repeats of glycine, e.g., (Gly)6 or (Gly)8. On the other hand, a rigid linker may be used, for example, a linker (EAAAK)x, where x is an integer, 1, 2, 3, 4 etc. gives rise to a rigid linker. Various linker lengths and flexibilities between domains or subunits of the fusion proteins provided herein can be employed, e.g., ranging from very flexible linkers of the form (GGGS)n, (GGGGS)n, and (G)n to more rigid linkers of the form (EAAAK)n, (SGGS)n, SGSETPGTSESATPES (see, e.g., Guilinger JP, Thompson DB, Liu DR. Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nat. Biotechnol. 2014; 32(6): 577-82; the entire contents are incorporated herein by reference) and (XP)n) in order to achieve the optimal length. In some embodiments, n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15. In some embodiments, the linker comprises a (GGS)n motif, wherein n is 1, 3, or 7. In some embodiments, the linker comprises amino acid sequence SGGGGSG. In some embodiments, the linker comprises amino acid sequence GSGS. [0389] In some embodiments the linkers are flexible. In some embodiments the linkers comprise a hinge region. Where included, such a spacer or linker domain may position the binding domain away from the host cell surface to further enable proper cell/cell contact, binding, and activation. The length of the extracellular spacer may be varied to optimize target molecule binding based on the selected target molecule, selected binding epitope, binding domain size and affinity. In certain embodiments, an extracellular spacer domain is an immunoglobulin hinge region (e.g., IgGl, IgG2, IgG3, IgG4, IgA, IgD). An immunoglobulin hinge region may be a wild type immunoglobulin hinge region or an altered wild type immunoglobulin hinge region. In some embodiments, a linker or a spacer used herein comprises an IgG4 hinge region, having a sequence: ESKYGPPCPPCP. In some embodiments, the hinge region comprises a hinge or a spacer comprising a sequence present in the extracellular regions of type 1 membrane proteins, such as CD8a, CD4, CD28 and CD7, which may be wild-type or variants thereof. In some embodiments, an extracellular spacer domain comprises all or a portion of an immunoglobulin Fc domain selected from: a CH1 domain, a CH2 domain, a CH3 domain, or combinations thereof. In some embodiments the spacer or the linker may be further modified by post-translation modifications, such as glycosylation. [0390] In some embodiments, an extracellular spacer domain may comprise a stalk region of a type II C-lectin (the extracellular domain located between the C-type lectin domain and the transmembrane domain). Type II C-lectins include CD23, CD69, CD72, CD94, NKG2A, and NKG2D. In yet further embodiments, an extracellular spacer domain may be derived from scavenger receptor MERTK. [0391] In some embodiments, the linker comprises at least 2, or at least 3 amino acids. In some embodiments, the linker comprises 4 amino acids. In some embodiments, the linker comprises 5 amino acids. In some embodiments, the linker comprises 6 amino acids. In some embodiments, the linker comprises 7 amino acids. In some embodiments, the linker comprises 8 amino acids. In some embodiments, the linker comprises 9 amino acids. In some embodiments, the linker comprises 8 amino acids. In some embodiments, the linker comprises 10 amino acids. In some embodiments the linker comprises greater than 10 amino acids. In some embodiments, the linker comprises 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids. In some embodiments there are 12 or more amino acids in the linker. In some embodiments, there are 14 or more amino acids in the linker. In some embodiments, there are 15 or more amino acids in the linker. Other Fusion proteins for Enhancement of Phagocytosis [0392] In one aspect of the disclosure, recombinant nucleic acids are prepared which encode one or more chimeric receptors that enhance phagocytosis in myeloid cells, such as macrophages, principally by blocking inhibitory signals. Myeloid cells, such as macrophages, especially in the tumor environment encounter phagocytosis dampening or inhibitory signals, such as CD47 mediated anti-phagocytic activity on target cells, e.g., cancer cells. Chimeric receptors are generated which when expressed in a phagocytic cell blocks CD47 signaling. [0393] In some embodiments, other CAR fusion protein may be designed for expression in a phagocytic cell that may enhance phagocytosis. In one embodiment, provided herein is a composition comprising a recombinant nucleic acid encoding a chimeric antigen receptor (CAR) fusion protein (CFP) comprising: (a) a subunit comprising: (i) an extracellular domain; and (ii) a transmembrane domain; (b) an extracellular antigen binding domain specific to CD47 of a target cell; wherein: the extracellular domain of the subunit and the extracellular antigen binding domain are operatively linked; and the subunit does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase. In some embodiments, the extracellular antigen binding domain is derived from signal- regulatory protein alpha (SIRPα). In some embodiments, the extracellular antigen binding domain is derived from signal-regulatory protein alpha (SIRPβ). In some embodiments, the transmembrane domain is derived from SIRPα. In some embodiments, the transmembrane domain is derived from SIRPβ. [0394] In some embodiments, the additional CAR fusion protein (CFP) may be co-transfected with the recombinant PFP described above. In some embodiments, the scavenger receptor intracellular domain comprises a second intracellular domain comprising a signaling domain that activates phagocytosis; or a proinflammatory domain at the cytoplasmic terminus, which are operably linked. The signaling domain that activates phagocytosis is derived from a receptor selected from the group consisting of the receptors listed in Table 2. [0395] In some embodiments, the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs. ITAMs are conserved sequences present in the cytoplasmic tails of several receptors of the immune system, such as T cell receptors, immunoglobulins (Ig) and FcRs. They have a conserved amino acid sequence motif consisting of paired YXXL/I motifs (Y= Tyrosine, L= Lysine and I= Isoleucine) separated by a defined interval (YXXL/I-X6-8-YXXL/I). In addition, most ITAMs contain a negatively charged amino acid (D/E) in the +2 position relative to the first ITAM tyrosine. Phosphorylation of residues within the ITAM recruits several signaling molecules that activate phagocytosis. ITAM motifs are also present in the intracellular adapter protein, DNAX Activating Protein of 12 kDa (DAP12). [0396] In some embodiments, the phagocytic signaling domain in the intracellular region comprises a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain). The PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR. PI3 kinase recruitment to the binding domain leads to the Akt mediated signaling cascade and activation of phagocytosis. The PI3K-Akt signaling pathway is important in phagocytosis, regulation of the inflammatory response, and other activities, including vesicle trafficking and cytoskeletal reorganization. The PI3kinase recruitment domain is an intracellular domain in a plasma membrane protein, which has tyrosine residues that can be phosphorylated, and which can in turn be recognized by the Src homology domain (SH2) domain of PI3Kp85. The SH2 domain of p85 recognizes the phosphorylated tyrosines on the cytosolic domain of the receptor. This causes an allosteric activation of p110 and the production of phosphatidylinositol-3,4,5-trisphosphate (PIP3) that is recognized by the enzymes Akt and the constitutively active 3′-phosphoinositide-dependent kinase 1 (PDK1) through their plekstrin homology domains. The interaction of Akt with PIP3 causes a change in the Akt conformation and phosphorylation of the residues Thr308 and Ser473 by PDK1 and rictor-mTOR complex, respectively. Phosphorylation of these two residues causes the activation of Akt which in turn phosphorylates, among other substrates, the enzyme glycogen synthase kinase- 3 (GSK-3). GSK-3 has two isoforms, GSK-3α and GSK-3β both of which are constitutively active. The isoforms are structurally related but functionally nonredundant. Inactivation of GSK-3 is observed when the residues Ser21 in GSK-3α or Ser9 in GSK-3β, located in their regulatory N- terminal domains, are phosphorylated by Akt and other kinases. Inhibition of GSK-3 by phosphorylation is important for the modulation of the inflammation and in phagocytosis processes. [0397] In some embodiments, a recombinant PFP comprises (a) an extracellular CD47 binding domain SIRPα, (b) a SIRPβ transmembrane domain, and (c) an intracellular domain of SIRPβ. SIRPβ signaling can activate pro-phagocytic signaling by engaging DAP12 activation. [0398] Various members of the family transduce checkpoint signal upon contact with sialylated glycans on membrane proteins. In some members, the intracellular domains of the Siglec proteins comprise multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs). ITIMs share a consensus amino acid sequence in their cytoplasmic tail, namely (I/V/L/S)-X-Y-X-X-(L/V), where X denotes any amino acid, I= Isoleusine, V=valine, L=Lysine, S=Serine, Y=Tyrosine. Phosphorylation of the Tyrosine residues at the ITIM motif recruit either of two SH2 domain– containing negative regulators: the inositol phosphatase SHIP (Src homology 2–containing inositol polyphosphate 5-phosphatase) or the tyrosine phosphatase SHP-1 (Src homology 2–containing protein tyrosine phosphatase-1). A leucine in the (Y+2) position favors binding to SHIP, whereas an isoleucine in the (Y–2) position favors SHP-1 binding. ITIMs can also bind to another tyrosine phosphatase, SHP-2, but evidence for SHP-2 playing a functional role in ITIM-mediated inhibition is less clear than for the other mediators. Therefore, activation of the Siglec membrane proteins at the extracellular ligand binding domain by binding with a sialic acid residue, (e.g. in sialylated membrane glycan proteins), the ITIMs receive the intracellular signals, which are phosphorylated, and initiate the SHP mediated signaling for immunomodulation, including reduction in phagocytic potential. [0399] In some embodiments the composition described herein comprises a recombinant nucleic acid construct encoding a chimeric Siglec receptor (SgR) fusion protein (SgFP), comprising: (a) a SgR subunit which comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (a) an extracellular domain comprising an antigen binding domain specific to a sialylated glycan of a cell surface protein of a target cell; (b) wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the SgFP does not comprise a functional intracellular domain of an endogenous receptor that binds a sialylated glycan, or (ii) the SgFP comprises an intracellular signaling domain that activates phagocytosis or an inflammatory pathway. In some embodiments, the chimeric receptor is deficient in an intracellular domain, and therefore acts as a blocker for Siglec induced immunoregulatory intracellular signaling. Such is achieved by deletion of the nucleic acid region encoding the intracellular domain and cloning the remainder of the coding sequence of the Siglec receptor. This construct can be designated as a siglec intracellular domain deletion construct [SiglecΔICD]. In some embodiments, the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a cancer antigen specific scFv fused with the extracellular domain (ECD) of a siglec receptor. This allows targetability of the construct to the cancer cell. The chimeric receptor comprises the TM and the ICD of the siglec receptor, which can be the endogenous ICD, or the ICD fused with additional phagocytosis promoting domains, such as PI3K binding domain or the domains. In some embodiment, a chimeric receptor comprising an extracellular siglec domain, is co- expressed with a sialidase. The nucleic acid encoding a sialidase may be incorporated in the expression vector expressing the chimeric domain with a signal sequence for secretion. Since the sialidase is expressed by the same cell that expressed the CAR-siglec receptor, expression of sialidase deprives the ECD of the siglec from binding to its natural ligand, but is activated by the scFv binding to its receptor, thereby ensuring the specificity of action of the chimeric receptor on a cancer-antigen expressing cell. [0400] In some embodiments, the chimeric receptors comprise one or more domains from TREM proteins, fused at the extracellular region with an antigen binding domain that can specifically bind to a cancer antigen, such as a cancer antigen-specific antibody or part or fragment thereof. In some embodiments, recombinant nucleic acids encoding a TREM chimeric antigen receptor encode a fusion proteins that comprises: (a) the at least a TREM transmembrane domain (TM) and a TREM intracellular domain (ICD); and (b) an extracellular domain (ECD) comprising an antigen binding domain that can specifically bind to a cancer antigen. The fusion proteins are designed to target cancer cells and bind to the target cancer cells via the ECD comprising the antigen binding domain, and the binding triggers and enhance phagocytosis via signaling through the TREM TM and/or the intracellular domains. The transmembrane domain of TREM trimerizes with DAP12 transmembrane domains and trigger intracellular pro-phagocytosis signaling cascade. In some embodiments, the TREM domains are contributed by TREM1, or by TREM2, or by TREM3 members. The extracellular antigen binding domain is fused to the extracellular terminus of the TREM domains through a short spacer or linker. [0401] In some embodiments, the extracellular antigen binding domain comprises an antibody, specific to a cancer antigen. In some embodiments, the extracellular antigen binding domain comprises an antibody or an antigen binding part thereof that binds specifically to an antigen on the surface of a cancer cell. [0402] In some embodiments the extracellular antigen binding domain is an antibody specific for a cancer antigen. In some embodiments, the extracellular antigen binding domain is a fraction of an antibody, wherein the fragment can bind specifically to the cancer antigen on a cancer cell. In some embodiments the antigen binding domain comprises a single chain variable fraction (scFv) specific for a cancer antigen binding domain. [0403] In some embodiments, the chimeric fusion protein (CFP) comprises an extracellular domain (ECD) targeted to bind to CD5 (CD5 binding domain), for example, comprising a heavy chain variable region (VH) having an amino acid sequence as set forth in SEQ ID NO: 1. In some embodiments, the chimeric CFP comprises a CD5 binding heavy chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 1. In some embodiments, the extracellular domain (ECD) targeted to bind to CD5 (CD5 binding domain) comprises a light chain variable domain (VL) having an amino acid sequence as set forth in SEQ ID NO: 2. In some embodiments, the chimeric CFP comprises a CD5 binding light chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 2. [0404] In some embodiments, the CFP comprises an extracellular domain targeted to bind to HER2 (HER2 binding domain) having for example a heavy chain variable domain amino acid sequence as set forth in SEQ ID NO: 8 and a light chain variable domain amino acid sequence as set forth in SEQ ID NO: 9. In some embodiments, the CFP comprises a HER2 binding heavy chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 8. In some embodiments, the CFP comprises a HER2 binding light chain variable domain comprising an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 9. [0405] In some embodiments, the CFP comprises a hinge connecting the ECD to the transmembrane (TM). In some embodiments the hinge comprises the amino acid sequence of the hinge region of a CD8 receptor. In some embodiments, the CFP may comprise a hinge having the amino acid sequence set forth in SEQ ID NO: 7 (CD8α chain hinge domain). In some embodiments, the PFP hinge region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 7. [0406] In some embodiments, the CFP comprises a CD8 transmembrane region, for example having an amino acid sequence set forth in SEQ ID NO: 6. In some embodiments, the CFP TM region comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 6. [0407] In some embodiments, the CFP comprises an intracellular domain having an FcR domain. In some embodiments, the CFP comprises an FcR domain intracellular domain comprises an amino acid sequence set forth in SEQ ID NO: 3, or at least a sequence having 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 3. [0408] In some embodiments, the CFP comprises an intracellular domain having a PI3K recruitment domain. In some embodiments the PI3K recruitment domain comprises an amino sequence set forth in SEQ ID NO: 4. In some embodiments the PI3K recruitment domain comprises an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 4. [0409] In some embodiments, the CFP comprises an intracellular domain having a CD40 intracellular domain. In some embodiments the CD40 ICD comprises an amino sequence set forth in SEQ ID NO: 5. In some embodiments the CD40 ICD comprises an amino acid sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 5. Table 4 - Sequences of chimeric antigen receptors and domains thereof [0410] Table 4 shows exemplary sequences of chimeric fusion protein domains and/or fragments thereof that are meant to be non-limiting for the disclosure. Underlines denote the CDR sequences in sequential order of CDR1, CDR2 and CDR3 for the respective heavy and light chains in accordance to the Kabat numbering system.
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Table 5 - Linker sequences
Figure imgf000099_0002
[0411] The instant disclosure therefore provides all the necessary building blocks for a construct design that contemplates using any one of the domains in combination with another to build a chimeric protein with the information provided herein. Any such domain can be used to generate a phagocytic receptor fusion protein, i..e., a CFP, or a CAR or a dual CFP-CAR construct. Characteristics of the PFP: [0412] The PFP structurally incorporates into the cell membrane of the cell in which it is expressed. Specific leader sequences in the nucleic acid construct, such as the signal peptide directs plasma membrane expression of the encoded protein. The transmembrane domain encoded by the construct incorporates the expressed protein in the plasma membrane of the cell. [0413] In some embodiments the transmembrane domain comprises a TM domain of an FcR- alpha receptor, which dimerizes with endogenous FcRγ receptors in the myeloid cells, such as macrophages, ensuring myeloid cell specific expression. [0414] In some embodiments, the PFP renders the cell expressing it as potently phagocytic. When the recombinant nucleic acid encoding the PFP is expressed in a cell, the cell exhibits an increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid. When the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid. In some embodiments, the recombinant nucleic acid when expressed in a cell, the cell exhibits at least 2-fold increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid. In some embodiments, the recombinant nucleic acid when expressed in a cell, the cell exhibits at least 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold 30- fold or at least 5-fold increased phagocytosis of a target cell having the antigen of a target cell, compared to a cell not expressing the recombinant nucleic acid. In some embodiments, the composition comprises a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the PFP to the antigen of the target cell, the killing or phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 11% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 12% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 13% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 14% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 15% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 16% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 17% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 18% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 19% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 30% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 40% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 50% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 60% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 70% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 80% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 90% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 100% compared to a cell not expressing the PFP. [0415] In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 4-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 6-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 8-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 10-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by at least greater than 50-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis activity of a cell expressing the PFP is increased by about 50-fold compared to a cell not expressing the PFP. [0416] In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 10% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 30% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 40% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 50% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 60% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 70% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 80% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 90% compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 100% compared to a cell not expressing the PFP. [0417] In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 4-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 6-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 8-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 10-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 20-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 30-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 40-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 50-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 100-fold compared to a cell not expressing the PFP. In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by about 100-fold compared to a cell not expressing the PFP. [0418] In some embodiments, the phagocytosis associated killing activity of a cell expressing the PFP is increased by at least greater than 2-fold compared to a cell not expressing the PFP. [0419] In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased cytokine production. The cytokine can comprise any one of: IL-1, IL-6, IL-12, IL-23, TNF, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27 and interferons. [0420] In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased cell migration. Enhanced cell migration may be detected in cell culture by standard motility assays. In some embodiments, actin filament rearrangements may be detected and monitored using phalloidin staining and fluorescent microscopy. In some instances, time-lapsed microscopy is used for the purpose. [0421] In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased immune activity. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of MHC II. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of CD80. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased expression of CD86. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits an increased iNOS production. [0422] In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits increased trogocytosis of a target cell expressing the antigen of a target cell compared to a cell not expressing the recombinant nucleic acid. In some embodiments, when the recombinant nucleic acid is expressed in a cell, the cell exhibits less trogocytosis of a target cell expressing the antigen of a target cell as compared to phagocytosis of the cell expressing the recombinant nucleic acid. [0423] In yet other embodiments, a design of a CAR is directed to enhancing a cytotoxic potential of a T cell in which the CAR is expressed. In some embodiments, the cytolytic activity of a T cell expressing a CAR may be at least 2-fold higher than a T cell that does not express a CAR in presence of a target cell. A target cell can be a cell that comprises the target antigen, wherein the extracellular antigen binding domain of the CAR is designed to specifically bind to the target antigen. In some embodiments, the cytolytic activity of a T cell expressing a CAR may be at least 3- fold higher, 4-fold higher, 5-fold higher, 6-fold higher, 7-fold higher, 8-fold higher, 9-fold higher, 10-fold higher, 15-fold higher, 20-fold higher or 50 fold higher than a T cell that does not express a CAR in presence of a target cell. A target cell is a cell that comprises the target antigen, wherein the extracellular antigen binding domain of the CAR is designed to specifically bind to the target antigen. In some embodiments, a target cell is a cancer cell, which expresses a cancer antigen to which the extracellular antigen binding domain of the CAR binds to, thereby activating the T cell and enhancing cytotoxicity. In absence of a CAR, an unmodified T cell, for example can also bind to an potentially destroy the same target cell as a CAR-expressing T cell, provided that the unmodified T cell has a TCR that binds to the target antigen. On the other hand, an unmodified T cell that binds to the same target as the CAR-expressing T cells of the invention, but at least shows a lower potential for target cell cytotoxicity compared to a CAR-expressing cell. [0424] In embodiments, the chimeric receptors may be glycosylated, pegylated, and/or otherwise post-translationally modified. In further embodiments, glycosylation, pegylation, and/or other posttranslational modifications may occur in vivo or in vitro and/or may be performed using chemical techniques. In additional embodiments, any glycosylation, pegylation and/or other posttranslational modifications may be N-linked or O-linked. In embodiments any one of the chimeric receptors may be enzymatically or functionally active such that, when the extracellular domain is bound by a ligand, a signal is transduced to polarize myeloid cells, such as macrophages. Methods for preparing CFPs and engineered myeloid cells [0425] The method for preparing CAR-Ps comprise the steps of (1) screening for PSR subunit framework; (2) screening for antigen binding specificity; (3) CAR-P recombinant nucleic acid constructs; (4) engineering cells and validation. [0426] Screening for PSR subunit framework: As described above, the design of the receptor comprises at least of one phagocytic receptor domain, which enables the enhanced signaling of phagocytosis. In essence a large body of plasma membrane proteins can be screened for novel phagocytic functions or enhancements domains. Methods for screening phagocytic receptor subunits are known to one of skill in the art. Additional information can be found in The Examples section. In general, functional genomics and reverse engineering is often employed to obtain a genetic sequence encoding a functionally relevant protein polypeptide or a portion thereof. In some embodiments, primers and probes are constructed for identification, and or isolation of a protein, a polypeptide or a fragment thereof or a nucleic acid fragment encoding the same. In some embodiments, the primer or probe may be tagged for experimental identification. In some embodiments, tagging of a protein or a peptide may be useful in intracellular or extracellular localization. [0427] Potential antibodies are screened for selecting specific antigen binding domains of high affinity. Methods of screening for antibodies or antibody domains are known to one of skill in the art. Specific examples provide further information. Examples of antibodies and fragments thereof include, but are not limited to IgAs, IgDs, IgEs, IgGs, IgMs, Fab fragments, F(ab')2 fragments, monovalent antibodies, scFv fragments, scRv-Fc fragments, IgNARs, hcIgGs, VHH antibodies, nanobodies, and alphabodies. [0428] Commercially available antibodies can be adapted to generate extracellular domains of a chimeric receptor. Examples of commercially available antibodies include, but are not limited to: anti-HGPRT, clone 13H11.1 (EMD Millipore), anti-ROR1 (ab135669) (Abcam), anti-MUC1 [EP1024Y] (ab45167) (Abcam), anti-MUC16 [X75] (ab1107) (Abcam), anti-EGFRvIII [L8A4] (Absolute antibody), anti-Mesothelin [EPR2685 (2)] (ab134109) (Abcam), HER2 [3B5] (ab16901) (Abcam), anti-CEA (LS-C84299-1000) (LifeSpan BioSciences), anti-BCMA (ab5972) (Abcam), anti-Glypican 3 [9C2] (ab129381) (Abcam), anti-FAP (ab53066) (Abcam), anti-EphA2 [RM-0051- 8F21] (ab73254) (Abcam), anti-GD2 (LS-0546315) (LifeSpan BioSciences), anti-CD19 [2E2B6B10] (ab31947) (Abcam), anti-CD20 [EP459Y] (ab78237) (Abcam), anti-CD30 [EPR4102] (ab134080) (Abcam), anti-CD33 [SP266](ab199432) (Abcam), anti-CD123 (ab53698) (Abcam), anti-CD133 (BioLegend), anti-CD123 (1A3H4) ab181789 (Abcam), and anti-CD171 (L1.1) (Invitrogen antibodies). Techniques for creating antibody fragments, such as scFvs, from known antibodies are routine in the art. [0429] The recombinant nucleic acid can be generated following molecular biology techniques known to one of skill in the art. The methods include but are not limited to designing primers, generating PCR amplification products, restriction digestion, ligation, cloning, gel purification of cloned product, bacterial propagation of cloned DNA, isolation and purification of cloned plasmid or vector. General guidance can be found in: Molecular Cloning of PCR Products: by Michael Finney, Paul E. Nisson, Ayoub Rashtchian in Current Protocols in Molecular Biology, Volume 56, Issue 1 (First published: 01 November 2001); Recombinational Cloning by Jaehong Park, Joshua LaBaer in Current Protocols in Molecular Biology Volume 74, Issue 1 (First published: 15 May 2006) and others. In some embodiments specific amplification techniques may be used, such as TAS technique (Transcription-based Amplification System), described by Kwoh et al. in 1989; the 3SR technique, which are hereby incorporated by reference. (Self-Sustained Sequence Replication), described by Guatelli et al. in 1990; the NASBA technique (Nucleic Acid Sequence Based Amplification), described by Kievitis et al. in 1991; the SDA technique (Strand Displacement Amplification) (Walker et al., 1992); the TMA technique (Transcription Mediated Amplification). [0430] In some embodiments the recombinant nucleic acid sequence is optimized for expression in human. [0431] DNA, mRNA and Circular RNA: In some embodiments, naked DNA or messenger RNA (mRNA) may be used to introduce the nucleic acid inside the cell. In some embodiments, DNA or mRNA encoding the PFP is introduced into the phagocytic cell by lipid nanopaticle (LNP) encapsulation. mRNA is single stranded and may be codon optimized. In some embodiments the mRNA may comprise one or more modified or unnatural bases such as 5’-Methylcytosine, or Pseudouridine. mRNA may be 50-10,000 bases long. In one aspect the transgene is delivered as an mRNA. The mRNA may comprise greater than about 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10,000 bases. In some embodiments, the mRNA may be more than 10,000 bases long. In some embodiments, the mRNA may be about 11,000 bases long. In some embodiments, the mRNA may be about 12,000 bases long. In some embodiments, the mRNA comprises a transgene sequence that encodes a fusion protein. LNP encapsulated DNA or RNA can be used for transfecting myeloid cells, such as macrophages, or can be administered to a subject. [0432] In some embodiments, circular RNA (circRNAs) encoding the PFP is used. In circular RNAs (circRNAs) the 3′ and 5′ ends are covalently linked, constitute a class of RNA. CircRNA may be delivered inside a cell or a subject using LNPs. MECHANISMS OF FURTHER ENHANCEMENT MYELOID CELL FUNCTION: MYELOID CELLS EXPRESSING CHIMERIC ANTIGEN RECEPTOR PROTEIN [0433] Myeloid cells, such as macrophages, especially in the tumor microenvironment encounter phagocytosis dampening or inhibitory signals, such as CD47 mediated anti-phagocytic activity by target cells, e.g., cancer cells. Cluster of Differentiation 47 (CD47) is a receptor belonging to the immunoglobulin superfamily. It can bind to integrins, and thrombospondin 1 (TSP- 1), and is ubiquitously expressed in human cells. Target cells, including tumor cells express CD47 as the ‘don’t eat me’ signals to evade phagocytosis mediated killing and removal. Phagocytic cells can express signal regulatory protein alpha receptors, (SIRP) which bind to CD47. SIRP family members are receptor-type transmembrane glycoproteins involved in the negative regulation of receptor tyrosine kinase-coupled signaling processes. SIRP-α can be phosphorylated by tyrosine kinases. The phospho-tyrosine residues of this PTP have been shown to recruit SH2 domain containing tyrosine phosphatases (PTP), and serve as substrates of PTPs. SIRP-β is another member of the SIRP family, which is found to interact with TYROBP/DAP12, a protein bearing immunoreceptor tyrosine-based activation motifs. It can trigger activation of myeloid cells when associated with TYROBD. This protein was also reported to participate in the recruitment of tyrosine kinase SYK. [0434] In one aspect, provided herein are chimeric receptors generated to functionally block CD47 signaling when expressed in a phagocytic cell. [0435] In another aspect, provided herein are compositions and methods for phagocytic enhancement of the engineered myeloid cells, such as macrophages, by blocking CD47 signal. In some embodiments the recombinant nucleic acids encoding the CFP receptors described herein are transfected or transduced into myeloid cells, such as phagocytic cells, alone or in combination with other recombinant phagocytic receptors for creating engineered macrophages for use in immunotherapy. In some other embodiments, the recombinant phagocytic receptors comprise an intracellular domain of a scavenger receptor. [0436] In some embodiments, an additional binding domain or an additional therapeutic agent comprises a SIRPα antagonist, a SIRPα blocker, an antibody, a chimeric SIRPα receptor, a cytokine, a proinflammatory gene, a procaspase, or an anti-cancer agent. [0437] In some embodiments, an additional binding domain or an additional therapeutic agent comprises a PD1 antagonist, a PD1 blocker, an antibody, a chimeric PD1 receptor, a cytokine, a proinflammatory gene, a procaspase, or an anti-cancer agent. [0438] In some embodiments, provided herein is a composition comprising a recombinant nucleic acid encoding a CFP comprising: (a) a subunit comprising: (i) an extracellular domain that can specifically bind to CD47 on a target cell; and (ii) a transmembrane domain; wherein the extracellular domain of the subunit and the extracellular antigen binding domain are operably linked; and the subunit does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase. In some embodiments, the extracellular antigen binding domain is derived from signal-regulatory protein alpha (SIRPα). In some embodiments, the extracellular antigen binding domain is derived from signal-regulatory protein beta (SIRPβ). SIRPβ does not bind to CD47. In some embodiments, the transmembrane domain is derived from SIRPα. In some embodiments, the transmembrane domain is derived from SIRPβ. The recombinant nucleic acid of this category lacks any intracellular domain, and is therefore a non-signaling receptor and blocker of CD47 signaling. This construct is referred to as SIRP- ^ICD. With the extracellular ligand binding domain of either the SIRPα or the SIRPβ ECD of this chimeric receptor, the receptor binds to CD47 on the target cell, but renders it’s signaling inert by not having a functional SIRPα intracellular domain, thereby reducing CD47 signaling and inhibition of phagocytosis of the CD47+ cells by myeloid cells, such as macrophages, that express the CFP. Over-expression of this construct can largely reduce CD47 mediated anti-phagocytic activity of myeloid cells, such as macrophages, by cancer cells. [0439] In some embodiments endogenous SIRPα may be further inhibited, in addition to overexpression of the SIRP- ^ICD. siRNA can be designed specifically targeting the portion of the mRNA encoding the ICD of SIRPα, such that the siRNA does not reduce or affect the expression of the SIRP- ^ICD. [0440] In some embodiments the SIRP- ^ICD and or the inhibitor of endogenous SIRPα may be expressed in a cell expressing a fusion protein comprising a phagocytic receptor and an extracellular antigen binding domain specific for cancer antigen. Chimeric antigen receptor for blocking anti-phagocytic signal and activating phagocytosis A. Alteration of CD47-binding Signal Transduction [0441] In one aspect, a recombinant nucleic acid is generated, comprising a recombinant nucleic acid encoding a chimeric antigen receptor (CAR) fusion protein (CFP) comprising: (a) a transmembrane domain; (b) an extracellular antigen binding domain specific to CD47 of a target cell; wherein: the transmembrane domain and extracellular antigen binding domain specific to CD47 are operatively linked; and the CFP does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, or does not comprise an intracellular domain that activates a phosphatase, and, (b) an intracellular domain from a phagocytic receptor, that is capable of activating intracellular signaling to enhance phagocytosis. In some embodiments the recombinant nucleic acid construct comprises a nucleic acid sequence encoding an intracellular signaling domain of a scavenger receptor. This class of receptors are termed herein the “switch receptor”, because they are designed to convert (or switch) a phagocytosis inhibitory signal to a phagocytosis promoting signal. In some embodiments, the intracellular domain of the chimeric receptor may comprise the intracellular domain of a scavenger receptor, selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor, which is fused at the extracellular terminus with the extracellular domain comprising the SIRPα CD47 binding domain. [0442] In some embodiments, the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs. ITAMs are conserved sequences present in the cytoplasmic tails of several receptors of the immune system, such as T cell receptors, immunoglobulins (Ig) and FcRs. They have a conserved amino acid sequence motif consisting of paired YXXL/I motifs (Y= Tyrosine, L= Lysine and I= Isoleucine) separated by a defined interval (YXXL/I-X6-8-YXXL/I). In addition, most ITAMs contain a negatively charged amino acid (D/E) in the +2 position relative to the first ITAM tyrosine. Phosphorylation of residues within the ITAM recruits several signaling molecules that activate phagocytosis. ITAM motifs are also present in the intracellular adapter protein, DNAX Activating Protein of 12 kDa (DAP12). [0443] In some embodiments, the phagocytic signaling domain in the intracellular region can comprise a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain). The PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR. PI3 kinase recruitment to the binding domain leads to the Akt mediated signaling cascade and activation of phagocytosis. The PI3K-Akt signaling pathway is important in phagocytosis, regulation of the inflammatory response, and other activities, including vesicle trafficking and cytoskeletal reorganization. The PI3kinase recruitment domain is an intracellular domain in a plasma membrane protein, which has tyrosine residues that can be phosphorylated, and which can in turn be recognized by the Src homology domain (SH2) domain of PI3Kp85. The SH2 domain of p85 recognizes the phosphorylated tyrosines on the cytosolic domain of the receptor. This causes an allosteric activation of p110 and the production of phosphatidylinositol-3,4,5-trisphosphate (PIP3) that is recognized by the enzymes Akt and the constitutively active 3′-phosphoinositide-dependent kinase 1 (PDK1) through their plekstrin homology domains. The interaction of Akt with PIP3 causes a change in the Akt conformation and phosphorylation of the residues Thr308 and Ser473 by PDK1 and rictor-mTOR complex, respectively. Phosphorylation of these two residues causes the activation of Akt which in turn phosphorylates, among other substrates, the enzyme glycogen synthase kinase- 3 (GSK-3). GSK-3 has two isoforms, GSK-3α and GSK-3β both of which are constitutively active. The isoforms are structurally related but functionally nonredundant. Inactivation of GSK-3 is observed when the residues Ser21 in GSK-3α or Ser9 in GSK-3β, located in their regulatory N- terminal domains, are phosphorylated by Akt and other kinases. Inhibition of GSK-3 by phosphorylation is important for the modulation of the inflammation and in phagocytosis processes. [0444] In some embodiments, a recombinant PFP comprises (a) an extracellular CD47 binding domain SIRPα, (b) a SIRPβ transmembrane domain, and (c) an intracellular domain of SIRPβ. SIRPβ signaling can activate pro-phagocytic signaling by engaging DAP12 activation. B. Alteration of Sialic Acid-binding Signal Transduction [0445] In one aspect, disclosed herein are compositions and methods of switching a phagocytosis regulatory signal transduction by members of the Siglec family of membrane proteins that are expressed on immune cells. Various members of the family transduce checkpoint signal upon contact with sialylated glycans on membrane proteins. In some members, the intracellular domains of the Siglec proteins comprise multiple immunoreceptor tyrosine-based inhibitory motifs (ITIMs). ITIMs share a consensus amino acid sequence in their cytoplasmic tail, namely (I/V/L/S)- X-Y-X-X-(L/V), where X denotes any amino acid, I= Isoleusine, V=valine, L=Lysine, S=Serine, Y=Tyrosine. Phosphorylation of the Tyrosine residues at the ITIM motif recruit either of two SH2 domain–containing negative regulators: the inositol phosphatase SHIP (Src homology 2–containing inositol polyphosphate 5-phosphatase) or the tyrosine phosphatase SHP-1 (Src homology 2– containing protein tyrosine phosphatase-1). A leucine in the (Y+2) position favors binding to SHIP, whereas an isoleucine in the (Y–2) position favors SHP-1 binding. ITIMs can also bind to another tyrosine phosphatase, SHP-2, but evidence for SHP-2 playing a functional role in ITIM-mediated inhibition is less clear than for the other mediators. Therefore, activation of the Siglec membrane proteins at the extracellular ligand binding domain by binding with a sialic acid residue, (e.g. in sialylated membrane glycan proteins), the ITIMs receive the intracellular signals, which are phosphorylated, and initiate the SHP mediated signaling for immunomodulation, including reduction in phagocytic potential. [0446] In some embodiments the composition described herein comprises a recombinant nucleic acid construct encoding a chimeric Siglec receptor (SgR) fusion protein (SgFP), comprising: (a) a SgR subunit which comprises: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (a) an extracellular domain comprising an antigen binding domain specific to a sialylated glycan of a cell surface protein of a target cell; (b) wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the SgFP does not comprise a functional intracellular domain of an endogenous receptor that binds a sialylated glycan, or (ii) the SgFP comprises an intracellular signaling domain that activates phagocytosis or an inflammatory pathway. [0447] Siglec family receptors comprise the membrane proteins, siglec 1 (CD169), siglec 2 (CD22), siglec 3 (CD33), siglec 4 (MAG), siglec 5, siglec 6, siglec 7, siglec 8, siglec 9, siglec 10, siglec 11, siglec 12, siglec 13, siglec 14, siglec 15, siglec 16. [0448] In some embodiments the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising an extracellular domain (ECD) of a Siglec receptor that can bind to sialylated residues on membrane proteins of a target cell, which comprises any one of the siglec family members. In some embodiments, the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a transmembrane protein (TM) domain of a Siglec receptor. In some embodiments, the chimeric receptor is deficient in an intracellular domain, and therefore acts as a blocker for Siglec induced immunoregulatory intracellular signaling. Such is achieved by deletion of the nucleic acid region encoding the intracellular domain and cloning the remainder of the coding sequence of the Siglec receptor. This construct can be designated as a siglec intracellular domain deletion construct [Siglec ^ICD]. [0449] In some embodiments the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising an extracellular domain (ECD) of a Siglec receptor that can bind to sialylated residues on membrane proteins of a target cell. In some embodiments, the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a transmembrane protein (TM) domain of a Siglec receptor. [0450] In some embodiments, the chimeric receptor comprises a TM domain of an unrelated membrane protein, for example CD8 TM or CD2 TM domains. In some embodiments the chimeric antigenic receptor comprising the Siglec ECD and/or Siglec TM is deficient in endogenous Siglec intracellular domain (ICD) (e.g., achieved by a deletion of the intracellular domain [SiglecΔICD]), and wherein an intracellular domain of an unrelated protein is fused to the cytoplasmic end of the construct. [0451] Of note, Siglec 2, 3, 5, 6, 7, 8, 9, 10, 11, and 12 family members comprise 2 or more intracellular ITIM motifs. In some embodiments, the intracellular domains of the siglec proteins comprising the ITIM motifs are deleted to generate SiglecΔICD, and fused with an ICD of a phagocytosis promoting protein, thereby altering the inhibitory signal generated by binding of the siglec to its ligand (sialylated glycan) on a cancer cell, into a pro-inflammatory and phagocytosis promoting signal. [0452] The unrelated protein can comprise an intracellular domain that can generate phagocytosis activation signals or pro-inflammatory signals, such as the intracellular domains of the proteins: MRC1, ItgB5, MERTK, ELMO, BAIL Tyro3, Axl, Traf6, Syk, MyD88, Zap70, PI3K, FcyR1, FcyR2A, FcyR2B2, FcyR2C, FcyR3A, FcER1, FcaRl, BAFF-R, DAP12, NFAM1, and CD79b intracellular domains. [0453] In some embodiments, the intracellular domain of the chimeric receptor may comprise the intracellular domain of a scavenger receptor, selected from lectin, dectin 1, mannose receptor (CD206), scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169 receptor, which is fused at the extracellular terminus with the extracellular domain comprising the Siglec sialylated glycan binding domain. [0454] In some embodiments, the phagocytic signaling domain in the intracellular region can comprise a PI3kinase (PI3K) recruitment domain (also called PI3K binding domain). The PI3K binding domains used herein can be the respective PI3K binding domains of CD19, CD28, CSFR or PDGFR. [0455] In some embodiments, the intracellular domain with a phagocytosis signaling domain comprises a domain having one or more Immunoreceptor Tyrosine-based Activation Motif (ITAM) motifs. C. Chimeric Siglec Constructs and Sialidase Co-expression [0456] In some embodiments, the recombinant nucleic acid construct encodes a recombinant chimeric antigenic receptor comprising a cancer antigen specific scFv fused with the extracellular domain (ECD) of a siglec receptor. This allows targetability of the construct to the cancer cell. The chimeric receptor comprises the TM and the ICD of the siglec receptor, which can be the endogenous ICD, or the ICD fused with additional phagocytosis promoting domains, such as PI3K binding domain or the domains. The siglec ECD region is prevented from activation by a concurrently expressed sialidase. The sialidase may be encoded by the construct in a monocistronic design, where the entire construct is expressed as a single polypeptide and is readily cleaved by an endogenous T2A cleavage; or may be bicistronic, where the coding sequence of the sialidase enzyme is transcribed as a separate mRNA. [0457] The siglec receptors bind to sialylated residues ubiquitously present on membrane proteins, and can activate the downstream signals. In some embodiments, the chimeric construct described herein encodes an extracellular domain, a transmembrane domain and an intracellular domain of the siglec protein, but is fused with a cancer targeting scFv at the extracellular terminus, and encodes for a sialidase at the intracellular terminus. [0458] In some embodiments, irrespective of whether the recombinant construct is mono- or polycistronic, the sialidase coding sequence is fused with the coding sequence of an N-terminal signal peptide that signals for secretion of the protein sialidase upon translation. Upon expression of the sialidase and its release into the extracellular environment, the enzyme removes the digests sialic acid residues from the extracellular membrane proteins in the environment proximal to the membrane of the cell expressing the construct. Since the sialidase is expressed by the same cell that expressed the CAR- siglec receptor, expression of sialidase deprives the ECD of the siglec from binding to its natural ligand, but is activated by the scFv binding to its receptor, thereby ensuring the specificity of action of the chimeric receptor on a cancer-antigen expressing cell. [0459] In some embodiments, the chimeric antigenic receptor functions as an anti-inflammatory and phagocytosis regulatory signaling protein by activation of the endogenous ICD domains of the siglec receptor through activation of the ITIMs. [0460] In some embodiments, the chimeric antigenic receptor comprises endogenous ICD domains that have a pro-inflammatory signaling moiety, and/or a depletion of the endogenous siglec ITIM comprising ICDs, each of which are described in the previous section, and can be combined in modular ways with the scFv comprising CAR described herein. Chimeric Antigenic Receptor Domains for Enhancing Phagocytosis TREM Domains [0461] In one aspect, presented herein are recombinant nucleic acids encoding chimeric receptors, having one or more domains from a Triggering Receptor Expressed on Myeloid cells (TREM) receptor, a TREM chimeric receptor. TREM receptors are important regulators of the immune response, due to their ability to either amplify or decrease PRR-induced signals. This family of proteins includes the members: TREM 1, 2, 3. TREMs share common structural properties, including the presence of a single extracellular immunoglobulin-like domain of the V-type, a trans- membrane domain and a short cytoplasmic tail. In particular, the TREM trans-membrane domain (TM) possesses negatively charged residues that interact with the positively charged residues of the DNAX Activating Protein of 12 kDa (DAP12), a trans-membrane adaptor containing an immunoreceptor tyrosine-based activation motif (ITAM). [0462] In some embodiments, recombinant nucleic acids encoding a chimeric antigen receptor comprises sequences that encode at least the TREM TM domain, such that the chimeric receptor interacts with DAP12 and enhance phagocytosis via phosphorylation of residues within the ITAM in DAP12, which recruits several signaling molecules that activate phagocytosis. [0463] In some embodiments, the chimeric receptors comprise one or more domains from TREM proteins, fused at the extracellular region with an antigen binding domain that can specifically bind to a cancer antigen, such as a cancer antigen-specific antibody or part or fragment thereof. [0464] In some embodiments, recombinant nucleic acids encoding a TREM chimeric antigen receptor encode a fusion proteins that comprises: (a) the at least a TREM transmembrane domain (TM) and a TREM intracellular domain (ICD); and (b) an extracellular domain (ECD) comprising an antigen binding domain that can specifically bind to a cancer antigen. The fusion proteins are designed to target cancer cells and bind to the target cancer cells via the ECD comprising the antigen binding domain, and the binding triggers and enhance phagocytosis via signaling through the TREM TM and/or the intracellular domains. The transmembrane domain of TREM trimerizes with DAP12 transmembrane domains and trigger intracellular pro-phagocytosis signaling cascade. In some embodiments, the TREM domains are contributed by TREM1, or by TREM2, or by TREM3 members. The extracellular antigen binding domain is fused to the extracellular terminus of the TREM domains through a short spacer or linker. [0465] In some embodiments, the extracellular antigen binding domain comprises an antibody, specific to a cancer antigen. In some embodiments, the extracellular antigen binding domain comprises an antibody or an antigen binding part thereof that binds specifically to an antigen on the surface of a cancer cell. [0466] In some embodiments the extracellular antigen binding domain is an antibody specific for a cancer antigen. In some embodiments, the extracellular antigen binding domain is a fraction of an antibody, wherein the fragment can bind specifically to the cancer antigen on a cancer cell. In some embodiments the antigen binding domain comprises a single chain variable fraction (scFv) specific for a cancer antigen binding domain. FcR domains [0467] In some aspects, described herein are recombinant nucleic acids encoding chimeric antigen receptors that comprise an FcR domain. In some embodiments, the chimeric receptors described herein may comprise a FcRα (FcRα1) domain. The FcRα1 transmembrane domain heterodimerizes with FcRγ transmembrane domains in the myeloid cells, such as macrophages, and other phagocytic cells, such as mast cells, and the heterodimerization is required for expression of the protein on the cell surface. In some embodiments, expression of a recombinant protein that comprises the FcRα1 TM domain is restricted to expression in cells that naturally express the FcγR. In this respect, a recombinant chimeric protein having the FcRα1 TM domain is precluded for expressing in any cells other than the phagocytic cells that express the FcRγ. Similarly, FcRβ expression is restricted to mast cells. In some embodiments, the chimeric receptors are designed with one or more domains comprising the FcRα1 TM domain for myeloid cell specific expression of the chimeric protein. In some embodiments, the chimeric receptors are designed with one or more domains comprising the FcRβ TM domain for mast-cell specific expression of the chimeric protein. Pro-caspases domains [0468] In some aspects, described herein are recombinant nucleic acids encoding chimeric antigen receptors and a fusion protein comprising: an Src Homology 2 domain (SH2) l linked at the C terminus with a caspase cleavage sequence and a sequence encoding a Procaspase (SH2-ccs- Procaspase). In some embodiments, the recombinant nucleic acid encodes chimeric antigen receptor which comprises a first polypeptide (a) comprising: an extracellular antigen binding domain that can specifically bind to a cancer antigen, a transmembrane domain and an intracellular signaling domain comprising an ITAM motif that can trigger or enhance phagocytosis, and a second polypeptide (b) an intracellular polypeptide comprising the Src Homology 2 domain (SH2) l linked at the C terminus with a caspase cleavage sequence and a sequence encoding a Procaspase (SH2-ccs-Procaspase). The recombinant nucleic acid may be monocistronic or polycistronic. In some embodiments the recombinant nucleic acid is monocistronic and the sequence encoding (a) and the sequence encoding (b) are separated by a T2A sequence that cleaves the two polypeptides endogenously after translation. The Procaspase can be a Procaspase 1, Procaspase 2 or Procaspase 3. Upon expression of and cleavage of (a) and (b), the SH2 domain of the SH2-ccs-Procaspase directs the Procaspase to the phosphorylated intracellular domain of the ITAM motif of (a), and is activated. Activation of the SH2 domain in contact with the phosphorylated ITAM residues activates the proteolytic cleavage of Procaspase to generate activated caspase which are required to digest phagocytosed cells. Ubiquitylation Activation Domains [0469] In some embodiments, intracellular signaling domains may be added to the chimeric receptor to enhance phagocytotic signaling. Ubiquitin signaling is involved in triggering of phagocytosis. Monoubiquitylation of endocytic receptors can target them for lysosomal degradation. IL-4 signaling mediated polyubiquitination of scavenger receptor MSR1 at the intracellular domain can lead to activation of the receptor, and trigger its interaction with MAP kinase pathway proteins that are involved in inducing inflammatory gene activation. For example, K63 polyubiquitylation of the MSR1 protein leads to its interaction with TAK1/MKK7/JNK in the phagosomes. Ubiquitylation of K27 residue of MSR1 is also implicated in MSR signaling and pro-inflammatory gene activation, such as expression of pro-inflammatory cytokines. Ubiquitin E3 ligase can promote ligation of ubiquitin to specific lysine residues on the intracellular signaling domain of MSR1, which is activated upon IL4 activation, and which in turn can activate intracellular signaling by binding of TAK1, MKK7 and JNK, and triggering expression of pro-inflammatory genes, such as TNF-alpha and IL-1 ^. [0470] In some embodiments the recombinant chimeric antigen receptors described herein may comprise an intracellular domain which comprises residues that can be ubiquitylated and activated to generate a proinflammatory signal. [0471] In some embodiments the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain of MSR1 comprising the residues that can undergo E3 ligase mediated ubiquitylation. [0472] In some embodiments, the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to TAK1. In some embodiments, the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a MAP kinase protein, such as MKK7. In some embodiments, the intracellular domain of a chimeric antigen receptors described herein comprises the intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that comprises a MAP kinase protein, such as MKK7. In some embodiments, the intracellular domain of a chimeric antigen receptors described herein comprises an intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that comprises a JNK. In some embodiments, the intracellular domain of a chimeric antigen receptors described herein comprises an intracellular domain which can be ubiquitylated, and which upon ubiquitylation can bind to a kinase or a protein complex that can activate pro-inflammatory gene transcription. [0473] As contemplated herein, a suitable ubiquitylation domain can be ligated at the intracellular portion of any one of the CARP receptors described in the disclosure. Coexpression with other chimeric receptors for phagocytosis [0474] In some embodiments, one or more CFPs described herein may be co-expressed with a recombinant phagocytic receptor fusion protein (PFP) that has an extracellular domain comprising an antigen binding domain that can specifically bind to a cancer antigen on the surface of a cancer cell. The fusion protein further comprises a transmembrane domain and an intracellular phagocytosis receptor domain, may comprise and further phagocytosis signaling enhancement domains, such as kinase binding domains. The PFP is specifically designed to target a cancer cell and activate phagocytosis upon binding to the target. The phagocytosis enhancement by PFP is augmented by co- expression of the CFP. For example, cells coexpressing the PFP with the SIRP-CFPs enhance the phagocytic potential of the cells, wherein additionally the PFPs provide specific cancer targeting to the cells. For example, cells coexpressing the PFP with SH2-ccs-Procaspase have specific targeting to cancer cells with the PFP, whereas the phagocytic activity is enhanced by the PFP intracellular domains, and cancer cell killing activity is enhanced due to clearance of apoptotic cells by SH2-ccs- Procaspase via Procaspase activation to caspase. [0475] In some embodiments, the scavenger receptor intracellular domain comprises a second intracellular domain comprising a signaling domain that activates phagocytosis; or a proinflammatory domain at the cytoplasmic terminus, which are operably linked. As the CD47 binding domain is operably linked with the one or more intracellular signaling domains of the phagocytic receptor, the signaling event originating from the engagement of the CD47 ligand at the extracellular end is altered upon passage through the intracellular domains to phagocytosis enhancing signal at the intracellular end of the recombinant receptor instead of the phagocytosis inhibition signal of a native CD47-binding SIRPα receptor. The phagocytic potential of the cell expressing the recombinant receptor is highly enhanced, or over a cell expressing SIRP- ^ICD. [0476] In some embodiments, the intracellular phagocytosis signaling domain may comprise domains selected from MRC1, ItgB5, MERTK, ELMO, BAIL Tyro3, Axl, Traf6, Syk, MyD88, Zap70, PI3K, FcyR1, FcyR2A, FcyR2B2, FcyR2C, FcyR3A, FcER1, FcaRl, BAFF-R, DAP12, NFAM1, and CD79b intracellular domains. [0477] In one aspect, the function of the chimeric receptor may be further augmented by expressing an additional recombinant protein in the myeloid cell concurrently with or independent of the CFP expression. In some embodiments the additional recombinant protein is co-expressed with the CFP described above, wherein the CFP can bind to an antigen that is expressed in the target cell, and the CFP enhances phagocytosis and killing of the target cell by the myeloid cell expressing the CFP. In some embodiments, the additional recombinant protein is designed to further enhance the functioning of the CFP expressing cell. [0478] In some embodiments, the additional recombinant protein is a second chimeric protein, such as a chimeric fusion protein, for example, a second chimeric protein. In some embodiments, the second chimeric protein is expressed in population of myeloid cells that expresses the cancer antigen targeting CFP. In some embodiments the additional recombinant protein is a second chimeric fusion protein or phagocytosis receptor fusion protein (PFP). In some embodiments, the second chimeric protein is a truncated protein. Exemplary additional recombinant constructs for augmenting phagocytosis and killing by myeloid cells: [0479] In one aspect, provided herein is a composition comprising a recombinant nucleic acid encoding a chimeric CD47 receptor (CR) fusion protein comprising: (a) a CR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to CD47 of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein: (i) the CR does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, (ii) the CR comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase, or (iii) the CR comprises an intracellular signaling domain derived from a phagocytic receptor. In some embodiments, the CR does not comprise a functional intracellular domain of an endogenous receptor that binds CD47. [0480] In some embodiments, the CR comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase. In some embodiments, the CR comprises an intracellular signaling domain derived from a phagocytic or tethering receptor. [0481] In some embodiments, the antigen binding domain specific to CD47 comprises an antibody domain specific to CD47. [0482] In some embodiments, the antigen binding domain specific to CD47 comprises an extracellular domain derived from SIRPα or SIRPβ. [0483] In some embodiments, the CR comprises an intracellular domain comprising an intracellular signaling domain. [0484] In one aspect, provided herein is a composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; wherein the intracellular signaling domain is derived from a phagocytic receptor; and wherein the recombinant nucleic acid encodes: (A) encodes a pro-inflammatory polypeptide, (B) comprises a binding motif for a molecule, such that upon binding of the molecule to the binding motif translation of the recombinant nucleic acid is inhibited (C) a procaspase domain, or (D) a procaspase binding domain, or (E) a sialidase. [0485] In one aspect, provided herein is a composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: a transmembrane domain, and an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; an wherein the PFP forms a functional complex with FcRγ when expressed in a cell. [0486] In one aspect, provided herein is a composition comprising a recombinant nucleic acid encoding a phagocytic receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; an (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the PFP forms a functional complex with DAP12 when expressed in a cell. [0487] In some embodiments, the recombinant nucleic acid encodes: (A) encodes a pro- inflammatory polypeptide, (B) comprises a binding motif for a molecule, such that upon binding of the molecule to the binding motif translation of the recombinant nucleic acid is inhibited, (C) a procaspase domain, or (D) a procaspase binding domain. [0488] In some embodiments, the PFP forms a functional complex with FcRγ when expressed in a cell. [0489] In some embodiments, the PFP forms a functional complex with DAP12 when expressed in a cell. [0490] In some embodiments, the PFP comprises an antigen binding domain that binds to a CD47 ligand, but does not comprise a functional intracellular domain of an endogenous receptor that binds CD47, (i) the PFP comprises a phosphatase inactivation domain or does not comprise an intracellular domain that activates a phosphatase, or (ii) the PFP comprises an intracellular signaling domain derived from a phagocytic receptor. [0491] In some embodiments, the CFP or the PFP forms a functional complex with an FcR. [0492] In some embodiments, the CFP or the PFP forms a functional complex with FcRγ. [0493] In some embodiments, the CFP or the PFP comprises a transmembrane domain of FcR- alpha or FcRβ. [0494] In some embodiments, the CFP or the PFP forms a functional complex with a TREM. [0495] In some embodiments, the CFP or the PFP comprises an intracellular domain comprising an ITAM. [0496] In some embodiments, the CFP or the PFP comprises an intracellular domain comprising an ITIM. [0497] In some embodiments, a cell expressing the CFP or the PFP exhibits inhibits CD47 mediated anti-phagocytosis activity. [0498] In some embodiments, the truncated SIRPα binds to CD47 and inhibits. [0499] In some embodiments, the binding motif is in a UTR region of the recombinant nucleic acid. In some embodiments, the binding motif is an ARE sequence. [0500] In some embodiments, the transmembrane domain binds to a transmembrane domain of DAP12. [0501] In some embodiments, the transmembrane domain that binds to a transmembrane domain of DAP12 is derived from TREM. [0502] In some embodiments, the CFP or the PFP comprises an intracellular domain derived from a DAP12 monomer. [0503] In some embodiments, the recombinant polynucleotide further comprises a sequence encoding a first modified signal regulatory protein α (SIRPα). [0504] In some embodiments, the first modified SIRPα lacks an intracellular signaling domain of a wild type SIRPα. [0505] In some embodiments, the first modified SIRPα lacks an intracellular signaling domain of a wild type SIRPβ. [0506] In some embodiments, the CFP or the PFP does not transmit a signal to block phagocytosis. [0507] In some embodiments, the composition further comprises a polynucleotide encoding a second modified signal regulatory protein α (SIRPα), wherein the second modified SIRPα comprises a PI3K binding domain [0508] In some embodiments, the PI3K binding domain is derived from CD19, CD28, CSFR or PDGFR. [0509] In some embodiments, the composition further comprises a polynucleotide encoding a third modified signal regulatory protein α (SIRPα), wherein the third modified SIRPα comprises a pro-inflammatory domain. [0510] In some embodiments, the composition further comprises a polynucleotide encoding a recombinant SIRP, wherein the recombinant SIRP comprises an extracellular domain derived from SIRPα, a transmembrane domain derived from SIRPβ, and an intracellular domain derived from SIRPβ. [0511] In some embodiments, the composition further comprises when the recombinant SIRP binds to an antigen CD47, the third modified SIRPα does not transmit a signal to block phagocytosis. [0512] In some embodiments, the anti-CD47 binding domain is derived from signal-regulatory protein alpha (SIRPα) or signal-regulatory protein beta (SIRPβ). [0513] In some embodiments, upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. [0514] In some embodiments, the intracellular signaling domain is derived from a phagocytic or tethering receptor. [0515] In some embodiments, the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from any one of the receptors listed in Table 2. [0516] In some embodiments, the intracellular signaling domain is derived from a receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), and CD169. [0517] In some embodiments, wherein the intracellular signaling domain comprises a pro- inflammatory signaling domain. [0518] In some embodiments, the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. [0519] In some embodiments, the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. [0520] In some embodiments, upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. [0521] In some embodiments, the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of any one of the proteins listed in Table 1. In some embodiments, the intracellular signaling domain of any one of the PFPs described herein is derived from a T cell receptor component or tethering receptor or wherein the intracellular signaling domain comprises a T cell activation domain. [0522] In some embodiments, the intracellular signaling domain comprises a PI3K recruitment domain. [0523] In some embodiments, the CFP or the PFP functionally incorporates into a cell membrane of a cell when the CFP or the PFP is expressed in the cell. [0524] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP. [0525] In some embodiments, a cell expressing the CFP or the PFP exhibits at least a 1.1-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP. [0526] In some embodiments, a cell expressing the CFP or the PFP exhibits at least a 2-fold, 3- fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP. [0527] In some embodiments, the target cell expressing the antigen is a cancer cell. [0528] In some embodiments, the target cell expressing the antigen is at least 0.8 microns in diameter. [0529] In some embodiments, the intracellular signaling domain is derived from a scavenger receptor. [0530] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in production of a cytokine compared to a cell not expressing the CFP or the PFP. [0531] In some embodiments, the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof. [0532] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in effector activity compared to a cell not expressing the CFP or the PFP. [0533] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in cross- presentation compared to a cell not expressing the CFP or the PFP. [0534] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the CFP or the PFP. [0535] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD80 compared to a cell not expressing the CFP or the PFP. [0536] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD86 compared to a cell not expressing the CFP or the PFP. [0537] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the CFP or the PFP. [0538] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of TRAIL/TNF Family death receptors compared to a cell not expressing the CFP or the PFP. [0539] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of B7-H2 compared to a cell not expressing the CFP or the PFP. [0540] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of LIGHT compared to a cell not expressing the CFP or the PFP. [0541] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of HVEM compared to a cell not expressing the CFP or the PFP. [0542] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD40 compared to a cell not expressing the CFP or the PFP. [0543] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of TL1A compared to a cell not expressing the CFP or the PFP. [0544] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of 41BBL compared to a cell not expressing the CFP or the PFP. [0545] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of OX40L compared to a cell not expressing the CFP or the PFP. [0546] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of GITRL death receptors compared to a cell not expressing the CFP or the PFP. [0547] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD30L compared to a cell not expressing the CFP or the PFP. [0548] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of TIM4 compared to a cell not expressing the CFP or the PFP. [0549] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of TIM1 Ligand compared to a cell not expressing the CFP or the PFP. [0550] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of SLAM compared to a cell not expressing the CFP or the PFP. [0551] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD48 compared to a cell not expressing the CFP or the PFP. [0552] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD58 compared to a cell not expressing the CFP or the PFP. [0553] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD155 compared to a cell not expressing the CFP or the PFP. [0554] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of CD112 compared to a cell not expressing the CFP or the PFP. [0555] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in expression of PDL1compared to a cell not expressing the CFP or the PFP. [0556] In some embodiments, provided herein is the composition of any of B7-DC compared to a cell not expressing the CFP or the PFP. [0557] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in respiratory burst compared to a cell not expressing the CFP or the PFP. [0558] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in ROS production compared to a cell not expressing the CFP or the PFP. [0559] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in iNOS production compared to a cell not expressing the CFP or the PFP. [0560] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in iNOS production compared to a cell not expressing the CFP or the PFP. [0561] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in extra- cellular vesicle production compared to a cell not expressing the CFP or the PFP. [0562] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in trogocytosis with a target cell expressing the antigen compared to a cell not expressing the CFP or the PFP. [0563] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in resistance to CD47 mediated inhibition of phagocytosis compared to a cell not expressing the CFP or the PFP. [0564] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the CFP or the PFP. [0565] In some embodiments, the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain. [0566] In some embodiments, the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the CFP or the PFP. [0567] In some embodiments, the intracellular domain comprises an F-actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain. [0568] In some embodiments, a cell expressing the CFP or the PFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production. [0569] In some embodiments, the extracellular domain comprises an Ig binding domain. [0570] In some embodiments, the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, FcγRI, FcγRIIA, FcγRIIB, FcγRIIC, FcγRIIIA, FcγRIIIB, FcRn, TRIM21, FcRL5 binding domain. [0571] In some embodiments, the extracellular domain comprises an FcR extracellular domain. [0572] In some embodiments, the extracellular domain comprises an FcRα, FcRβ, FcRε or FcRγ extracellular domain. [0573] In some embodiments, the extracellular domain comprises FcRα (FCAR) extracellular domain. [0574] In some embodiments, the extracellular domain comprises an FcRβ extracellular domain. [0575] In some embodiments, the extracellular domain comprises an FcRε (FCER1A) extracellular domain. [0576] In some embodiments, the extracellular domain comprises an FcRγ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, FCGR3B) extracellular domain The composition of any one of the preceding claims, wherein the extracellular domain comprises an integrin domain. [0577] In some embodiments, the extracellular domain comprises one more integrin α1,α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αIIb, αD, αE, αL, αM, αV, αX, β1, β2, β3, β4, β5, β6, β7, β8 domain. [0578] In some embodiments, the intracellular domain comprises a CD47 inhibition domain. [0579] In some embodiments, the PSR subunit further comprises an extracellular domain operatively linked to the transmembrane domain and the extracellular antigen binding domain. In some embodiments, the extracellular domain further comprises an extracellular domain of a receptor, a hinge, a spacer or a linker. In some embodiments, the extracellular domain comprises an extracellular portion of a PSR. In some embodiments, the extracellular portion of the PSR is derived from the same PSR as the PSR intracellular signaling domain. In some embodiments, the extracellular domain comprises an extracellular domain of a scavenger receptor or an immunoglobulin domain. [0580] In some embodiments, the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region. In some embodiments, the extracellular domain comprises a phagocytic engulfment marker. [0581] In some embodiments, the extracellular domain comprises a structure capable of multimeric assembly. In some embodiments, the extracellular domain comprises a scaffold for multimerization. [0582] In some embodiments, the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. In some embodiments, the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length. [0583] In some embodiments, the extracellular antigen binding domain specifically binds to the antigen of a target cell. In some embodiments, the extracellular antigen binding domain comprises an antibody domain. In some embodiments, the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single-domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof. [0584] In some embodiments, the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor. [0585] In some embodiments, the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen. [0586] In some embodiments, the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen. In some embodiments, the antigen is a cancer antigen or a pathogenic antigen or an autoimmune antigen. In some embodiments, the antigen comprises a viral antigen. In some embodiments, the antigen is a T-lymphocyte antigen. In some embodiments, the antigen is an extracellular antigen. In some embodiments, the antigen is an intracellular antigen. [0587] In some embodiments, the antigen is selected from the group consisting of Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), Mesothelin, EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B-Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2, a Natural Killer Group 2D (NKG2D) ligand, Disialoganglioside 2 (GD2), CD2, CD3, CD4, CD5, CD7, CD8, CD19, CD20, CD22, CD24, CD30, CD33, CD38, CD44v6, CD45, CD56CD79b, CD97, CD117, CD123, CD133, CD138, CD171, CD179a, CD213A2, CD248, CD276, PSCA, CS-1, CLECL1, GD3, PSMA, FLT3, TAG72, EPCAM, IL-1, an integrin receptor, PRSS21, VEGFR2, PDGFR-β, SSEA-4, EGFR, NCAM, prostase, PAP, ELF2M, GM3, TEM7R, CLDN6, TSHR, GPRC5D, ALK, IGLL1 and combinations thereof. In some embodiments, the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56. [0588] In some embodiments, the antigen is an ovarian cancer antigen or a T lymphoma antigen. In some embodiments, the antigen is an integrin receptor. In some embodiments, the antigen is an integrin receptor selected from the group consisting of α1, α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αD, αE, αL, αM, αV, αX, β 1, β 2, β 3, β 4, β 5, β 6, β 7, and β8. [0589] In some embodiments, the transmembrane domain and the extracellular antigen binding domain is operably linked through a linker. In some embodiments, the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker such as the hinge region of CD8α, IgG1 or IgG4. [0590] In some embodiments, the extracellular domain comprises a multimerization scaffold. [0591] In some embodiments, the transmembrane domain comprises an FcR transmembrane domain. [0592] In some embodiments, the transmembrane domain comprises an FcR-alpha, FcRβ or FcRγ transmembrane domain. [0593] In some embodiments, the transmembrane domain comprises an FcαR (FCAR) transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRε (FCER1A) transmembrane domain. In some embodiments, the transmembrane domain comprises an FcRγ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B) transmembrane domain. [0594] In some embodiments, the transmembrane domain comprises a T cell Receptor subunit, CD3 epsilon, CD3 gamma and CD3 delta, CD45, CD2 CD4, CD5, CD8, CD9, CD16, CD19, CD22, CD33, CD28, CD30, CD37, CD64, CD80, CD86, CD134, CD137 and CD 154, or a functional fragment thereof, or an amino acid sequence having at least one, two or three modifications but not more than 20, 10 or 5 modifications transmembrane domain. [0595] In some embodiments, the transmembrane domain comprises a transmembrane domain from a syntaxin such as syntaxin 3 or syntaxin 4 or syntaxin 5. [0596] In some embodiments, the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the CR or the PFP is expressed in a cell. [0597] In some embodiments, the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the CR or the PFP is expressed in a cell. [0598] In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an endogenous receptor when the CFP or the PFP is expressed in a cell. [0599] In some embodiments, the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the CFP or the PFP is expressed in a cell. [0600] In some embodiments, the transmembrane domain is derived from a protein that is different than the protein from which the intracellular signaling domain is derived. [0601] In some embodiments, the transmembrane domain is derived from a protein that is different than the protein from which the extracellular domain is derived. [0602] In some embodiments, the transmembrane domain comprises a transmembrane domain of a phagocytic receptor. [0603] In some embodiments, the transmembrane domain and the extracellular domain are derived from the same protein. [0604] In some embodiments, the transmembrane domain is derived from the same protein as the intracellular signaling domain. [0605] In some embodiments, the recombinant nucleic acid encodes a DAP12 recruitment domain. [0606] In some embodiments, the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12. [0607] In some embodiments, the transmembrane domain is at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. [0608] In some embodiments, the transmembrane domain is at most 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. [0609] In some embodiments, the intracellular domain comprises a phosphatase inhibition domain. [0610] In some embodiments, the intracellular domain comprises an ARP2/3 inhibition domain. [0611] In some embodiments, the intracellular domain comprises at least one ITAM domain. [0612] In some embodiments, the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 ITAM domains. [0613] In some embodiments, the intracellular domain further comprises at least one ITAM domain. In some embodiments, the intracellular domain further comprises at least one ITAM domain select from a group CD3 zeta TCR subunit, CD3 epsilon TCR subunit, CD3 gamma TCR subunit, CD3 delta TCR subunit, TCR zeta chain, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278, CD66d, functional fragments thereof, and amino acid sequences thereof having at least one but not more than 20 modifications thereto. [0614] In some embodiments, the at least one ITAM domain comprises a Src-family kinase phosphorylation site. In some embodiments, the at least one ITAM domain comprises a Syk recruitment domain. In some embodiments, the intracellular domain comprises an F-actin depolymerization activation domain. [0615] In some embodiments, the intracellular domain comprises residues that can be ubiquitylated. In some embodiments, the intracellular domain can bind to an E3 ubiquitin ligase. [0616] In some embodiments, the intracellular domain can bind to a TAK1 kinase. [0617] In some embodiments, the intracellular domain can bind to a MAP kinase or a MAP kinase family member. [0618] In some embodiments, the intracellular domain can be activated upon ubiquitylation and can activate intracellular signaling resulting in pro-inflammatory gene transcription. [0619] In some embodiments, the intracellular domain lacks enzymatic activity. [0620] In some embodiments, the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. [0621] In some embodiments, the intracellular domain comprises a CD47 inhibition domain [0622] In some embodiments the intracellular signaling domain comprises a domain that activates integrin such as the intracellular region of PSGL-1. [0623] In some embodiments, the intracellular signaling domain comprises a domain that activates Rap1 GTPase, such as that from EPAC and C3G. [0624] In some embodiments, the intracellular signaling domain is from Paxillin. In some embodiments, the intracellular signaling domain activates focal adhesion kinase. In some embodiments, the intracellular signaling domain is derived from a single phagocytic receptor. In some embodiments, the intracellular signaling domain is derived from a single scavenger receptor. In some embodiments, the intracellular domain further comprises a phagocytosis enhancing domain. In some embodiments, the intracellular domain comprises a pro-inflammatory signaling domain. In some embodiments, the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain. In some embodiments, the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain. In some embodiments, the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, an NLRP family member, NLRP1- 14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40 or any combination thereof. In some embodiments, the CFP or the PFP does not comprise a full length intracellular signaling domain. In some embodiments, the pro-inflammatory signaling domain comprises an intracellular signaling domain from CD3-zeta, CD3-epsilon, CD3-delta or CD3-gamma. In some embodiments, the pro-inflammatory signaling domain comprises an intracellular signaling domain from TCR- alpha, TCR-beta, TCR-gamma or TCR-delta. [0625] In some embodiments, the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. [0626] In some embodiments, the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. [0627] In some embodiments, the recombinant nucleic acid encodes an FcRα chain extracellular domain, an FcRα chain transmembrane domain and/or an FcRα chain intracellular domain. In some embodiments, the recombinant nucleic acid encodes an FcRβ chain extracellular domain, an FcRβ chain transmembrane domain and/or an FcRβ chain intracellular domain. In some embodiments, the FcRα chain or the FcRβ chain forms a complex with FcRγ when expressed in a cell. In some embodiments, the FcRα chain or FcRβ chain forms a complex with endogenous FcRγ when expressed in a cell. In some embodiments, the FcRα chain or the FcRβ chain does not incorporate into a cell membrane of a cell that does not express FcRγ. In some embodiments, the CFP or the PFP does not comprise an FcRα chain intracellular signaling domain. In some embodiments, the CFP or the PFP does not comprise an FcRβ chain intracellular signaling domain. [0628] In some embodiments, the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain. In some embodiments, the TREM is TREM1, TREM 2 or TREM 3. [0629] In some embodiments, the recombinant nucleic acid comprises a sequence encoding a pro-inflammatory polypeptide. In some embodiments, the composition further comprises a pro- inflammatory polypeptide. [0630] In some embodiments, the pro-inflammatory polypeptide is a chemokine, cytokine and nucleotides. In some embodiments, In some embodiments, the chemokine is selected from the group consisting of CCL2, CXCL1, CXCL12, CXCL9, CXCL10, CXCL11. [0631] In some embodiments, the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon. [0632] In some embodiments, the recombinant nucleic acid comprises a homeostatic regulator of inflammation. [0633] In some embodiments, the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA. In some embodiments, the sequence in the UTR is a sequence that binds to an RNA binding protein. In some embodiments, translation is inhibited or prevented upon binding of the RNA binding protein to the sequence in an untranslated region (UTR). In some embodiments, the sequence in the UTR comprises a consensus sequence of WWWU(AUUUA)UUUW, wherein W is A or U. [0634] In some embodiments, the recombinant nucleic acid is expressed on a bicistronic vector. [0635] In some embodiments, the target cell is a mammalian cell. In some embodiments, the target cell is a human cell. In some embodiments, the target cell comprises a cell infected with a pathogen. In some embodiments, the target cell is a cancer cell. In some embodiments, the target cell is a cancer cell that is a lymphocyte. In some embodiments, the target cell is a cancer cell that is an ovarian cancer cell. In some embodiments, the target cell is a cancer cell that is an ovarian pancreatic cell. In some embodiments, the target cell is a cancer cell that is a glioblastoma cell. [0636] In some embodiments, the recombinant nucleic acid is DNA. In some embodiments, the recombinant nucleic acid is RNA. In some embodiments, the recombinant nucleic acid is mRNA. In some embodiments, the recombinant nucleic acid is a circRNA. In some embodiments, the recombinant nucleic acid is a tRNA. In some embodiments, the recombinant nucleic acid is a microRNA. [0637] Provided herein is a vector comprising the composition described above. In some embodiments, vector is viral vector. In some embodiments, the viral vector is retroviral vector or a lentiviral vector. In some embodiments, the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector is polycistronic. In some embodiments, each of the at least one nucleic acid sequence is operably linked to a separate promoter. In some embodiments, the vector further comprises one or more internal ribosome entry sites (IRESs). In some embodiments, the vector further comprises a 5’UTR and/or a 3’UTR flanking the at least one nucleic acid sequence encoding one or more polypeptides. In some embodiments, the vector further comprises one or more regulatory regions. [0638] Provided herein is a polypeptide encoded by the recombinant nucleic acid of the composition described above. [0639] In one aspect, provided herein is a cell comprising a vector described above or the polypeptide described above. In some embodiments, the cell is a phagocytic cell. In some embodiments, the cell is a stem cell derived cell, myeloid cell, macrophage, a dendritic cell, lymphocyte, mast cell, monocyte, neutrophil, microglia, or an astrocyte. In some embodiments, the cell is an autologous cell. In some embodiments, the cell is an allogeneic cell. In some embodiments, the cell is an M1 myeloid cell, such as a macrophage. In some embodiments, the cell is an M2 myeloid cell, such as a macrophage. In some embodiments, the cell is a precursor cell of myeloid lineage. In some embodiments, the myeloid cell is a CD14+ cell. In some embodiments, the myeloid cell is a CD16- cell. In some embodiments, the myeloid cell is a CD14+ and CD16- cell. [0640] In some embodiments, the therapeutic agent comprises a T cell such as described herein. In some embodiments, the T cell is a T cell precursor. In some embodiments, the T cell is an undifferentiated and/or unpolarized. In some embodiments, the T cell is a CD4 T cells. In some embodiments, T cell is a CD8 T cell. In some embodiments, T cell is gamma delta T cell. In some embodiments, the cell is a NK T cell. [0641] Provided herein is a pharmaceutical composition comprising (a) the nucleic acid composition or the vector or the polypeptide or the cell as described above; and (b) a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition further comprising an additional therapeutic agent. In some embodiments, the pharmaceutical composition comprises additional therapeutic agent which is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the CFP or the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. [0642] In some embodiments, the pharmaceutically acceptable excipient comprises serum free media, a lipid, or a nanoparticle. [0643] Provided herein is a method of treating a disease in a subject in need thereof comprising administering to the subject the pharmaceutical composition described herein. [0644] In some embodiments, the disease is cancer. In some embodiments, the cancer is a solid cancer. In some embodiments, the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer. In some embodiments, the cancer is a liquid cancer. In some embodiments, the liquid cancer is a leukemia or a lymphoma. In some embodiments, the liquid cancer is a T cell lymphoma. In some embodiments, the disease is a T cell malignancy. In some embodiments the method further comprises administering an additional therapeutic agent to the subject. In some embodiments, the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F- actin disassembly, an agent that promotes PI3K recruitment to the CFP or the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5- trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. [0645] In some embodiments, administering comprises infusing or injecting. In some embodiments, administering comprises administering directly to the solid cancer. [0646] In some embodiments, administering comprises a circRNA, mRNA, viral-, particle-, liposome-, or exosome-based delivery procedure. [0647] In some embodiments, a CD4+ T cell response or a CD8+ T cell response is elicited in the subject. [0648] In some embodiments, method comprising contacting a cell with the composition described above, the vector or the polypeptide described above. In some embodiments, contacting comprises transducing. In some embodiments, transducing comprises chemical transfection, electroporation, nucleofection, or viral infection. [0649] In some embodiments, provided herein is a method of preparing a pharmaceutical composition comprising contacting a lipid to the composition described herein or the vector described herein. [0650] In some embodiments, contacting comprises forming a lipid nanoparticle. [0651] Also provided is a method of preparing a pharmaceutical composition comprising contacting an antibody to the composition as described herein or the vector described herein. In some embodiments, contacting comprises forming a lipid nanoparticle. [0652] In some embodiments, the recombinant nucleic comprises one or more regulatory elements within the noncoding regions that can be manipulated for desired expression profiles of the encoded proteins. In some embodiments, the noncoding region may comprise suitable enhancer. In some embodiments, the enhancer comprises a binding region for a regulator protein or peptide may be added to the cell or the system comprising the cell, for commencement of expression of the protein encoded under the influence of the enhancer. Conversely, a regulatory element may comprise a protein binding domain that remains bound with the cognate protein and continue to inhibit transcription and/or translation of recombinant protein until an extracellular signal is provided for the protein to decouple from the bound position to allow commencement of the protein synthesis. Examples include but are not limited to Tetracycline-inducible (Tet-Inducible or Tet-on) and Tetracycline repressible (Tet-off) systems known to one of skill in the art. Construct comprising metabolic switch: [0653] In some embodiments the 5’ and 3’ untranslated regions flanking the coding regions of the construct may be manipulated for regulation of expression of the recombinant protein encoded by the nucleic acid constructs described above. For instance, the 3’UTR may comprise one or more elements that are inserted for stabilizing the mRNA. In some embodiments, AU-Rich Elements (ARE) sequences are inserted in the 3’ UTR that result in binding of RNA binding proteins that stabilize or destabilize the mRNA, allowing control of the mRNA half-life. [0654] In some embodiments the 3’UTR may comprise a conserved region for RNA binding proteins (e.g. GAPDH) binding to mature mRNA strand preventing translation. In some embodiments, glycolysis results in the uncoupling of the RNA binding proteins (e.g. GAPDH) allowing for mRNA strand translation. The principle of the metabolic switch is to trigger expression of target genes when a cell enters a certain metabolic state. In resting cells, for example, GAPDH is a RNA binding protein (RBP). It binds to ARE sequences in the 3’UTR, preventing translation of mRNA. When the cell enters glycolysis, GAPDH is required to convert glucose into ATP, coming off the mRNA allowing for translation of the protein to occur. In some embodiments, the environment in which the cell comprising the recombinant nucleic acid is present, provides the metabolic switch to the gene expression. For example, hypoxic condition can trigger the metabolic switch inducing the disengaging of GAPDH from the mRNA. The expression of the mRNA therefore can be induced when the myeloid cell, such as a macrophage, leaves the circulation and enters into a tumor environment, which is hypoxic. This allows for systemic administration of the nucleic acid or a cell comprising the nucleic acid, but ensures a local expression, specifically targeting the tumor environment. [0655] In some embodiments the nucleic acid construct can be a split construct, for example, allowing a portion of the construct to be expressed under the control of a constitutive expression system whereas another portion of the nucleic acid is expressed under control of a metabolic switch, as described above. In some embodiments the nucleic acid may be under bicistronic control. In some embodiments, the bicistronic vector comprises a first coding sequence under a first regulatory control, comprising the coding sequence of a target recognition moiety which may be under constitutive control; and a second coding sequence encoding an inflammatory gene expression which may be under the metabolic switch. In some embodiments the bicistronic vector may be unidirectional. In some embodiments the bicistronic vector may be bidirectional. [0656] In some embodiments, the ARE sequences comprise protein binding motifs for binding ARE sequence that bind to ADK, ALDH18A1, ALDH6A1, ALDOA, ASS1, CCBL2, CS, DUT, ENO1, FASN, FDPS, GOT2,HADHB, HK2, HSD17B10, MDH2, NME1, NQ01, PKM2, PPP1CC, SUCLG1, TP11, GAPDH, or LDH. [0657] Recombinant mRNA and modifications [0658] Provided herein is a recombinant nucleic acid that comprises a sequence encoding a gene of interest, often referred to as a cargo, that is incorporated into a myeloid cell, e.g., a CD14+ cell ex vivo, in vitro or in vivo to produce an engineered myeloid cell (e.g., a myeloid cell expressing a chimeric receptor that can bind to and phagocytose a target cell in vivo, e.g., an ATAK cell). The engineered cell is effective as an immunotherapeutic cell. The recombinant nucleic acid may be a DNA or an RNA. In some preferred embodiments the recombinant nucleic acid is an mRNA. Provided herein is a recombinant mRNA, comprising a sequence encoding a gene of interest, e.g., a chimeric fusion protein that can be expressed in a myeloid cell. In some embodiments the recombinant mRNA is variously modified to enhance stability, translatability, reduce any toxicity, reduce any unintended TLR activation or innate immune reaction to the foreign RNA. Various optimization efforts are in progress as described herein. In some embodiments, an mRNA comprising about 3000 nucleotides or more (e.g., 4kb, 5 kb, 6 kb, 7 kb, 8 kb, 10kb etc.,) may be modified in one or more ways to confer stability in vivo. The 5′ and 3′ UTR elements flanking the coding sequence profoundly influence the stability and translation of mRNA, both of which are critical concerns for ensuring stability of the mRNA. In some embodiments, the 5’-UTR comprises an anti-reverse cap analogues (ARCA) cap. In some embodiments, the 3’ UTR comprises a poly A tail, which may be 50-200 nucleotides long. In some embodiments, the mRNA coding region comprises one or more modified nucleotides, such as methylcytosine, or pseudouridine. Replacing rare codons with frequently used synonymous codons that have abundant cognate tRNA in the cytosol is a common practice to increase protein production from mRNA. In some embodiments, the G:C content of the coding region may be altered for stability optimization. In some embodiments, the immunostimulatory property of the mRNA cargo may be enhanced or complemented by co- administering the mRNA with a suitable adjuvant. In some embodiments the mRNA is purified to obtain greater than 90%, greater than 95%, greater than 97%, 98% or 99% purity, with less than 5%, or less than 1% or less than 0.5% contaminants from the mRNA generation procedure. In some cases, the mRNA is prepared in vitro translation (IVT). [0659] IVT, capping of the 5’ end [0660] mRNA is prepared by in vitro transcription (IVT). The transcription reaction is typically carried out by bacteriophage RNA polymerase (T7, T3, SP6) in the presence of a single (ss) or double stranded (ds) DNA template containing an appropriate promoter sequence and the mix of four NTPs. The polymerase initiates the transcription by the attack of the 3 -OH of the first transcribed nucleotide defined by the promoter sequence on the -phosphate of the upcoming NTP, to yield 5 -triphosphate RNA. The first transcribed nucleotide is usually a purine nucleotide; the most common is the use of T7 polymerase and 6.5 promoter, which imposes the initiation with GTP if an m7GpppG derived cap analog is added to the transcription mixture. [0661] mRNA generated in vitro is 5’capped and a 3’-polyA tail is added. [0662] In some embodiments, the 5’Cap is modified from a native mRNA 5’Cap. [0663] In some embodiments, the 5’ Cap is Cap 0 ((m7GpppN1pG). [0664] IN some embodiments the 5’ Cap is Cap 1 (m7GpppN1 mpG). [0665] One aspect of this disclosure involves having an efficient Cap structure using suitable capping analogs. [0666] In some embodiments, the RNA products prepared by IVT mRNA are co-transcriptional capped with m7GpppN-derived dinucleotides. In some embodiments, the 5’-Cap is a trinucleotide cap analog. In some embodiments the first nucleotide base following a 5’-Cap plays a crucial role in mRNA stability. In some embodiments, the base is adenine (A). In some embodiments, the base is cytosine (C). In some embodiments, the base is guanine (G). In some embodiments, the base is uracil (U). In some embodiments, cap-proximal sequence is crucial for mRNA stability and function. [0667] In some embodiments, the Cap analog is m7GpppApG. [0668] In some embodiments, the Cap analog is m7GpppAmpG. [0669] In some embodiments, the Cap analog is m7Gpppm6ApG. [0670] In some embodiments, the Cap analog is m7Gpppm6AmpG. [0671] In some embodiments, the Cap analog is m7GpppCpG. [0672] In some embodiments, the Cap analog is m7GpppCmpG. [0673] In some embodiments, the Cap analog is m7GpppGpG. [0674] In some embodiments, the Cap analog is m7GpppGmpG. [0675] In some embodiments, the Cap analog is m7GpppUpG. [0676] In some embodiments, the Cap analog is m7GpppUmpG [0677] In some embodiments, the 5’-Cap is anti-reverse cap analogs (ARCAs) having chemically modified 3 -O or 2 -O positions of m7G (m27,3 -OGpppG). [0678] In some embodiments, the 5’-Cap is N7-methylguanosine 5 -diphosphate (m7GDPIm). [0679] In some embodiments, the caping involves post-transcriptional enzymatic capping of 5 - triphosphates RNA using Vaccinia virus capping complex. In some embodiments, the 5’capping confers stability and translational efficiency to the mRNA. [0680] In some embodiments, the 5’ Cap analog is a trinucleotide analog, and is 2’-O- methyladenosine (Am). In some embodiments, the 5’ Cap analog is a trinucleotide analog, and N6,2’-O-dimethyladenosine (m6Am) (FIG.3). [0681] Dinucleotides pNpG and trinucleotide cap analogs are isolated from the reaction mixtures by ion-exchange chromatography on a DEAE Sephadex A-25 (HCO3 – form) column a described in Sikorski et al., Nucleic Acids Research, 2020, Vol.48, No.4. [0682] In some embodiments, the m7GpppAmpG -capped mRNA shows higher stability, as well as higher translational efficiency that a related Cap analog. [0683] In some embodiments, m7GpppAmpG capped mRNA shows at least 1.2-fold higher protein yield when transfected in suitable cells. [0684] Transcription of the RNA may be performed in vitro using the linearized DNA template in the presence of the corresponding RNA polymerase and adenosine, guanosine, uridine and cytidine ribonucleoside triphosphates (rNTPs) under conditions that support polymerase activity while minimizing potential degradation of the resultant mRNA transcripts. In vitro transcription can be performed using a variety of commercially available kits including, but not limited to RiboMax Large Scale RNA Production System (Promega), MegaScript Transcription kits (Life Technologies) as well as with commercially available reagents including RNA polymerases and rNTPs. The methodology for in vitro transcription of mRNA is well known in the art. (see, e.g. Losick, R., 1972, In vitro transcription, Ann Rev Biochem v. 41409-46; Kamakaka, R. T. and Kraus, W. L. 2001. In Vitro Transcription. Current Protocols in Cell Biology. 2:11.6:11.6.1-11.6.17; Beckert, B. And Masquida, B., (2010) Synthesis of RNA by In Vitro Transcription in RNA in Methods in Molecular Biology v. 703 (Neilson, H. Ed), New York, N.Y. Humana Press, 2010; Brunelle, J. L. and Green, R., 2013, Chapter Five—In vitro transcription from plasmid or PCR-amplified DNA, Methods in Enzymology v. 530, 101-114; all of which are incorporated herein by reference). [0685] The desired in vitro transcribed mRNA is then purified from the undesired components of the transcription or associated reactions (including unincorporated rNTPs, protein enzyme, salts, short RNA oligos etc). Techniques for the isolation of the mRNA transcripts are well known in the art. Well known procedures include phenol/chloroform extraction or precipitation with either alcohol (ethanol, isopropanol) in the presence of monovalent cations or lithium chloride. Additional, non- limiting examples of purification procedures which can be used include size exclusion chromatography (Lukavsky, P. J. and Puglisi, J. D., 2004, Large-scale preparation and purification of polyacrylamide-free RNA oligonucleotides, RNA v.10,889-893), silica-based affinity chromatography and polyacrylamide gel electrophoresis (Bowman, J. C., Azizi, B., Lenz, T. K., Ray, P., and Williams, L. D. in RNA in vitro transcription and RNA purification by denaturing PAGE in Recombinant and in vitro RNA syntheses Methods v. 941 Conn G. L. (ed), New York, N.Y. Humana Press, 2012). Purification can be performed using a variety of commercially available kits including, but not limited to SV Total Isolation System (Promega) and In Vitro Transcription Cleanup and Concentration Kit (Norgen Biotek). RNA glycosylation [0686] In an embodiment, the recombinant nucleic acid is an mRNA, wherein the mRNA is a glycosylated mRNA. Glycans are the fourth fundamental biopolymer, next to proteins, polynucleotides, and polyalkanes (lipids). In mammals, glycans are composed of roughly 10 monomeric carbohydrate units, which are strung together and appended to proteins or lipids. Just like RNA, glycans are present in every cell studied to date, across the kingdoms of life. They perform a myriad of essential functions, especially in the context of cell surface events, where complex glycans facilitate the folding and purposeful trafficking of proteins and lipids for secretion or membrane presentation. Thus, processes such as embryogenesis, host-pathogen recognition, and tumor-immune interactions rely on proper glycosylation. It was recently observed that specific RNAs in mammalian cells are modified with glycans (Flynn et al., bioRxiv 2019). Chemical, genetic, and metabolic labeling coupled biorthogonal chemistry revealed sialylated glycans are conjugated to RNAs in mammalian cells. These conjugates are termed glycoRNAs. GlycoRNA assembly depends on canonical N-glycan biosynthetic machinery and results in structures enriched in sialic acid and fucose. The glycoRNA conjugate is present in all cell types and tissues tested across human, mouse, and hamster. Sequencing revealed a distinct set of small RNAs including Y RNAs, small nuclear RNAs, and small nucleolar RNAs as modified putatively at guanosine. Chemical, genetic, and enzymatic approaches revelated that sialylated N-glycans, produced by the oligosaccharyltransferase (OST) complex in the ER lumen, modify these RNA transcripts. Overall, these findings point to a common strategy for RNA glycosylation in mammals. In some embodiments, the mRNA comprising the gene of interest is subjected to glycosylation in a process that recapitulates an intracellular glycosylation process to ensure proper intracellular trafficking, translation, interaction with intracellular proteins, attachment to a cellular organelle structure or scaffolding. Delivery of nucleic acids into a cell: [0687] Nucleic acids encoding the CFP or PFP as described herein may be introduced to a cell, e.g. a myeloid cell, via different delivery approaches. A recombinant nucleic acid as described herein may be introduced to a cell in vitro, ex vivo or in vivo. In some embodiments, a nucleic acid is introduced into a myeloid cell in the form of a plasmid or a vector. In some embodiments, the vector is a viral vector. In some embodiments, the vector is an expression vector, for example, a vector comprising one or more promoters, and other regulatory components, including enhancer binding sequence, initiation and terminal codons, a 5’UTR, a 3’UTR comprising a transcript stabilization element, optional conserved regulatory protein binding sequences and others. In some embodiments, the vector is a phage, a cosmid, or an artificial chromosome. [0688] In some embodiments, a vector is introduced or incorporated in the cell by known methods of transfection, such as using lipofectamine, or calcium phosphate, or via physical means such as electroporation or nucleofection. In some embodiments the vector is introduced or incorporated in the cell by infection, a process commonly known as viral transduction. [0689] In some embodiments, the vector for expression of the CFP is of a viral origin. In some embodiments, the recombinant nucleic acid is encoded by a viral vector capable of replicating in non-dividing cells. In some embodiments, the nucleic acid encoding the recombinant nucleic acid is encoded by a lentiviral vector, e.g. HIV and FIV-based vectors. In some embodiments the lentiviral vector is prepared in-house and manufactured in large scale for the purpose. In some embodiments, commercially available lentiviral vectors are utilized, as is known to one of skill in the art. In some embodiments, the recombinant nucleic acid is encoded by a herpes simplex virus vector, a vaccinia virus vector, an adenovirus vector, or an adeno-associated virus (AAV) vector. In some embodiments, the myeloid cell may be modified by expressing a transgene via incorporation of the transgene in a transient expression vector. In some embodiments expression of the transgene may be temporally regulated by a regulator from outside the cell. Examples include the Tet-on Tet-off system, where the expression of the transgene is regulated via presence or absence of tetracycline. Genome-integrating mRNA [0690] In some embodiments, a stable integration of transgenes into myeloid cells, such as macrophages, and other phagocytic cells may be accomplished via the use of a transposase and transposable elements, in particular, mRNA-encoded transposase. In one embodiment, Long Interspersed Element-1 (L1) RNAs may be contemplated for retrotransposition of the transgene and stable integration into myeloid cells, such as macrophages or phagocytic cells. Retrotransposon may be used for stable integration of a recombinant nucleic acid encoding a gene of interest, e.g. a fusion protein, such as a chimeric fusion protein or a chimeric antigen receptor for myeloid cells. In some embodiments, recombinant nucleic acid is an mRNA comprising a sequence encoding human ORF2 protein and/or a human ORF1 protein with the recombinant nucleic acid sequence as the payload. In some embodiments, the mRNA payload may be about 6kb in length, or about 10 kb in length. Self-amplifying RNA (saRNA) [0691] In one aspect, the recombinant nucleic acid is a self-amplifying mRNA. Self-amplifying or self-replicating RNA molecules (e.g., described in US20110300205A1), which replicate in host cells leading to an amplification of the amount of RNA encoding the desired gene product, can enhance efficiency of RNA delivery and expression of the encoded gene products. See, e.g., Johanning, F. W., et al., Nucleic Acids Res., 23(9):1495-1501 (1995); Khromykh, A. A., Current Opinion in Molecular Therapeutics, 2(5):556-570 (2000); Smerdou et al., Current Opinion in Molecular Therapeutics, 1(2):244-251 (1999). Self-replicating RNAs have been produced as virus particles and as free RNA molecules. [0692] saRNAs are genetically engineered replicons derived from self-replicating single- stranded RNA viruses. In some embodiments, the saRNA may be polycistronic. They can be delivered as viral replicon particles (VRPs) with the saRNA packaged into the viral particle, or as a completely synthetic saRNA produced after in vitro transcription. To generate replication-defective VRPs, envelope proteins are provided in trans as defective helper constructs during production. Resulting VRPs therefore lack the ability to form infectious viral particles following a first infection, and only the RNA is capable of further amplification. VRPs may be derived from both positive- sense and negative-sense RNA viruses, however the latter are more complex and require reverse genetics to rescue the VRPs. In some embodiments, the pharmaceutical composition described herein comprises an saRNA that can be produced and delivered in a similar manner to conventional mRNA vaccines. Positive-sense alphavirus genomes that have been commonly used for saRNA design include the Venezuelan equine encephalitis virus (VEE), Sindbis virus (SINV), and Semliki forest virus (SFV). In one embodiment, the saRNA may be used to engineer a myeloid cell in vitro, and the resultant engineered cell comprising the saRNA that comprises the gene of interest may be formulated in a pharmaceutical composition and administered to a subject in need thereof. Alternatively, in some embodiments, the saRNA may be delivered directly in vivo. In some embodiments the saRNA may be delivered inside a suitable cell either in vitro or in vivo as a naked RNA. In some embodiments the saRNA may be delivered inside a suitable cell either in vitro or in vivo as a nucleic acid and lipid complex (LN complex). In some embodiments, the saRNA may be delivered as a cationic nano-emulsion. In some embodiments, the saRNA may be delivered inside a suitable cell either in vitro or in vivo in liposomes. In some embodiments, saRNA may be delivered inside a suitable cell either in vitro or in vivo as a lipid nanoparticle (LNPs). Exemplary saRNA vaccines currently in development are discussed in Bloom et al., Gene Therapy (2021) 28:117–129. [0693] Various designs of LNP compositions are contemplated herein, as described in the later section. Circ-RNA [0694] In some embodiments, the recombinant nucleic acid described herein is a circular RNA (circRNA). A circular RNA comprises a RNA molecule where the 5’ end and the 3’ end of the RNA molecule are joined together. Without wishing to be bound by any theory, circRNAs have no free ends and may have longer half-life as compared to some other forms of RNAs or nucleic acid and may be resistant to digestion with RNase R exonuclease and turn over more slowly than its counterpart linear RNA in vivo. In some embodiments, the half-life of a circRNA is more than 20 hours. In some embodiments, the half-life of a circRNA is more than 30 hours. In some embodiments, the half-life of a circRNA is more than 40 hours. In some embodiments, the half-life of a circRNA is more than 48 hours. In certain embodiments, a circRNA comprises an internal ribosome entry site (IRES) element that engages a eukaryotic ribosome and an RNA sequence element encoding a polypeptide operatively linked to the IRES for insertion into cells in order to produce a polypeptide of interest. [0695] circRNAs can be prepared by methods known to those skilled in the art. For example, circRNAs may be chemically synthesized or enzymatically synthesized. In some embodiments, a linear primary construct or linear mRNA may be cyclized, or concatemerized to create a circRNA. The mechanism of cyclization or concatemerization may occur through methods such as, but not limited to, chemical, enzymatic, or ribozyme catalyzed methods. The newly formed 5′-/3′-linkage may be an intramolecular linkage or an intermolecular linkage. In some embodiments, a linear primary construct or linear mRNA may be cyclized, or concatemerized using the chemical method to form a circRNA. In the chemical method, the 5′-end and the 3′-end of the nucleic acid (e.g., linear primary construct or linear mRNA) contain chemically reactive groups that, when close together, form a new covalent linkage between the 5′-end and the 3′-end of the molecule. The 5′-end may contain an NHS-ester reactive group and the 3′-end may contain a 3′-amino-terminated nucleotide such that in an organic solvent the 3′-amino-terminated nucleotide on the 3′-end of a linear RNA molecule will undergo a nucleophilic attack on the 5′-NHS-ester moiety forming a new 5′-/3′-amide bond. In some embodiments, a DNA or RNA ligase, e.g. a T4 ligase, may be used to enzymatically link a 5′-phosphorylated nucleic acid molecule (e.g., a linear primary construct or linear mRNA) to the 3′-hydroxyl group of a nucleic acid forming a new phosphodiester linkage. In some embodiments, a linear primary construct or linear mRNA may be cyclized or concatemerized by using at least one non-nucleic acid moiety. For example, the at least one non-nucleic acid moiety may react with regions or features near the 5′ terminus and/or near the 3′ terminus of the linear primary construct or linear mRNA in order to cyclize or concatemerized the linear primary construct or linear mRNA. In some embodiments, a linear primary construct or linear mRNA may be cyclized or concatemerized due to a non-nucleic acid moiety that causes an attraction between atoms, molecules surfaces at, near or linked to the 5′ and 3′ ends of the linear primary construct or linear mRNA. For example, a linear primary construct or linear mRNA may be cyclized or concatemerized by intermolecular forces or intramolecular forces. Non-limiting examples of intermolecular forces. In some embodiments, a linear primary construct or linear mRNA may comprise a ribozyme RNA sequence near the 5′ terminus and near the 3′ terminus. In some embodiments, a circRNA may be synthesized by inserting DNA fragments into a plasmid containing sequences having the capability of spontaneous cleavage and self-circularization. In some embodiments, a circRNA is produced by making a DNA construct encoding an RNA cyclase ribozyme, expressing the DNA construct as an RNA, and then allowing the RNA to self-splice, which produces a circRNA free from intron in vitro. In some embodiments, a circRNA is produced by synthesizing a linear polynucleotide, combining the linear nucleotide with a complementary linking oligonucleotide under hybridization conditions, and ligating the linear polynucleotide. [0696] The circRNA may be modified or unmodified. In some embodiments, the circRNA is chemically modified. For example, an A, G, U or C ribonucleotide of a circRNA may comprise chemical modifications. In some embodiments, any region of a circRNA, e.g. the coding region of the CFP or PFP, the flanking regions and/or the terminal regions may contain one, two, or more (optionally different) nucleoside or nucleotide modifications. In some embodiments, a modified circRNA introduced to a cell may exhibit reduced degradation in the cell, as compared to an unmodified circRNA. Modifications such as to the sugar, the nucleobase, or the internucleoside linkage (e.g. to a linking phosphate/to a phosphodiester linkage/to the phosphodiester backbone) are also encompassed. In some embodiments, one or more atoms of nucleobase, e.g. a pyrimidine nucleobase may be replaced or substituted with optionally substituted amino, optionally substituted thiol, optionally substituted alkyl (e.g., methyl or ethyl), or halo (e.g., chloro or fluoro). In certain embodiments, modifications (e.g., one or more modifications) are present in each of the sugar and the internucleoside linkage. Additional modifications to circRNAs are described in US20170204422, the entire content of which is incorporated herein by reference. [0697] In some embodiments, the circRNA is conjugated to other polynucleotides, dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases, proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell, hormones and hormone receptors, non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, or cofactors. [0698] In some embodiments, the circRNA is administered directly to tissues of a subject. Additional description of circRNAs in U.S. Patent Nos 5,766,903, 5,580,859, 5,773,244, 6,210,931, PCT publication No. WO1992001813, Hsu et al., Nature (1979) 280:339-340, Harland & Misher, Development (1988) 102:837-852, Memczak et al. Nature (2013) 495:333-338, Jeck et al., and RNA (2013) 19:141-157, each of which is incorporated herein by reference in its entirety. Cell-specific expression of constructs: structural, transcriptional and post-transcriptional controls: [0699] In particular embodiments, where the therapeutic comprises a recombinant nucleic acid encoding a polypeptide of interest, e.g., a fusion protein for expression in a myeloid cell, and the recombinant nucleic acid is administered systemically, an additional safety layer may be designed to reduce or prevent expression of the polypeptide of interest in a non-intended cell type, e.g., in a non- myeloid cell. For example, Brown et al. demonstrated higher efficacy of a lentiviral delivery and expression of Factor IX by incorporating within the vector a miR142-3p microRNA binding site to reduce off-target expression in hematopoietic cells in mouse model of hemophilia (Brown B. et al., Blood. 2007; 110: 4144-4152). In some embodiments, cell specific expression may be incorporated in the design of the recombinant nucleic acid encoding the polypeptide of interest. For example, in one embodiment, as also described elsewhere in the disclosure, the fusion protein may comprise (i) an extracellular domain comprising, e.g. a target binding domain, and (ii) a FcR-alpha domain, which when expressed, the FcR-alpha domain pairs with an endogenous Fcgamma receptor for membrane expression, ensuring that only cells that express an FcR gamma, e.g., myeloid cells, can properly express the chimeric fusion protein. [0700] For example, in one embodiment, the expression construct may comprise a regulatory control unit for cell-specific expression, such as a cell-specific promoter or an enhancer that controls the expression of the polypeptide of interest. Exemplary myeloid cell-specific promoters may be promoter elements of the lysM, csf1r, CD11c, CD68, macrophage SRA, and CD11b genes. Such controls may additionally be under regulatable control, for example, by incorporating an inducible element. In one embodiment, the myeloid cell-specific promoter or enhancer may be human myeloid cell-specific gene promoters, enhancers or portions thereof. [0701] In another embodiment, cell specific expression of the construct may be incorporated in the design by incorporating one or more microRNA binding sites. MicroRNAs (miRNAs) are endogenous small non-coding single stranded RNAs that control diverse biological processes. MiRNAs are well suited to suppress previously active programs and, thereby, provide robustness to cell fate decisions (Babiarz, J. E. & Blelloch, R. Small RNAs—their biogenesis, regulation and function in embryonic stem cells. StemBook, ed. The Stem Cell Research Community, StemBook, doi/10.3824/stembook.1.47.1. (2009). Hornstein, E. & Shomron, N. Canalization of development by microRNAs. Nat. Genet 38, S20-24 (2006). They regulate the expression of complementary target mRNAs by post-transcriptional gene silencing, which leads to mRNA cleavage or translational repression and major signaling pathways, like developmental timing, hematopoietic cell differentiation, apoptosis, cell proliferation, and organ development. MiRNAs identify their targets via base pairing of nucleotides 2-8 of the miRNA (the seed sequence) with complementary sequences within the target mRNA's open reading frame (ORF) and 3′ untranslated region (UTR) (The Stem Cell Research Community, StemBook, doi/10.3824/stembook. 1.47.1. (2009)). MicroRNAs (miRNAs) are single-stranded RNA molecules, typically between 21 and 23 nucleotides in length. [0702] Removal of genes required for maturation of all miRNAs has shown that miRNAs play essential roles in the proliferation and differentiation of Embryonic Stem Cells (ESCs)(Wang, Y. et al., Nat Genet 39:380-5 (2007); Kanellopoulou, C. et al. Genes Dev 19:489-501 (2005); Murchison, E. P. et al., Proc Natl Acad Sci USA 102:12135-40 (2005)). For example, the loss of the RNA binding protein DGCR8, which is required for the production of all canonical miRNAs, results in a cell cycle defect and an inability to silence the self-renewal program of ESCs when they are placed in differentiation-inducing conditions (Wang, Y. et al., Nat Genet 39:380-5 (2007). Frequently, one miRNA can target multiple mRNAs and one mRNA can be regulated by multiple miRNAs targeting different regions of the 3′ UTR. Once bound to an mRNA, miRNA can modulate gene expression and protein production by affecting, e.g., mRNA translation and stability (Baek et al., Nature 455(7209):64 (2008); Selbach et al., Nature 455(7209):58 (2008); Ambros, 2004, Nature, 431, 350-355; Bartel, 2004, Cell, 116, 281-297; Cullen, 2004, Virus Research., 102, 3-9; He et al., 2004, Nat. Rev. Genet., 5, 522-531; and Ying et al., 2004, Gene, 342, 25-28). [0703] Uchida et al. (WO2015066042A1) demonstrated use of oncolytic HSV in treating Glioblastoma multiforme, by inhibition of vector replication by a cellular microRNA that is highly expressed in normal brain but virtually absent in tumor cells. [0704] Many of miRNAs are specific to or over-expressed in certain organs/tissues/cells (see, e.g., Hua et al., BMC Genomics, 2009, 10:214; Liang et al., BMC Genomics. 2007, 8:166; Landgraf et al., Cell.2007, 129:1401-1414; Lee et al., RNA.2008, 14:35-42). [0705] In some aspects of the invention, provided herein is a method of incorporating one or more binding sites (sequences) for particular microRNA(s) that are specifically expressed in non- myeloid cells. This method can be utilized herein to reduce off-target expression of the gene of interest encoded by the recombinant nucleic acid. A binding site for miRNA, for example is a short polynucleotide sequence within the recombinant mRNA that comprises a region that hybridizes with an miRNA present in a cell. Upon hybridization, the miRNA will trigger the inhibition and destruction of the mRNA, thereby the mRNA will not express in the cell, where expression of the recombinant mRNA is not desired or intended, e.g., a non-myeloid cell. [0706] In some embodiments, non-myeloid cells discussed in this concept include, but are not limited to blood, bone, brain, kidney, muscle, spinal cord, nerve, endocrine system, uterine, ear, foreskin, liver, intestine, bladder or skin. In some embodiments the cells can include neural cells, lymphocytes, epidermal cells, islet cells, intestinal cells or fibroblasts. [0707] Exemplary non-myeloid cells may include embryonic stem cells. Accordingly, some miRNA binding sites that may be incorporated in the recombinant mRNA of the invention may include for example, hsa-mir-302a, hsa-miR-302b, hsa-miR-302c, hsa-miR-302d, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373. (See, e.g., Wang, Y. et al. Embryonic stem cell-specific microRNAs regulate the G1-S transition and promote rapid proliferation Nat Genet 40:1478-1483 (2008)). [0708] Exemplary non-myeloid cells may include neuronal cells. Accordingly, some miRNA binding sites that may be incorporated in the recombinant mRNA of the invention may include those are enriched in certain cellular compartments, particularly in axons, dendrites and synapses (see, e.g., Schratt et al., Nature.439:283-289, 2006; Lugli et al., J Neurochem.106:650-661, 2008; Bicker and Schratt, J Cell Mol Med., 12:1466-1476, 2008; Smalheiser and Lugli, Neuromolecular Med. 11:133-140, 2009; Rajasethupathy, Neuron. 63:714-716, 2009; Kye, RNA 13:1224-1234, 2007; Yu et al., Exp Cell Res. 314:2618-2633, 2008; Cougot, et al., J Neurosci. 28:13793-13804, 2008; Kawahara, Brain Nerve.60:1437-1444, 2008; Schratt G. Rev Neurosci.2009; 10:842-849; Pichardo- Casas et al. Brain Research.1436:20-33, 2012). [0709] Exemplary non-myeloid cells may include endothelial cells. Exemplary endothelial cell- specific miRNA may include miR-10a/b, miR-24/27, miR-125a, miR-126 and miR-221/222. [0710] In some embodiments, binding sites for more than one miRNAs are incorporated in the recombinant construct. In some embodiments, the miRNA binding sites are incorporated in more than one copy numbers, in tandem. In some embodiments, the recombinant nucleic acid may incorporate binding sites for two or more different miRNAs. In some embodiments, the recombinant nucleic acid may incorporate binding sites for 1, 2, 3, 4, 5, 6 or more different miRNAs. [0711] In some embodiments, the recombinant nucleic acid may incorporate multiple binding sites for each miRNA. For example, the recombinant nucleic acid may incorporate 1, 2, 3, 4, 5, 6 or binding sites for one or more miRNAs, in tandem. [0712] MicroRNAs may upregulate the expression of a gene and the miRNA binding site may be utilized accordingly. Nanoparticles for nucleic acid delivery [0713] In some embodiments, a nucleic acid is introduced into a myeloid cell with a nanoparticle (NP). A nanoparticle may be of various shapes or sizes and may harbor the nucleic acid encoding the CFP or PFP. In some embodiments, the NP is a lipid nanoparticle (LNP). In some embodiments, the NP comprises poly(amino acids), polysaccharides and poly(alpha-hydroxy acids), gold, silver, carbon, iron, silica, or any combination thereof. In some embodiments, the NP comprises a polylactide-co-glycolide (PGLA) particle. In some embodiments, the nucleic acid is encapsulated in the NP, for example, via water/oil emulsion or water-oil-water emulsion. In some embodiments, the nucleic acid is conjugated to a component of or complexed with components of the NP. In some embodiments, a protein corona may form around NPs. A protein corona may form in a two-step process. In the first step, high-affinity proteins rapidly bind to NPs to form a primary corona. In the second step, proteins of lower affinity bind either directly to the NP or to the proteins in the primary corona forming a secondary corona. Constituents of the protein corona may thus be impacted by the protein content of the serum and thus by the homeostatic or immune responses that regulate it. In some embodiments, proteins with high abundance, such as albumin, comprise a significant proportion of the primary corona. In some embodiments, NPs with different charges bind significant amounts of less-abundant proteins in particular environments, e.g. in plasma with certain antigen or antibody. In vivo formation of a protein corona may alter NP charge or mask functional groups important for NP targeting to certain receptors and/or enhance clearance of NPs by phagocytes. In some embodiments, NPs are engineered to reduce changes to NP charges or masking of functional groups, and/or increase the serum half-life of the NPs. In some embodiments, NP surface coating are designed to modulate opsonization events. For example, the NP’s surface may be coated with polymeric ethylene glycol (PEG) or its low molecular weight derivative polyethylene oxide (PEO). Without wishing to be bound by any theory, PEG increases surface hydrophilicity, resulting in improved circulating NP half-life due to reduced serum protein binding. In some embodiments, the NP coated with PEG or PEO are engineered to result in reduced toxicity or increased biocompatibility of the NPs. Additional NP design and NP targeting for myeloid cells are described in Getts et al., Trends Immunol. 36(7): 419-427 (2015), the entirety of which is incorporated herein by reference. [0714] NPs described herein may be used to introduce the recombinant nucleic acid into a cell in in vitro/ex vivo cell culture or administered in vivo. In some embodiments, the NP is modified for in vivo administration. For example, the NP may comprise surface modification or attachment of binding moieties to bind specific toxins, proteins, ligands, or any combination thereof, before being taken up by liver or spleen phagocytes. In recent rodent proof-of-concept studies, infused highly negatively charged ‘immune-modifying NPs’ (IMPs) can absorb certain blood proteins, including S100 family and heat shock proteins, before finally being removed and destroyed by cells of the mononuclear phagocyte system. Furthermore, this mechanism may also be used to capture and concentrate certain circulating proteins. IMPs have been shown to bind Annexin 1. The accumulation of Annexin 1 and its presentation to particular leukocyte subsets can have broad immune outcomes. For example, Annexin 1-loaded NPs may reduce neutrophils via induction of apoptosis and/or promote T cell activation. In some embodiments, the NP is designed to target a cell surface receptor, e.g. a scavenger receptor. In some embodiments, an NP is a particle with highly negative surface charge. [0715] In some embodiments, the NP encapsulates the nucleic acid wherein the nucleic acid is a naked DNA molecule. In some embodiments, the NP encapsulates the nucleic acid wherein the nucleic acid is an mRNA molecule. In some embodiments, the NP encapsulates the nucleic acid wherein the nucleic acid is a circular RNA (circRNA) molecule. In some embodiments, the NP encapsulates the nucleic acid wherein the nucleic acid is a vector, a plasmid, or a portion or fragment thereof. [0716] NPs may be delivered to a cell in vitro, ex vivo or in vivo. In some embodiments, a NP is delivered to a phagocytic cell ex vivo. In some embodiments, a NP is delivered to a phagocytic cell in vivo. In some embodiments, the NP is less than 100nm in diameter. In some embodiments, the NP is more than 100nm in diameter. In some embodiments, the NP is a rod-shaped NP. In some embodiments, the NP is a spherical NP. In particular embodiments, the NP is a spherical NP for delivery to a phagocytic cell. In additional embodiments, the NP is at least 100nm in diameter and does not trigger or triggers reduced toxicity when delivered to a cell. [0717] In some embodiments, the NP is positively charged. In some embodiments, the NP is negatively charged. In some embodiments, the NP is a cationic NP that is delivered and taken up by a myeloid cell ex vivo or in vivo. [0718] Stiffness may affect the biological impact of NPs. NPs made of rigid materials may be associated with increased potential for embolism, while flexible polymer-based NPs that can more easily deform may gain better access to tissues during the complex vascular changes associated with inflammation. The fluidity of NPs, too, affects the ability of antigen-loaded NP to stimulate immune responses. Thus, intramuscular, solid-phase, antigen-containing liposome immunization may elicit a more robust Th1/Th17 response than similarly administered fluid-phase liposomes. Without wishing to be bound by any theory, solid-phase particles may result from the formation of an immobilized antigen particle depot and may result in a prolonged supply of antigen for APCs also associated with upregulation of positive costimulatory molecules such as CD80, which support efficient T cell priming. [0719] In some embodiments, the lipid nanoparticle comprises a charged lipid, e.g., a cationic lipid. Charged lipids may be synthetic or naturally derived. Examples of charged lipids include phosphatidylserines, phosphatidic acids, phosphatidylglycerols, phosphatidylinositols, sterol hemisuccinates, dialkyl trimethylammonium-propanes, (e.g. DOTAP, DOTMA), dialkyl dimethylaminopropanes, ethyl phosphocholines, dimethylaminoethane carbamoyl sterols (e.g. DC- Chol). [0720] In some embodiments, the lipid nanoparticle comprises a neutral lipid. A neutral lipid is a lipid that exists in either an uncharged state or as a zwitterionic form at a selected pH. At physiological pH, such lipids include, but are not limited to, phosphotidylcholines such as 1,2- Distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-Dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine (POPC), 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), phophatidylethanolamines such as 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), sphingomyelins (SM), ceramides, steroids such as sterols and their derivatives. In some embodiment, a neutral lipid is selected from the neutral lipid is selected from DSPC, DPPC, DMPC, DOPC, POPC, DOPE and SM. In some embodiments, the neutral lipid may be DSPC. [0721] Neutral lipids may be synthetic or naturally derived. [0722] In some embodiments, a steroid or steroid analog may be incorporated into the LNP. [0723] In various embodiments, the molar ratio of the compound (e.g., the mRNA) to the neutral lipid ranges from about 2:1 to about 8:1. [0724] In various embodiments, the compositions further comprise a steroid or steroid analogue. In certain embodiments, the steroid or steroid analogue is cholesterol. In some of these embodiments, the molar ratio of the compound to cholesterol ranges from about 2:1 to 1:1. In various embodiments, the polymer conjugated lipid is a pegylated lipid. For example, some embodiments include a pegylated diacylglycerol (PEG-DAG) such as 1-(monomethoxy- polyethyleneglycol)-2,3-dimyristoylglycerol (PEG-DMG), a pegylated phosphatidylethanoloamine (PEG-PE), a PEG succinate diacylglycerol (PEG-S-DAG) such as 4-O-(2′,3′- di(tetradecanoyloxy)propyl-1-O-(cw-methoxy(polyethoxy)ethyl)butanedioate (PEG-S-DMG), a pegylated ceramide (PEG-cer), or a PEG dialkoxypropylcarbamate such as ω- methoxy(polyethoxy)ethyl-N-(2,3-di(tetradecanoxy)propyl)carbamate or 2,3- di(tetradecanoxy)propyl-N-(ω-methoxy(polyethoxy)ethyl)carbamate. In various embodiments, the molar ratio of the compound to the pegylated lipid ranges from about 100:1 to about 25:1. In some embodiments, the lipid components of the LNP described herein comprise components that have been previously described in US20170342442A1. [0725] In some embodiments, the lipid nanoparticle for the polynucleic acid delivery in myeloid cells ex vivo or in vivo comprises one or more ionizable cationic lipids as described in US patent 9,593,077. [0726] In some embodiments, the lipid nanoparticle for the polynucleic acid delivery in myeloid cells ex vivo or in vivo comprises one or more phospholipids selected from phospholipid is selected from the group consisting of phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyloleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidylethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidylcholine, di stearoylphosphatidylcholine, and dilinoleoylphosphatidylcholine, as described in US patent 9,504,651. Delivery of nucleic acids in vivo [0727] In some embodiments, the subject is administered a pharmaceutical composition comprising a mRNA encoding the CFP or PFP as described herein. In some embodiments, the mRNA is co-formulated into nanoparticles (NPs), such as lipid nanoparticles (LNPs). For example, the LNP may comprise cationic lipids or ionizable lipids. In some embodiments, the mRNA is formulated into polymeric particles, for example, polyethyleneimine particles, poly(glycoamidoamine), ly(β-amino)esters (PBAEs), PEG particles, ceramide-PEGs, polyamindoamine particles, or polylactic-co-glycolic acid particles (PLGA). In some embodiments, the mRNA is administered by direct injection. In some embodiments, the mRNA is complexed with transfection agents, e.g. Lipofectamine 2000, jetPEI, RNAiMAX, or Invivofectamine. [0728] In one embodiment, provided herein is a method of in vivo delivery of a composition comprising mRNA, wherein the mRNA may be complexed with a cationic molecule wherein the delivery comprises systemic delivery, e.g., delivery via intravenous, intraperitoneal, intramuscular or subcutaneous, or any other suitable delivery course. In some embodiments, the delivery comprises local delivery, for example intratumoral or in a specific tissue or site of a disease, such as a site of an inflammation, e.g. arthritic joints. In some embodiments, the cationic molecule may be a cationic lipid, a cationic polymer, a cationic dendrimer, or a synthetic molecule. In one embodiment, the mRNA is at least 5 kb in length. In one embodiment, the mRNA is about 5.5 kb, about 6.0, about 6.5, about 7.0, about 7.5, about 8, about 8.5, about 9, about 9.5, about 10 kb in length. In some embodiments, the mRNA is at least 95% pure. In some embodiments, the mRNA is at least 98% pure. In some embodiments, the mRNA is at least99% pure. In some embodiments, the mRNA is sufficiently free of salts. In some embodiments, the mRNA is at least 99% pure. In some embodiments, the mRNA is effectively free of salts. In some embodiments, the mRNA is about 0.1 microgram/microliter in concentration. In some embodiment, the mRNA is complexed with a cationic molecule, e.g., a cationic polymer and is formulated in a microsome or a nanoparticle. In some embodiments, the nanoparticle is a lipid nanoparticle, comprising one or more cationic lipids and one or more neutral lipids. In one embodiment, the mRNA of a length greater than 5kb is complexed with a cationic molecule, and is administered systemically. [0729] The mRNA may be a naked mRNA. The mRNA may be modified or unmodified. For example, the mRNA may be chemically modified. In some embodiments, nucleobases and/or sequences of the mRNA are modified to increase stability and half-life of the mRNA. In some embodiments, the mRNA is glycosylated. Additional mRNA modification and delivery approaches as described in Flynn et al., BioRxiv 787614 (2019) and Kowalski et al. Mol. Ther. 27(4): 710-728 (2019) are each incorporated herein by reference in its entirety. [0730] In some embodiments, the mRNA is a self-amplifying mRNA. In some embodiments, the mRNA is a genome-integrating mRNA. In some embodiments, the mRNA is a circular RNA. In some embodiments, the mRNA comprises a sequence comprising a binding site for an miRNA that is expressed in a non-myeloid cell, but is not expressed in a myeloid cell. [0731] In some embodiments, the NP is a Lipid nanoparticle (LNP). LNPs may comprise a polar and or a nonpolar lipid. In some embodiments cholesterol is present in the LNPs for efficient delivery. LNPs are 100-300 nm in diameter provide efficient means of mRNA delivery to various cell types, including myeloid cells, such as macrophages. In some embodiments, LNP may be used to introduce the recombinant nucleic acids into a cell in in vitro cell culture. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is a naked DNA molecule. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is an mRNA molecule. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is inserted in a vector, such as a plasmid vector. In some embodiments, the LNP encapsulates the nucleic acid wherein the nucleic acid is a circRNA molecule. [0732] In some embodiments, the LNP is used to deliver the nucleic acid into the subject. LNP can be used to deliver nucleic acid systemically in a subject. It can be delivered by injection. In some embodiments, the LNP comprising the nucleic acid is injected by intravenous route. In some embodiments the LNP is injected subcutaneously. Pharmaceutical Composition [0733] Provided herein is a pharmaceutical composition, comprising engineered myeloid cells, such as macrophages, comprising a recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient. [0734] Also provided herein is a pharmaceutical composition, comprising a recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient. The pharmaceutical composition may comprise DNA, mRNA, self-amplifying mRNA, self-integrating mRNA (e.g., retrotransposons), or circRNA or a liposomal composition of any one of these. In some embodiments, the liposome is an LNP. [0735] Also provided herein is a pharmaceutical composition comprising a vector comprising the recombinant nucleic acid encoding the CFP and a pharmaceutically acceptable excipient. The pharmaceutical composition may comprise DNA, mRNA or circRNA inserted in a plasmid vector or a viral vector. [0736] In some embodiments the engineered myeloid cells, such as macrophages, are grown in cell culture sufficient for a therapeutic administration dose, and washed, and resuspended into a pharmaceutical composition comprising a pharmaceutically acceptable solution. “Pharmaceutically acceptable solution” may refer to those solutions or comprising an acid or aqueous salt of the acid, which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, gluconic acid, glucuronic acid, glutamic acid, glutaric acid, 2-oxo- glutaric acid, glycerophosphoric acid, glycolic acid, hippuric acid, isobutyric acid, lactic acid, lactobionic acid, lauric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, mucic acid, naphthalene-1,5-disulfonic acid, naphthalene-2-sulfonic acid, 1-hydroxy-2- naphthoic acid, nicotinic acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, propionic acid, pyroglutamic acid, pyruvic acid, salicylic acid, 4-aminosalicylic acid, sebacic acid, stearic acid, succinic acid, tartaric acid, thiocyanic acid, p-toluenesulfonic acid, trifluoroacetic acid, undecylenic acid, and the like. [0737] In some embodiments, the pharmaceutically acceptable solution may comprise a pharmaceutically acceptable base or a salt thereof. “Pharmaceutically acceptable base” refers to those basic solutions or salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline and caffeine. [0738] Often crystallizations produce a solvate of the compound of the invention. As used herein, the term “solvate” may refer to an aggregate that comprises one or more molecules of a compound of the invention with one or more molecules of solvent. The solvent may be water, in which case the solvate may be a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compound of the invention may be true solvates, while in other cases, the compound of the invention may merely retain adventitious water or be a mixture of water plus some adventitious solvent. [0739] In some embodiments the excipient comprises a sterile buffer, (e.g. HEPES or PBS) at neutral pH. In some embodiment, the pH of the pharmaceutical composition is at 7.5. In some embodiments, the pH may vary within an acceptable range. In some embodiments, the engineered cells may be comprised in sterile enriched cell suspension medium comprising complement deactivated or synthetic serum. In some embodiments the pharmaceutic composition further comprises cytokines, chemokines or growth factors for cell preservation and function. [0740] In some embodiments, the pharmaceutical composition may comprise additional therapeutic agents, co-administered with the engineered cells. [0741] Also provided herein is a pharmaceutical composition comprising one or more polynucleic acids, the one or more polynucleic acids comprising a first sequence encoding a chimeric fusion protein (CFP), the CFP comprising: a CFP extracellular domain comprising a first antigen binding domain, and a CFP transmembrane domain operatively linked to the CFP extracellular domain, wherein the CFP transmembrane domain is a transmembrane domain from a protein that dimerizes with endogenous FcR-gamma receptors in myeloid cells; and a second sequence encoding a chimeric antigen receptor (CAR), the CAR comprising: a CAR extracellular domain comprising a second antigen binding domain, a CAR transmembrane domain operatively linked to the CAR extracellular domain, wherein the CAR transmembrane domain is a transmembrane domain from a protein that functionally interacts with an endogenous T cell receptor (TCR) complex and/or at least one endogenous TCR polypeptide, and a CAR intracellular domain operatively linked to the transmembrane domain. [0742] In some embodiments, the one or more polynucleic acids comprises a polynucleic acid molecule comprising both the first sequence and the second sequence. In some embodiments, the first sequence and the second sequence are operatively linked by a linker sequence. In some embodiments, the linker sequence comprises a protease cleavage site. In some embodiments, the protease cleavage site is a T2A cleavage site or a P2A cleavage site [0743] In some embodiments, the one or more polynucleic acids is encapsulated within a nanoparticle delivery vehicle. In some embodiments, the one or more polynucleic acids is encapsulated within the same nanoparticle delivery vehicle. [0744] In some embodiments, the one or more polynucleic acids comprises a first polynucleic acid molecule comprising the first sequence and a second polynucleic acid molecule comprising the second sequence. [0745] In some embodiments, the first polynucleic acid molecule is encapsulated within a first nanoparticle delivery vehicle and the second polynucleic acid molecule is encapsulated within a second nanoparticle delivery vehicle. In some embodiments, the first polynucleic acid molecule and the second polynucleic acid molecule are encapsulated within the same nanoparticle delivery vehicle. [0746] In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is expressed on the surface of myeloid cells of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is expressed on the surface of T cells of the human subject. [0747] In some embodiments, the CAR functionally integrates into an endogenous TCR complex of a T cell of the human subject. In some embodiments, the CAR extracellular domain is a TCR extracellular domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR transmembrane domain is a TCR transmembrane domain from TCR-alpha, TCR-beta, TCR-delta, TCR-gamma, CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, the CAR intracellular domain is a TCR intracellular domain from CD3-zeta, CD3-gamma, CD3-delta or CD3-epsilon. In some embodiments, at least two of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. In some embodiments, each of the TCR extracellular domain, the TCR transmembrane domain and the TCR intracellular domain are from the same TCR subunit. [0748] In some embodiments, the CAR intracellular domain further comprises a costimulatory domain. In some embodiments, the costimulatory domain is a functional signaling domain from a protein selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). [0749] In some embodiments, the first and/or second first and/or second antigen binding domain comprises a Fab fragment, an scFv domain or an sdAb domain. [0750] In some embodiments, the CFP extracellular domain is an extracellular domain from CD8, CD16a, CD64, CD68 or CD89. In some embodiments, the CFP extracellular domain further comprises a hinge domain derived from CD8, wherein the hinge domain is operatively linked to the CFP transmembrane domain and the first antigen binding domain. [0751] In some embodiments, after administration of the pharmaceutical composition to a human subject the CFP is specifically expressed in myeloid cells, monocytes or macrophages of the human subject. In some embodiments, after administration of the pharmaceutical composition to a human subject the CAR is specifically expressed in T cells of the human subject. [0752] In some embodiments, the CFP transmembrane domain is a transmembrane domain from CD16a, CD64, CD68 or CD89. In some embodiments, the CFP further comprises a CFP intracellular domain operatively linked to the CFP transmembrane domain. In some embodiments, the CFP intracellular domain comprises one or more intracellular signaling domains, and wherein the one or more intracellular signaling domains comprises an intracellular signaling domain from FcγR, FcαR, FcεR, CD40 or CD3zeta. In some embodiments, the one or more intracellular signaling domains further comprises a phosphoinositide 3-kinase (PI3K) recruitment domain. In some embodiments, the PI3K recruitment domain comprises a sequence with at least 90% sequence identity to SEQ ID NO: 4. In some embodiments, the CFP intracellular domain comprises an intracellular domain from CD16a, CD64, CD68 or CD89. [0753] In some embodiments, the one or more polynucleic acids is an mRNA. [0754] In some embodiments, the nanoparticle delivery vehicle comprises a lipid nanoparticle. In some embodiments, the lipid nanoparticle comprises a polar lipid. In some embodiments, the polar lipid is a cationic lipid. In some embodiments, the lipid nanoparticle comprises a non-polar lipid. In some embodiments, the nanoparticle comprises polyethylene glycol (PEG). In some embodiments, the nanoparticle comprises cholesterol as one of the lipid components. In some embodiments, the lipid nanoparticle is from 100 to 300 nm in diameter. [0755] In some embodiments, the first antigen binding domain and the second antigen binding domain bind to the same antigen. In some embodiments, the first antigen binding domain and the second antigen binding domain bind to different antigens. In some embodiments, the first antigen binding domain and/or the second antigen binding domain binds to an antigen selected from the group consisting of TROP2, GPC3, CD5, HER2, CD137, CD70, Claudin 3, Claudin 18.2, TMPRSS, CD19, CD22, CD7, PSMA, MSLN and GP75. [0756] In some embodiments, the pharmaceutical composition is a lipid nanoparticle (LNP) encapsulated mRNA encoding a CAR, (alternatively, or interchangeably, a monocyte CAR, a chimeric fusion protein (CFP) or a phagocytic fusion protein (PFP)), where the CAR comprises an antigen binding domain that can bind to an antigen on a cell surface, a transmembrane domain and an intracellular signaling domain, and the mRNA comprises a 5’UTR, a 3’UTR, further comprising a 5’CAP at the terminal, where the 5’ CAP has a general formula 5’-m7GpppAmpG –, described as CC1 cap throughout the specification provided herein. The second nucleotide is methylated. The second nucleotide is Adenine. This is a Cap 1 structure, and is identical to most eukaryotic mRNA capping structure. In some embodiments, the CAR of the pharmaceutical composition comprises a TROP2 binding domain. [0757] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula m7GpppAmpG; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a) and an intracellular cytoplasmic sequence from the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a) respectively from which the TMD is derived. In some embodiments, the TMD has a sequence that is identical to the sequence of the TMD of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a). In some embodiments, the second nucleotide of the 5’Cap is further modified. In some embodiments, the adenosine at the second position is modified at the N6 position. The mRNA further comprises a poly A tail. The poly A tail is at least 80 nucleotides long. The poly A tail is about 100, about 110, about 120 or about 150 nucleotides long. [0758] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine comprising a further modification in the N6 position; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a) and an intracellular cytoplasmic sequence from the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a) respectively from which the TMD is derived. In some embodiments, the TMD has a sequence that is identical to the sequence of the TMD of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a). In some embodiments, the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., TROP2. In some embodiments, the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., GPC3. In some embodiments, the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., HER2. In some embodiments, the CFP comprises an antigen binding domain that binds to a cancer antigen, e.g., CD5. In some embodiments, the antigen binding domain of the CAR is an scFv. In some embodiments, the antigen binding domain of the CAR is a VHH domain. The mRNA further comprises a poly A tail. The poly A tail is at least 80 nucleotides long. The poly A tail is about 100, about 110, about 120 or about 150 nucleotides long. [0759] Myeloid cell-specific CFP constructs are designed to comprise a transmembrane domain that dimerizes with an endogenous Fc-gamma receptor that is specifically expressed in a myeloid cell, thereby becoming functionally active as a result of dimerization. [0760] In some embodiments, myeloid cell-specific CFP constructs comprises an Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a human Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a mouse Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a rodent Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a equine Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a porcine Fc ^R1 (CD89) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a ovine Fc ^R1 (CD89) transmembrane domain. In some embodiments, the CD89 TMD is at least 80% homologous to a human or a mouse CD89 TMD. In some embodiments, the CD89 TMD is at least 85% homologous to a human or a mouse CD89 TMD. In some embodiments, CD89 TMD is at least 90% homologous to a human or a mouse CD89 TMD. In some embodiments, CD89 TMD is at least 95% homologous to a human or a mouse CD89 TMD. [0761] In some embodiments, myeloid cell-specific CFP constructs comprises an Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a human Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a mouse Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a rodent Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a equine Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a porcine Fc γRIII (CD16) transmembrane domain. In some embodiments, myeloid cell-specific CFP constructs comprises a ovine Fc γRIII (CD16) transmembrane domain. In some embodiments, the CD16 TMD is at least 80% homologous to a human or a mouse CD16 TMD. In some embodiments, the CD16TMD is at least 85% homologous to a human or a mouse CD16 TMD. In some embodiments, CD16 TMD is at least 90% homologous to a human or a mouse CD16 TMD. In some embodiments, CD16 TMD is at least 95% homologous to a human or a mouse CD16 TMD. In one embodiment, the Fc γRIII is Fc γRIIIA (CD16a). [0762] It is contemplated herein that any myeloid cell specific CFP can be designed or generated using a TMD from a CD89 molecule, a CD16 molecule, a CD64 molecule or a CD32a molecule, and a myeloid cell specific CFP having the desired antigen binding domain can be created based on the compositions and methods described in the instant disclosure. In some embodiments, the CFP exhibits myeloid cell-specific expression. In some embodiments, the myeloid cell-specific expression is an Fc-gamma dependent expression, that is, if the cell that comprises the nucleic acid comprising the CFP construct expresses an Fc-gamma endogenously, the CFP will co-express in the cell. In some embodiments, the CFP, even though it expresses in a cell other than a myeloid cell, does so only transiently, and may not be detectable. In some embodiments, only a myeloid cell expresses a functional CFP in an Fc-gamma dependent manner, that is, the CFP is not functional in a cell that does not express Fc-gamma endogenously. In some embodiments, the myeloid cell specific CFP comprises an antigen binding domain that binds to a cancer cell-specific target antigen, and an intracellular region comprising 1, 2, 3 or more domain, e.g., intracellular cell signaling domain, for example, a PI3 kinase recruitment domain. For example, the myeloid cell specific CFP comprises an antigen binding domain that binds to a CD5 target antigen. The respective CFP is designed herein comprising : an extracellular domain comprising an anti-CD5 antibody or a part thereof, e.g., an anti-CD5 ScFv, a hinge domain, e.g. a CD8 hinge domain, a transmembrane domain capable of dimerizing with an Fc-gamma receptor endogenously expressed by a myeloid cell and an intracellular region comprising 1, 2, 3 or more domain, e.g., intracellular cell signaling domain including, for example, a CD40 intracellular signaling domain, and/or a PI3kinase recruitment domain. [0763] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position and the encoded CFP is an anti-CD5 CFP that comprises sequence that is at least 80% identical to the sequence of
Figure imgf000153_0001
Figure imgf000154_0001
G S NQ QGSGS G Q S GQ G N S N . (SEQ ID NO. 14, wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to Kabat scheme). [0764] In one embodiment, the anti-CD5 heavy chain variable domain comprises an amino acid sequence that is at least 80% identical to the sequence
Figure imgf000154_0003
[0765] In one embodiment, the anti-CD5 light chain variable domain comprises an amino acid sequence that is at least 80% identical to the sequence DIQMTQSPSSLSASVGDRVTITCRASQDINSYLSWFQQKPGKAPKTLIYRANRLESGVPSRFS GSGSGTDYTLTISSLQYEDFGIYYCQQYDESPWTFGGGTKLEIK. [0766] In one embodiment, the anti-CD5 binding domain comprises a heavy chain CDR1 sequence, a CDR2 sequence and a CDR3 sequence, wherein the CDR1 may be denoted by a sequence of NYGMN, a CDR2 may be denoted by a sequence of WINTHTGEPTYADSFKG and a CDR3 may be denoted by a sequence of RGYDWYFDV; or the anti-CD5 binding domain may comprise a sequence of
Figure imgf000154_0002
wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to Kabat scheme. [0767] In some embodiments, the anti-CD5 binding domain comprises a light chain CDR1 sequence, a CDR2 sequence and a CDR3 sequence, wherein the CDR1 sequence may be RASQDINSYLS; the CDR2 sequence may be RANRLES, and the CDR3 sequence may be QQYDESPWT; or wherein the anti-CD5 binding domain may comprise a sequence of DIQMTQSPSSLSASVGDRVTITCRASQDINSYLSWFQQKPGKAPKTLIYRANRLESGVPSRFS GSGSGTDYTLTISSLQYEDFGIYYCQQYDESPWTFGGGTKLEIK wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to Kabat scheme. [0768] In some embodiments, an exemplary anti-CD5 CAR sequence comprises a signal sequence of MWLQSLLLLGTVACSIS, or a sequence that is at least 90% identical to the above sequence. [0769] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and the encoded CFP is an anti- HER2 CFP that comprises a sequence that is at least 80% identical to :
Figure imgf000155_0001
SDGVYTGLSTRNQETYETLKHEKPPQGSYEDMRGILYAAPQLRSIRGQPGPNHEEDADSYE
Figure imgf000155_0002
(wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to the Kabat numbering scheme). [0770] In some embodiments, the anti-HER2 CFP antigen binding domain comprises a heavy chain variable sequence that is at least 80% identical to :
Figure imgf000155_0003
Q Q [0771] In some embodiments, the anti-HER2 CFP antigen binding domain comprises a light chain variable sequence that is at least 80% identical to :
Figure imgf000155_0004
[0772] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a); wherein, the encoded CFP is a TROP2 binding CFP, and comprises a sequence that has at least 80%, or at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence:
Figure imgf000155_0005
EASADVAEPSWSQQMCQPGLTFARTPSVCK (wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to the Kabat numbering scheme). [0773] In some embodiments, the antigen binding domain binds to TROP2 antigen, and comprises a heavy chain variable domain having a sequence that has at least 80%, or at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence QVQLQQSGSELKKPGASVKVSCKASGYTFTNYGMNWVKQAPGQGLKWMGWINTYTGEP TYTDDFKGRFAFSLDTSVSTAYLQISSLKADDTAVYFCARGGFGSSYWYFDVWGQGSLVT VSS. [0774] In some embodiments, the antigen binding domain binds to TROP2 antigen, and comprises a light chain variable domain having a sequence that has at least 80%, or at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence DIQLTQSPSSLSASVGDRVSITCKASQDVSIAVAWYQQKPGKAPKLLIYSASYRYTGVPDRF SGSGSGTDFTLTISSLQPEDFAVYYCQQHYITPLTFGAGTKVEIKR. [0775] In some embodiments, wherein the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the CFP comprises a TROP2 binding domain that comprises a heavy chain CDR3 sequence, GGFGSSYWYFDV. In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a); and wherein the antigen binding domain binds to TROP2 antigen, and wherein the binding domain comprises a heavy chain CDR3 sequence, GGFGSSYWYFDV. In some embodiment, the mRNA encodes a TROP2-binding CFP that comprises an antigen binding domain having a heavy chain (VH) CDR1 sequence of NYGMN and a VH CDR2 sequence of WINTYTGEPTYTDDFKG. [0776] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a); wherein, the encoded CFP is a GPC3 binding CFP, and comprises a sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence:
Figure imgf000157_0001
Figure imgf000157_0002
(wherein underlined residues represent a CDR1, a CDR2 and a CDR3 in successive order according to the Kabat numbering scheme). [0777] In some embodiments, the pharmaceutical composition comprises an LNP comprising a cationic lipid, a non-cationic lipid, optionally, a neutral lipid and/or cholesterol; encapsulating an mRNA comprising a sequence encoding a CFP, wherein the mRNA comprises a 5’-Cap1 having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position; and wherein the encoded CFP comprises a transmembrane domain from a sequence from the transmembrane domain of the protein Fc γR1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a); wherein, the encoded chimeric antigen receptor comprises GPC3 binding variable heavy chain (VH), and a variable light chain (VL), wherein the variable heavy chain comprises a HCDR1, a HCDR2 and a HCDR3, the HCDR3 having a sequence selected from a group consisting of ATACADTTQYAYDY, ATACADTTLYEYDY, ATACVDTTQYEYDY, ATACADATQHEYDY, ATACADTTQYDYDY, ATACADTTQYEYDY, ATACADTTHYEYDY, ATACVITTLYEYDY, ATACAETTLYEYDY, ATACADTTQHEYDY, ATACVDTTHYEYDY, ATACASTTLYEYDY, ATACVVTTLYEYDY, ATACGGATGPYDY, ATACAGAIGPYDY, ATACVVVGDQNDY, ATACVVVGDRNDY, ATDCAGGTSTPYDY, ATDCAGGTATPYDY, ATACVVADRNEYDY, ATSCVVVTKNEYDY, ATACSGLTHEYDY, ATTCSGLTHEYDY, ATACANWSSLGPYDY, ATACANWSTLGPYDY, ATACSDPRVYEYDY, ATTCASPEKYEYDY, ATHCGGTSWGTSYDY, ATHCGGSSWSNEYDY, YARYSGRTY, ASSAWPAGPKHQVEYDY, ATACGSLVGMYDY, ATACGSAVHEYDY, ATDCVGFGSNWFDY, ATACASPVIYEYDY, ATDCAGGVGHEYDY, ATDCSLHGSDYPYDY and AVRIYSGSFDNTLAYDY. [0778] In some embodiments, the pharmaceutical composition comprises an mRNA having a sequence that is at least 95% identical to a polynucleotide sequence:
Figure imgf000157_0003
Figure imgf000158_0001
The 5’ UTR sequence and the
Figure imgf000158_0002
3’ UTR sequence is underlined. [0779] In some embodiments, provided herein is a composition comprising an polynucleotide, having a sequence that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to
Figure imgf000158_0003
Figure imgf000159_0001
C UG G GGG UUUU U UGCCGCGCCCC GCUGC C GC U C UCGGGCCC
Figure imgf000159_0002
In some embodiments, the polynucleotide is an mRNA, further comprising a 5’ UTR, a 3’ UTR, a poly A tail, and a 5’- cap, wherein the cap is a cap-1 analog. In some embodiments, the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’-m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position. In some embodiments, the polynucleotide, e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non-cationic lipid, and PEG. [0780] In some embodiments, the mRNA encodes a chimeric fusion protein having an anti- CD70 binding domain, comprising a sequence that is at least 80% identical to, or a sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence :
Figure imgf000159_0003
QQ Q Q Q
Figure imgf000159_0004
Q ; wherein, in some embodiments, the mRNA comprises a 5’ UTR, a 3’ UTR, a poly A tail, and a 5’-cap, wherein the cap is a cap-1 analog. In some embodiments, the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position. In some embodiments, the polynucleotide, e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non- cationic lipid, and PEG. [0781] In some embodiments, the mRNA encodes a chimeric fusion protein having an anti- CD137 binding domain, comprising a sequence that is at least 80% identical to, or a sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence :
Figure imgf000160_0001
; wherein, in some embodiments, the mRNA comprises a 5’ UTR, a
Figure imgf000160_0002
3’ UTR, a poly A tail, and a 5’-cap, wherein the cap is a cap-1 analog. In some embodiments, the cap-1 analog is a CC1 cap; wherein the cap is characterized as having a structural formula 5’- m7GpppAmpG, wherein the second nucleotide is a methylated adenine, optionally comprising a further modification in the N6 position. In some embodiments, the polynucleotide, e.g., the mRNA as described herein is encapsulated in a lipid nanoparticle, having a cationic lipid, a non- cationic lipid, and PEG. [0782] In some embodiments, the pharmaceutical composition inhibits growth of a cancer when administered to a human subject with the cancer. Treatment Methods [0783] Provided herein are methods for treating cancer in a subject using a pharmaceutical composition comprising engineered myeloid cells, such as phagocytic cells (e.g., macrophages), expressing a recombinant nucleic acid encoding a CFP, such as a phagocytic receptor (PR) fusion protein (PFP), to target, attack and kill cancer cells directly or indirectly. The engineered myeloid cells, such as phagocytic cells, are also designated as CAR-P cells in the descriptions herein. [0784] Cancers include, but are not limited to T cell lymphoma, cutaneous lymphoma, B cell cancer (e.g., multiple myeloma, Waldenstrom's macroglobulinemia), the heavy chain diseases (such as, for example, alpha chain disease, gamma chain disease, and mu chain disease), benign monoclonal gammopathy, and immunocytic amyloidosis, melanomas, breast cancer, lung cancer, bronchus cancer, colorectal cancer, prostate cancer (e.g., metastatic, hormone refractory prostate cancer), pancreatic cancer, stomach cancer, ovarian cancer, urinary bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, cancer of the oral cavity or pharynx, liver cancer, kidney cancer, testicular cancer, biliary tract cancer, small bowel or appendix cancer, salivary gland cancer, thyroid gland cancer, adrenal gland cancer, osteosarcoma, chondrosarcoma, cancer of hematological tissues, and the like. Other non-limiting examples of types of cancers applicable to the methods encompassed by the present disclosure include human sarcomas and carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, colorectal cancer, pancreatic cancer, breast cancer, ovarian cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, liver cancer, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, bone cancer, brain tumor, testicular cancer, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, retinoblastoma; leukemias, e.g., acute lymphocytic leukemia and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphoma (Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, and heavy chain disease. In some embodiments, the cancer is an epithelial cancer such as, but not limited to, bladder cancer, breast cancer, cervical cancer, colon cancer, gynecologic cancers, renal cancer, laryngeal cancer, lung cancer, oral cancer, head and neck cancer, ovarian cancer, pancreatic cancer, prostate cancer, or skin cancer. In other embodiments, the cancer is breast cancer, prostate cancer, lung cancer, or colon cancer. In still other embodiments, the epithelial cancer is non-small-cell lung cancer, nonpapillary renal cell carcinoma, cervical carcinoma, ovarian carcinoma (e.g., serous ovarian carcinoma), or breast carcinoma. The epithelial cancers can be characterized in various other ways including, but not limited to, serous, endometrioid, mucinous, clear cell, or undifferentiated. In some embodiments, the present disclosure is used in the treatment, diagnosis, and/or prognosis of lymphoma or its subtypes, including, but not limited to, mantle cell lymphoma. Lymphoproliferative disorders are also considered to be proliferative diseases. [0785] In general, cellular immunotherapy comprises providing the patient a medicament comprising live cells. In some embodiments a patient or a subject having cancer, is treated with autologous cells, the method comprising, isolation of PBMC-derived myeloid cells, such as macrophages, modifying the cells ex vivo to generate phagocytic myeloid cells capable of tumor lysis by introducing into the cells a recombinant nucleic acid encoding a CFP, and administering the modified myeloid cells into the subject. [0786] In some embodiments, a subject is administered one or more doses of a pharmaceutical composition comprising therapeutic myeloid cells, such as phagocytic cells, wherein the cells are allogeneic. An HLA may be matched for compatibility with the subject, and such that the cells do not lead to graft versus Host Disease, GVHD. A subject arriving at the clinic is HLA typed for determining the HLA antigens expressed by the subject. [0787] HLA‐typing is conventionally carried out by either serological methods using antibodies or by PCR‐based methods such as Sequence Specific Oligonucleotide Probe Hybridization (SSOP), or Sequence Based Typing (SBT). [0788] The sequence information may be identified by either sequencing methods or methods employing mass spectrometry, such as liquid chromatography—mass spectrometry (LC-MS or LC- MS/MS, or alternatively HPLC-MS or HPLC-MS/MS). These sequencing methods may be well- known to a skilled person and are reviewed in Medzihradszky KF and Chalkley RJ. Mass Spectrom Rev.2015 Jan-Feb;34(1):43-63. [0789] In some embodiments, the phagocytic cell is derived from the subject, transfected or transduced with the recombinant nucleic acid in vitro, expanded in cell culture in vitro for achieving a number suitable for administration, and then administered to the subject. In some embodiments, the steps of transfected or transduced with the recombinant nucleic acid in vitro, expanded in cell culture in vitro for achieving a number suitable for administration takes 2 days, or 3 days, or 4 days or 5 days or 6 days or 7 days or 8 days or 9 days or 10 days. [0790] In some embodiments, sufficient quantities of transfected or transduced myeloid cells, such as macrophages, comprising the recombinant nucleic acid are preserved aseptically, which are administered to the subject as “off the shelf” products after HLA typing and matching the product with the recipients HLA subtypes. In some embodiments, the engineered phagocytes are cryopreserved. In some embodiments, the engineered phagocytes are cryopreserved in suitable media to withstand thawing without considerable loss in cell viability. [0791] In some embodiment, the subject is administered a pharmaceutical composition comprising the DNA, or the mRNA or the circRNA in a vector, or in a pharmaceutically acceptable excipient described above. [0792] In some embodiments the administration of the off the shelf cellular products may be instantaneous, or may require 1 day, 2 days or 3 days or 4 days or 5 days or 6 days or 7 days or more prior to administration. The pharmaceutical composition comprising cell, or nucleic acid may be preserved over time from preparation until use in frozen condition. In some embodiments, the pharmaceutical composition may be thawed once. In some embodiments, the pharmaceutical composition may be thawed more than once. In some embodiments, the pharmaceutical composition is stabilized after a freeze-thaw cycle prior administering to the subject. In some embodiments the pharmaceutical composition is tested for final quality control after thawing prior administration. [0793] In some embodiments, a composition comprising 10γ6 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10^7 engineered cells are administered per administration dose. In some embodiments, a composition comprising 5X 10^7 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10^8 engineered cells are administered per administration dose. In some embodiments, a composition comprising 2x10^8 engineered cells are administered per administration dose. In some embodiments, a composition comprising 5x10^8 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10^9 engineered cells are administered per administration dose. In some embodiments, a composition comprising 10^10 engineered cells are administered per administration dose. [0794] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once. [0795] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered more than once. [0796] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are twice, thrice, four times, five times, six times, seven times, eight times, nine times, or ten times or more to a subject over a span of time comprising a few months, a year or more. [0797] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered twice weekly. [0798] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once weekly. [0799] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once every two weeks. [0800] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once every three weeks. [0801] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered once monthly. [0802] In some embodiments, the engineered phagocytic cells are administered once in every 2 months, once in every 3 months, once in every 4 months, once in every 5 months or once in every 6 months. [0803] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by injection. [0804] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by infusion. [0805] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by intravenous infusion. [0806] In some embodiments, the engineered myeloid cells, such as phagocytic cells, are administered by subcutaneous infusion. [0807] The pharmaceutical composition comprising the recombinant nucleic acid or the engineered cells may be administered by any route which results in a therapeutically effective outcome. These include, but are not limited to enteral (into the intestine), gastroenteral, epidural (into the dura mater), oral (by way of the mouth), transdermal, peridural, intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intravenous bolus, intravenous drip, intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection (into a pathologic cavity), intracavitary (into the base of the penis), intravaginal administration, intrauterine, extra-amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), transvaginal, insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), in ear drops, auricular (in or by way of the ear), buccal (directed toward the cheek), conjunctival, cutaneous, dental (to a tooth or teeth), electro-osmosis, endocervical, endosinusial, endotracheal, extracorporeal, hemodialysis, infiltration, interstitial, intra-abdominal, intra-amniotic, intra-articular, intrabiliary, intrabronchial, intrabursal, intracartilaginous (within a cartilage), intracaudal (within the cauda equine), intracisternal (within the cisterna magna cerebellomedularis), intracorneal (within the cornea), dental intracornal, intracoronary (within the coronary arteries), intracorporus cavernosum (within the dilatable spaces of the corporus cavernosa of the penis), intradiscal (within a disc), intraductal (within a duct of a gland), intraduodenal (within the duodenum), intradural (within or beneath the dura), intraepidermal (to the epidermis), intraesophageal (to the esophagus), intragastric (within the stomach), intragingival (within the gingivae), intraileal (within the distal portion of the small intestine), intralesional (within or introduced directly to a localized lesion), intraluminal (within a lumen of a tube), intralymphatic (within the lymph), intramedullary (within the marrow cavity of a bone), intrameningeal (within the meninges), intraocular (within the eye), intraovarian (within the ovary), intrapericardial (within the pericardium), intrapleural (within the pleura), intraprostatic (within the prostate gland), intrapulmonary (within the lungs or its bronchi), intrasinal (within the nasal or periorbital sinuses), intraspinal (within the vertebral column), intrasynovial (within the synovial cavity of a joint), intratendinous (within a tendon), intratesticular (within the testicle), intrathecal (within the cerebrospinal fluid at any level of the cerebrospinal axis), intrathoracic (within the thorax), intratubular (within the tubules of an organ), intratumor (within a tumor), intratympanic (within the aurus media), intravascular (within a vessel or vessels), intraventricular (within a ventricle), iontophoresis (by means of electric current where ions of soluble salts migrate into the tissues of the body), irrigation (to bathe or flush open wounds or body cavities), laryngeal (directly upon the larynx), nasogastric (through the nose and into the stomach), occlusive dressing technique (topical route administration which is then covered by a dressing which occludes the area), ophthalmic (to the external eye), oropharyngeal (directly to the mouth and pharynx), parenteral, percutaneous, periarticular, peridural, perineural, periodontal, rectal, respiratory (within the respiratory tract by inhaling orally or nasally for local or systemic effect), retrobulbar (behind the pons or behind the eyeball), soft tissue, subarachnoid, subconjunctival, submucosal, topical, transplacental (through or across the placenta), transtracheal (through the wall of the trachea), transtympanic (across or through the tympanic cavity), ureteral (to the ureter), urethral (to the urethra), vaginal, caudal block, diagnostic, nerve block, biliary perfusion, cardiac perfusion, photopheresis or spinal. In specific embodiments, compositions may be administered in a way which allows them to cross the blood-brain barrier, vascular barrier, or other epithelial barrier. [0808] In some embodiments, the subject is administered a pharmaceutical composition comprising the nucleic acid encoding the CFP or PFP as described herein. In some embodiments, the subject is administered a pharmaceutical composition comprising DNA, mRNA, or circRNA. In some embodiments, the subject is administered a vector harboring the nucleic acid, e.g., DNA, mRNA, or circRNA. In some embodiments, the nucleic acid is administered or in a pharmaceutically acceptable excipient described above. [0809] In some embodiments, the subject is administered a nanoparticle (NP) associated with the nucleic acid, e.g. a DNA, an mRNA, or a circRNA encoding the CFP or PFP as described herein. In some embodiments, the nucleic acid is encapsulated in the nanoparticle. In some embodiments, the nucleic acid is conjugated to the nanoparticle. In some embodiments, the NP is a polylactide-co- glycolide (PGLA) particle. In some embodiments, the NP is administered subcutaneously. In some embodiments, the NP is administered intravenously. In some embodiments, the NP is engineered in relation to the administration route. For example, the size, shape, or charges of the NP maybe engineered according to the administration route. In some embodiments, subcutaneously administered NPs are less than 200nm in size. In some embodiments, subcutaneously administered NPs are more than 200nm in size. In some embodiments, subcutaneously administered NPs are at least 30nm in size. In some embodiments, the NPs are intravenously infused. In some embodiments, intravenously infused NPs are at least 5nm in diameter. In some embodiments, intravenously infused NPs are at least 30nm in diameter. In some embodiments, intravenously infused NPs are at least 100nm in diameter. In certain embodiments, the administered NPs, e.g. intravenously administered NPs, are engulfed by circulating monocytes. Additional NP design and administration approaches are described in Getts et al., Trends Immunol. 36(7): 419-427 (2015), the entirety of which is incorporated herein by reference. [0810] In some embodiments, the subject is administered a pharmaceutical composition comprising a circRNA encoding the CFP or PFP as described herein. The circRNA may be administered in any route as described herein. In some embodiments, the circRNA may be directly infused. In some embodiments, the circRNA may be in a formulation or solution comprising one or more of sodium chloride, calcium chloride, phosphate and/or EDTA. In some embodiment, the circRNA solution may include one or more of saline, saline with 2 mM calcium, 5% sucrose, 5% sucrose with 2 mM calcium, 5% Mannitol, 5% Mannitol with 2 mM calcium, Ringer's lactate, sodium chloride, sodium chloride with 2 mM calcium and mannose. In some embodiments, the circRNA solution is lyophilized. The amount of each component may be varied to enable consistent, reproducible higher concentration saline or simple buffer formulations. The components may also be varied in order to increase the stability of circRNA in the buffer solution over a period of time and/or under a variety of conditions. In some embodiments, the circRNA is formulated in a lyophilized gel- phase liposomal composition. In some embodiments, the circRNA formulation comprises a bulking agent, e.g. sucrose, trehalose, mannitol, glycine, lactose and/or raffinose, to impart a desired consistency to the formulation and/or stabilization of formulation components. Additional formulation and administration approaches for circRNA as described in US Publications No. US2012060293, and US20170204422 are herein incorporated by reference in entirety. EMBODIMENTS 1. A composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein upon binding of the PFP to the antigen of the target cell, the killing or phagocytosis activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. 2. The composition of embodiment 1, wherein the intracellular signaling domain is derived from a phagocytic or tethering receptor or wherein the intracellular signaling domain comprises a phagocytosis activation domain. 3. The composition of embodiment 1 or 2, wherein the intracellular signaling domain is derived from a receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. 4. The composition of any one of embodiments 1-3, wherein the intracellular signaling domain is derived from a receptor selected from the group consisting of the receptors listed in Table 2. 5. The composition of any one of embodiments 1-4, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain. 6. The composition of embodiment 5, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. 7. A composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. 8. The composition of embodiment 7, wherein upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. 9. The composition of embodiment 7 or 8, wherein the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim-1, Tim-4 and CD169. 10. The composition of any one of embodiments 7-9, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain. 11. A composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim-1, Tim-4 and CD169. 12. The composition of embodiment 11, wherein upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 55% compared to a cell not expressing the PFP. 13. The composition of embodiment 11 or 12, wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. 14. The composition of any one of embodiments 11-13, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain. 15. The composition of embodiment 14, wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. 16. A composition comprising a recombinant nucleic acid encoding a phagocytic or tethering receptor (PR) fusion protein (PFP) comprising: (a) a PR subunit comprising: (i) a transmembrane domain, and (ii) an intracellular domain comprising an intracellular signaling domain; and (b) an extracellular domain comprising an antigen binding domain specific to an antigen of a target cell; wherein the transmembrane domain and the extracellular domain are operatively linked; and wherein the intracellular signaling domain comprises a pro-inflammatory signaling domain that is not a PI3K recruitment domain. 17. The composition of embodiment 16, wherein upon binding of the PFP to the antigen of the target cell, the killing activity of a cell expressing the PFP is increased by at least greater than 20% compared to a cell not expressing the PFP. 18. The composition of embodiment 16 or 17, wherein the intracellular signaling domain is derived from a phagocytic receptor. 19. The composition of any one of embodiments 16-18, wherein the intracellular signaling domain is derived from a phagocytic receptor other than a phagocytic receptor selected from Megf10, MerTk, FcR-alpha, or Bai1. 20. The composition of any one of embodiments 16-19, wherein the intracellular signaling domain is derived from a phagocytic receptor selected from the group consisting of lectin, dectin 1, CD206, scavenger receptor A1 (SRA1), MARCO, CD36, CD163, MSR1, SCARA3, COLEC12, SCARA5, SCARB1, SCARB2, CD68, OLR1, SCARF1, SCARF2, CXCL16, STAB1, STAB2, SRCRB4D, SSC5D, CD205, CD207, CD209, RAGE, CD14, CD64, F4/80, CCR2, CX3CR1, CSF1R, Tie2, HuCRIg(L), CD64, CD32a, CD16a, CD89, Fc-alpha receptor I, CR1, CD35, CD3ζ, CR3, CR4, Tim- 1, Tim-4 and CD169. 21. The composition of any one of embodiments 1-15, wherein the intracellular signaling domain comprises a PI3K recruitment domain. 22. The composition of any one of the preceding embodiments, wherein the PFP functionally incorporates into a cell membrane of a cell when the PFP is expressed in the cell. 23. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the PFP. 24. The composition of embodiment 23, wherein a cell expressing the PFP exhibits at least a 1.1- fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the PFP. 25. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits at least a 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30- fold or 50-fold increase in phagocytosis of a target cell expressing the antigen compared to a cell not expressing the PFP. 26. The composition of any one of the preceding embodiments, wherein the target cell expressing the antigen is a cancer cell. 27. The composition of any one of the preceding embodiments, wherein the target cell expressing the antigen is at least 0.8 microns in diameter. 28. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain is derived from a scavenger receptor. 29. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in production of a cytokine compared to a cell not expressing the PFP. 30. The composition according to embodiment 29, wherein the cytokine is selected from the group consisting of IL-1, IL3, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon and combinations thereof. 31. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in effector activity compared to a cell not expressing the PFP. 32. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in cross-presentation compared to a cell not expressing the PFP. 33. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of an MHC class II protein compared to a cell not expressing the PFP. 34. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD80 compared to a cell not expressing the PFP. 35. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD86 compared to a cell not expressing the PFP. 36. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of MHC class I protein compared to a cell not expressing the PFP. 37. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of TRAIL/TNF Family death receptors compared to a cell not expressing the PFP. 38. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of B7-H2 compared to a cell not expressing the PFP. 39. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of LIGHT compared to a cell not expressing the PFP. 40. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of HVEM compared to a cell not expressing the PFP. 41. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD40 compared to a cell not expressing the PFP. 42. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of TL1A compared to a cell not expressing the PFP. 43. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of 41BBL compared to a cell not expressing the PFP. 44. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of OX40L compared to a cell not expressing the PFP. 45. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of GITRL death receptors compared to a cell not expressing the PFP. 46. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD30L compared to a cell not expressing the PFP. 47. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of TIM4 compared to a cell not expressing the PFP. 48. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of TIM1 ligand compared to a cell not expressing the PFP. 49. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of SLAM compared to a cell not expressing the PFP. 50. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD48 compared to a cell not expressing the PFP. 51. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD58 compared to a cell not expressing the PFP. 52. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD155 compared to a cell not expressing the PFP. 53. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of CD112 compared to a cell not expressing the PFP. 54. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of PDL1 compared to a cell not expressing the PFP. 55. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in expression of B7-DC compared to a cell not expressing the PFP. 56. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in respiratory burst compared to a cell not expressing the PFP. 57. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in ROS production compared to a cell not expressing the PFP. 58. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in iNOS production compared to a cell not expressing the PFP. 59. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in iNOS production compared to a cell not expressing the PFP. 60. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in extra-cellular vesicle production compared to a cell not expressing the PFP. 61. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in trogocytosis with a target cell expressing the antigen compared to a cell not expressing the PFP. 62. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in resistance to CD47 mediated inhibition of phagocytosis compared to a cell not expressing the PFP. 63. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in resistance to LILRB1 mediated inhibition of phagocytosis compared to a cell not expressing the PFP. 64. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a Rac inhibition domain, a Cdc42 inhibition domain or a GTPase inhibition domain. 65. The composition of embodiment 64, wherein the Rac inhibition domain, the Cdc42 inhibition domain or the GTPase inhibition domain inhibits Rac, Cdc42 or GTPase at a phagocytic cup of a cell expressing the PFP. 66. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises an F-actin disassembly activation domain, a ARHGAP12 activation domain, a ARHGAP25 activation domain or a SH3BP1 activation domain. 67. The composition of any one of the preceding embodiments, wherein a cell expressing the PFP exhibits an increase in phosphatidylinositol 3,4,5-trisphosphate production. 68. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an Ig binding domain. 69. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an IgA, IgD, IgE, IgG, IgM, FcRγI, FcRγIIA, FcRγIIB, FcRγIIC, FcRγIIIA, FcRγIIIB, FcRn, TRIM21, FcRL5 binding domain. 70. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcR extracellular domain. 71. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcR-alpha, FcRβ , FcRε or FcRγ extracellular domain. 72. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcRα (FCAR) extracellular domain. 73. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcRβ extracellular domain. 74. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcRε (FCER1A) extracellular domain. 75. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an FcRγ (FDGR1A, FCGR2A, FCGR2B, FCGR2C, FCGR3A, FCGR3B) extracellular domain. 76. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises an integrin domain. 77. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises one or more integrin α1, α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αD, αE, αL, αM, αV, αX, β1, β2, β3, β4, β5, β6, β7, or β8 domains. 78. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a CD47 inhibition domain. 79. The composition of any one of the preceding embodiments, wherein the PSR subunit further comprises an extracellular domain operatively linked to the transmembrane domain and the extracellular antigen binding domain. 80. The composition of embodiment 79, wherein the extracellular domain further comprises an extracellular domain of a receptor, a hinge, a spacer or a linker. 81. The composition of embodiment 80, wherein the extracellular domain comprises an extracellular portion of a PSR. 82. The composition of embodiment 81, wherein the extracellular portion of the PSR is derived from the same PSR as the PSR intracellular signaling domain. 83. The composition of any one of the embodiments 79-82, wherein the extracellular domain comprises an extracellular domain of a scavenger receptor or an immunoglobulin domain. 84. The composition of embodiment 83, wherein the immunoglobulin domain comprises an extracellular domain of an immunoglobulin or an immunoglobulin hinge region. 85. The composition of any one of the embodiments 79-84, wherein the extracellular domain comprises a phagocytic engulfment marker. 86. The composition of any one of the embodiments 79-85, wherein the extracellular domain comprises a structure capable of multimeric assembly. 87. The composition of any one of the embodiments 79-86, wherein the extracellular domain comprises a scaffold for multimerization. 88. The composition of any one of the preceding embodiments, wherein the extracellular domain is at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. 89. The composition of any one of the preceding embodiments, wherein the extracellular domain is at most 500, 400, 300, 200, or 100 amino acids in length. 90. The composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain specifically binds to the antigen of a target cell. 91. The composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises an antibody domain. 92. The composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises a receptor domain, antibody domain, wherein the antibody domain comprises a functional antibody fragment, a single chain variable fragment (scFv), an Fab, a single- domain antibody (sdAb), a nanobody, a VH domain, a VL domain, a VNAR domain, a VHH domain, a bispecific antibody, a diabody, or a functional fragment or a combination thereof. 93. The composition of any one of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises a ligand, an extracellular domain of a receptor or an adaptor. 94. The composition of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises a single extracellular antigen binding domain that is specific for a single antigen. 95. The composition of any one of any one of the preceding embodiments, wherein the extracellular antigen binding domain comprises at least two extracellular antigen binding domains, wherein each of the at least two extracellular antigen binding domains is specific for a different antigen. 96. The composition of any one of the preceding embodiments, wherein the antigen is a cancer antigen or a pathogenic antigen or an autoimmune antigen. 97. The composition of any one of the preceding embodiments, wherein the antigen comprises a viral antigen. 98. The composition of any one of the preceding embodiments, wherein the antigen is a T- lymphocyte antigen. 99. The composition of any one of the preceding embodiments, wherein the antigen is an extracellular antigen. 100. The composition of any one of the preceding embodiments, wherein the antigen is an intracellular antigen. 101. The composition of any one of the preceding embodiments, wherein the antigen is selected from the group consisting of Thymidine Kinase (TK1), Hypoxanthine-Guanine Phosphoribosyltransferase (HPRT), Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1), Mucin-1, Mucin-16 (MUC16), MUC1, Epidermal Growth Factor Receptor vIII (EGFRvIII), Mesothelin, Human Epidermal Growth Factor Receptor 2 (HER2), Mesothelin, EBNA-1, LEMD1, Phosphatidyl Serine, Carcinoembryonic Antigen (CEA), B-Cell Maturation Antigen (BCMA), Glypican 3 (GPC3), Follicular Stimulating Hormone receptor, Fibroblast Activation Protein (FAP), Erythropoietin-Producing Hepatocellular Carcinoma A2 (EphA2), EphB2, a Natural Killer Group 2D (NKG2D) ligand, Disialoganglioside 2 (GD2), CD2, CD3, CD4, CD5, CD7, CD8, CD19, CD20, CD22, CD24, CD30, CD33, CD38, CD44v6, CD45, CD56CD79b, CD97, CD117, CD123, CD133, CD138, CD171, CD179a, CD213A2, CD248, CD276, PSCA, CS-1, CLECL1, GD3, PSMA, FLT3, TAG72, EPCAM, IL-1, an integrin receptor, PRSS21, VEGFR2, PDGFR-β, SSEA-4, EGFR, NCAM, prostase, PAP, ELF2M, GM3, TEM7R, CLDN6, TSHR, GPRC5D, ALK, IGLL1 and combinations thereof. 102. The composition of any one of the preceding embodiments, wherein the antigen is selected from the group consisting of CD2, CD3, CD4, CD5, CD7, CCR4, CD8, CD30, CD45, CD56. 103. The composition of any one of the preceding embodiments, wherein the antigen is an ovarian cancer antigen or a T lymphoma antigen. 104. The composition of any one of the preceding embodiments, wherein the antigen is an integrin receptor. 105. The composition of any one of the preceding embodiments, wherein the antigen is an integrin receptor selected from the group consisting of α1, α2, αIIb, α3, α4, α5, α6, α7, α8, α9, α10, α11, αD, αE, αL, αM, αV, αX, β 1, β 2, β 3, β 4, β 5, β 6, β 7, and β8. 106. The composition of any one of the preceding embodiments, wherein the antigen comprises 2 or more antigens. 107. The composition of any one of the preceding embodiments, wherein the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker. 108. The composition of any one of the preceding embodiments, wherein the transmembrane domain and the extracellular antigen binding domain is operatively linked through a linker such as the hinge region of CD8α, IgG1 or IgG4. 109. The composition of any one of the preceding embodiments, wherein the extracellular domain comprises a multimerization scaffold. 110. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises an FcR transmembrane domain. 111. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises an FcR-ε with no more than 20, 10 or 5 modifications transmembrane domain. 112. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises a transmembrane domain from a syntaxin such as syntaxin 3 or syntaxin 4 or syntaxin 5. 113. The composition of any one of the preceding embodiments, wherein the transmembrane domain oligomerizes with a transmembrane domain of an endogenous receptor when the PFP is expressed in a cell. 114. The composition of any one of the preceding embodiments, wherein the transmembrane domain oligomerizes with a transmembrane domain of an exogenous receptor when the PFP is expressed in a cell. 115. The composition of any one of the preceding embodiments, wherein the transmembrane domain dimerizes with a transmembrane domain of an endogenous receptor when the PFP is expressed in a cell. 116. The composition of any one of the preceding embodiments, wherein the transmembrane domain dimerizes with a transmembrane domain of an exogenous receptor when the PFP is expressed in a cell. 117. The composition of any one of the preceding embodiments, wherein the transmembrane domain is derived from a protein that is different than the protein from which the intracellular signaling domain is derived. 118. The composition of any one of the preceding embodiments, wherein the transmembrane domain is derived from a protein that is different than the protein from which the extracellular domain is derived. 119. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises a transmembrane domain of a phagocytic receptor. 120. The composition of any one of the preceding embodiments, wherein the transmembrane domain and the extracellular domain are derived from the same protein. 121. The composition of any one of the preceding embodiments, wherein the transmembrane domain is derived from the same protein as the intracellular signaling domain. 122. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes a DAP12 recruitment domain. 123. The composition of any one of the preceding embodiments, wherein the transmembrane domain comprises a transmembrane domain that oligomerizes with DAP12. 124. The composition of any one of the preceding embodiments, wherein the transmembrane domain is at least 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. 125. The composition of any one of the preceding embodiments, wherein the transmembrane domain is at most 12, 13, 14, 15, 16, 17,18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 or 32 amino acids in length. 126. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a phosphatase inhibition domain. 127. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises an ARP2/3 inhibition domain. 128. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises at least one ITAM domain. 129. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 ITAM domains. 130. The composition of any one of the preceding embodiments, wherein the intracellular domain further comprises at least one ITAM domain. 131. The composition of any one of the preceding embodiments, wherein the intracellular domain further comprises at least one ITAM domain select from a group CD3 zeta TCR subunit, CD3 epsilon TCR subunit, CD3 gamma TCR subunit, CD3 delta TCR subunit, TCR zeta chain, Fc epsilon receptor 1 chain, Fc epsilon receptor 2 chain, Fc gamma receptor 1 chain, Fc gamma receptor 2a chain, Fc gamma receptor 2b 1 chain, Fc gamma receptor 2b2 chain, Fc gamma receptor 3a chain, Fc gamma receptor 3b chain, Fc beta receptor 1 chain, TYROBP (DAP12), CD5, CD16a, CD16b, CD22, CD23, CD32, CD64, CD79a, CD79b, CD89, CD278, CD66d, functional fragments thereof, and amino acid sequences thereof having at least one but not more than 20 modifications thereto. 132. The composition of embodiment 129, wherein the at least one ITAM domain comprises a Src- family kinase phosphorylation site. 133. The composition of embodiment 129, wherein the at least one ITAM domain comprises a Syk recruitment domain. 134. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a F-actin depolymerization activation domain. 135. The composition of any one of the preceding embodiments, wherein the intracellular domain lacks enzymatic activity. 136. The composition of any one of the preceding embodiments, wherein the intracellular domain does not comprise a domain derived from a CD3 zeta intracellular domain. 137. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a CD47 inhibition domain. 138. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain comprises a domain that activate integrin such as the intracellular region of PSGL-1. 139. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain comprises a domain that activate Rap1 GTPase, such as that from EPAC and C3G. 140. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain are from paxillin. 141. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain activates focal adhesion kinase. 142. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain is derived from a single phagocytic receptor. 143. The composition of any one of the preceding embodiments, wherein the intracellular signaling domain is derived from a single scavenger receptor. 144. The composition of any one of the preceding embodiments, wherein the intracellular domain further comprises a phagocytosis enhancing domain. 145. The composition of any one of the preceding embodiments, wherein the intracellular domain comprises a pro-inflammatory signaling domain. 146. The composition of embodiment 145, wherein the pro-inflammatory signaling domain comprises a kinase activation domain or a kinase binding domain. 147. The composition of embodiment 145 or 146, wherein the pro-inflammatory signaling domain comprises an IL-1 signaling cascade activation domain. 148. The composition of any one of embodiments 145-147, the pro-inflammatory signaling domain comprises an intracellular signaling domain derived from TLR3, TLR4, TLR7, TLR 9, TRIF, RIG-1, MYD88, MAL, IRAK1, MDA-5, an IFN-receptor, an NLRP family member, NLRP1-14, NOD1, NOD2, Pyrin, AIM2, NLRC4, FCGR3A, FCERIG, CD40, a caspase domain or a procaspase binding domain or any combination thereof. 149. The composition of any one of the preceding embodiments, wherein the PFP does not comprise a full length intracellular signaling domain. 150. The composition of any one of the preceding embodiments, wherein the intracellular domain is at least 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. 151. The composition of any one of the preceding embodiments, wherein the intracellular domain is at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 300, 400, or 500 amino acids in length. 152. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes an FcRα chain extracellular domain, an FcRα chain transmembrane domain and/or an FcRα chain intracellular domain. 153. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes an FcRβ chain extracellular domain, an FcRβ chain transmembrane domain and/or an FcRβ chain intracellular domain. 154. The composition of embodiment 152 or 153, wherein the FcRα chain or the FcRβ chain forms a complex with FcRγ when expressed in a cell. 155. The composition of embodiment 154, wherein the FcRα chain or FcRβ chain forms a complex with endogenous FcRγ when expressed in a cell. 156. The composition of any one of embodiments 152-155, wherein the FcRα chain or the FcRβ chain does not incorporate into a cell membrane of a cell that does not express FcRγ. 157. The composition of any one of embodiments 152-156, wherein the PFP does not comprise an FcRα chain intracellular signaling domain. 158. The composition of any one of embodiments 152-157, wherein the PFP does not comprise an FcRβ chain intracellular signaling domain. 159. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid encodes a TREM extracellular domain, a TREM transmembrane domain and/or a TREM intracellular domain. 160. The composition of embodiment 159, wherein the TREM is TREM1, TREM 2 or TREM 3. 161. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid comprises a sequence encoding a pro-inflammatory polypeptide. 162. The composition of any one of the preceding embodiments, wherein the composition further comprises a pro-inflammatory polypeptide. 163. The composition of embodiment 162, wherein the pro-inflammatory polypeptide is a chemokine, cytokine and nucleotides. 164. The composition of embodiment 163, wherein the chemokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, il8, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon. 165. The composition of embodiment 163, wherein the cytokine is selected from the group consisting of IL-1, IL3, IL5, IL-6, IL-12, IL-13, IL-23, TNF, CCL2, CXCL9, CXCL10, CXCL11, IL-18, IL-23, IL-27, CSF, MCSF, GMCSF, IL17, IP-10, RANTES, an interferon. 166. The composition of embodiment 163, wherein the nucleotide is selected from ATP, ADP, UTP, UDP, and/or UDP-glucose. 167. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid comprises a sequence encoding a homeostatic regulator of inflammation. 168. The composition of embodiment 167, wherein the homeostatic regulator of inflammation is a sequence in an untranslated region (UTR) of an mRNA. 169. The composition of embodiment 168, wherein the sequence in the UTR is a sequence that binds to an RNA binding protein. 170. The composition of embodiment 168 or 169, wherein translation is inhibited or prevented upon binding of the RNA binding protein to the sequence in an untranslated region (UTR). 171. The composition of embodiment 169 or 170, wherein the sequence in the UTR comprises a consensus sequence of WWWU(AUUUA)UUUW, wherein W is A or U. 172. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is expressed on a bicistronic vector. 173. The composition of any one of the preceding embodiments, wherein the target cell is a mammalian cell. 174. The composition of any one of the preceding embodiments, wherein the target cell is a human cell. 175. The composition of any one of the preceding embodiments, wherein the target cell comprises a cell infected with a pathogen. 176. The composition of any one of the preceding embodiments, wherein the target cell is a cancer cell. 177. The composition of any one of the preceding embodiments, wherein the target cell is a cancer cell that is a lymphocyte. 178. The composition of any one of the preceding embodiments, wherein the target cell is a cancer cell that is an ovarian cancer cell. 179. The composition of any one of the preceding embodiments, wherein the target cell is a cancer cell that is an ovarian pancreatic cell. 180. The composition of any one of the preceding embodiments, wherein the target cell is a cancer cell that is an glioblastoma cell. 181. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is DNA. 182. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is RNA. 183. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is mRNA. 184. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a circRNA. 185. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a tRNA. 186. The composition of any one of the preceding embodiments, wherein the recombinant nucleic acid is a microRNA. 187. A vector comprising the composition of any one of embodiments 1-186. 188. The vector of embodiment 187, wherein the vector is viral vector. 189. The vector of embodiment 188, wherein the viral vector is retroviral vector or a lentiviral vector. 190. The vector of any one of embodiments 187-189, wherein the vector further comprises a promoter operably linked to at least one nucleic acid sequence encoding one or more polypeptides. 191. The vector of any one of embodiments 187-190, wherein the vector is polycistronic. 192. The vector of embodiment 190 or 191, wherein each of the at least one nucleic acid sequence is operably linked to a separate promoter. 193. The vector of any one of embodiments 187-192, wherein the vector further comprises one or more internal ribosome entry sites (IRESs). 194. The vector of any one of embodiments 187-192, wherein the vector further comprises a 5’UTR and/or a 3’UTR flanking the at least one nucleic acid sequence encoding one or more polypeptides. 195. The vector of any one of embodiments 187-192, wherein the vector further comprises one or more regulatory regions. 196. A polypeptide encoded by the recombinant nucleic acid of the composition of any one of embodiments 1-186. 197. A cell comprising the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195 or the polypeptide of embodiment 196. 198. The cell of embodiment 197, wherein the cell is a phagocytic cell. 199. The cell of embodiment 197 or 198, wherein the cell is a stem cell derived cell, myeloid cell, macrophage, a dendritic cell, lymphocyte, mast cell, monocyte, neutrophil, microglia, or an astrocyte. 200. The cell of any one of embodiments 197-199, wherein the cell is an autologous cell. 201. The cell of any one of embodiments 197-199, wherein the cell is an allogeneic cell. 202. The cell of any one of embodiments 197-201, wherein the cell is an M1 cell. 203. The cell of any one of embodiments 197-201, wherein the cell is an M2 cell. 204. A pharmaceutical composition comprising (a) the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195, the polypeptide of embodiment 196 or the cell of any one of embodiments 197-203; and (b) a pharmaceutically acceptable excipient. 205. The pharmaceutical composition of embodiment 204, further comprising an additional therapeutic agent. 206. The pharmaceutical composition of embodiment 204 or 205, wherein the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. 207. The pharmaceutical composition of any one of embodiments 204-206, wherein the pharmaceutically acceptable excipient comprises serum free media, a lipid, or a nanoparticle. 208. A method of treating a disease in a subject in need thereof comprising administering to the subject the pharmaceutical composition of any one of embodiments 204-207. 209. The method of embodiment 208, wherein the disease is cancer. 210. The method of embodiment 209, wherein the cancer is a solid cancer. 211. The method of embodiment 210, wherein the solid cancer is selected from the group consisting of ovarian cancer, suitable cancers include ovarian cancer, renal cancer, breast cancer, prostate cancer, liver cancer, brain cancer, lymphoma, leukemia, skin cancer, pancreatic cancer, colorectal cancer, lung cancer. 212. The method of embodiment 209, wherein the cancer is a liquid cancer. 213. The method of embodiment 212, wherein the liquid cancer is leukemia or a lymphoma. 214. The method of embodiment 212, wherein the liquid cancer is a T cell lymphoma. 215. The method of embodiment 208, wherein the disease is a T cell malignancy. 216. The method of any one of embodiments 208-215, wherein the method further comprises administering an additional therapeutic agent to the subject. 217. The method of embodiment 216, wherein the additional therapeutic agent is selected from the group consisting of a CD47 agonist, an agent that inhibits Rac, an agent that inhibits Cdc42, an agent that inhibits a GTPase, an agent that promotes F-actin disassembly, an agent that promotes PI3K recruitment to the PFP, an agent that promotes PI3K activity, an agent that promotes production of phosphatidylinositol 3,4,5-trisphosphate, an agent that promotes ARHGAP12 activity, an agent that promotes ARHGAP25 activity, an agent that promotes SH3BP1 activity and any combination thereof. 218. The method of any one of embodiments 208-217, wherein administering comprises infusing or injecting. 219. The method of any one of embodiments 208-218, wherein administering comprises administering directly to the solid cancer. 220. The method of any one of embodiments 208-219, wherein administering comprises a circRNA, mRNA, viral-, particle-, liposome-, or exosome-based delivery procedure. 221. The method of any one of embodiments 208-220, wherein a CD4+ T cell response or a CD8+ T cell response is elicited in the subject. 222. A method of preparing a cell, the method comprising contacting a cell with the composition of any one of embodiments 1-186, the vector of any one of embodiments 187-195 or the polypeptide of embodiment 196. 223. The method of embodiment 222, wherein contacting comprises transducing. 224. The method of embodiment 223, where transducing comprises chemical transfection, electroporation, nucleofection, or viral infection. 225. A method of preparing a pharmaceutical composition comprising contacting a lipid to the composition of any one of embodiments 1-186 or the vector of any one of embodiments 187-195. 226. The method of embodiment 225, where contacting comprises forming a lipid nanoparticle. 227. A method of preparing a pharmaceutical composition comprising contacting an antibody to the composition of any one of embodiments 1-186 or the vector of any one of embodiments 187-195. 228. The method of embodiment 225, where contacting comprises forming a lipid nanoparticle. EXAMPLES Example 1. Generation of novel chimeric receptors fusion proteins (CFP) constructs [0811] In this section, an exemplary design for identification of useful CFP ECD, TM, ICD and antigen binding domains for the generation of novel CFPs is described. Briefly, a large number of potential candidate proteins are screened for enhanced phagocytic properties and their respective phagocytosis related intracellular signaling. The useful domains are then used for generation of novel CFPs. The screen can be divided in two parts: A. Screening for the PR domains; B. Screening for the extracellular antigen binding domains. Screening for the PR domains: [0812] 5,800 plasma membrane proteins are screened for their phagocytic potential. J774 macrophage cells are transiently transfected with the library of 5800 plasma proteins. High- throughput multiplex assays (ranging from 6-well plate assay set up to up to 384-well plate assay with robotic handling) are set up to evaluate various potential functions of the plasma membranes. Exemplary assays include, but are not limited to phagocytosis assay, cytokine production assay, inflammasome activation assay, and iNOS activation assay. Exemplary simplified methods are described in the following paragraphs. Variations of each method are also used and are understood by a skilled artisan. Variations of each method are also used and are understood by a skilled artisan. Exemplary intracellular signaling domains tested for include but are not limited to CD40-FcRγ; FcRγ-CD40; NLRP3; FcRγ-SH2-Procaspase; FcRγ-Myd88; FcRγ-IFN receptor; FcR-TNFR1; FcRγ- TNFR2; FcR-AIM2; FcRγ-TRIFN; FcRγ-Procaspase; TRIFC; RIG1; MDA5; TBK; CD64; CD16A; CD89; FcRε; SIRPβ; (two consecutive intracellular domains are represented as hyphenated terms, for example, FcRγ-Myd88 refers to an intracellular domain comprising an FcRγ intracellular signaling domain as signaling domain 1; and an Myd88 intracellular signaling domain as signaling domain 2). The extracellular linker domains screened include but are not limited to CD64, CD16A, CD89, SIRPα, FcRε, CD8 hinge. The transmembrane domains tested include but are not limited to CD8, CD64, CD16A, CD89, FcRε, SIRPα, TNFR1 and CD40. MDA5 domains were also screened. Phagocytosis assay: [0813] Antigen-linked silica or polysterene beads ranging in diameters 1 nm, 5 nm or 10 nm were used for a screen of macrophages. Inert beads are coated in a supported lipid bilayer and the antigens are ligated to the lipid bilayer. J774 macrophage cell lines are prepared, each cell line expressing a cloned recombinant plasma membrane protein. The recombinant plasma membrane protein may also express a fluorescent tag. The cell lines are maintained and propagated in complete RPMI media with heat inactivated serum and antibiotics (Penicillin/Streptomycin). On the day of the assay, cells are plated at a density of 1x10^6 cells/ml per well in 6 well plates or in a relative proportion in 12 or 24 well plates, and incubated for 2-6 hours. The cells are then washed once in Phosphate Buffer Saline, and the beads are added in serum depleted or complement depleted nutrient media. Cells are visualized by light microscopy at 30 minutes and 2 hours after addition of the beads. Immunofluorescence reaction may be performed using tagged antibody, and fluorescent confocal microscopy is used to detect the interaction and co-localization of cellular proteins at engulfment. Confidence levels are determined by Kruskal-Wallis test with Dunn’s multiple comparison correction. [0814] In some examples, dye loaded tumor cells are fed to macrophage cell lines and phagocytosis is assessed by microscopy. Cytokine production: [0815] Macrophage cell lines are cultured as above. In one assay, each J774 cell line expressing a plasma membrane protein is plated in multi-wells and challenged with antigen-linked beads and cytokine production was assayed by collecting the supernatants at 4 hours and 24 hours. Cytokines are assayed from the supernatant by ELISA. In another fraction, cells are collected at 4 and 24 hours after incubation with the beads and flow cytometry is performed for detection of cytokines. In each case, multiple cytokines are assayed in a multiplex format, which can be selected from: IL-1α, IL-1β, IL-6, IL-12, IL-23, TNF-α, GMCSF, CXCL1, CXCL3, CXCL9, CXCL-10, MIP1-α and MIP-2. Macrophage inflammatory cytokine array kit (R&D Systems) is used. [0816] Intracellular signaling pathway for inflammatory gene and cytokine activation can be identified by western blot analysis for phosphorylation of MAP kinases, JNK, Akt signaling pathway, Interferon activation pathway including phosphorylation and activation of STAT-1. Inflammasome activation assay: [0817] Activation of NLRP3 inflammasome is assayed by ELISA detection of increased IL-1 production and detection caspase-1 activation by western blot, detecting cleavage of procaspase to generate the shorter caspase. In a microwell plate multiplex setting, Caspase-Glo (Promega Corporation) is used for faster readout of Caspase 1 activation. iNOS activation assay: [0818] Activation of the oxidative burst potential is measured by iNOS activation and NO production using a fluorimetric assay NOS activity assay kit (AbCAM). Cancer cell killing assay: [0819] Raji B cells are used as cancer antigen presenting cells. Raji cells are incubated with whole cell crude extract of cancer cells, and co-incubated with J774 macrophage cell lines. The macrophages can destroy the cells after 1 hour of infection, which can be detected by microscopy or detected by cell death assay. Screening for high affinity antigen binding domains: [0820] Cancer ligands are subjected to screening for antibody light chain and heavy chain variable domains to generate extracellular binding domains for the CFPs. Human full length antibodies or scFv libraries are screened. Also potential ligands are used for immunizing llama for development of novel immunoglobulin binding domains in llama, and preparation of single domain antibodies. [0821] Specific useful domains identified from the screens are then reverse transcribed, and cloned into lentiviral expression vectors to generate the CFP constructs. A recombinant nucleic acid encoding a CFP can generated using one or more domains from the extracellular, TM and cytoplasmic regions of the highly phagocytic receptors generated from the screen. Briefly plasma membrane receptors showing high activators of pro-inflammatory cytokine production and inflammasome activation are identified. Bioinformatics studies are performed to identify functional domains including extracellular activation domains, transmembrane domains and intracellular signaling domains, for example, specific kinase activation sites, SH2 recruitment sites. These screened functional domains are then cloned in modular constructions for generating novel CFPs. These are candidate CFPs, and each of these chimeric construct is tested for phagocytic enhancement, production of cytokines and chemokines, and/or tumor cell killing in vitro and/or in vivo. A microparticle based phagocytosis assay was used to examine changes in phagocytosis. Briefly, streptavidin coupled fluorescent polystyrene microparticles (6 µm diameter) are conjugated with biotinylated recombinantly expressed and purified cancer ligand. Myeloid cells expressing the novel CFP were incubated with the ligand coated microparticles for 1-4 h and the amount of phagocytosis was analyzed and quantified using flow cytometry. Plasmid or lentiviral constructions of the designer CFPs are then prepared and tested in macrophage cells for cancer cell lysis. [0822] Exemplary functional domain containing CFPs are described in the following sections. Example 2. Manufacturing Protocol for Myeloid and Macrophage Cell Preparation from a subject Myeloid/Macrophage cell isolation from PBMCs: [0823] Peripheral blood mononuclear cells are separated from normal donor buffy coats by density centrifugation using Histopaque 1077 (Sigma). After washing, CD14+ monocytes are isolated from the mononuclear cell fraction using CliniMACS GMP grade CD14 microbeads and LS separation magnetic columns (Miltenyi Biotec). Briefly, cells are resuspended to appropriate concentration in PEA buffer (phosphate-buffered saline [PBS] plus 2.5 mmol/L ethylenediaminetetraacetic acid [EDTA] and human serum albumin [0.5% final volume of Alburex 20%, Octopharma]), incubated with CliniMACS CD14 beads per manufacturer’s instructions, then washed and passed through a magnetized LS column. After washing, the purified monocytes are eluted from the demagnetized column, washed and re-suspended in relevant medium for culture. Isolation of CD14+ cells from leukapheresis: PBMCs are collected by leukapheresis from cirrhotic donors who gave informed consent to participate in the study. Leukapheresis of peripheral blood for mononuclear cells (MNCs) is carried out using an Optia apheresis system by sterile collection. A standard collection program for MNC is used, processing 2.5 blood volumes. Isolation of CD14 cells is carried out using a GMP-compliant functionally closed system (CliniMACS Prodigy system, Miltenyi Biotec). Briefly, the leukapheresis product is sampled for cell count and an aliquot taken for pre-separation flow cytometry. The percentage of monocytes (CD14+) and absolute cell number are determined, and, if required, the volume is adjusted to meet the required criteria for selection (≤ 20 x 109 total white blood cells; < 400 x 106 white blood cells/mL; ≤ 3.5 x 109 CD14 cells, volume 50–300 mL). CD14 cell isolation and separation is carried out using the CliniMACS Prodigy with CliniMACS CD14 microbeads (medical device class III), TS510 tubing set and LP-14 program. At the end of the process, the selected CD14+ positive monocytes are washed in PBS/EDTA buffer (CliniMACS buffer, Miltenyi) containing pharmaceutical grade 0.5% human albumin (Alburex), then re-suspended in TexMACS (or comparator) medium for culture. Cell count and purity: [0824] Cell counts of total MNCs and isolated monocyte fractions are performed using a Sysmex XP-300 automated analyzer (Sysmex). Assessment of macrophage numbers is carried out by flow cytometry with TruCount tubes (Becton Dickinson) to determine absolute cell number, as the Sysmex consistently underestimated the number of monocytes. The purity of the separation is assessed using flow cytometry (FACSCanto II, BD Biosciences) with a panel of antibodies against human leukocytes (CD45-VioBlue, CD15-FITC, CD14-PE, CD16-APC), and product quality is assessed by determining the amount of neutrophil contamination (CD45int, CD15pos). Cell culture—development of cultures with healthy donor samples [0825] Optimal culture medium for macrophage differentiation is investigated, and three candidates are tested using for the cell product. In addition, the effect of monocyte cryopreservation on deriving myeloid cells and macrophages for therapeutic use is examined. Functional assays are conducted to quantify the phagocytic capacity of myeloid cells and macrophages and their capacity for further polarization, and phagocytic potential as described elsewhere in the disclosure. Full-scale process validation with subject samples [0826] Monocytes cultured from leukapheresis from Prodigy isolation are cultured at 2 x 106 monocytes per cm2 and per mL in culture bags (MACS GMP differentiation bags, Miltenyi) with GMP-grade TexMACS (Miltenyi) and 100 ng/mL M-CSF. Monocytes are cultured with 100 ng/mL GMP-compliant recombinant human M-CSF (R&D Systems). Cells are cultured in a humidified atmosphere at 37°C, with 5% CO2 for 7 days. A 50% volume media replenishment is carried out twice during culture (days 2 and 4) with 50% of the culture medium removed, then fed with fresh medium supplemented with 200 ng/mL M-CSF (to restore a final concentration of 100 ng/mL). Cell harvesting: [0827] For normal donor-derived macrophages, cells are removed from the wells at day 7 using Cell Dissociation Buffer (Gibco, Thermo Fisher) and a pastette. Cells are resuspended in PEA buffer and counted, then approximately 1x106 cells per test are stained for flow cytometry. Leukapheresis- derived macrophages are removed from the culture bags at day 7 using PBS/EDTA buffer (CliniMACS buffer, Miltenyi) containing pharmaceutical grade 0.5% human albumin from serum (HAS; Alburex). Harvested cells are resuspended in excipient composed of two licensed products: 0.9% saline for infusion (Baxter) with 0.5% human albumin (Alburex). Flow cytometry characterization: [0828] Monocyte and macrophage cell surface marker expression is analyzed using either a FACSCanto II (BD Biosciences) or MACSQuant 10 (Miltenyi) flow cytometer. Approximately 20,000 events are acquired for each sample. Cell surface expression of leukocyte markers in freshly isolated and day 7 matured cells is carried out by incubating cells with specific antibodies (final dilution 1:100). Cells are incubated for 5 min with FcR block (Miltenyi) then incubated at 4°C for 20 min with antibody cocktails. Cells are washed in PEA, and dead cell exclusion dye DRAQ7 (BioLegend) is added at 1:100. Cells are stained for a range of surface markers as follows: CD45- VioBlue, CD14-PE or CD14-PerCP-Vio700, CD163-FITC, CD169-PE and CD16-APC (all Miltenyi), CCR2-BV421, CD206-FITC, CXCR4-PE and CD115-APC (all BioLegend), and 25F9- APC and CD115-APC (eBioscience). Both monocytes and macrophages are gated to exclude debris, doublets and dead cells using forward and side scatter and DRAQ7 dead cell discriminator (BioLegend) and analyzed using FlowJo software (Tree Star). From the initial detailed phenotyping, a panel is developed as Release Criteria (CD45-VB/CD206-FITC/CD14-PE/25F9 APC/DRAQ7) that defined the development of a functional macrophage from monocytes. Macrophages are determined as having mean fluorescence intensity (MFI) five times higher than the level on day 0 monocytes for both 25F9 and CD206. A second panel is developed which assessed other markers as part of an Extended Panel, composed of CCR2-BV421/CD163-FITC/CD169-PE/CD14-PerCP- Vio700/CD16-APC/DRAQ7), but is not used as part of the Release Criteria for the cell product. [0829] Both monocytes and macrophages from buffy coat CD14 cells are tested for phagocytic uptake using pHRodo beads, which fluoresce only when taken into acidic endosomes. Briefly, monocytes or macrophages are cultured with 1–2 uL of pHRodo Escherichia coli bioparticles (LifeTechnologies, Thermo Fisher) for 1 h, then the medium is taken off and cells washed to remove non-phagocytosed particles. Phagocytosis is assessed using an EVOS microscope (Thermo Fisher), images captured and cellular uptake of beads quantified using ImageJ software (NIH freeware). The capacity to polarize toward defined differentiated macrophages is examined by treating day 7 macrophages with IFNγ (50 ng/mL) or IL-4 (20 ng/mL) for 48 h to induce polarization to M1 or M2 phenotype (or M[IFNγ] versus M[IL-4], respectively). After 48 h, the cells are visualized by EVOS bright-field microscopy, then harvested and phenotyped as before. Further analysis is performed on the cytokine and growth factor secretion profile of macrophages after generation and in response to inflammatory stimuli. Macrophages are generated from healthy donor buffy coats as before, and either left untreated or stimulated with TNFα (50 ng/mL, Peprotech) and polyinosinic:polycytidylic acid (poly I:C, a viral homolog which binds TLR3, 1 g/mL, Sigma) to mimic the conditions present in the inflamed liver, or lipopolysaccharide (LPS, 100 ng/mL, Sigma) plus IFNγ (50 IU/mL, Peprotech) to produce a maximal macrophage activation. Day 7 macrophages are incubated overnight and supernatants collected and spun down to remove debris, then stored at −80°C until testing. Secretome analysis is performed using a 27-plex human cytokine kit and a 9-plex matrix metalloprotease kit run on a Magpix multiplex enzyme linked immunoassay plate reader (BioRad). Product stability: [0830] Various excipients are tested during process development including PBS/EDTA buffer; PBS/EDTA buffer with 0.5% HAS (Alburex), 0.9% saline alone or saline with 0.5% HAS. The 0.9% saline (Baxter) with 0.5% HAS excipient is found to maintain optimal cell viability and phenotype (data not shown). The stability of the macrophages from cirrhotic donors after harvest is investigated in three process optimization runs, and a more limited range of time points assessed in the process validation runs (n = 3). After harvest and re-suspension in excipient (0.9% saline for infusion, 0.5% human serum albumin), the bags are stored at ambient temperature (21–22°C) and samples taken at 0, 2, 4, 6, 8, 12, 24, 30 and 48 h postharvest. The release criteria antibody panel is run on each sample, and viability and mean fold change from day 0 is measured from geometric MFI of 25F9 and CD206. Statistical analysis: [0831] Results are expressed as mean ± SD. The statistical significance of differences is assessed where possible with the unpaired two-tailed t-test using GraphPad Prism 6. Results are considered statistically significant when the P value is < 0.05. Example 3. CD5-FcR-PI3K CFP construct [0832] In this example, a CD5-targeted CFP was constructed using known molecular biology techniques. The CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8α chain hinge and CD8α chain TM domain via a short linker. The TM domain is fused at the cytosolic end with an FcRγ cytosolic portion, and a PI3K recruitment domain. The construct was prepared in a vector having a fluorescent marker and a drug (ampicillin) resistance and amplified by transfecting a bacterial host. The sequence is provided below: CD5-FcR-PI3K MWLQSLLLLGTVACSISEIQLVQSGGGLVKPGGSVRISCAASGYTFTNYGMNWVRQAPGK GLEWMGWINTHTGEPTYADSFKGRFTFSLDDSKNTAYLQINSLRAEDTAVYFCTRRGYDW YFDVWGQGTTVTVSSGGGGSGGGGSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQDINSY LSWFQQKPGKAPKTLIYRANRLESGVPSRFSGSGSGTDYTLTISSLQYEDFGIYYCQQYDESP WTFGGGTKLEIKSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRP EACRPAAGGAVHTRGLDIYIWAPLAGTCGVLLLSLVITLYCRRLKIQVRKAAITSYEKSDGV YTGLSTRNQETYETLKHEKPPQGSGSYEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM (SEQ ID NO: 14). [0833] mRNA was generated by in vitro reverse transcription of the purified plasmids using suitable primers. The purified mRNA was transduced into a cell line for expression analysis. Example 4. HER2-FcR-PI3K CFP construct [0834] In this example, a HER2-targeted CFP was constructed using known molecular biology techniques. The CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to HER2 on a target cell, attached to a CD8α chain hinge and CD8α chain TM domain via a short linker. The TM domain is fused at the cytosolic end with an FcRγ cytosolic portion, and a PI3K recruitment domain as in the previous example. The sequence is provided below: HER2-FcR-PI3K MWLQSLLLLGTVACSISDIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAP KLLIYSASFLYSGVPSRFSGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKVEIKR TGSTSGSGKPGSGEGSEVQLVESGGGLVQPGGSLRLSCAASGFNIKDTYIHWVRQAPGKGL EWVARIYPTNGYTRYADSVKGRFTISADTSKNTAYLQMNSLRAEDTAVYYCSRWGGDGFY AMDVWGQGTLVTVSSSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPL SLRPEACRPAAGGAVHTRGLDIYIWAPLAGTCGVLLLSLVITLYCRRLKIQVRKAAITSYEKS DGVYTGLSTRNQETYETLKHEKPPQGSGSYEDMRGILYAAPQLRSIRGQPGPNHEEDADSYE NM (SEQ ID NO: 15). Example 5. CD5-FcR-CD40 CFP construct [0835] In this example, a CD5-targeted CFP was constructed using known molecular biology techniques having an intracellular domain comprising CD40 sequence. The CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8α chain hinge and CD8α chain TM domain via a short linker. The TM domain is fused at the cytosolic end with an FcRγ cytosolic portion, followed by a CD40 cytosolic portion. The sequence is provided below: CD5-FcR-CD40 MWLQSLLLLGTVACSISEIQLVQSGGGLVKPGGSVRISCAASGYTFTNYGMNWVRQAPGK GLEWMGWINTHTGEPTYADSFKGRFTFSLDDSKNTAYLQINSLRAEDTAVYFCTRRGYDW YFDVWGQGTTVTVSSGGGGSGGGGSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQDINSY LSWFQQKPGKAPKTLIYRANRLESGVPSRFSGSGSGTDYTLTISSLQYEDFGIYYCQQYDESP WTFGGGTKLEIKSGGGGSGALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRP EACRPAAGGAVHTRGLDIYIWAPLAGTCGVLLLSLVITLYCRLKIQVRKAAITSYEKSDGVY TGLSTRNQETYETLKHEKPPQKKVAKKPTNKAPHPKQEPQEINFPDDLPGSNTAAPVQETLH GCQPVTQEDGKESRISVQERQ (SEQ ID NO: 16). [0836] Table 6 describes the domains of the constructs generated for testing. Table 6 – Exemplary CD5-CFP constructs and HER2 CFP constructs
Figure imgf000189_0001
Figure imgf000190_0001
Example 6. CD5-FcR-MDA5 CFP construct [0837] In this example, a CD5-targeted CFP was constructed using known molecular biology techniques having an intracellular domain comprising two caspase activation (CARD) domains of MDA-5 intracellular domain sequence (Tandem CARD ICD sequence as in SEQ ID NO: 23). The CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8α chain hinge and CD8α chain TM domain via a short linker. The TM domain is fused at the cytosolic end with an FcRγ cytosolic portion, followed by a MDA5 cytosolic portion (SEQ ID NO: 23). [0838]
Figure imgf000191_0003
Q Q Q
Figure imgf000191_0002
(SEQ ID NO: 24). Example 7. CD5-FcR-TNFR1 or TNFR2 CFP construct [0839] In this example, a CD5-targeted CFP was constructed using known molecular biology techniques having a TNFR1 or TNFR2 intracellular domain. As shown graphically in FIG. 18A, the CFP has an extracellular domain comprising a signal peptide fused to an scFv containing a heavy chain variable domain linked to a light chain variable domain that binds to CD5 on a target cell, attached to a CD8α chain hinge and CD8α chain TM domain via a short linker. The TM domain is fused at the cytosolic end with an FcRγ cytosolic portion, followed by an intracellular signaling domain (ICD) sequence TNFR1 (SEQ ID NO: 21) or an ICD of TNFR2 (SEQ ID NO: 22). The full length sequence of the CFP having a TNFR1 intracellular signaling domain is depicted below:
Figure imgf000191_0001
(SEQ ID NO: 25) . [0840] The full length sequence of the CFP having a TNFR2 intracellular signaling domain is depicted below:
Figure imgf000192_0001
Example 8. Bicistronic constructs for in vivo delivery [0841] In this example, design and expression of bicistronic construct is demonstrated in which a single mRNA comprises sequence encoding a CAR and a CFP for expression in a T cell and a myeloid cell respectively, each of which can comprise an extracellular binding domain that binds to the same (or two different) cancer cell surface antigen. The design is directed towards in vivo delivery and cell specific expression of either the CAR or the CFP, depending on whether the cell is a T cell lineage cell or a myeloid lineage cell. The single mRNA can be delivered via an LNP. Design and structural elements of a bicistronic CFP-CAR dual construct: The molecular design of a bicistronic CFP-CAR dual construct comprises (a) a sequence encoding a CAR-T and a sequence encoding a CFP, separated by an autocleavage sequence, such as a T2A, or a P2A sequence. A CAR-T sequence comprises a CD3 epsilon transmembrane region and cytoplasmic region that facilitate TCR complex assembly and is expressed in a T cell. This design promotes a T cell-specific expression of a CAR with a binder (the extracellular antigen binding domain) that specifically binds to a desired antigen, e.g., CD19 cell surface antigen on a cancer cell. Such CD19 binder CAR multimerizes with members of the TCR endogenously expressed by T cells. The design not only facilitates T cell specific expression, but also the CAR activates a T cell upon binding of the extracellular antigen binding domain to the antigen that it specifically binds to. Therefore a CAR, that is functional specifically in a T cell is designed. A CFP sequence, as designed here for specific expression on a myeloid cell comprises a transmembrane and intracellular domain of Fc-alpha R, which assembles in a myeloid cell with endogenously expressed Fc-gamma receptors and expresses on a myeloid cell surface. The CFP comprises an extracellular binding domain specific for a cancer cell antigen, and can be a binder that binds the antigen that the CAR binds. Each of the binders can be an scFv or can be any other form of binder as described elsewhere in the specification. The design can include a CAR with a binder that binds to a different antigen in a CFP but is directed to the same tumor tissue. By design a CFP as described here will exhibit myeloid cell specific expression and function as the Fc-alpha R domain promotes expression and function of the CFP in a cell that naturally expresses Fcgamma receptor, and myeloid cells specifically express Fcgamma receptor. [0842] Sequences: Sequence for `MYL222
Figure imgf000193_0001
QQ Q Sequence for MYL223 [0843]
Figure imgf000193_0003
Figure imgf000193_0002
[0844] Example 9.5’-Cap structures for stability and efficient translation. [0845] In this prophetic example, several Cap structures are tested for efficiency in mRNA translation. In general, mRNA constructs comprising a sequence encoding a CFP as described herein is transcribed in vitro and 5’capping is performed using a different Cap analog for each test set. The IVT mRNAs are purified by HPLC and electroporated into a myeloid or suitable cell line. The Cap analogs are listed in Table 7. [0846] Table 7. Cap analogues. G, nucleotide base Guanine; C, nucleotide base Cytosine; A, nucleotide base Adenine; U, nucleotide base Uracil; m, methylation; p-phosphate.
Figure imgf000194_0001
[0847] From this study a projected result is that m7Gpppm6AmpG analog shows the highest protein expression in the test cell line. This test is repeated for different constructs of varying mRNA length and starting nucleotide – thereby generating a better understanding and confidence in the use of the Cap analog. [0848] In one set of experiments, the Cap analogs are designed for suitable expression of the construct comprise an anti-CD5 scFv, a CD8 hinge, a CD8 transmembrane domain, an FcR intracellular domain and an intracellular PI3kinase recruitment domain (anti-CD5-CD8hinge- CD8TM-FcR-PI3K) having a translated amino acid sequence (SEQ ID NO: 50):
Figure imgf000194_0002
[0849] In another set of experiments, the Cap analogs are designed for suitable expression of the construct comprise an anti-HER2 scFv, a CD8 hinge, a CD8 transmembrane domain, a CD40 intracellular domain, an FcR intracellular domain and an intracellular PI3kinase recruitment domain (anti-HER2-CD8hinge-CD8TM-CD40-FcR-PI3K) having a translated amino acid sequence (SEQ ID NO: 51): [0850]
Figure imgf000195_0002
Q Q Q Q QQ
Figure imgf000195_0001
[0851] Example 10. Functional evaluation of 5’-Cap structures: expression [0852] In this experiment, three sets of Cap analogues were tested for expression of the mRNA constructs. The structures of the three sets of the Cap analogues are graphically represented in FIG. 4, the ARCA, the CC1 Cap and the EC1 Cap. EC1 comprises a novel modification at N6 adenosine position. The full length mRNA constructs encoding anti-HER2 CARs and comprising the respective 5’-Caps were electroporated (EP) into primary monocytes derived from human donors. FIG. 5 illustrates graphically a simplified generalized work plan for evaluating the functional aspects. In general, 0.8 pmol RNA/1x10^6 cells (100ug/ml) was used for electroporation of the cells. The mRNA sets were: Uncapped AG-anti-HER2-CAR-polyA; CC1-Capped-anti-HER2-CAR- polyA; m6A-Capped-anti-HER2-CAR-polyA; EC1-Capped-anti-HER2-CAR-polyA RNA was prepared by in vitro transcription (IVT) in IVT buffer at pH 6.5, as described elsewhere in the examples. Transfection using lipofectamine was also used in one exemplary set. FIGs. 6 and 7 demonstrate representative results of these experiments. As shown in FIG. 6, at 24h, the expression of the encoded protein (anti- HER2-CD8h-CD8TM-FcR-PI3K) from constructs having either CC1 or EC1 caps (both are Cap 1 analog structures) were significantly higher than the constructs having ARCA caps (which is a Cap 0 analog). No difference was observed between the CC1 and EC1 capped mRNA expressions. The ARCA mRNA had about 3-fold lower expression than either of the CC1 and EC1 capped mRNAs. The expression was measured by binding to HER2, indicated by flow cytometric analysis. FIG.7 shows the duration of the CAR expression, measured at 24 and 48h after electroporation. The data demonstrates the high expression levels that peak at 24h with the CC1 and EC1 5’ capped mRNA compared to the ARCA capped mRNA. FIGs. 14A and its continuing figure FIG. 14B show flow cytometry sorting of electroporated cells from a single donor (Donor 23B), using 0.1 microM RNA. FIG. 15 shows HER2 binder (CAR) expression of monocytes for the same subject for the experiment at 4h, 24h and 48h respectively. FIG. 16 shows a summary graph plot, indicating the increase and the duration of expression of the anti-HER2 CAR in these cells over the indicated time. Similar results were observed in case of cells from donor PD-004-04, as shown in FIGs. 17-19, when cells from this donor were electroporated (EP). FIGs. 20-22 demonstrate data showing results from using cells from the donor PD-004-04 (same donor’s cells used in results shown in FIGs 17-19), where the cells were transfected with lipofectamine instead of electroporation. Additional data from cells from Donor MYE-101-0024A (Donor 24A) electroporated with 0.1 microM RNA – Mock RNA, uncapped AG-anti-HER2 CAR-polyA; CC1- cap-anti-HER2 CAR-polyA; EC1-cap-anti-HER2 CAR-polyA or ARCA-cap-anti-HER2 CAR- polyA are shown in FIGs. 23-25. The table below provides a summary of results. The anti-HER2 CAR comprises an antigen binding domain having an scFV, having a heavy chain variable domain sequence of
Figure imgf000196_0002
and a light chain variable domain having a sequence of:
Figure imgf000196_0003
The sequence additionally has a CD8 hinge domain having an amino acid sequence of ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLD; a CD8 TM domain having a sequence of: IYIWAPLAGTCGVLLLSLVITLYC; an FcR intracellular domain having a sequence of:
Figure imgf000196_0004
and intracellular PI3 K recruitment domain having a sequence of: YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM. [0853] Table 8. A summary of data from a representative example from three monocyte donor’s cells is presented in the Table below.
Figure imgf000196_0001
Figure imgf000197_0001
[0854] Example 11. Functional evaluation of 5’-Cap structures: immune activation by capped RNA [0855] An investigation of activation of the RNA recognition pathway after lipofection of mRNA constructs encoding a CAR but each construct differing in the Cap analogue present at the 5’-UTR was also undertaken. The protocol workflow is outlined in FIG. 8. Following transfection (e.g., lipofection) of the respective mRNA constructs with the different Cap analogues the Type 1 interferons, Interferon alpha and beta (IFN-a and IFN-b) were measured at 4 and 20h. mRNA constructs, with or without HPLC purification were used for the study. Monocytes from a single human donor were used per assay. 0.6 x 10^6 cells were plated in 24 well plate in 0.5ml recovery media/0.5 ug RNA/ul transfection reagent (TransIT reagent) and 1 ul Boost reagent in 50 ul final Opti-Mem. Cells were also collected for assessing IFN recognition pathways via qPCR. In the representative experiments shown, the mRNA encoded by the constructs was a TROP2 binder CAR, having anti-TROP2 antigen binding domain, a CD89 TM domain, with short CD89 extracellular and intracellular regions. FIG. 9A shows that IFN-a and IFN-b were induced upon transfection with an in house ARCA construct and commercial ARCA construct, both at 4h and 20h. ARCA in house construct was HPLC purified. With HPLC purification, in-house ARCA RNA was comparable to the commercial ARCA. However, in-house generated and HPLC purified CC1 and EC1 caps did not result in IFN response. As is well understood by one skilled in the art that the activation of interferons can lead to an inflammatory sequel which is harmful. Additionally, presence of double stranded RNA in the RNA preparation, even in small amounts, is likely to activate the antiviral innate immune response, marked by interferon type I response. IFN alpha and beta are exemplary type I interferons. Release of these interferons in turn activate a number of inflammatory cytokines and chemokines. FIG. 9B shows induction of CXCL10 and IFITM, which followed similar pattern as the interferons. These chemokines are interferon responsive and potentiate the inflammatory response. These results indicate that with HPLC purification, in-house ARCA RNA is comparable to the commercial ARCA. With HPLC purification, in-house CC1 and EC1 Caps induced significantly lower IFN Type 1 activation than the commercial ARCA. These results indicate that the Cap0 structure is recognized by the innate immune system which is activated. In contrast CC1 and EC1 caps do not activate the immune system, thereby making them a safer option for in vivo mRNA therapeutic development. FIGs 26-28 demonstrate the flow cytometry data of an experiment with monocyte cells from donor PD-004-04, and comparing the different mRNA preparations with the caps with or without HPLC purification, as shown in the figures. [0856] Example 12. Large scale ARCA- versus CC1-capped GP75-FcR-PI3K RNA production for comparison studies in vivo [0857] In this example, ARCA and CC1 cap structures were evaluated for their efficacy in an in vivo tumor model. Both ARCA capped and CC1-capped RNA were successfully generated in large scale for the studies. [0858] An exemplary GP75-FcR-PI3K mRNA sequence including 5’-UTR (underlined) and 3’- UTR (underlined) is shown below:
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000199_0002
[0859] Pre-capped in vitro transcribed mRNA and CC1-capped mRNA before and after HPLC purification were resolved in gel electrophoresis, a representative is shown in FIG. 10, left image. A dot blot analysis was performed for detecting dsRNA content. A representative dot blot analysis for the dsRNA impurities is shown in FIG.10, right. [0860] Example 13. Comparison of effectiveness of anti-gp75 myeloid cell CAR mRNA with ARCA and CC1 Cap in in vivo tumor model [0861] In this example, a comparison of the activities of two mRNAs encoding gp75-targeting CAR for myeloid cells, one with ARCA 5’-Cap and one with CC1-5’Cap in a mouse murine model was evaluated. In the B16-F10 OVA syngeneic tumor model, tumor cells were inoculated in the mice and allowed to grow to a certain size, and the treatment was initiated with intravenous (i.v.) administration of anti-gp75-CAR monocytes (2 x 106), generated from the bone marrow of C57BL/6 mice from day 10 at intervals for 6 doses, spread across 8 days till day 18. Anti-gp75 CAR expressing murine monocytes were prepared as follows: Fresh murine monocytes were isolated at different days corresponding to different doses and mRNA (gp75-FLAG-FcR-PI3K) capped with ARCA or CC1 cap was nucleofected. Before the anti-gp-75-CAR monocytes (2 x 10^6) were administrate through i.v., the cells were assessed for viability (FIG. 11, upper panel, based on the 7- AAD staining); and anti-gp75 expression was verified as shown in lower panel, anti-FLAG was used for the detection of the CAR. [0862] Upper panel shows comparable cell viability after nucleofection of anti-gp75-CAR, which indicates that the same cell number take up the mRNA at different doses. FIG. 11 lower panel shows consistent high CAR expression across all injection. In each case, the CC1-capped mRNA had a higher expression compared to ARCA-capped mRNA. However, measurement of tumor volume over time shows interesting differences. Mice injected with CC1-capped anti-gp75 CAR mRNA showed the least increase in tumor volume, and the tumor volume was about half the size of the tumor size of untreated mice. ARCA-capped mRNA encoding the same anti-gp75 CAR led to a lesser suppression of tumor, compared to the CC1-Capped anti-gp75 CAR. This was in keeping with the previously described data in FIGs. 5-7, showing efficiency of the CCI-Capped mRNA in expression of the mRNA, and also FIGs. 9A and 9B showing reduced immune activation. The results indicate that the beneficial effect of using CC1-Cap for the mRNA is also evident in vivo, and that CC1 is a superior design over the ARCA. This indicates great promise for use of CC1-capped mRNA in mRNA constructs for therapeutic development. [0863] Example 14. Capping Efficiency [0864] In this example, capping efficiency is determined and compared across CC1, EC1 and m6A Cap structures. An improved IVT buffer was used, composed of 40mM Bis-Tris pH 6.5, 25 mM MgCl2, 10mM DTT and 2 mM spermidine. FIG. 13 shows gel electrophoresis data showing post capping samples. The indicated ratios on the figure represents a ratio of Cap: ATP if an AG initiator; or the ratio of Cap: GTP if an GG initiator. EC1- cap was incorporated with 100% efficiency, when the IVT buffer (pH= 6.5) and 2.5 fold excess of the cap was used. CC1 and m6A showed a higher capping efficiency compared to EC1 for Cap:ATP ratio: 1:1.25. A summary of the capping efficiencies of ARCA and CC1 and EC1 capping is shown in the table below. From the data it is understood that 2.5-fold more EC1 cap is needed to obtain 100% capping efficiency compared to CC1. [0865] Table 9. A comparison of benefits of CC1/EC1 vs ARCA.
Figure imgf000200_0001
[0866] Thus, from the above examples, it is shown that compared to ARCA, CC1 and EC1 mRNA express 3-fold higher. No difference was observed in the expression between CC1 and EC1 mRNAs. EC1 mRNA has the lowest dsRNA content. ARCA-capped RNA produced in-house requires HPLC purification to remove double stranded DNA (dsRNA) to a level compared to the commercial ARCA-GMP lot. It was further observed that even after removal of dsRNA, ARCA mRNA triggers higher levels of IFN type I upregulation compared to CC1 and EC1 cap, suggesting remaining innate immune recognition by IFIT family of proteins, which recognize Cap 0.

Claims

CLAIMS What is claimed is: 1. A composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a CD5 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least three intracellular signaling domains, wherein the at least two intracellular signaling domains comprise: (A) a first intracellular signaling domain from Fc epsilon receptor Ig (FCER1G), and (B) a second intracellular signaling domain comprising a PI3K recruitment domain.
2. The composition of claim 1, wherein the CFP has the sequence according to SEQ ID NO:50.
3. The composition of claim 1, wherein the Cap structure is a trinucleotide cap analog.
4. A composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a Cap structure selected from the Cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a HER2 binding domain, (ii) a transmembrane domain operatively linked to the extracellular domain; and (iii) an intracellular domain comprising at least two intracellular signaling domains, wherein the at least three intracellular signaling domains comprise: (A) a first intracellular signaling domain from CD40, (B) a second intracellular signaling domain from Fc epsilon receptor Ig (FCER1G), (C) a third intracellular signaling domain comprising a PI3K recruitment domain.
5. The composition of claim 4, wherein the CFP has the sequence according to SEQ ID NO:51.
6. The composition of claim 4, wherein the Cap structure is a trinucleotide cap analog.
7. The composition of any one of the claims 1-6, wherein the Cap comprises a 2’-O-methyl modification.
8. The composition of any one of the claims 1-7, wherein the Cap comprises an N6 methyl modification.
9. The composition of any one of the claims 1-7, wherein the Cap comprises both a 2’-O- methyl modification and an N6 methyl modification.
10. The composition of any one of the claims 1-9, wherein the mRNA is in vitro transcribed.
11. The composition of any one of the claims 1-10, wherein the mRNA is HPLC purified.
12. A composition comprising an mRNA comprising a sequence encoding a CFP; a 5’UTR, a 3’UTR, the 5’UTR terminating in a 5’cap analog represented by the formula 5’- m7GpppAmpG, wherein, G represents Guanine, A represents Adenine, p represents phosphate group, and m represents methylation; and wherein, the CFP comprises an antigen binding domain having a variable heavy chain sequence with at least 95% sequence identity to
Figure imgf000202_0001
13. The composition of claim 12, wherein the CFP comprises an antigen binding domain having a variable light chain sequence with at least 95% sequence identity to
Figure imgf000202_0002
14. The composition of claim 12 or 13, wherein the CFP comprises an scFv having a sequence that is at least 95% identical to the sequence
Figure imgf000202_0003
15. The composition of any one of claims 12-14, wherein the CFP further comprises a transmembrane domain comprising a CD8 transmembrane domain, the sequence comprising at least 95% identity to IYIWAPLAGTCGVLLLSLVITLYC.
16. The composition of any one of claims 12-15, wherein the CFP further comprises a CD8 hinge domain having a sequence that has at least 95% sequence identity to the sequence ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTR GLD.
17. The composition of any one of claims 12-16, wherein the CFP further comprises an intracellular signaling domain encoding an FCER1G intracellular signaling domain having a sequence: RRLKIQVRKAAITSYEKSDGVYTGLSTRNQETYETLKHEKPPQ.
18. The composition of any one of claims 12-17, wherein the CFP further comprises an intracellular signaling domain encoding an intracellular signaling domain from PI3kinase recruitment domain, having the sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM.
19. The composition of any one of claims 12-18, wherein the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
20. The composition of any one of claims 12-19, wherein the CFP specifically binds to CD5 antigen.
21. The composition of any one of claims 1-11, wherein the cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
22. The composition of claim 21, wherein a nucleotide in the second position of the 5’cap analog is methylated.
23. The composition of claim 21 or 22, wherein the nucleotide in the second position of the 5’cap is further modified at the N6 position.
24. The composition of any one of claims 1-23, wherein expression of the sequence encoded by the mRNA in a cell is enhanced compared to an mRNA that does not comprise the 5’cap of the claim.
25. The composition of claim 4 or 5, wherein the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to
Figure imgf000203_0002
26. The composition of claim 4 or 5, wherein the CFP comprises an antigen binding domain having a variable domain that has a sequence of at least 95% identity to DIQMTQSPSSLSASVGDRVTITCRASQDVNTAVAWYQQKPGKAPKLLIYSASFLYSG VPSRFSGSRSGTDFTLTISSLQPEDFATYYCQQHYTTPPTFGQGTKVEIK.
27. The composition of any one of claims 4-5, 25-26, wherein the CFP further comprises an antigen binding domain that is an scFv having a sequence that is at least 95% identical to the sequence
Figure imgf000203_0001
28. The composition of any one of claims 4-5, 25-27, wherein the CFP further comprises a CD8 hinge domain having a sequence ALSNSIMYFSHFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTR GLD.
29. The composition of any one of claims 4-5, 25-28, wherein the CFP further comprises a CD8 transmembrane domain having a sequence, IYIWAPLAGTCGVLLLSLVITLYC.
30. The composition of any one of claims 4-5, 25-29, wherein the intracellular signaling domain from CD40 comprises a sequence KKVAKKPTNKAPHPKQEPQEINFPDDLPGSNTAAPVQETLHGCQPVTQEDGKESRIS VQERQ.
31. The composition of any one of claims 4-5, 25-30, wherein the intracellular signaling domain from FCER1G comprises a sequence RLKIQVRKAAITSYEKSDGVYTGLSTRNQETYETLKHEKPPQ.
32. The composition of any one of claims 4-5, 25-31, wherein the intracellular signaling domain from PI3K recruitment domain comprises a sequence YEDMRGILYAAPQLRSIRGQPGPNHEEDADSYENM.
33. A composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; and wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a GPC3 binding domain and (ii) a CD89 transmembrane domain.
34. The composition of claim 33, wherein the CD89 transmembrane domain is operably linked to the extracellular domain.
35. The composition of claim 33 or 34, wherein the CFP further comprises (iii) a CD89 intracellular domain.
36. The composition of claim 33 or 34, wherein the CFP further comprises an extracellular domain from CD89.
37. The composition of any one of the claims 33-36, wherein the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
38. The composition of any one of the claims 33-37, wherein the GPC3 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is FYSYTY.
39. The composition of claim 38, wherein the HCDR1 sequence is DYEMH; and the HCDR2 sequence is ALDPKTGDTAYSQKFKG.
40. The composition of any one of claims 33-39, wherein the GPC3 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is SQNTHVPPT.
41. The composition of any one of claims 33-40, wherein the LCDR1 sequence is RSSQSLVHSNRNTYLH; and the LCDR2 sequence is KVSNRFS.
42. The composition of any one of claims 33-41, wherein the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence QVQLVQSGAEVKKPGASVKVSCKASGYTFTDYEMHWVRQAPGQGLEWMGALDP KTGDTAYSQKFKGRVTLTADKSTSTAYMELSSLTSEDTAVYYCTRFYSYTYWGQG TLVTVSS.
43. The composition of any one of claims 33-42, wherein the GPC3 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000205_0001
44. The composition of any one of claims 33-43, wherein the CD89 transmembrane domain is
Figure imgf000205_0002
45. The composition of any one of claims 33-44, wherein CFP comprises a CD89 sequence that is at least 95 % identical to the sequence
Figure imgf000205_0003
46. The composition of any one of claims 33-44, wherein the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
47. A composition comprising a recombinant polynucleic acid or a myeloid cell comprising the recombinant polynucleic acid, wherein the recombinant polynucleic acid is an mRNA with a 5’cap structure selected from the cap structures of Table 7; wherein the mRNA comprises a sequence encoding a chimeric fusion protein (CFP), the CFP comprising: (i) an extracellular domain comprising a TROP2 binding domain and (ii) a CD89 transmembrane domain.
48. The composition of claim 47, wherein the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV.
49. The composition of claim 38, wherein the HCDR1 sequence is NYGMN; and the HCDR2 sequence is WINTYTGEPTYTDDFKG.
50. The composition of any one of claims 33-39, wherein the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLTF.
51. The composition of any one of claims 33-40, wherein the LCDR1 sequence is KASQDVSIAVA; and the LCDR2 sequence is SASYRYT.
52. The composition of any one of claims 33-41, wherein the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000206_0001
53. The composition of any one of claims 33-42, wherein the TROP2 binding variable domain comprises a sequence that is at least 95% identical to the sequence
Figure imgf000206_0002
54. The composition of any one of claims 33-43, wherein the CD89 transmembrane domain is LIRMAVAGLVLVALLAILV.
55. The composition of any one of claims 33-44, wherein the CFP comprises a CD89 extracellular sequence DSIHQDYTTQN.
56. The composition of any one of claims 33-44, wherein the CFP comprises a CD89 intracellular sequence ENWHSHTALNKEASADVAEPSWSQQMCQPGLTFARTPSVCK.
57. The composition of any one of claims 33-44, wherein the CFP further comprises a signal sequence MWLQSLLLLGTVACSIS.
58. A composition comprising: (a) a lipid nanoparticle comprising (i) a cationic lipid (ii) a non-cationic lipid, and optionally, (iii) a neutral lipid; the lipid nanoparticle encapsulating: (b) a recombinant mRNA encoding a chimeric fusion protein (CFP), wherein the recombinant mRNA comprises a 5’cap analog represented by the formula 5’- m7GpppAmpG; the CFP comprise an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain; wherein the transmembrane domain of the protein CD89, Fc ^R1 (CD64), Fc γRIIIA (CD16), or Fc γRIIA (CD32a).
59. The composition of claim 58, wherein the 5’cap is a cap 1 structure.
60. The composition of claim 58, wherein a nucleotide in the second position of the 5’cap is methylated.
61. The composition of any one of claims 58-60, wherein the nucleotide in the second position of the 5’cap is further modified.
62. The composition of any one of claims 58-61, wherein the nucleotide in the second position of the 5’cap is modified at the N6 position.
63. The composition of any one of claims 58-62, wherein the lipid nanoparticle further comprises cholesterol.
64. The composition of any one of claims 58-63, wherein the lipid nanoparticle is less than 200 nM in diameter.
65. The composition of any one of claims 58-64, wherein the chimeric antigen receptor comprises an antigen binding domain that binds to a cancer antigen.
66. The composition of any one of claims 58-65, wherein the chimeric antigen receptor comprises an antigen binding domain that is an scFv, and that specifically binds to a cancer antigen.
67. The composition of any one of claims 58-66, wherein the chimeric antigen receptor comprises an antigen binding domain that binds to TROP2.
68. The composition of claim 47, wherein the TROP2 binding domain comprises a variable heavy chain (VH) domain, having an HCDR1, an HCDR2 and an HCDR3 sequence, wherein the HCDR3 sequence is GGFGSSYWYFDV.
69. The composition of claim 38, wherein the HCDR1 sequence is NYGMN; and the HCDR2 sequence is WINTYTGEPTYTDDFKG.
70. The composition of any one of claims 33-39, wherein the TROP2 binding domain comprises a variable light chain (VL) domain, having an LCDR1, an LCDR2 and an LCDR3 sequence, wherein the LCDR3 sequence is QQHYITPLT.
71. The composition of any one of claims 33-40, wherein the LCDR1 sequence is KASQDVSIAVA; and the LCDR2 sequence is SASYRYT.
72. The composition of any one of claims 58-66, wherein the chimeric antigen receptor comprises GPC3 binding variable heavy chain (VH), and a variable light chain (VL), wherein the variable heavy chain comprises a HCDR1, a HCDR2 and a HCDR3, the HCDR3 having a sequence of ATACADTTQYAYDY, ATACADTTLYEYDY, ATACVDTTQYEYDY, ATACADATQHEYDY, ATACADTTQYDYDY, ATACADTTQYEYDY, ATACADTTHYEYDY, ATACVITTLYEYDY, ATACAETTLYEYDY, ATACADTTQHEYDY, ATACVDTTHYEYDY, ATACASTTLYEYDY, ATACVVTTLYEYDY, ATACGGATGPYDY, ATACAGAIGPYDY, ATACVVVGDQNDY, ATACVVVGDRNDY, ATDCAGGTSTPYDY, ATDCAGGTATPYDY, ATACVVADRNEYDY, ATSCVVVTKNEYDY, ATACSGLTHEYDY, ATTCSGLTHEYDY, ATACANWSSLGPYDY, ATACANWSTLGPYDY, ATACSDPRVYEYDY, ATTCASPEKYEYDY, ATHCGGTSWGTSYDY, ATHCGGSSWSNEYDY, YARYSGRTY, ASSAWPAGPKHQVEYDY, ATACGSLVGMYDY, ATACGSAVHEYDY, ATDCVGFGSNWFDY, ATACASPVIYEYDY, ATDCAGGVGHEYDY, ATDCSLHGSDYPYDY and AVRIYSGSFDNTLAYDY.
73. The composition of any one of claims 58-72, wherein the chimeric antigen receptor comprises an antigen binding domain that is an variable heavy chain (VHH), and that specifically binds to a cancer antigen.
74. A pharmaceutical composition comprising the composition of any one of claims 1-73; and a pharmaceutically acceptable carrier.
75. A cell comprising the recombinant mRNA of any one of claims 1-73.
76. A method of expressing a chimeric fusion protein in a cell, the method comprising in vitro transcribing a recombinant mRNA encoding the chimeric fusion protein, 5’capping the mRNA with a Cap structure selected from the Cap structures of Table 7, thereby generating a 5’-capped recombinant mRNA; contacting the cell with a composition comprising the 5’- capped recombinant mRNA encoding the chimeric fusion protein, wherein the mRNA is incorporated into the cell.
77. The method of claim 76, wherein the 5’cap structure is a 5’cap analog represented by the formula 5’-m7GpppAmpG.
78. The method of claim 76, wherein a nucleotide in the second position of the 5’cap is methylated, and is further modified at the N6 position.
79. The method of any one of claims 76-78, wherein the mRNA expression is enhanced compared to a recombinant mRNA that does not comprise the 5’cap structure of the claim.
80. The method of any one of claims 76-79, wherein the composition further comprises a lipid nanoparticle encapsulating the 5’-capped recombinant mRNA.
81. A method of treating a cancer in a subject, the method comprising administering to the subject the composition of any one of claims 1-73.
82. A method of treating a cancer in a subject, the method comprising administering to the subject a composition comprising the cell of claim 75.
83. The method of claim 82, wherein the cell is a CD14+ cell.
84. The method of claim 83, wherein the cell does not express CD16.
85. The method of claim 83, wherein the cell is CD14+ and CD16+.
PCT/US2023/024049 2022-05-31 2023-05-31 Engineered chimeric fusion protein compositions and methods of use thereof WO2023235422A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263347183P 2022-05-31 2022-05-31
US63/347,183 2022-05-31

Publications (2)

Publication Number Publication Date
WO2023235422A2 true WO2023235422A2 (en) 2023-12-07
WO2023235422A3 WO2023235422A3 (en) 2024-04-18

Family

ID=89025522

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/024049 WO2023235422A2 (en) 2022-05-31 2023-05-31 Engineered chimeric fusion protein compositions and methods of use thereof

Country Status (1)

Country Link
WO (1) WO2023235422A2 (en)

Also Published As

Publication number Publication date
WO2023235422A3 (en) 2024-04-18

Similar Documents

Publication Publication Date Title
RU2768829C2 (en) Anticancer rna vaccines
US20230027864A1 (en) Compositions and methods for delivery of agents to immune cells
AU2020266595A1 (en) Engineered chimeric fusion protein compositions and methods of use thereof
US11026973B2 (en) Engineered phagocytic receptor compositions and methods of use thereof
ES2948133T3 (en) Methods to improve the efficiency and expansion of cells expressing a chimeric antigen receptor
US20220241428A1 (en) Macrophage specific engager compositions and methods of use thereof
US20220001031A1 (en) Engineered chimeric fusion protein compositions and methods of use thereof
CA3204211A1 (en) Chimeric receptor therapy
US20230374538A1 (en) Methods and Compositions for Expression of Nucleic Acids in Cells
JP2022513372A (en) Chimeric antigen receptor targeting Sialyl Lewis A and its use
CN114901304A (en) Treatment involving immune effector cells genetically modified to express antigen receptors
WO2023235422A2 (en) Engineered chimeric fusion protein compositions and methods of use thereof
US20220378824A1 (en) Engineered chimeric fusion protein compositions and methods of use thereof
AU2022280016A1 (en) Engineered chimeric fusion protein compositions and methods of use thereof
CN117836327A (en) Engineered chimeric fusion protein compositions and methods of use thereof
US20240139241A1 (en) Compositions and methods for conditioning patients for cell therapy
AU2022261141A1 (en) Compositions and methods for conditioning patients for cell therapy
WO2024039683A1 (en) Compositions and methods for conditioning patients for cell therapy
WO2024064366A2 (en) Engineered chimeric fusion protein compositions and methods of use thereof
WO2024050138A2 (en) Compositions for cell-specific expression and uses thereof
TW202208418A (en) T cell receptors with vgll1 specificity and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23816707

Country of ref document: EP

Kind code of ref document: A2