Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PARTICLES DISPLAYING ADHESION-MOLECULE FUSIONS
Document Type and Number:
WIPO Patent Application WO/2024/097992
Kind Code:
A2
Abstract:
Provided herein are particles comprising a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule or an activation molecule, as well as vectors, such as lentiviral vectors, comprising the same, cells comprising the same, and methods of using the same. The particle may be a lentiviral particle that displays on the surface of the particle a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 or CD86 extracellular domain, or a functional fragment thereof, and a viral glycoprotein (G protein).

Inventors:
NICOLAI CHRISTOPHER (US)
TANG WEILIANG (US)
RYU BYOUNG (US)
LARSON RYAN (US)
QIN JIM (US)
GOTTSCHALK REBECCA (US)
LEUNG WAI-HANG (US)
PARK LISA (US)
Application Number:
PCT/US2023/078688
Publication Date:
May 10, 2024
Filing Date:
November 03, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UMOJA BIOPHARMA INC (US)
International Classes:
C12N15/867; A61P35/00; C07K14/725; C07K16/30; C07K19/00; C12N5/0783; C12N5/10; C12N15/62
Domestic Patent References:
WO2015057852A12015-04-23
WO2016139463A12016-09-09
WO2020106992A12020-05-28
WO1995014785A11995-06-01
WO1996022378A11996-07-25
WO1994019478A11994-09-01
WO2022072885A12022-04-07
WO2019217954A12019-11-14
WO2020123649A12020-06-18
WO2009072003A22009-06-11
WO2019217964A12019-11-14
Foreign References:
US7741465B12010-06-22
US7446190B22008-11-04
US9856322B22018-01-02
US8399964B22013-03-19
US20160152723A12016-06-02
US10736918B22020-08-11
US10357514B22019-07-23
US20200246381A12020-08-06
US10918665B22021-02-16
US20190161553A12019-05-30
US20220033509A12022-02-03
US20180118803A12018-05-03
US20200231686A12020-07-23
US20210393689A12021-12-23
US20170275362A12017-09-28
US20220096651A12022-03-31
US20160348073A12016-12-01
US20180085399A12018-03-29
US20190256597A12019-08-22
US20140349402A12014-11-27
US20170240612A12017-08-24
US9624276B22017-04-18
US20160272718A12016-09-22
US20150320799A12015-11-12
US20190224237A12019-07-25
US20200023009A12020-01-23
US20200405760A12020-12-31
US20150044229W2015-08-07
US20170290900A12017-10-12
US20190091308A12019-03-28
US20200216502A12020-07-09
US6013516A2000-01-11
US5994136A1999-11-30
US5882877A1999-03-16
US4861719A1989-08-29
US5278056A1994-01-11
US20170240612A12017-08-24
US9624276B22017-04-18
US20160272718A12016-09-22
US20200123224A12020-04-23
US5543158A1996-08-06
US5641515A1997-06-24
US5399363A1995-03-21
US5466468A1995-11-14
US20190269727A12019-09-05
US20160122782A12016-05-05
US20210163893A12021-06-03
US20170037369A12017-02-09
US20210244871A12021-08-12
Other References:
IKEMIZU ET AL., PNAS USA, vol. 96, no. 8, 1999, pages 4289 - 94
IKEMIZU ET AL., IMMUNITY, vol. 12, 2000, pages 51 - 60
SCHWARTZ ET AL., NATURE, vol. 410, 2001, pages 604 - 608
SUSAC ET AL., CELL, vol. 185, no. 17, 2022, pages 3201 - 3213
GUEDAN SCALDERON HPOSEY ADMAUS MV, MOLECULAR THERAPY - METHODS & CLINICAL DEVELOPMENT., vol. 12, 2019, pages 145 - 156
CARTELLIERI ET AL., BLOOD CANCER JOURNAL, vol. 6, 2016, pages 458
ARCANGELI ET AL., TRANSL CANCER RES, vol. 5, 2016, pages 174 - 177
KOLMAR H. ET AL., THE FEBS JOURNAL., vol. 275, no. 11, 2008, pages 26684 - 90
REICHERT, J.M. MABS, vol. 1, no. 3, 2009, pages 190 - 209
KULARATNE, S.A. ET AL., MOL PHARM., vol. 6, no. 3, 2009, pages 780 - 9
WAYUA. C. ET AL., MOLECULAR PHARMACEUTICS, 2013
SEGA, E.I. ET AL., CANCER METASTASIS REV, vol. 27, no. 4, 2008, pages 655 - 64
HENNIG ET AL., INTERNATIONAL JOURNAL OF CANCER, vol. 61, pages 786 - 792
NALDINI ET AL., SCIENCE, vol. 272, 1996, pages 263 - 7
ZUFFEREY ET AL., J. VIROL., vol. 72, 1998, pages 8150 - 8471
HUMBERT ET AL., MOLECULAR THERAPY, vol. 24, 2016, pages 1237 - 1246
GAERERTS ET AL., BMC BIOTECHNOL., vol. 6, 2006, pages 34
CARTELIIERI ET AL., BLOOD CANCER J., vol. 6, 2016, pages 458
"Remington's Pharmaceutical Sciences", 1980
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
ABOU-EL-ENEIN, M. ET AL., BLOOD CANCER DISCOV, vol. 2, no. 5, 2021, pages 408 - 422
ARCANGELI, S. ET AL., FRONT. IMMUNOL, vol. 11, no. 1217, 19 June 2020 (2020-06-19), pages 1 - 13
GHASSEMI, S. ET AL., NAT BIOMED ENG, vol. 6, no. 2, February 2022 (2022-02-01), pages 118 - 128
VORMITTAG, P. ET AL., CURR OPIN BIOTECHNOL, vol. 54, October 2018 (2018-10-01), pages 164 - 181
Attorney, Agent or Firm:
MIRON, Anna et al. (US)
Download PDF:
Claims:
Attorney Ref. No.061479-506001WO CLAIMS 1. A particle, comprising, displayed on the surface of the particle: a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule or an activation molecule. 2. The particle of claim 1, wherein the adhesion molecule is linked to the costimulatory molecule and the activation molecule. 3. The particle of any one of claim 1-2, wherein the adhesion molecule comprises an adhesion protein. 4. The particle of any one of claims 1-3, wherein the adhesion molecule comprises CD58, a CD58 extracellular domain, or a functional fragment of CD58; optionally wherein the fusion molecule comprises a CD58 extracellular domain, or a functional fragment thereof, a CD80 or CD86 extracellular domain, or a functional fragment thereof, and an antigen-binding fragment of an anti-CD3 antibody. 5. The particle of any one of claims 1-4, wherein the adhesion molecule comprises ICAM-1, ICAM-2, ICAM-3, ICAM-4, ICAM-5, JAM-A, CD155 or CD112; an extracellular domain thereof; or a functional fragment thereof. 6. The particle of any one of claims 1-5, wherein the adhesion molecule comprises an antibody or antigen-binding fragment thereof. 7. The particle of any one of claims 1-6, wherein the adhesion molecule specifically binds CD2, LFA-1, or DNAM-1. 8. The particle of any one of claims 1-7, wherein the costimulatory molecule comprises costimulatory protein. 9. The particle of any one of claims 1-8, wherein the costimulatory molecule comprises CD80, CD86,CD40L, GITRL, OX40L, 41BBL, ICOSL, CD27, CD30L, LIGHT, LTalpha, MICA, or MICB; an extracellular domain thereof; or a functional fragment thereof. 10. The particle of any one of claims 1-9, wherein the costimulatory molecule comprises a CD80, a CD80 extracellular domain thereof, or a functional fragment of CD80. 11. The particle of any one of claims 1-10, wherein the costimulatory molecule comprises a CD86, a CD86 extracellular domain thereof, or a functional fragment of CD86. Attorney Ref. No.061479-506001WO 12. The particle of any one of claims 1-11, wherein the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: a CD80, a CD80 extracellular domain, or a functional fragment of CD80; a polypeptide linker; and a CD58, a CD58 extracellular domain; or a functional fragment of CD58. 13. The particle of any one of claims 1-12, wherein the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: a CD86, a CD86 extracellular domain, or a functional fragment of CD86; a polypeptide linker; and a CD58, a CD58 extracellular domain; or a functional fragment of CD58. 14. The particle of any one of claims 1-13, wherein the activation molecule comprises a TCR-binding molecule. 15. The particle of any one of claims 1-14, wherein the fusion molecule comprises the adhesion molecule, the costimulatory molecule, and the TCR-binding molecule, each component linked directly or indirectly to the other components. 16. The particle of any one of claims 1-15, wherein the TCR-binding molecule comprises an antibody, or antigen-binding fragment thereof, that specifically binds CD3. 17. The particle of any one of claims 1-16, wherein the TCR-binding molecule comprises a single chain variable fragment that specifically binds CD3. 18. The particle of any one of claims 1-17, wherein the TCR-binding molecule comprises a variable domain comprising complementarity determining regions: a. an antibody VL domain comprising L-CDR1, L-CDR2 and L-CDR3, wherein: L-CDR1 comprises the sequence SASSSVSYMN (SEQ ID NO: 57); L-CDR2 comprises the sequence DTSKLASG (SEQ ID NO: 58); and L-CDR3 comprises the sequence QQWSSNPFT (SEQ ID NO: 59); and b. an antibody VH domain comprising H-CDR1, H-CDR2 and H-CDR3, wherein: H-CDR1 comprises the sequence RYTMH (SEQ ID NO: 54); H-CDR2 comprises the sequence YINPSRGYTNYNQKVKD (SEQ ID Attorney Ref. No.061479-506001WO NO: 55); and H-CDR3 comprises the sequence YYDDHYCLDY (SEQ ID NO: 56). 19. The particle of any one of claims 1-18, wherein the fusion molecule is a fusion protein comprising, in any order: a. CD80, a CD80 extracellular domain, or a functional fragment of CD80; b. CD58, a CD58 extracellular domain; or a functional fragment of CD58; c. a TCR-binding molecule; and d. polypeptide linkers. 20. The particle of any one of claims 1-19, wherein the CD58 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 1 or 10. 21. The particle of any one of claims 1-20, wherein the CD80 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to one or more of SEQ ID NO: 12 or 25-26. 22. The particle of any one of claims 1-21, wherein the CD86 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to one or more of SEQ ID NO: 13 or 27-28. 23. The particle of any one of claims 1-22, wherein the TCR-binding molecule comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 31. 24. The particle of any one of claims 1-23, wherein the fusion protein comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 32 or 33. 25. The particle of any one of claims 1-24, wherein the particle comprises a viral glycoprotein. 26. The particle of claim 25, wherein the viral glycoprotein comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 247. 27. The particle of any one of claims 1-24, wherein the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen Attorney Ref. No.061479-506001WO receptor, a small molecule-inducible cytokine receptor, and/or an immunosuppresion-resistance protein, optionally wherein the particle comprises a polynucleotide encapsulated therein. 28. The particle of any one of claims 1-27, wherein the particle is a viral particle, optionally a lentiviral particle. 29. A pharmaceutical composition comprising the particle of any one of claims 1- 28, and a pharmaceutically acceptable carrier. 30. An ex vivo method of transducing target cells, comprising contacting the target cells with the particle of any one of claims 1-28. 31. An in vivo method of transducing target cells in a subject in need thereof, comprising administering to the subject the particle of any one of claims 1-28. 32. The method of claim 31, wherein the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen receptor, and wherein the chimeric antigen receptor is expressed on the target cells after administration of the particle. 33. The method of claim 31 or 32, wherein the particle is administered by intranodal, intravenous, or subcutaneous injection. 34. The method of any one of claims 31-33, wherein the particle is contacted with a target cell by extracorporeal incubation. 35. The method of any one of claims 31-34, where the subject suffers from or is at risk for a B-cell malignancy, relapsed/refractory CD19-expressing malignancy, diffuse large B-cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma. 36. A kit comprising the particle of any one of claims 1-28, the particle comprising the fusion molecule or a polynucleotide encoding the fusion molecule, and instructions for use in transduction of target cells and/or treatment of a subject. Attorney Ref. No.061479-506001WO 37. The kit of claim 36, further comprising a pharmaceutically acceptable carrier. 38. The kit of claim 36 or claim 37, further comprising an injection device. 39. A polynucleotide encoding the fusion molecule of any one of claims 1-28. 40. A host cell comprising the polynucleotide of claim 39. 41. A method of making a particle, comprising introducing a polynucleotide encoding a vector genome into the host cell of claim 40, wherein the fusion molecule and the vector genome are expressed by the host cell and wherein the host cell packages the vector genome into a viral particle comprising the fusion molecule. 42. A lentiviral particle, comprising, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody c) a CD80 or CD86 extracellular domain, or a functional fragment thereof; and a viral glycoprotein (G protein), wherein the lentiviral particle comprises a polynucleotide encoding a chimeric antigen receptor. 43. The lentiviral particle of claim 42, whererin a), b), and c) are in N- to C- terminal order. 44. The lentiviral particle of claim 42 or claim 43, wherein the fusion molecule comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to any one of SEQ ID NOs: 72 or 33. 45. The lentiviral particle of any one of claims 42-44, wherein the fusion molecule comprises a CD58 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 248. 46. The lentiviral particle of any one of claims 42-45, wherein the fusion molecule comprises a CD80 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 250. Attorney Ref. No.061479-506001WO 47. The lentiviral particle of any one of claims 42-46, wherein the fusion molecule comprises a anti-CD3 scFv polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 249. 48. A pharmaceutical composition comprising the particle of any one of claims 42- 47, and a pharmaceutically acceptable carrier. 49. An ex vivo method of transducing target cells, comprising contacting the target cells with the particle of any one of claims 42-47. 50. An in vivo method of transducing target cells in a subject in need thereof, comprising administering to the subject the particle of any one of claims 42-47. 51. The method of claim 50, wherein the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen receptor, and wherein the chimeric antigen receptor is expressed on the target cells after administration of the particle. 52. A polynucleotide encoding the particle of any one of claims 42-47. 53. A host cell comprising the polynucleotide of claim 52. 54. A method of making a particle, comprising introducing a polynucleotide encoding a vector genome into the host cell of claim 53, wherein the fusion molecule and the vector genome are expressed by the host cell and wherein the host cell packages the vector genome into a viral particle comprising the fusion molecule. 55. A composition or method as described herein comprisng the particle of any of the preceding claims. 56. A method of treating cancer in a subject in need thereof, the method comprsing administering to the subject a therapeutically effective amount of the lentiviral particle of any of the preceing claims, where the subject suffers from or is at risk for a B-cell malignancy, relapsed/refractory CD19-expressing malignancy, diffuse large B-cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B Attorney Ref. No.061479-506001WO cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma. 57. A method of administering a lentiviral particle to a subject, the method comprising: a) obtaining whole blood from a subject; b) collecting the fraction of blood containing peripheral blood mononuclear cells (PBMCs) or a subset thereof; c) contacting the collected PBMCs or subset with a composition comprising lentiviral particles to create a transfection mixture; and d) reinfusing the transfection mixture to the subject, thereby administering the lentiviral particle to the subject, wherein the lentiviral particle comprises, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 or CD86 extracellular domain, or a functional fragment thereof; and a viral glycoprotein (G protein), and wherein the lentiviral particle comprises a polynucleotide encoding a chimeric antigen receptor. 58. The method of claim 57, wherein the method is carried out in a single in-line procedure to maintain a closed or functionally closed fluid circuit. 59. The method of any one of claims 57-58, wherein: two or more of steps (a)-(d) are carried out in-line in a closed fluid circuit; three or more of steps (a)-(d) are carried out in-line in a closed fluid circuit; or wherein all of steps (a)-(d) are carried out in-line in a closed fluid circuit. 60. A fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, c) a CD80 or CD86 extracellular domain, or a functional fragment thereof; and a viral glycoprotein (G protein), Attorney Ref. No.061479-506001WO wherein the lentiviral particle comprises a polynucleotide encoding a chimeric antigen receptor. 61. The fusion molecule of claim 60, whererin a), b), and c) are in N- to C-terminal order. 62. The fusion molecule of claim 60 or claim 61, wherein the fusion molecule comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to any one of SEQ ID NOs: 72 or 33. 63. The fusion molecule of any one of claims 60-62, wherein the fusion molecule comprises a CD58 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 248. 64. The fusion molecule of any one of claims 60-63, wherein the fusion molecule comprises a CD80 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 250. 65. The fusion molecule of any one of claims 60-64, wherein the fusion molecule comprises a anti-CD3 scFv polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 249.
Description:
Attorney Ref. No.061479-506001WO PARTICLES DISPLAYING ADHESION-MOLECULE FUSIONS CROSS-REFERENCE [0001] This application claims the benefit of U.S. Provisional Application No. 63/422,678, filed November 4, 2022, U.S. Provisional Application No.63/487,784, filed March 1, 2023, U.S. Provisional Application No. 63/466,471, filed May 15, 2023, and U.S. Provisional Application No.63/579,188, filed Aug 28, 2023, which applications are incorporated herein by reference in their entireties. INCORPORATION BY REFERENCE OF SEQUENCE LISTING [0002] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled 061479- 506001WO_SeqList_ST26.xml, created November 2, 2023, which is 340 kilobytes in size. The information in the electronic format of the Sequence Listing is incorporated by reference in its entirety. FIELD OF THE INVENTION [0003] This disclosure relates generally to cell biology, immunology, and medicine - more particularly to lentiviral particles for use as medical treatments. BACKGROUND [0004] T cells may be genetically engineered for use as therapeutic agents. For example, some chimeric antigen receptor (CAR) T cells have been approved as treatments for liquid tumors. Improved methods and compositions for enhancing T cells are needed. Genetic engineering of T cells may require delivery of polynucleotides into the T cells selected for engineering, a procedure termed transduction. Transduction of T cells may be achieved using various viral and non-viral delivery vehicles. In one example, a recombinant lentivirus is used for transduction. The lentiviral particle may be engineered to display, on its surface, molecules that enhance transduction. An antibody or antibody fragment against a component of a T cell receptor, such as CD3, may be surface-displayed on the lentivirus to target the virus to T cells. Additionally, engagement of CD3 by a lentivirus comprising a binding domain targeting CD3 may cause the T cells to activate via a primary activation signal. Surface display of one or more ligands for a co-receptor, such as CD28, a molecule expressed by T cells, may cause the T cells to activate via a secondary activation signal. Activation of a T cell, via the primary and optionally secondary activation signals, which Attorney Ref. No.061479-506001WO may make them more susceptible to transduction. Ligands for CD28 may include, for example, CD80 and CD86. [0005] There remains a need in the art for new compositions and methods that further improve transduction of T cells by delivery vehicles such as recombinant lentiviruses. The present disclosure addresses this need. SUMMARY [0006] The present disclosure relates, in part, to the recognition by the present inventors that, by engineering a particle used as a delivery vehicle to display an adhesion molecule on its surface, transduction of target cells (such as T cells) by the particle may be enhanced. More specifically, without being bound by theory, surface engineering of a particle with an adhesion molecule, a co-stimulation molecule, and optionally an activation molecule (such as an anti-CD3 antibody fragment) may generate a macromolecular complex at the interface of the particle and cell that acts as artificial supramolecular activation cluster. [0007] The present inventors have further recognized that engineered particles may be enhanced by fusing an adhesion molecule to a costimulatory molecule, an activation molecule, or both. Surprisingly, the present inventors discovered that a fusion molecule as disclosed herein, e.g. comprising adhesion molecule domain(s), costimulatory molecule domain(s), and optionally activation molecule domain(s) were able to bind, activate, and permit transduction of target cells indicating that each domain not only was appropriately positioned to enable binding its cognate ligand, but maintained its functional characteristics (e.g. folding and structure). [0008] Accordingly, in one aspect, the disclosure provides a lentiviral particle for transduction of target cells, comprising, displayed on the surface of the lentiviral particle, a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule, an activation molecule, or both. The particle may be a viral particle, such as a lentiviral particle. The adhesion molecule, costimulatory molecule, and activation molecule may each be proteins and may collectively be fused into one (or more) fusion proteins. In one aspect, the present disclosure provides a lentiviral particle comprising a polycistronic construct comprising a polynucleotide sequence encoding a chimeric antigen receptor. [0009] In other aspects, the disclosure provides ex vivo and in vivo uses of the lentiviral particles (such as for cell manufacturing and medical treatments), pharmaceutical compositions, and kits, as well as methods of making the particles, polynucleotides, and host cells. Attorney Ref. No.061479-506001WO [0010] The present disclosure provides a particle, comprising, displayed on the surface of the particle: a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule or an activation molecule. [0011] In some embodiments, the particle is a viral particle. In some embodiments, the particle is a lentiviral particle. [0012] In some embodiments, the adhesion molecule is linked to the costimulatory molecule and the activation molecule. [0013] In some embodiments, the adhesion molecule comprises an adhesion protein. [0014] In some embodiments, the adhesion molecule comprises CD58, a CD58 extracellular domain, or a functional fragment of CD58; optionally wherein the fusion molecule comprises a CD58 extracellular domain, or a functional fragment thereof, a CD80 or CD86 extracellular domain, or a functional fragment thereof, and an activation domain, for example, an antigen-binding fragment of an anti-CD3 antibody. [0015] In some embodiments, the adhesion molecule comprises ICAM-1, ICAM-2, ICAM- 3, ICAM-4, ICAM-5, JAM-A, CD155 or CD112; an extracellular domain thereof; or a functional fragment thereof. [0016] In some embodiments, the adhesion molecule comprises an antibody or antigen- binding fragment thereof. [0017] In some embodiments, the adhesion molecule specifically binds CD2, LFA-1, or DNAM-1. [0018] In some embodiments, the costimulatory molecule comprises costimulatory protein. [0019] The particle of any one of claims 1-8, wherein the costimulatory molecule comprises CD80, CD86,CD40L, GITRL, OX40L, 41BBL, ICOSL, CD27, CD30L, LIGHT, LTalpha, MICA, or MICB; an extracellular domain thereof; or a functional fragment thereof. [0020] In some embodiments, the costimulatory molecule comprises a CD80, a CD80 extracellular domain thereof, or a functional fragment of CD80. [0021] In some embodiments, the costimulatory molecule comprises a CD86, a CD86 extracellular domain thereof, or a functional fragment of CD86. [0022] In some embodiments, the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: i) a CD80, a CD80 extracellular domain, or a functional fragment of CD80; ii) a polypeptide linker; and iii) a CD58, a CD58 extracellular domain; or a functional fragment of CD58. Attorney Ref. No.061479-506001WO [0023] In some embodiments, the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: i) a CD86, a CD86 extracellular domain, or a functional fragment of CD86; ii) a polypeptide linker; and iii) a CD58, a CD58 extracellular domain; or a functional fragment of CD58. [0024] In some embodiments, the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: i) a CD58, a CD58 extracellular domain; or a functional fragment of CD58; ii) a polypeptide linker; and iii) a CD80, a CD80 extracellular domain, or a functional fragment of CD80. [0025] In some embodiments, the fusion molecule comprises a fusion protein comprising, in N- to C-terminal order or in C- to N-terminal order: i) a CD58, a CD58 extracellular domain; or a functional fragment of CD58; ii) a polypeptide linker; and iii) a CD86, a CD86 extracellular domain, or a functional fragment of CD86. [0026] In some embodiments, the activation molecule comprises a TCR-binding molecule. [0027] In some embodiments, the fusion molecule comprises the adhesion molecule, the costimulatory molecule, and the TCR-binding molecule, each component linked directly or indirectly to the other components. [0028] In some embodiments, the TCR-binding molecule comprises an antibody, or antigen-binding fragment thereof, that specifically binds CD3. [0029] In some embodiments, the TCR-binding molecule comprises a single chain variable fragment that specifically binds CD3. [0030] In some embodiments, the TCR-binding molecule comprises a variable domain comprising complementarity determining regions: an antibody VL domain comprising L- CDR1, L-CDR2 and L-CDR3, wherein: L-CDR1 comprises the sequence SASSSVSYMN (SEQ ID NO: 57); L-CDR2 comprises the sequence DTSKLASG (SEQ ID NO: 58); and L- CDR3 comprises the sequence QQWSSNPFT (SEQ ID NO: 59); and an antibody VH domain comprising H-CDR1, H-CDR2 and H-CDR3, wherein: H-CDR1 comprises the sequence RYTMH (SEQ ID NO: 54); H-CDR2 comprises the sequence YINPSRGYTNYNQKVKD (SEQ ID NO: 55); and H-CDR3 comprises the sequence YYDDHYCLDY (SEQ ID NO: 56). Attorney Ref. No.061479-506001WO [0031] In some embodiments, the fusion molecule is a fusion protein comprising, in any order: i) CD80, a CD80 extracellular domain, or a functional fragment of CD80; ii) CD58, a CD58 extracellular domain; or a functional fragment of CD58; iii) a TCR-binding molecule; and iv) polypeptide linkers. [0032] In some embodiments, the CD58 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 1 or 10. [0033] In some embodiments, the CD80 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to one or more of SEQ ID NO: 12 or 25-26. [0034] In some embodiments, the CD86 comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to one or more of SEQ ID NO: 13 or 27-28. [0035] In some embodiments, the TCR-binding molecule comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 31. [0036] In some embodiments, the fusion protein comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 32 or 33. [0037] In some embodiments, the particle comprises a viral glycoprotein. [0038] In some embodiments, the viral glycoprotein comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 247. [0039] In some embodiments, the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen receptor, a small molecule-inducible cytokine receptor, and/or an immunosuppresion-resistance protein. [0040] The present disclosure provides a pharmaceutical composition comprising a particle of the present disclosure, and a pharmaceutically acceptable carrier. [0041] An ex vivo method of transducing target cells, comprising contacting the target cells with a particle of the present disclosure. Attorney Ref. No.061479-506001WO [0042] An in vivo method of transducing target cells in a subject in need thereof, comprising administering to the subject a particle of the present disclosure. [0043] In some embodiments, the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen receptor, and wherein the chimeric antigen receptor is expressed on the target cells after administration of the particle. [0044] In some embodiments, the particle is administered by intranodal, intravenous, or subcutaneous injection. [0045] In some embodiments, the particle is contacted with a target cell by extracorporeal incubation. [0046] In some embodiments, the subject suffers from or is at risk for a B-cell malignancy, relapsed/refractory CD19-expressing malignancy, diffuse large B-cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma. [0047] The present disclosure provides a kit comprising a particle of the present disclosure, the particle comprising the fusion molecule or a polynucleotide encoding the fusion molecule, and instructions for use in transduction of target cells and/or treatment of a subject. [0048] In some embodiments, the kit comprises a pharmaceutically acceptable carrier. [0049] In some embodiments, the kit comprises an injection device. [0050] The present disclosure provides a polynucleotide encoding a fusion molecule of the present disclosure. [0051] The present disclosure provides a host cell comprising a polynucleotide of the present disclosure. [0052] The present disclosure provides a method of making a particle, comprising introducing a polynucleotide encoding a vector genome into a host cell, wherein the fusion molecule and the vector genome are expressed by the host cell and wherein the host cell packages the vector genome into a viral particle comprising the fusion molecule. Attorney Ref. No.061479-506001WO [0053] The present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 or a CD86 extracellular domain, or a functional fragment thereof; and a viral glycoprotein (G protein), wherein the lentiviral particle comprises a polynucleotide encoding a chimeric antigen receptor. [0054] In some embodiments, the present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 or a CD86 extracellular domain, or a functional fragment thereof. [0055] In some embodiments, the present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding gragment of an anti-CD3 antibody, and c) a CD80 extracellular domain, or a functional fragment thereof. [0056] In some embodiments, the present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD86 extracellular domain, or a functional fragment thereof. [0057] In all such embodiments, the lentiviral particle may further comprise a viral glycoprotein (G protein). In an exemplary embodiment, the present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: (1) a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 extracellular domain, or a functional fragment thereof; and (2) a viral glycoprotein. [0058] In an exemplary embodiment, the present disclosure provides a lentiviral particle, comprising, displayed on the surface of the particle: Attorney Ref. No.061479-506001WO (1) a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD86 extracellular domain, or a functional fragment thereof; and (2) a viral glycoprotein. In some embodiments, the G protein is a cocal glycoprotein. [0059] In some embodiments, the G protein is a VSV-G protein. In additional embodiments, the lentiviral particle may further comprise a payload comprising a polynucleotide encoding a protein, such as a chimeric antigen receptor. [0060] In some embodiments, a), b), and c) are in N- to C-terminal order. [0061] In some embodiments, the fusion molecule comprises a polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to any one of SEQ ID NOs: 72 or 33. [0062] In some embodiments, the fusion molecule comprises a CD58 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 248. [0063] In some embodiments, the fusion molecule comprises a CD80 polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 250. [0064] In some embodiments, the fusion molecule comprises a anti-CD3 scFv polypeptide having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to SEQ ID NO: 249. [0065] The present disclosure provides a pharmaceutical composition comprising a particle of the present disclosure, and a pharmaceutically acceptable carrier. [0066] The present disclosure provides an ex vivo method of transducing target cells, comprising contacting the target cells with a particle of the present disclosure. [0067] The present disclosure provides an in vivo method of transducing target cells in a subject in need thereof, comprising administering to the subject a particle of the present disclosure. [0068] In some embodiments, the particle comprises a polynucleotide having a polynucleotide sequence encoding a chimeric antigen receptor, and wherein the chimeric antigen receptor is expressed on the target cells after administration of the particle. [0069] The present disclosure provides a polynucleotide encoding the fusion molecule of the present disclosure. Attorney Ref. No.061479-506001WO [0070] The present disclosure provides a host cell comprising the polynucleotide of the present disclosure. [0071] The present disclosure provides a method of making a particle, comprising introducing a polynucleotide encoding a vector genome into the host cell, wherein the fusion molecule and the vector genome are expressed by the host cell and wherein the host cell packages the vector genome into a viral particle comprising the fusion molecule. [0072] The present disclosure provides a composition or method as described herein comprisng the lentiviral particle disclosed herein. [0073] The present disclosure provides a method of treating cancer in a subject in need thereof, the method comprsing administering to the subject a therapeutically effective amount of the lentiviral particle disclosed herein, where the subject suffers from or is at risk for a B-cell malignancy, relapsed/refractory CD19-expressing malignancy, diffuse large B- cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma. [0074] The present disclosure provides a method of administering a lentiviral particle to a subject, the method comprising: a) obtaining whole blood from a subject; b) collecting the fraction of blood containing peripheral blood mononuclear cells (PBMCs) or a subset thereof; c) contacting the collected PBMCs or subset with a composition comprising lentiviral particles to create a transfection mixture; and d) reinfusing the transfection mixture to the subject, thereby administering the lentiviral particle to the subject, wherein the lentiviral particle comprises, displayed on the surface of the particle: a fusion molecule comprising: a) a CD58 extracellular domain, or a functional fragment thereof, b) an antigen-binding fragment of an anti-CD3 antibody, and c) a CD80 or CD86 extracellular domain, or a functional fragment thereof; and Attorney Ref. No.061479-506001WO a viral glycoprotein (G protein), and wherein the lentiviral particle comprises a polynucleotide encoding a chimeric antigen receptor. [0075] In some embodiments, the method is carried out in a single in-line procedure to maintain a closed or functionally closed fluid circuit. [0076] In some embodiments, two or more of steps (a)-(d) are carried out in-line in a closed fluid circuit; three or more of steps (a)-(d) are carried out in-line in a closed fluid circuit; or wherein all of steps (a)-(d) are carried out in-line in a closed fluid circuit. BRIEF DESCRIPTION OF THE DRAWINGS [0077] FIG. 1 depicts T-cell activation by a lentiviral particle displaying a single-chain variable fragment specific for CD3, a viral envelope protein (Cocal G), and two costimulatory molecules. [0078] FIG. 2A shows activation of CD8+ T cells as measured by % CD25+ cells with a lentiviral particle displaying CD3scfv or CD3scfv+CD80. [0079] FIG. 2B shows activation of CD8+ T cells as measured by % CD25+ cells with a lentiviral particle displaying CD3scfv only, CD3scfv+CD80 or CD3scfv+CD58. [0080] FIGs. 2C-2D show levels of CAR expression in CD8+ T cells as determined by %CAR expression (FIG. 2C) or total CAR+ CD8+ T cells (FIG. 2D) generated using lentiviral particles with CD3scfv only or CD3scfv+CD80. [0081] FIGs. 2E-2F show levels of CAR expression in CD3+ T cells as determined by %CAR expression (FIG. 2E) or total CAR+ CD3+ T cells (FIG. 2F) generated using lentiviral particles with CD3scfv only, CD3scfv+CD80 or CD3scfv+CD58. [0082] FIGs.2G-2H show fold expansion of CAR+CD8+ T cells generated with lentiviral particles with CD3scfv only or CD3scfv+CD80 stimulated with IL-2 (FIG. 2G) or rapamycin (FIG.2H). [0083] FIG. 3A shows percentages of CD25(+) CD8 T cells after incubation with a lentiviral particle displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58. [0084] FIG. 3B shows the geometric mean fluorescent intensity (gMFI) of CD25(+) CD8 T cells after incubation with a lentiviral particle displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58. Attorney Ref. No.061479-506001WO [0085] FIGs. 3C-3E show production of cytokines 3 days after incubation with particles displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58. ?<C(l $FIG.3C), IL-2 (FIG.3D%' KWN IC<(h $FIG.3E) levels were measured. [0086] FIGs. 3F-3G show CAR expression in CD3+ T cells generated with lentiviral particles displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 (mixed particles). Percentage (%) CAR expression (FIG. 3F) and total CAR+ T cells (FIG.3G) were measured. [0087] FIGs. 3H-3I show CAR expression in CD8+ T cells generated with lentiviral particles displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 (same particle). Percentage (%) CAR expression (FIG. 3H) and total CAR+ T cells (FIG.3I) were measured. [0088] FIGs. 3J-3L show staining of Cocal (FIG. 3J), CD80 (FIG. 3K) or CD58 (FIG. 3L) on CD8+ T cells incubated with lentiviral particles displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58. [0089] FIG. 3M shows a principal components analysis with three main clusters of differentiation based on particle costimulatory-molecule makeup using CCR7, CD45RO, CD45RA, CD27, CD25, CAR+, CD4, and CD8 markers and total cells. [0090] FIG. 3N shows CD3scfv+CD80 particles generate CAR+ T cells with a predominantly central memory (Tcm) phenotype compared to CD3scfv only, which produced effector T cells (Teff). [0091] FIG. 3O shows CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles generate CAR+ T cells with a predominantly central memory (Tcm) phenotype compared to CD3scfv only, which produced effector T cells (T eff ) central memory T cells (T cm ). [0092] FIG. 4A shows the number of K562.CD19 cells over several days after incubation with anti-CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles. The particles were added to PBMCs at an MOI of 10 along with Tumor cells at PBMC:Tumor ratio of 5:1 and put directly on the Incucyte® live- cell imaging system. CD3scfv+CD80+CD58 CAR T cells were generated using a mixture of individual particles. [0093] FIG.4B shows the number of Raji cells over several days after incubation with anti- CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and Attorney Ref. No.061479-506001WO displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles. The particles were added to PBMCs at an MOI of 10 along with Tumor cells at PBMC:Tumor ratio of 5:1 and put directly on the incucyte. CD3scfv+CD80+CD58 CAR T cells were generated using a mixture of individual particles. [0094] FIG. 4C shows the number of K562.CD19 cells over several days after incubation with anti-CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles.7 days after transduction at an MOI of 10, the total CAR+ cells were calculated and incubated with either K562.CD19 at E:T ratios of 0.5 and 1, respectively. CD3scfv+CD80+CD58 CAR T cells were generated using a mixture of individual particles. [0095] FIG.4D shows the number of Raji cells over several days after incubation with anti- CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles. 7 days after transduction at an MOI of 10, the total CAR+ cells were calculated and incubated with either Raji cells at E:T ratios of 0.5 and 1, respectively. CD3scfv+CD80+CD58 CAR T cells were generated using a mixture of individual particles. [0096] FIG. 4E shows the number of K562.CD19 cells over several days after incubation with anti-CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles.7 days after transduction at an MOI of 10, the total CAR+ cells were calculated and incubated with K562.CD19 cells at E:T ratios of 1:1, respectively. CD3scfv+CD80+CD58 CAR T cells were generated using a single particle with both costimulatory and adhesion molecules. [0097] FIG. 4F shows the number of Nalm6 cells over several days after incubation with anti-CD19 CAR+ T cells generated with lentiviral particles encoding an anti-CD19 CAR and displaying CD3scfv only, CD3scfv+CD80, CD3scfv+CD58, or CD3scfv+CD80+CD58 particles. 7 days after transduction at an MOI of 10, the total CAR+ cells were calculated and incubated with Nalm6 cells at E:T ratios of 1:1, respectively. CD3scfv+CD80+CD58 CAR T cells were generated using a single particle with both costimulatory and adhesion molecules. FIG. 4A-4F are labeled with a key to the right of each plot with labels that correspond (in order) to the right end of each line in the plot. Attorney Ref. No.061479-506001WO [0098] FIG. 5A shows the number of CAR T cells in blood samples of NSG MHCI/II KO mice 11 days after injection of PMBCs and lentiviral particles displaying CD3scfv only or CD3scfv+CD80 particles. [0099] FIGs.5B-5C show the tumor burden in NSG MHCI/II KO mice over 100 days after administration with lentiviral particles displaying CD3scfv only (FIG. 5B) or CD3scfv+CD80 (FIG.5C). [0100] FIGs. 6A-6B show number of cells expressing a CAR 3 days (FIG. 6A) or 7 days (FIG. 6B) after transduction of PBMCs from three healthy donors with lentiviral particles displaying CD3scfv only or CD3scfv+CD80+CD58 particles. [0101] FIGs. 7A-7C show expression of CAR in cells transduced with lentiviral particles pseudotyped with mutant VSV-G envelope proteins. SupT1 cells (FIG.7A) or PBMCs from two healthy donors (FIGs. 7B-7C) were cultured with lentiviral particles having an anti- CD19 CAR payload and displaying mutant VSV-G envelope proteins with or without CD3scfv+CD80+CD58. CAR expression was assessed in CD4+ T cells (FIG. 7B) and CD8+ T cells (FIG. 7C) after transduction of the PBMCs. [0102] FIG. 8 shows the number of CAR negative T cells in the blood of mice after administration of particles at indicated doses encoding an anti-CD19 CAR and displaying CD3scfv only or CD3scfv+CD80+CD58. [0103] FIG.9A is a schematic that shows an illustrative fusion protein comprising a CD58 Oa][KMOUU^UK[ [OQSXW KWN h(9:. \M<_ P^\ON ]X ]RO C(]O[VSW^\ XP K 9:3+ _SK K USWTO[) IRO construct is termed “498.” [0104] FIG. 9B is a schematic that shows an illustrative fusion protein comprising a CD58 extracellular region fused to the N-terminus of a CD80 via a linker. The construct is termed e/00)f h(9:. \M<_ S\ OaY[O\\ON K\ K \OYK[K]O YXUbYOY]SNO SW ]RO Y[XN^MO[ MOUU\) [0105] FIG. 10 shows staining of Cocal in CD8+ T cells generated with lentiviral particles NS\YUKbSWQ h(9:. \M<_' 9:3+' KWN 9:03 `RSMR `O[O OaY[O\\ON Lb ]RO UOW]S_S[KU YK[]SMUO Y[XN^MO[ MOUU\ K\ \OYK[K]O YXUbYOY]SNO\ $eHOYK[K]Of%6 UOW]S_S[KU YK[]SMUO\ NS\YUKbSWQ h(9:. scFv, CD80, and CD58 which were expressed by the lentiviral particle producer cells as P^\SXW YXUbYOY]SNO MXVY[S\SWQ 9:03 P^\ON ]X 9:3+' `S]R h(9:. \M<_ OaY[O\\ON K\ K separate polypeptide (“455”); and lentiviral particles displaying a fusion protein comparing 9:03' h(9:. \M<_' KWN 9:3+ $e/43f%' X[ MXW][XU `S]RX^] UOW]S_S[KU YK[]SMUO\ $eBD? +f%) [0106] FIG. 11A shows the percent CD25(+) CD4+ T cells after incubation with lentiviral particles, labelled as in FIG. 10. Attorney Ref. No.061479-506001WO [0107] FIG. 11B shows the percent of CD25(+) CD8+ T cells after incubation with lentiviral particles, labelled as in FIG. 10. [0108] FIG. 11C shows the geometric mean fluorescent intensity (gMFI) CD25(+) CD4+ T cells after incubation with lentiviral particles, labelled as in FIG. 10. [0109] FIG. 11D shows the geometric mean fluorescent intensity (gMFI) of CD25(+) CD8+ T cells after incubation with lentiviral particles, labelled as in FIG.10. [0110] FIGs.12A-12C show production of cytokines 3 days after incubation with lentiviral YK[]SMUO\' UKLOUUON K\ SW <?=) ,+) ?<C(l $FIG. 12A), IL-2 (FIG. 12B%' KWN IC<(h $FIG. 12C) levels were measured. Particles comprised an anti-CD19-FRB-RACR payload. FRB =FKBP-rapamycin complex binding domain; RACR = rapamycin-activated cell-surface receptor. [0111] FIGs. 13A-13D show “#498” displaying particles generate CAR+ T cells with a larger proportion of memory-like CD4+ (FIG. 13A and FIG. 13B) or memory-like CD8+ (FIG. 13C and FIG. 13D) CAR T cells compared to “#455” or “Separate” displaying particles. [0112] FIG. 14A is a schematic that shows an illustrative experimental timeline. [0113] FIG. 14B shows the percent CD25(+) CD3+ T cells in blood after incubation with lentiviral particles, labelled as in FIG.10. [0114] FIG. 14C shows the percent CD71(+) CD3+ T cells in blood after incubation with lentiviral particles, labelled as in FIG.10. [0115] FIG. 14D \RX`\ UO_OU XP ?<C(l Mb]XTSWO VOK\^[ON / NKb\ KP]O[ SWM^LK]SXW `S]R lentiviral particles, labelled as in FIG.10. [0116] FIG. 15A-15C is a panel of graphs showing geometric mean fluorescent intensity (gMFI) of Cocal in cells generated with lentiviral particles via extracorporeal in vivo incubation. Cells were stained prior to incubation with lentiviral particles “pre-particle”, following lentiviral particle incubation with cell but before washing (“particle, pre-wash”), or following lentiviral particle incubation with cell and after washing (“Final”). Lentiviral particles display CD58 and CD80 expressed as a fusion polypeptide are labeled as in FIG. 10. Lentiviral particle incubation with CD4+ T cells, CD8+ T cells, NK T cells, NK cells, CD56+ NK cells, monocytes, B cells, and other mean fluorescent intensity (MFI) was evaluated. Attorney Ref. No.061479-506001WO [0117] FIG. 16A shows CAR+ T cells in the blood of mice injected with PBMCs from Donor 1 either after Lupagen™ wash or after incubation with lentiviral particles labeled as in FIG.10. [0118] FIG. 16B shows CAR+ T cells in the blood of mice injected with PBMCs from Donor 2 either after Lupagen™ wash or after incubation with lentiviral particles labeled as in FIG.10. [0119] FIG. 16C shows total tumor burden (Total flux) over the course of 21 days of the study in the blood of mice injected with PBMCs from Donor 1, either after Lupagen™ wash or after incubation labeled as in FIG. 10. [0120] FIG. 16D shows total tumor burden (Total flux) over the course of 21 days of the study in the blood of mice injected with PBMCs from Donor 2 either after Lupagen™ wash or after incubation labeled as in FIG. 10. [0121] FIG. 16E shows Bioluminescence imaging using the IVIS™ spectrum system depicting total tumor burden quantitated in FIG.16C and FIG.16D. [0122] FIGs. 17A-17B show expression of CD25 in CD4+ (FIG. 17A) cells transduced with lentiviral particles produced using the indicated surface plasmids encoding variations of CD58 and CD80 fusion polypeptide expression or expression of CD25 in CD8+ cells (FIG. 17B). [0123] FIG.17C shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured labeled as in FIG. 10. T cell early activation measured by CD25 expression level on Day 3 post PBMC culturing was analyzed by flow cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 2 and 5. [0124] FIGs. 18A-18B show CAR expression in CD4+ (FIG. 18A) cells transduced with lentiviral particles produced using the indicated surface plasmids encoding variations of CD58 and CD80 fusion polypeptide expression or expression of CD25 in CD8+ cells (FIG. 18B). [0125] FIG.18C shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured with lentiviral particles displaying variations of CD58 and CD80 fusion polypeptide. CAR expression as measured by FMC63 expression level on Day 7 post PBMC culturing was analyzed by flow cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 2 and 5. Attorney Ref. No.061479-506001WO [0126] FIGs. 19A-19D show the effects of indicated lentiviral surface proteins on FMC63 CAR-T induced cytotoxicity in the presence of NucLight™ Red-labeled Nalm6 target cells expressing hCD19 antigen. The IncuCyte™ kinetic killing curves for each CAR-T variant transduced using lentivirus displaying variations of CD58 and CD80 fusion polypeptides at MOI 2. FIG. 19A shows a CAR-T to target cell ratio of 0.25:1. FIG. 19B shows killing curves when CAR-T to target cell ratio is 0.5:1. FIG.19C shows killing curves when CAR- T to target cell ratio is 1:1. FIG.19D shows target-cell lytic capabilities of CAR-T cells by integrating the area under the normalized target cell killing curves (AUC) when CAR-T cells to target cell ratio range from 0.25 to 4. Percent (%) Antigen specific, CAR mediated Killing = (1-(AUC/AUC Mock ))*100. [0127] FIG. 20 S\ K LK[ Q[KYR \RX`SWQ ]K[QO](NOYOWNOW] ?<C(l' ?A(-' KWN IC<h \OM[O]SXW in FMC63 CAR-T cells, which were transduced lentiviral particles displaying indicated variations of CD58 and CD80 fusion polypeptides at MOI 2. The transduced T cells were co-cultured with Nalm6 target cells at CAR-T cell to target cell ratio of 1:1. [0128] FIGs. 21A-21B show CD25 expression in CD4+ (FIG. 21A) cells transduced with lentiviral particles produced using the indicated surface plasmids encoding variations of CD58 and CD80 fusion polypeptide expression or expression of CD25 in CD8+ cells (FIG. 21B). [0129] FIG.21C shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured with lentiviral particles displaying variations of CD58 and CD80 fusion polypeptide. T cell early activation measured by CD25 expression level on Day 3 post PBMC culturing was analyzed by flow cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 0.5 and 1. [0130] FIGs. 22A-22B show CAR expression in CD4+ (FIG. 22A) cells transduced with lentiviral particles produced using the indicated surface plasmids encoding variations of CD58 and CD80 fusion polypeptide expression or expression of CD25 in CD8+ cells (FIG. 22B). [0131] FIG.22C shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured with lentiviral particles displaying variations of CD58 and CD80 fusion polypeptide. CAR expression as measured by FMC63 expression level on Day 7 post PBMC culturing was analyzed by flow Attorney Ref. No.061479-506001WO cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 0.5 and 1. [0132] FIG. 23 shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured with lentiviral particles displaying variations of CD58, CD80, and anti-CD3 scFv fusion polypeptides. T cell early activation measured by CD25 expression level on Day 3 post PBMC culturing was analyzed by flow cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 1 and 10. [0133] FIG. 24 shows a bar graph of the effects of lentivirus particles produced using the indicated surface plasmids. Non-stimulated human PBMCs were cultured with lentiviral particles displaying variations of CD58, CD80, and anti-CD3 scFv fusion polypeptides. CAR expression as measured by FMC63 expression level on Day 7 post PBMC culturing was analyzed by flow cytometry. NTC (non-transduced cells) are included for comparison. Lentiviral particles were added at multiplicity of infection (MOI) 1 and 10. [0134] FIG. 25 shows a graph of CAR+ T cell expansion over 11 days post transduction. The CAR+ T cells were transduced with lentiviral particles displaying variations of CD58, CD80, and anti-CD3 scFv fusion polypeptides. On Day 3 post transduction, PBMCs are washed to remove lentiviral particles, and seeded in fresh culture media at 0.5E6 cells per well. CAR+ cells were determined by staining for surface expression of anti-FMC63 scFv, and analyzed by flow cytometry. [0135] FIG. 26 is a schematic of the fusion polypeptide screening approach depicted in FIGs.17-25. [0136] FIG. 27A shows diagrams of illustrative fusion proteins. [0137] FIG. 27B shows diagrams of illustrative fusion proteins. The 21aa linker may have the polypeptide sequence GSSGGSGGGGSGGGGSGGGGS (SEQ ID NO: 34). The 23aa linker may have the polypeptide sequence GSSGGSGGGGSGGGGSGGGGSSG (SEQ ID NO: 35). [0138] FIG. 28A shows a study design and timeline. [0139] FIG. 28B is a graph showing staining of Cocal on CD3+ T cells incubated with engineered particles displaying CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide. [0140] FIG.28C is a graph showing staining of Cocal on engineered particle bound T cells The left peak shows CD3- T cells and the right peak shows CD3+ T cells. The engineered particles display a CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide. Attorney Ref. No.061479-506001WO [0141] FIG.28D shows CD25 expression in CD8+ T cells on day 3 after transduction with lentiviral particles displaying CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide “Engineered particle”. [0142] FIG. 28E shows CAR expression in CD8+ T cells on day 7 after transduction with lentiviral particles displaying CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide “Engineered particle”. [0143] FIG.29 shows the number of Nalm6 cells after anti-CD19 CAR+ T cells were serial- stimulated with Nalm6 tumor cells every 2-3 days. anti-CD19 CAR+ T cells were generated with lentiviral particles encoding an anti-CD19 CAR transgene and displaying CD3scfv- CD80-CD58 tri-fusion polypeptide particles “Engineered particle”. Arrows denote stimulation with Nalm6 tumor cells. Error bars denote mean±SEM. [0144] FIG.30A shows the study design and timeline. FIG.30B shows the number of cells expressing activation marker CD25 in circulation four days after transduction with lentiviral particles displaying CD3scfv-CD80-CD58- tri-fusion polypeptide. FIG. 30C shows the number of cells expressing activation marker CD71 in circulation four days after transduction with lentiviral particles displaying CD3scfv-CD80-CD58 tri-fusion polypeptide. FIG. 30D \RX`\ Y[XN^M]SXW XP ?<C(l / NKb\ KP]O[ SWM^LK]SXW `S]R YK[]SMUO\ displaying CD3scfv-CD80-CD58 tri-fusion polypeptide “Engineered particle”. FIG. 30E shows the number of T cells expressing an anti-CD19 CAR in the blood 11 days after transduction with lentiviral particles displaying CD3scfv-CD80-CD58- tri-fusion polypeptide at a lentiviral dose of 10 Million or 50 Million transducing units (TU). FIG. 30F shows the tumor burden in NSG MHCI/II KO mice after administration of lentiviral particles displaying CD3scfv-CD80-CD58 tri-fusion polypeptide at a lentiviral dose of 10 Million or 50 Million transducing units (TU). [0145] FIG. 31A shows the study design and timeline. FIG. 31B shows the number of T cells from Donor 1 and Donor 2 expressing an anti-CD19 CAR in the blood 14 days after extracorporeal incubation with lentiviral particles. FIG. 31C shows the tumor burden in Donor 1 and Donor 2 NSG MHCI/II KO mice after administration of T cells produced by via extracorporeal incubation with lentiviral particles. N=7 animals; error bars denote mean±SEM. FIG. 31D shows the study design and timeline for re-challenge study. FIG. 31E shows the tumor burden in NSG MHCI/II KO mice after administration of T cells produced via extracorporeal incubation of PBMCs from Donor 1 or Donor 2 incubated with Attorney Ref. No.061479-506001WO lentiviral particles following tumor cell re-challenge at Day 49. Error bars denote mean±SEM. [0146] FIG. 32A shows the study design and timeline. FIG. 32B shows %CAR+ T cells (left panel) and total CAR+ T cells (right panel) in the blood of mice injected with PBMCs P[XV :XWX[ , KP]O[ A^YKQOWd SWM^LK]SXW `S]R UOW]S_S[KU YK[]SMUO\ NS\YUKbSWQ h(9:. \M<_' CD80, and CD58 which were expressed by the lentiviral particle producer cells as a bi- P^\SXW YXUbYOY]SNO MXVY[S\SWQ 9:03 P^\ON ]X 9:3+' `S]R h(9:. \M<_ OaY[O\\ON K\ K separate polypeptide (“#455”); and lentiviral particles displaying a tri-fusion protein MXVYK[SWQ 9:03' h(9:. \M<_' KWN 9:3+ $e"/43f%' X[ MXW][XU E8B9\ WX] SWM^LK]ON `S]R lentiviral particles. FIG. 32C shows %CAR+ T cells (left panel) and total CAR+ T cells (right panel) in the blood of mice injected with PBMCs from Donor 2 after Lupagen™ SWM^LK]SXW `S]R UOW]S_S[KU YK[]SMUO\ NS\YUKbSWQ h(9:. \M<_' 9:3+' KWN 9:03 `RSMR `O[O expressed by the lentiviral particle producer cells as a bi-fusion polypeptide comprising 9:03 P^\ON ]X 9:3+' `S]R h(9:. \M<_ OaY[O\\ON K\ K \OYK[K]O YXUbYOY]SNO$e"/00f%6 KWN UOW]S_S[KU YK[]SMUO\ NS\YUKbSWQ K ][S(P^\SXW Y[X]OSW MXVYK[SWQ 9:03' h(9:. \M<_' KWN CD80(“#498”), or control PBMCs not incubated with lentiviral particles. FIG. 32D shows Bioluminescence imaging using the IVIS™ spectrum system depicting total tumor burden. Images show mice injected with PBMCs from Donor 1 after Lupagen™ incubation with lentiviral particles displaying “#455” a dual fusion or “#498” a triple fusion (left panel), and mice injected with PBMCs from Donor 2 after Lupagen™ incubation with lentiviral particles displaying “#455” a dual fusion or “#498” a triple fusion (right panel). FIGs.32E- 32G show total tumor burden (Total flux) over the course of 45 days of the study in the blood of mice injected with PBMCs from Donor 1 (top row of panels) or Donor 2 (bottom row of panels) after Lupagen™ incubation with untreated PBMCs (FIG. 32E), lentiviral particles displaying “#455” a dual fusion (FIGs. 32F), or “#498” a triple fusion (FIGs. 32G). FIGs. 32H-32I show total tumor burden (Total flux) over the course of 28 days of the study in the blood of mice injected with PBMCs from Donor 1 (FIG. 32H) or Donor 2 (FIG. 32I) after Lupagen™ incubation with untreated PBMC control, lentiviral particles displaying “#455”, or “#498” a dual fusion or a triple fusion polypeptide, respectively. [0147] FIG. 33A shows the study design and timeline for re-challenge study. FIG. 33B shows the tumor burden in NSG MHCI/II KO mice after administration of T cells produced via extracorporeal incubation of PBMCs from Donor 1 (D1) or Donor 2 (D2) incubated with lentiviral particles displaying “#455” a dual fusion construct, or “#498” a triple fusion Attorney Ref. No.061479-506001WO construct following tumor cell re-challenge at Day 49. FIG. 33C shows Bioluminescence imaging using the IVIS™ spectrum system depicting total tumor burden. Images show mice injected with PBMCs from Donor 1 after Lupagen™ incubation with lentiviral particles displaying “#455” a dual fusion or “#498” a triple fusion (left panel), and mice injected with PBMCs from Donor 2 after Lupagen™ incubation with lentiviral particles displaying “#455” a dual fusion or “#498” a triple fusion (right panel) following tumor cell rechallenge at Day 49.FIG. 34A shows the study design. [0148] FIG. 34B is a graph showing staining of Cocal on CD3+ T cells incubated with no vector control (left panel), engineered particles displaying CD58, CD80, and anti-CD3 scFv individually expressed polypeptides (middle panel), and engineered particles displaying a CD58, CD80, and anti-CD3 scFv multi-domain fusion (MDF) polypeptide (right panel). [0149] FIG. 34C is a graph showing %Cocal on B cells, CD4+ T cells, CD8+ T cells, monocytes, and NK cells incubated with viral particles displaying individually expressed CD3scfv+CD80+CD58 (left panel) and viral particles displaying CD58, CD80, and anti- CD3 scFv multi-domain fusion (MDF) (right panel). [0150] FIG. 34D is a graph showing geometric mean fluorescence intensity (gMFI) Cocal on B cells, CD4+ T cells, CD8+ T cells, monocytes, and NK cells incubated with viral particles displaying individually expressed CD3scfv+CD80+CD58 (left panel) and viral particles displaying CD58, CD80, and anti-CD3 scFv multi-domain fusion (MDF) (right panel). [0151] FIG. 34E shows the study timeline. [0152] FIG. 34F shows activation of CD3+ T cells as measured by % CD25+ cells 3 days after transductions with a viral particle displaying CD58, CD80, and anti-CD3 scFv multi- domain fusion (MDF). [0153] FIG. 34G shows the level of CD3+ T cells transduction as measured by %CAR expression 7 days after transduction with a viral particle displaying CD58, CD80, and anti- CD3 scFv multi-domain fusion (MDF). [0154] . FIG. 34H is a flow cytometry staining showing CAR expression in CD3+ T cells on day 7 after 1 hour of contact with viral particles displaying CD58, CD80, and anti-CD3 scFv multi domain fusion polypeptide. [0155] FIG. 34I is a graph showing the percent of CAR positive T cells (left panel) and viral copy number (VCN) (right panel) on day 7 after 1 hour of contact with viral particles Attorney Ref. No.061479-506001WO displaying CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide multi domain fusion polypeptide (left panel). [0156] FIG.35A is a diagram of an illustrative surface engineered viral particle displaying a CD58, CD80, dual-fusion polypeptide and an anti-CD3 scFv that binds NHP CD3 and a payload comprising a human-specific anti-CD20 CAR. [0157] FIG. 35B is a diagram of an illustrative payload comprising a human-specific anti- CD20 CAR.The payload comprises: a. an MND promoter, b. an anti-CD20-based CAR comprising: i. Leu16 scFv – murine anti-human CD20 scFv (cross-reactive with NHP) ii. CD8a hinge and transmembrane domain iii. /(,88 MX(\]SV^UK]X[b NXVKSW KWN 9:.m NXVKSW c. low-affinity nerve growth factor receptor (LNGFR) molecule as a marker of transduction. [0158] FIG. 35C is a summary chart of the animals treated in an illustrative non-human primate study. Three animals were studied. * denotes at start of study. [0159] FIG. 36 is a diagram of an illustrative study design and timeline for an NHP study. The engineered viral particles were injected into auxiliary lymph nodes (LN) of Animal #1 due to difficulty injecting inguinal lymph nodes (LN) due to the small size of the animal. The engineered viral particles were injected into inguinal lymph nodes in Animal #2 and Animal #3. [0160] FIG. 37 is a graph showing the fraction of starting CD20+ cells in Animal #1, Animal #2, and Animal #3 over the course of the study to Day 56. [0161] FIG. 38 is a panel of graphs showing serum levels of IL-6, ferritin, and C-reactive protein (CRP) in Animal #1, Animal #2, and Animal #3 over the course of the study to Day 56 and body temperature to Day 28. [0162] FIG.39A is a diagram of an illustrative surface engineered viral particle displaying a CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide comprising an anti-CD3 scFv that binds NHP CD3 and a payload comprising a human-specific anti-CD20 CAR. [0163] FIG. 39B is a diagram of an illustrative payload comprising a human-specific anti- CD20 CAR. The payload comprises: a. an MND promoter, Attorney Ref. No.061479-506001WO b. an anti-CD20-based CAR comprising: i. Leu16 scFv – murine anti-human CD20 scFv (cross-reactive with NHP) ii. FLAG tag iii. CD8a hinge and transmembrane domain iv. /(,88 MX(\]SV^UK]X[b NXVKSW KWN 9:.m NXVKSW) [0164] FIG. 40 is a panel of flow cytometry staining graphs showing CAR expression in CD3+ T cells after 8 different donor PBMCs were transduced with viral particles displaying Human CD58-NHP-specific anti-CD3 scFv-Human CD80 multi-domain fusion (MDF) polypeptide at MOI=0.2. [0165] FIG.41A is a diagram of an illustrative study design and timeline for an NHP study. [0166] FIG. 41B is a summary chart of the animals treated in an illustrative non-human primate study. Four animals were studied. [0167] FIG. 42 is a panel of flow cytometry staining graphs showing anti-CD20 CAR expression in CD3+ T cells through to Day 111 of the study in Animal #1. [0168] FIG. 43 is a panel of flow cytometry staining graphs showing anti-CD20 CAR expression and CD25 expression in T cells through to Day 51 of the study in Animal #1. [0169] FIG.44 is a panel of flow cytometry staining graphs showing CD20+ cells (B cells) and CD3+ T cells through to Day 111 of the study in Animal #1. [0170] FIG.45 is a graph showing CD20+ B cells and CD3+ CAR+ T cells through to Day 111 of the study in Animal #1. [0171] FIG. 46A is a timeline of observed clinical symptoms over the course of the study. [0172] FIG.46B is a panel of graphs showing serum levels of IL-6, ferritin, and C-reactive protein (CRP) in Animal #1 over the course of the study to Day 56. The line depicting Anti- CD20 CAR T cells (CD3+ FLAG+) is identical in the 3 plots. [0173] FIG. 47 is a panel of flow cytometry staining graphs showing CD20+ cells (B cells – top panels) and CAR+ (FLAG+) CD3+ T cells (bottom panels) through to Day 56 of the study in Animal #2. [0174] FIG. 48 is a panel of flow cytometry staining graphs showing CD20+ cells (B cells – top panels) and CAR+ (FLAG+) CD3+ T cells (bottom panels) through to Day 37 of the study in Animal #3. Attorney Ref. No.061479-506001WO [0175] FIG. 49 is a panel of flow cytometry staining graphs showing CD20+ cells (B cells – top panels) and CAR+ (FLAG+) CD3+ T cells (bottom panels) through to Day 21 of the study in Animal #4. [0176] FIG. 50A-50J include examples of CD58 and CD80 dual fusion sequences. [0177] FIG. 51A-51F include examples of CD58, CD80 and CD3 scFV triple fusion sequences. [0178] FIG. 52A-52B illustrate details of assessment of engineered particle’s in vivo biodistribution. [0179] FIG. 53A-53B include plots showing vector DNA copies / µg genomic DNA (gDNA) in an experiment herein. [0180] [0181] FIG.54 includes a plot showing vector DNA copies / µg genomic DNA (gDNA) in an experiment herein. [0182] FIG. 55A-55B include data comparing the function of particles comprising two variations of fusion proteins (“V1” and “V2”) against control particles comprising a glycoprotein only. [0183] FIG. 56 depicts studies comparing the function of engineered lentiviral particles comprising an anti-CD3 scFv and cocal glycoprotein (“anti-CD3scFv”) with engineered lentiviral particles comprising an anti-CD3scFv, a CD58 protein, and a CD80 protein in addition to a cocal glycoprotein (“Tri protein”). FIG. 56A includes plots illustrating comparative activation data showing the dose-dependent activation of CD4 and CD8 T cells in response to incubation with each particle type. FIG. 56B includes plots illustrating comparative particle-T cell binding data. FIG. 56C includes plots illustrating comparative transduction data showing the dose-dependent transduction efficiency and total number of transduced CD4 and CD8 T cells after incubation with each particle type. FIG.56D includes plots illustrating comparative cytokine production data showing dose-dependent stimulation XP ?<C(b' ?A(-' KWN IC<(h KP]O[ SWM^LK]SXW `S]R OKMR YK[]SMUO ]bYO) FIG. 56E includes a plot illustrating comparative serial stimulation data. FIG.56F includes plots demonstrating that cells incubated with particles comprising an anti-CD3scFv, a CD58 protein, and a CD80 protein (“Tri protein”) produced more inflammatory cytokines than cells incubated with particles comprising an anti-CD3 scFv (“anti-CD3scFv”), but no costimulatory or adhesion molecules. FIG. 56G includes plots demonstrating that particles comprising an anti- CD3scFv, a CD58 protein, and a CD80 protein were able to generate a higher proportion of Attorney Ref. No.061479-506001WO CCR7+ and CD27+ CD4 and CD8 T cells compared with particles comprising an anti-CD3 scFv, but no costimulatory or adhesion molecules. [0184] FIG. 57 describes an in vivo mouse study to evaluate the function of particles comprising an anti-CD3 scFv, but no costimulatory or adhesion molecules and particles comprising an anti-CD3scFv, a CD58 protein, and a CD80 protein. FIG. 57A depicts the study design. FIG. 57B includes plots illustrating in vivo activation data at various dose levels of particles. FIG. 57C includes plots illustrating in vivo transduction of T cells at varying dose levels of particles. FIG. 57D includes plots demonstrating tumor growth and control across the study and in particular showing that the tri protein particles controlled tumor growth to a greater extent than the anti-CD3 scFv particles. [0185] FIG.58 includes data comparing engineered particles comprising CD58, CD80, and an anti-CD3 scFv separately expressed with engineered particles comprising a fusion protein comprising CD58, anti-CD3 scFv, and CD80 expressed together. FIG.58A includes plots illustrating particle-T cell binding data. FIG. 58B includes plots illustrating comparative activation data across varying MOIs. FIG. 58C includes plots illustrating transduction data across varying MOIs. FIG. 58D includes plots demonstrating cytokine production by cells following incubation with the two variations of engineered particles. [0186] FIG. 59 describes an in vivo mouse study to evaluate the function of particles comprising CD58, CD80, and an anti-CD3 scFv separately expressed with engineered particles comprising a fusion protein comprising CD58, anti-CD3 scFv, and CD80 expressed together. FIG. 59A includes plots illustrating in vivo activation data at various dose levels of particles. FIG. 59B includes plots illustrating in vivo transduction of T cells at varying dose levels of particles. FIG. 59C includes plots demonstrating tumor growth and control across the study and in particular showing that the fusion protein particles controlled tumor growth to a greater extent than the particles comprising CD58, CD80, and an anti-CD3 scFv separately expressed. [0187] FIG. 60A includes diagrams illustrating example fusion molecules. [0188] FIG. 60B includes diagrams illustrating example fusion molecules. DETAILED DESCRIPTION [0189] The present disclosure relates generally to particles and fusion molecules for use in transduction of target cells, such as immune cells, or specifically T cells. In one aspect, the disclosure provides, a particle for transduction of target cells, comprising, displayed on the Attorney Ref. No.061479-506001WO surface of the particle, a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule, an activation molecule, or both. [0190] The term “transduction” is used in its broadest sense to mean delivery of an agent to a cell, such as a therapeutic agent. The agent may be a small molecule, polynucleotide, or polypeptide. A combination of agents may be delivered, such as several polynucleotides or a protein-nucleic acid complex (e.g., a gene-editing nuclease in complex with guide nucleic acid). The term “particle” includes but is not limited to viral particles (i.e., a virion), lipid nanoparticles (LNPs), lipoplexes, liposomes, and nanocarriers. [0191] The fusion molecules of the disclosure combine an adhesion molecule with a costimulatory molecule, an activation molecule, or both. Without being bound by theory, it is believed that the inclusion of two or more of these types of molecule in a fusion molecule may cause such a particle, when it encounters a target cell, to form a macromolecular complex at the interface of the particle and cell that acts as artificial supramolecular activation cluster (SMAC). [0192] T cells that encounter an antigen-presenting cell (APC) form an immune synapse known as a SMAC. In natural SMACs, the APC presents an antigen in complex with a major histocompatibility complex (MHC) molecule to the T cell receptor (TCR) on a T cell; CD80 or CD86 interact with CD28 to provide a costimulatory signal; and CD58 interacts with CD2 to adhere the APC to the T cell. The interactions between CD58 and CD2 may also provide an activatory or costimulatory signal. The adhesion molecule displayed on a particle may be CD58, a ligand for LFA-1 (ICAM-1, ICAM-2, ICAM-3, ICAM-4, ICAM-5, or JAM-A), or a ligand for DNAM-1 (CD155 or CD112), which may bind cognate T cell molecules such as CD2, LFA-1, and DNAM-1. SMACs may further present costimulatory molecules. Costimulatory molecules that may be displayed on a particle include CD80 and CD86, CD40L (also known as CD154), GITRL, OX40L, 41BBL, ICOSL, CD27, CD30L, LIGHT, LTalpha, MICA, and MICB. [0193] As contemplated by the present disclosure, a particle may be engineered to display on its surface any of the foregoing adhesion molecules or costimulatory molecules; extracellular fragments thereof; or functional fragments thereof. Extracellular portions of these molecules may be identified in databases such as UniProt, which is available at www.uniprot.org, or may be predicted using methods, such as a method implemented by the TMHMM 2.0 program available at services.healthtech.dtu.dk. Furthermore, in some cases, functional fragments of each are identified in scientific literature or they may be Attorney Ref. No.061479-506001WO identified using laboratory methods. For example, one may predict the identify fragments of a protein likely to form well-folded domains. Fragments may be tested in binding assays against a cognate molecule, or used in pull-down assays compared to the full molecule. Functional assays, such as expression of a fluorescence reporter under the control of a promoter activated by T-cell signaling (e.g., the NKkB promoter) when a T cell is contacted with a cell or particle expressing a putative functional fragment. The sequence of the adhesion molecule, costimulatory molecule, or activation molecule may be varied to identify and use variants that retain function. For example, conservative mutations may be made to a molecule or a molecule may be randomly mutated with the function of the variant confirmed experimentally. [0194] In each case, a molecule may be displayed as a full-length form, including its native transmembrane portion. Alternatively, the extracellular portion of the molecule may be displayed with a heterologous transmembrane portion (e.g., a transmembrane portion from another membrane protein) or a membrane anchor (e.g., a glycosylphosphatidylinisotol anchor). Each fusion molecule may be associated with the particle, when the particle comprises a lipid envelope, directly or via the transmembrane portion or anchor connected to one of the other molecules in the fusion molecule. For example, an adhesion molecule, costimulatory molecule, and activation molecule may be linked in any order with only the most N-terminal or C-terminal of the molecules connected to a transmembrane region or anchor. In a variant, the fusion molecule comprises or is associated with another membrane- associated molecule, thereby displaying the fusion molecule on the particle. The term “display” is used, in a broad sense, to me position on the surface of the particle such that the molecule may contact cognate molecules on the target cell. It is further contemplated that for particles lacking an envelope (such as a non-enveloped viral particle) the fusion molecule may be displayed on the particle either by association with a component of the particle (e.g., a capsid protein) or by a direct linkage with the particle (e.g., as a fusion protein comprises a capsid protein). Adhesion Molecules [0195] Disclosed herein, in some embodiments, are adhesion molecules. The adhesion molecule may be included as part of a fusion molecule. The adhesion molecule may be included as part of a particle (e.g. on a particle surface). [0196] As used herein, the term “adhesion molecule” refers, in a broad sense, to a molecular component of a SMAC or other immune synapse, other than an activation molecule (e.g. Attorney Ref. No.061479-506001WO TCR-binding agent) or a costimulatory molecule, which in contributes to adhesion of a particle to target cells. Adhesion molecules from natural sources may be molecules expressed, natively, on antigen-presenting cells and adapted for use here on particles. Both naturally occurring adhesion molecules, and their variants, and artificial adhesion molecules, such as antibodies, or fragments thereof, are contemplated. An adhesion molecule, as the term is used herein, specifically binds a conjugate molecule with affinity sufficient to cause increased adhesion between the particle and the target cell compared to the adhesion of a reference particle lacking the adhesion molecule to the same or similar target cell. The term adhesion molecule includes but is not limited to CD58, a CD58 extracellular portion, and functional fragments of CD58. As described above, the term “functional fragment” is used herein to a fragment of a polypeptide, or other molecule, that retains the desired function of the polypeptide. For example, a functional fragment of CD58 is a fragment of CD58 that specifically binds CD2. The adhesion molecule may be a protein, termed herein an “adhesion protein.” [0197] In some embodiments, the costimulatory and/or adhesion molecule comprises an amino acid sequence 100% identical to a sequence in Table 1A or Table 1B. In some embodiments, the costimulatory and/or adhesion molecule shares at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to a sequence in Table 1A or Table 1B. In some embodiments, the costimulatory and/or adhesion molecule shares less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 96%, less than 97%, less than 98%, less than 99%, or less than 100% identity to a sequence in Table 1A or Table 1B. [0198] Polypeptide sequences of illustrative adhesion molecules are provided in Table 1A, with the “start” and “end” positions of the extracellular portion of each. In each case, the adhesion molecule may comprise a polypeptide at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to any sequence in Table 1A, or functional fragments thereof. Functional fragments may be or include any 10, 20, 30, 40, 50, 75, 100, 200, 300, 400, 500, or 600 (or any range thereof) amino acid portion that retains binding affinity to its cognate molecule, when measured using affinity assays such as biolayer interferometry or other assays that may be known in the art. Attorney Ref. No.061479-506001WO Table 1A Attorney Ref. No.061479-506001WO Table 1B Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO [0199] In some embodiments, the costimulatory and/or adhesion molecule is linked to a transmembrane domain. In some embodiments, the transmembrane domain may be the transmembrane domain of CD8, an alpha, beta or zeta chain of a T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1 BB (CD137), 4-1 BBL, GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRFI), CD160, CD19, IL-2R beta, IL-2R gamma, IL-7R a, ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRT AM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and/or NKG2C. In some embodiments, the transmembrane domain may be the transmembrane domain of CD28. In some embodiments, the transmembrane domain of may LO ]RO ][KW\VOVL[KWO NXVKSW XP 9:3' PX[ OaKVYUO' 9:3h) [0200] Without wishing to be bound by theory, reducing foreign junctions (i.e., between an adhesion molecule and a transmembrane domain) in the foreign nucleic acid incorporated into a lentiviral particle may reduce the immunogenicity of a subject to a lentiviral particle. In some embodiments, an engineered lentiviral particle displaying a multi-domain fusion polypeptide will comprise a transmembrane domain from the polypeptide domain which is membrane proximal. [0201] In some embodiments, the adhesion molecule is CD58. CD58 is also known as lymphocyte function-associated antigen 3 (LFA-3). CD58 binds to CD2 (LFA-2) on T cells. The extracellular portion of CD58 is residues 29-215 of SEQ ID NO: 1 (SEQ ID NO: 10): FSQQIYGVVYGNVTFHVPSNVPLKEVLWKKQKDKVAELENSEFRAFSSFKNRVY LDTVSGSLTIYNLTSSDEDEYEMESPNITDTMKFFLYVLESLPSPTLTCALTNGSIEV QCMIPEHYNSHRGLIMYSWDCPMEQCKRNSTSIYFKMENDLPQKIQCTLSNPLFN TTSSIILTTCIPSSGHSRHR (SEQ ID NO: 10) Attorney Ref. No.061479-506001WO [0202] In some embodiments, the polypeptide sequence of CD58 shares at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO: 248: FSQQIYGVVYGNVTFHVPSNVPLKEVLWKKQKDKVAELENSEFRAFSSFKNRVY LDTVSGSLTIYNLTSSDEDEYEMESPNITDTMKFFLYVLESL (SEQ ID NO: 248) [0203] A crystal structure of CD58 is described in Ikemizu et al. PNAS USA 96(8):4289-94 (1999). The extracellular portion of CD58 has a ligand-binding domain and a second extracellular domain. In embodiments, the ligand-binding domain may be used as the functional fragment of CD58—i.e., without the second extracellular domain. [0204] In some embodiments, the adhesion molecule (or the fusion protein) comprises the polypeptide sequence of SEQ ID NO: 1 or 10, or a sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO: 1 or 10. In some embodiments, the adhesion molecule (or the fusion protein) comprises a sequence having less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 96%, less than 97%, less than 98%, less than 99%, or less than 100% identity to SEQ ID NO: 1 or 10. The adhesion molecule may encoded by a polynucleotide (e.g. a DNA or RNA polynucleotide). [0205] The adhesion molecule may encoded by the polynucleotide sequence of CD58, SEQ ID NO: 11, or by a subsequence encoding the extracellular portion or a functional fragment. SEQ ID NO: 11 (5’ to 3’): ATGGTTGCTGGGAGCGACGCGGGGCGGGCCCTGGGGGTCCTCAGCGTGGTCT GCCTGCTGCACTGCTTTGGTTTCATCAGCTGTTTTTCCCAACAAATATATGGTG TTGTGTATGGGAATGTAACTTTCCATGTACCAAGCAATGTGCCTTTAAAAGAG GTCCTATGGAAAAAACAAAAGGATAAAGTTGCAGAACTGGAAAATTCTGAGT TCAGAGCTTTCTCATCTTTTAAAAATAGGGTTTATTTAGACACTGTGTCAGGTA GCCTCACTATCTACAACTTAACATCATCAGATGAAGATGAGTATGAAATGGAA TCGCCAAATATTACTGATACCATGAAGTTCTTTCTTTATGTGCTTGAGTCTCTT CCATCTCCCACACTAACTTGTGCATTGACTAATGGAAGCATTGAAGTCCAATG CATGATACCAGAGCATTACAACAGCCATCGAGGACTTATAATGTACTCATGGG ATTGTCCTATGGAGCAATGTAAACGTAACTCAACCAGTATATATTTTAAGATG GAAAATGATCTTCCACAAAAAATACAGTGTACTCTTAGCAATCCATTATTTAA Attorney Ref. No.061479-506001WO TACAACATCATCAATCATTTTGACAACCTGTATCCCAAGCAGCGGTCATTCAA GACACAGATATGCACTTATACCCATACCATTAGCAGTAATTACAACATGTATT GTGCTGTATATGAATGGTATTCTGAAATGTGACAGAAAACCAGACAGAACCA ACTCCAAT. [0206] The polynucleotide sequence may be varied by codon-optimization or other methods to generate polynucleotide sequences having at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO: 11, or a suitable subsequence, which may be used to express the adhesion molecule. [0207] It will be appreciated that further variants of CD58 may be used. For example, homologs of CD58 from other species (mice, ape, horse, etc.) may be identified and tested for use in transducing human, or non-human, target cells. It is expected that at least some non-human homologs will retain adhesion molecule function when used with human target cells. [0208] Further adhesion molecules useful in the practice of the present invention may include any molecule that specifically binds CD2, LFA-1, or DNAM-1. For example, the adhesion molecule may be a molecule that comprises an antibody, or antigen-binding fragment thereof, specific to CD2, LFA-1, or DNAM-1. [0209] In some embodiments, the adhesion molecule binds to CD2. CD2 is also known as T11, LFA-2, and the erythrocyte rosette receptor. In its native state, CD2 is a surface protein expressed on T lymphocytes and NK cells. CD2 is a natural ligand for CD58. In addition to performing adhesion functions, engagement of CD2 by CD58 provides a costimulatory signal that may enhance activation and effector functions. In some embodiments, the particle comprises an adhesion molecule that binds to CD2, which may be CD58 or a fragment thereof. In some embodiments, the lentiviral particle comprises an antibody, single domain antibody, antibody fragment, and/or nanobody specific for CD2. [0210] The foregoing description of CD58 and its derivatives as the adhesion molecule and CD2 as the cognate molecule may be extrapolated to the other adhesion molecules described herein. The adhesion molecule (or the fusion protein) may comprise any polypeptide sequence of in Table 1, to an extracellular portion thereof, or to a functional fragment thereof, or a sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to a sequence in Table 1, to an extracellular portion thereof, or to a functional fragment thereof. Attorney Ref. No.061479-506001WO Costimulatory Molecules [0211] Disclosed herein, in some embodiments, are costimulatory molecules. The costimulatory molecule may be included as part of a fusion molecule. The costimulatory molecule may be included as part of a particle (e.g. displayed on a particle surface). [0212] The fusion molecule displayed on the particle may include a costimulatory molecule. However, in some embodiments, the fusion molecule does not include a costimulatory molecule. The particle may display a costimulatory molecule as a separate molecule on the surface of the particle, or the particle may lack any costimulatory molecule. The costimulatory molecule may be a protein, termed herein a “costimulatory protein.” [0213] As used herein, the term “costimulatory molecule” refers to a molecule capable of providing a costimulatory signal to target cells. In T cell biology, the binding of the T cell receptor by an antigen can provide the primary stimulatory signal to the cell. So-called costimulatory signals are provided by accessory molecules. An example costimulatory signal is the signal provided by binding of CD28 on T cells by a ligand. Some examples of ligands of CD28 include CD80 and CD86. [0214] Illustrative costimulatory molecules include, but are not limited to, CD80, CD86, CD40L (also known as CD154), GITRL, OX40L, 41BBL, ICOSL, CD27, CD30L, LIGHT, LTalpha, MICA, and MICB. Each of the foregoing may be employed as a costimulatory molecules as a full-length protein, an extracellular domain, or functional fragment. [0215] Polypeptide sequences of illustrative costimulatory molecules are provided in Table 2, with the “start” and “end” positions of the extracellular portion of each. In each case, the costimulatory molecule may comprise a polypeptide at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to any sequence in Table 2, or functional fragments thereof. In some embodiments, the costimulatory molecule comprises a polypeptide having less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 96%, less than 97%, less than 98%, less than 99%, or 100% sequence identity to any sequence in Table 2, or a functional fragment thereof. Functional fragments may be or include any 10, 20, 30, 40, 50, 75, 100, 200, 300, 400, 500, or 600 amino acid portion that retains binding affinity to its cognate molecule, when measured using affinity assays such as biolayer interferometry or other assays known in the art. Attorney Ref. No.061479-506001WO Table 2 Attorney Ref. No.061479-506001WO [0216] In some embodiments, the costimulatory molecule is or includes CD80. In some embodiments, the costimulatory molecule is or includes a molecule that binds CD28. CD80 binds to CD28. The extracellular portion of CD80 includes residues 35-230 of SEQ ID NO: 12, which includes an Ig-like V-type domain (SEQ ID NO: 25) and an Ig-like C2-type Attorney Ref. No.061479-506001WO domain (SEQ ID NO: 26), either or both of which may be included to form the costimulatory molecule. [0217] VIHVTKEVKEVATLSCGHNVSVEELAQTRIYWQKEKKMVLTMMSGDMNI WPEYKNRTIFDITNNLSIVILALRPSDEGTYECVVLKYEKDAFKREHLAEVT (SEQ ID NO: 25) [0218] PSISDFEIPTSNIRRIICSTSGGFPEPHLSWLENGEELNAINTTVSQDPETELYA VSSKLDFNMTTNHSFMCLIKYGHLRVNQTFN (SEQ ID NO: 26) [0219] The crystal structure of CD80 (also known as B7-1) is described in Ikemizu et al. Immunity 12:51-60 (2000). The extracellular portion of CD80 has two domains, described above. In embodiments, one or both of the domains may be used as the functional fragment of CD80. [0220] In some embodiments, the costimulatory molecule is or includes CD86. CD86 binds to CD28. The extracellular portion of CD86 includes residues 33-225 of SEQ ID NO: 13, which includes an Ig-like V-type domain (SEQ ID NO: 27) and an Ig-like C2-type domain (SEQ ID NO: 28), either or both of which may be included to form the costimulatory molecule. [0221] NETADLPCQFANSQNQSLSELVVFWQDQENLVLNEVYLGKEKFDSVHSK YMGRTSFDSDSWTLRLHNLQIKDKGLYQCIIHHKKPTGMIRIHQMNSELS (SEQ ID NO: 27) [0222] NVYINLTCSSIHGYPEPKKMSVLLRTKNSTIEYDGVMQKSQDNVTELYDVS ISLSVSFPDVTSNMTIFCILETDKT (SEQ ID NO: 28) [0223] The crystal structure of CD86 (also known as B7-1) is described in Schwartz et al. Nature 410: 604-608 (2001). The extracellular portion of CD86 has two domains, described above. In embodiments, one or both of the domains may be used as the functional fragment of CD86. [0224] It will be appreciated that further variants of CD80 or CD86 may be used. For example, homologs of CD80 or CD86 from other species (mice, ape, horse, etc.) may be identified and tested for use in transducing human, or non-human, target cells. It is expected that at least some non-human homologs will retain costimulatory molecule function when used with human target cells. [0225] In some embodiments, the costimulatory molecule (or the fusion protein) comprises the polypeptide sequence of one or more of SEQ ID NO: 12-13 and 25-28, or a sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at Attorney Ref. No.061479-506001WO least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to one or more of SEQ ID NO: 12-13 and 25-28. [0226] In some embodiments, the costimulatory molecule CD80 comprises the polypeptide sequence of SEQ ID NO: 250, or a sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identity to SEQ ID NO: 250. [0227] In some embodiments, the costimulatory molecule (or the fusion protein) comprises a polypeptide sequence having less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 96%, less than 97%, less than 98%, less than 99%, or less than 100% identity to one or more of SEQ ID NO: 12-13 and 25-28. The costimulatory molecule may encoded by a polynucleotide (e.g. a DNA or RNA polynucleotide). The costimulatory molecule may encoded by the polynucleotide sequence of CD80 (SEQ ID NO: 29) or CD86 (SEQ ID NO: 30), or by a subsequence encoding the extracellular portion or a functional fragment. [0228] ATGGGCCACACACGGAGGCAGGGAACATCACCATCCAAGTGTCCATAC CTCAATTTCTTTCAGCTCTTGGTGCTGGCTGGTCTTTCTCACTTCTGTTCAGGTG TTATCCACGTGACCAAGGAAGTGAAAGAAGTGGCAACGCTGTCCTGTGGTCAC AATGTTTCTGTTGAAGAGCTGGCACAAACTCGCATCTACTGGCAAAAGGAGAA GAAAATGGTGCTGACTATGATGTCTGGGGACATGAATATATGGCCCGAGTACA AGAACCGGACCATCTTTGATATCACTAATAACCTCTCCATTGTGATCCTGGCTC TGCGCCCATCTGACGAGGGCACATACGAGTGTGTTGTTCTGAAGTATGAAAAA GACGCTTTCAAGCGGGAACACCTGGCTGAAGTGACGTTATCAGTCAAAGCTGA CTTCCCTACACCTAGTATATCTGACTTTGAAATTCCAACTTCTAATATTAGAAG GATAATTTGCTCAACCTCTGGAGGTTTTCCAGAGCCTCACCTCTCCTGGTTGGA AAATGGAGAAGAATTAAATGCCATCAACACAACAGTTTCCCAAGATCCTGAA ACTGAGCTCTATGCTGTTAGCAGCAAACTGGATTTCAATATGACAACCAACCA CAGCTTCATGTGTCTCATCAAGTATGGACATTTAAGAGTGAATCAGACCTTCA ACTGGAATACAACCAAGCAAGAGCATTTTCCTGATAACCTGCTCCCATCCTGG GCCATTACCTTAATCTCAGTAAATGGAATTTTTGTGATATGCTGCCTGACCTAC TGCTTTGCCCCAAGATGCAGAGAGAGAAGGAGGAATGAGAGATTGAGAAGGG AAAGTGTACGCCCTGTA (SEQ ID NO: 29) [0229] ATGGATCCCCAGTGCACTATGGGACTGAGTAACATTCTCTTTGTGATGG CCTTCCTGCTCTCTGGTGCTGCTCCTCTGAAGATTCAAGCTTATTTCAATGAGA Attorney Ref. No.061479-506001WO CTGCAGACCTGCCATGCCAATTTGCAAACTCTCAAAACCAAAGCCTGAGTGAG CTAGTAGTATTTTGGCAGGACCAGGAAAACTTGGTTCTGAATGAGGTATACTT AGGCAAAGAGAAATTTGACAGTGTTCATTCCAAGTATATGGGCCGCACAAGTT TTGATTCGGACAGTTGGACCCTGAGACTTCACAATCTTCAGATCAAGGACAAG GGCTTGTATCAATGTATCATCCATCACAAAAAGCCCACAGGAATGATTCGCAT CCACCAGATGAACTCTGAACTGTCAGTGCTTGCTAACTTCAGTCAACCTGAAA TAGTACCAATTTCTAATATAACAGAAAATGTGTACATAAATTTGACCTGCTCA TCTATACACGGTTACCCAGAACCTAAGAAGATGAGTGTTTTGCTAAGAACCAA GAACTCAACTATCGAGTATGATGGTGTTATGCAGAAATCTCAAGATAATGTCA CAGAACTGTACGACGTTTCCATCAGCTTGTCTGTTTCATTCCCTGATGTTACGA GCAATATGACCATCTTCTGTATTCTGGAAACTGACAAGACGCGGCTTTTATCTT CACCTTTCTCTATAGAGCTTGAGGACCCTCAGCCTCCCCCAGACCACATTCCTT GGATTACAGCTGTACTTCCAACAGTTATTATATGTGTGATGGTTTTCTGTCTAA TTCTATGGAAATGGAAGAAGAAGAAGCGGCCTCGCAACTCTTATAAATGTGG AACCAACACAATGGAGAGGGAAGAGAGTGAACAGACCAAGAAAAGAGAAAA AATCCATATACCTGAAAGGTCTGATGAAGCCCAGCGTGTTTTTAAAAGTTCGA AGACATCTTCATGCGACAAAAGTGATACATGTTTT (SEQ ID NO: 30) [0230] The polynucleotide sequence may be varied by codon-optimization or other methods to generate polynucleotide sequences having at least 50%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to SEQ ID NO: 29 or 30, or a suitable subsequence, which may be used to express the costimulatory molecule. [0231] Further costimulatory molecules useful in the practice of the present invention may include any molecule that specifically binds CD28. For example, the costimulatory molecule may be a molecule that comprises an antibody, or antigen-binding fragment thereof, specific to CD28. [0232] CD28 is a receptor expressed on T cells that provide costimulatory signal. T cell costimulation through CD28, resulting in, for example, the production of various interleukins (in particular IL-6). In some embodiments, the costimulatory molecule is an antibody, or fragment thereof, that specifically binds to CD28. Examples of such antibodies include 15E8, TGN1412, CD28.2, and 10F3, as well as humanized variants thereof. [0233] 15E8 is a mouse monoclonal antibody to human CD28. Its complementarity determining regions (CDRs) are as follows: Attorney Ref. No.061479-506001WO [0234] CDRH1 : GFSLTSY (SEQ ID NO: 36) [0235] CDRH2: WAGGS (SEQ ID NO. 37) [0236] CDRH3: DKRAPGKLYYGYPDY (SEQ ID NO.38) [0237] CDRL1: RASESVEYYVTSLMQ (SEQ ID NO.39) [0238] CDRL2: AASNYES (SEQ ID NO.40) [0239] CDRL3: QQTRKVPST (SEQ ID NO.41) [0240] TGN1412 (also known as CD28-SuperMAB) is a humanized monoclonal antibody that not only binds to, but also is a strong agonist for, the CD28 receptor. Its CDRs are as follows. [0241] CDRHl: GYTFSY (SEQ ID NO.42) [0242] CDRH2: YPGNVN (SEQ ID NO. 43) [0243] CDRH3: SHYGLDWNFDV (SEQ ID NO. 44) [0244] CDRLl: HASQNIYVLN (SEQ ID NO.45) [0245] CDRL2: KASNLHT (SEQ ID NO. 46) [0246] CDRL3: QQGQTYPYT (SEQ ID NO. 47) [0247] The foregoing description of CD80, CD86, and their derivatives as the costimulatory molecule and CD28 as the cognate molecule may be extrapolated to the other costimulatory molecules described herein, including but not limited to those listed in Table 2. The costimulatory molecule (or the fusion protein) may comprise any polypeptide sequence of in Table 2, to an extracellular portion thereof, or to a functional fragment thereof, or a sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to a sequence in Table 2, to an extracellular portion thereof, or to a functional fragment thereof. Activation Molecules [0248] Disclosed herein, in some embodiments, are activation molecules. The activation molecule may be included as part of a fusion molecule. The activation molecule may be included as part of a particle (e.g. displayed on a particle surface). An example of an activation molecule may include a TCR-binding molecule. [0249] The fusion molecule displayed on the particle may include an activation molecule (e.g. a TCR-binding molecule) or other subunit that provides an activation signal to a target cell. However, in some embodiments, the fusion molecule does not include a TCR-binding molecule or other activation domain. The particle may display a TCR-binding molecule as a separate molecule on the surface of the particle, or the particle may lack any TCR-binding Attorney Ref. No.061479-506001WO molecule. The TCR-binding molecule may be a protein, termed herein a “TCR-binding protein.” The activation molecule may be or include an activation protein. [0250] As used herein, the term “TCR-binding molecule” refers to a molecule capable of directly binding the extracellular portion of the T cell receptor (TCR) by contacting one or more components of the TCR or otherwise providing a primary or “signal 1” activation signal to a target cell (e.g. a T cell or NK cell). The structure of the TCR, its components, and function is described in Sušac et al. Cell 185(17):3201-3213.e19 (2022). Some examples of TCR-binding molecules may include an antibody, or antigen binding fragment, that specifically binds CD3 (an anti-CD3 monoclonal antibody, or antigen binding fragment thereof). In some embodiments, the activation molecule comprises an antibody, single domain antibody, antibody fragment, nanobody, or other binding protein specific for CD3. Illustrative antibodies include OKT3 (also known as Muromonab-CD3), otelixizumab, teplizumab and visilizumab. The complementarity determining regions of OKT3 are as follows: [0251] CDRH1: GYTFTRY (SEQ ID NO.48) [0252] CDRH2: NPSRGY (SEQ ID NO.49) [0253] CDRH3: YYDDHYCLDY (SEQ ID NO.50) [0254] CDRL1: SASSSVSYMN (SEQ ID NO.51) [0255] CDRL2: DTSKLAS (SEQ ID NO.52) [0256] CDRL3: QQWSSNPFT (SEQ ID NO.53) [0257] The activation molecule (e.g. TCR-binding molecule) may be a single chain variable fragment (scFv) displayed on the particle as linked to a transmembrane region or an anchor. OKT3 in scFv format may be used. [0258] In some embodiments, the activation molecule (e.g. TCR-binding molecule) is or includes an scFv comprising a polypeptide sequence at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identical to the anti-CD3 scFv of SEQ ID NO: 31, which includes a variable light (VL) and variable heavy (VH) domain with a 3 × GGGS linker: [0259] DIQMTQSPSSLSASVGDRVTITCSASSSVSYMNWYQQTPGKAPKRWIYDTS KLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSNPFTFGQGTKLQITRT SGGGGSGGGGSGGGGSQVQLVQSGGGVVQPGRSLRLSCKASGYTFTRYTMHWV RQAPGKGLEWIGYINPSRGYTNYNQKVKDRFTISRDNSKNTAFLQMDSLRPEDTG VYFCARYYDDHYCLDYWGQGTPVTVSSAAAKP (SEQ ID NO: 31) Attorney Ref. No.061479-506001WO [0260] In some embodiments, the activation molecule (e.g. TCR-binding molecule) is or includes an scFv comprising a polypeptide sequence at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identical to the anti-CD3 scFv of SEQ ID NO: 249, which includes a variable light (VL) and variable heavy (VH) domain with a 3 × GGGS linker: [0261] DIQMTQSPSSLSASVGDRVTITCSASSSVSYMNWYQQTPGKAPKRWIYDTS KLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSNPFTFGQGTKLQITRT SGGGGSGGGGSGGGGSQVQLVQSGGGVVQPGRSLRLSCKASGYTFTRYTMHWV RQAPGKGLEWIGYINPSRGYTNYNQKVKDRFTISRDNSKNTAFLQMDSLRPEDTG VYFCARYYDDHYCLDYWGQGTPVTVSSA (SEQ ID NO: 249) [0262] The complementarity determining regions of this scFv are as follows: [0263] CDRH1: RYTMH (SEQ ID NO: 54) [0264] CDRH2: YINPSRGYTNYNQKVKD (SEQ ID NO: 55) [0265] CDRH3: YYDDHYCLDY (SEQ ID NO: 56) [0266] CDRL1: SASSSVSYMN (SEQ ID NO: 57) [0267] CDRL2: DTSKLASG (SEQ ID NO: 58) [0268] CDRL3: QQWSSNPFT (SEQ ID NO: 59) [0269] Other activation molecules and/or domains may comprise the binding regions of other proteins commonly found in the supramolecular activation complex (SMAC) between T lymphocytes and antigen presenting cells. For example, CD3, CD2, CD4, CD8, CD28, LFA-1, CD45, CD43, CD40, ICAM-1, CTLA-4, CD80, CD86, MHC, LFA-3, AND CD40L are proteins that may be present within the SMAC. The fusion proteins disclosed herein may comprise portions of these proteins or domains that bind to these proteins. For example, without wishing to be bound by theory, T cells may express one or both of CD4 and/or CD8 and fusion molecules disclosed herein may comprise domains that engage with either or both of CD4 and/or CD8. [0270] When cells other than T cells are the intended target of the particles comprising a fusion molecule as disclosed herein, other binding domains may be more appropriate. For example, particles targeting NK cells may comprise domains that engage with proteins found on NK cells. In some embodiments, these proteins include CD2, CD16, NKp46, NKp30, and NKG2D. In some such embodiments, fusion proteins intended to target and/or activate NK cells may comprise domains that bind to CD2, CD16, NKp46, NKG2D, etc. Domains that bind to NKG2D may be derived from NKG2D ligands including, but not Attorney Ref. No.061479-506001WO limited to: MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, and ULBP6. In some embodiments, the fusion proteins described herein comprise a CD58 domain, a domain that binds NKG2D, and optionally a third domain which enhances activation of the target NK cell. [0271] The activation molecule may be encoded by a polynucleotide (e.g. a DNA or RNA polynucleotide). Fusion Molecule [0272] Disclosed herein, in some embodiments, are fusion molecules. The fusion molecule may include an adhesion molecule, a costimulatory molecule, or an activation molecule. The fusion molecule may include an adhesion molecule. The fusion molecule may include a costimulatory molecule. The fusion molecule may include an activation molecule. The fusion molecule may include an adhesion molecule, a costimulatory molecule, and an activation molecule. The fusion molecule may include an adhesion molecule and an activation molecule. The fusion molecule may include a costimulatory molecule and an activation molecule. The fusion molecule may be or include a fusion protein. The fusion molecule may be included as part of a particle. The fusion molecule may be used in a method described herein. [0273] In some embodiments, the disclosure provides a fusion molecule comprising a combination of an adhesion molecule, a costimulatory molecule, and an activation molecule (e.g. a TCR-binding molecule), thereof each component linked directly or indirectly to the other components. In some embodiments, the fusion molecule comprises adhesion molecule, a costimulatory molecule, and an activation molecule (e.g. a TCR-binding molecule). In some embodiments, the fusion molecule comprises adhesion molecule and a costimulatory molecule, but not a TCR-binding molecule. In some embodiments, the fusion molecule comprises adhesion molecule and an activation molecule (e.g. a TCR-binding molecule), but not a costimulatory molecule. The fusion molecule may further comprise one or more additional adhesion molecules, costimulatory molecules, or activation molecules (e.g. TCR-binding molecules). [0274] As used herein, the term “fusion molecule” refers to any molecule having multiple components link together, directly or indirectly, covalently or non-covalently. The fusion molecule may be made up of several proteins. When those proteins are linked together into a single molecule by peptide bonds, the fusion molecule is termed a “fusion protein.” Attorney Ref. No.061479-506001WO [0275] The fusion molecule may be made using various linkers, including chemical (covalent) bonds (e.g., by click chemistry) or by peptide bounds. When the fusion molecule is a fusion protein, the linker between each component of the fusion protein may be a single peptide bound (i.e., a direct C- to N- peptide bound in a polypeptide chain) or via a polypeptide linker. Illustrative polypeptide linkers may include, but are not limited to, the glycine-serine linkers, such as GGSGGS, GSSGSS, or others. [0276] In some embodiments, the fusion molecule is or includes a fusion protein. The fusion protein may comprise an adhesion protein, a polypeptide linker, and a costimulatory portion. In some embodiments, the fusion protein comprises an adhesion molecule, a costimulatory molecule, and an activation molecule. [0277] In some embodiments of the fusion protein, the adhesion molecule is N-terminal to the costimulatory molecule. In some embodiments, the adhesion molecule is N-terminal to the activation molecule. In some embodiments, the adhesion molecule is C-terminal to the costimulatory molecule. In some embodiments, the adhesion molecule is C-terminal to the activation molecule. [0278] In some embodiments of the fusion protein, the activation molecule is N-terminal to the costimulatory molecule. In some embodiments, the activation molecule is N-terminal to the adhesion molecule. In some embodiments, the activation molecule is C-terminal to the costimulatory molecule. In some embodiments, the activation molecule is C-terminal to the adhesion molecule. [0279] In some embodiments of the fusion protein, the costimulatory molecule is N- terminal to the activation molecule. In some embodiments, the costimulatory molecule is N-terminal to the adhesion molecule. In some embodiments, the costimulatory molecule is C-terminal to the activation molecule. In some embodiments, the costimulatory molecule is C-terminal to the adhesion molecule. [0280] Some embodiments of the fusion protein includes a linker. Some embodiments include multiple linkers. In some embodiments, a linker directly connects the costimulatory molecule with the adhesion molecule. In some embodiments, a linker directly connects the costimulatory molecule with the activation molecule. In some embodiments, a linker directly connects the adhesion molecule with the activation molecule. [0281] In some embodiments of the fusion protein, an N terminal end of the costimulatory molecule is juxtaposed (directly or via a linker) with an end of the adhesion molecule. In some embodiments of the fusion protein, a C terminal end of the costimulatory molecule is Attorney Ref. No.061479-506001WO juxtaposed (directly or via a linker) with an end of the adhesion molecule. In some embodiments of the fusion protein, an N terminal end of the costimulatory molecule is juxtaposed (directly or via a linker) with an end of the activation molecule. In some embodiments of the fusion protein, a C terminal end of the costimulatory molecule is juxtaposed (directly or via a linker) with an end of the activation molecule. [0282] In some embodiments of the fusion protein, an N terminal end of the activation molecule is juxtaposed (directly or via a linker) with an end of the adhesion molecule. In some embodiments of the fusion protein, a C terminal end of the activation molecule is juxtaposed (directly or via a linker) with an end of the adhesion molecule. In some embodiments of the fusion protein, an N terminal end of the activation molecule is juxtaposed (directly or via a linker) with an end of the costimulatory molecule. In some embodiments of the fusion protein, a C terminal end of the activation molecule is juxtaposed (directly or via a linker) with an end of the costimulatory molecule. [0283] In some embodiments of the fusion protein, an N terminal end of the adhesion molecule is juxtaposed (directly or via a linker) with an end of the costimulatory molecule. In some embodiments of the fusion protein, a C terminal end of the adhesion molecule is juxtaposed (directly or via a linker) with an end of the costimulatory molecule. In some embodiments of the fusion protein, an N terminal end of the adhesion molecule is juxtaposed (directly or via a linker) with an end of the activation molecule. In some embodiments of the fusion protein, a C terminal end of the adhesion molecule is juxtaposed (directly or via a linker) with an end of the activation molecule. [0284] Some non-limitng examples of fusion proteins are included in FIG. 60A-60B. For example, a fusion protein shown in FIG.60A-60B may further comprise a viral protein such as a viral glycoprotein. Any of such fusion proteins may be included as part of a particle (e.g. a viral particle such as a lentiviral particle), or may be displayed on a particle surface. [0285] The fusion protein may comprise, in any order, a CD80, a CD80 extracellular portion, or a functional fragment of CD80; a CD58, a CD58 extracellular portion; or a functional fragment of CD58; an activation molecule (e.g. a TCR-binding molecule); and polypeptide linkers. [0286] The fusion protein may comprise, in N- to C-terminal order, CD80, a CD80 extracellular portion, or a functional fragment of CD80; a polypeptide linker; and CD58, a CD58 extracellular portion; or a functional fragment of CD58. Attorney Ref. No.061479-506001WO [0287] The fusion protein may comprise, in N- to C-terminal order, CD58, a CD58 extracellular portion; or a functional fragment of CD58; a polypeptide linker; and CD80, a CD80 extracellular portion, or a functional fragment of CD80. [0288] The fusion protein may comprise, in N- to C-terminal order, an activation molecule (e.g. a TCR-binding protein); a polypeptide linker; CD80, a CD80 extracellular portion, or a functional fragment of CD80; a polypeptide linker; and CD58, a CD58 extracellular portion; or a functional fragment of CD58. [0289] The fusion protein may comprise, in N- to C-terminal order, CD80, a CD80 extracellular portion, or a functional fragment of CD80; a polypeptide linker; CD58, a CD58 extracellular portion; or a functional fragment of CD58; a polypeptide linker; and an activation molecule (e.g. a TCR-binding protein). [0290] The fusion protein may comprise, in N- to C-terminal order, an activation molecule (e.g. a TCR-binding protein); a polypeptide linker; CD58, a CD58 extracellular portion; or a functional fragment of CD58; a polypeptide linker; and CD80, a CD80 extracellular portion, or a functional fragment of CD80. [0291] The fusion protein may comprise, in N- to C-terminal order, CD58, a CD58 extracellular portion; or a functional fragment of CD58; a polypeptide linker; CD80, a CD80 extracellular portion, or a functional fragment of CD80; a polypeptide linker; and an activation molecule (e.g. a TCR-binding protein). [0292] 7W SUU^\][K]S_O P^\SXW Y[X]OSW MXVY[S\O\ K 9:03 Oa][KMOUU^UK[ [OQSXW KWN h(9:. scFv fused to the N-terminus of a CD80 via a linker; this construct is termed a tri-fusion polypepetide and/or termed “498.” [0293] An illustrative fusion protein comprises a CD58 extracellular region fused to the N- terminus of a CD80 via a linker; this construct is termed a bi-fusion polypeptide and/or ]O[VON e/00)f ?W ]RS\ MXW\][^M]' KW h9:. \M<_ S\ OaY[O\\ON K\ K \OYK[K]O YXUbYOY]SNO SW ]RO producer cells. [0294] In each case, the polypeptide linker may be optional. It may be omitted by directly linking protein molecule to the next via a peptide bound. Although one my generate fusion proteins through chemical synthesis, fusion protein are more made by expressing the fusion protein from a single polynucleotide comprising a polynucleotide sequence encoding the entire fusion protein. Methods for designing and cloning polynucleotides are known in the art. Attorney Ref. No.061479-506001WO [0295] The fusion molecule may encoded by a polynucleotide (e.g. a DNA or RNA polynucleotide). In some embodiments, the disclosure provides polynucleotides encoding such fusion proteins. The polynucleotide may be an isolated polynucleotide, or it may be part of a vector (e.g., a plasmid) or it may be introduced into and propagated in a host cell. [0296] Polypeptide sequences of illustrative dual CD58+CD80 fusion proteins are provided in Table 3. In each case, the fusion protein may comprise a polypeptide at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% sequence identity to any sequence in Table 3. In some embodiments, the fusion protein may comprise a polypeptide having less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to any sequence in Table 3. In each case, an optional signal peptide is shown in parentheses. The signal peptide is cleaved during expression of the sequence. Sequence identity to a reference sequence is determined without the optional residues. Diagrams of each fusion are provided in FIG.9A and 9B. Table 3 Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO [0297] EXUbYOY]SNO \OZ^OWMO\ XP SUU^\][K]S_O ][SYUO 9:03&9:3+&h9:.\M<_ P^\SXW Y[X]OSW\ are provided in Table 4A. In each case, the fusion protein may comprise a polypeptide at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% sequence identity to any sequence in Table 4A. In some embodiments, the fusion protein may comprise a polypeptide less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to any sequence in Table 4A. In each case, an optional signal peptide is shown in parentheses. The signal peptide is cleaved during expression of the sequence. Sequence identity to a reference sequence is determined without the optional residues. Diagrams of each fusion are provided in FIG.27B. Table 4A Attorney Ref. No.061479-506001WO [0298] FIG. 50A-50J include examples of CD58 and CD80 dual fusion sequences. Some embodiments include a nucleic acid sequence having at least at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, Attorney Ref. No.061479-506001WO at least 99%, or 100% sequence identity to a nucleic acid sequence in any of FIG.50A-50J or any of SEQ ID NOs: 215-234, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include a nucleic acid sequence having less than less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to a nucleic acid sequence in any of FIG.50A-50J or any of SEQ ID NOs: 215-234, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include an amino acid sequence having at least at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% sequence identity to an amino acid sequence in any of FIG.50A-50J or any of SEQ ID NOs: 215-234, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include an amino acid sequence having less than less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to an amino acid sequence in any of FIG.50A-50J or any of SEQ ID NOs: 215-234, or to a fragment or portion thereof such as may be identified in the figure keys. [0299] FIG. 51A-51F include examples of CD58, CD80 and CD3 scFV triple fusion sequences. Some embodiments include a nucleic acid sequence having at least at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% sequence identity to a nucleic acid sequence in any of FIG.51A-51F or any of SEQ ID NOs: 235-246, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include a nucleic acid sequence having less than less than 75%, less than 80%, less than 85%, less than 90%, less than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to a nucleic acid sequence in any of FIG.51A-51F or any of SEQ ID NOs: 235-246, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include an amino acid sequence having at least at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% sequence identity to an amino acid sequence in any of FIG.51A-51F or any of SEQ ID NOs: 235-246, or to a fragment or portion thereof such as may be identified in the figure keys. Some embodiments include an amino acid sequence having less than less than 75%, less than 80%, less than 85%, less than 90%, less Attorney Ref. No.061479-506001WO than 91%, less than 92%, less than 93%, less than 94%, less than 95%, less than 99%, or less than 100% sequence identity to an amino acid sequence in any of FIG.51A-51F or any of SEQ ID NOs: 235-246, or to a fragment or portion thereof such as may be identified in the figure keys. Chimeric Antigen Receptors (CARs) [0300] In some embodiments, a particle such as a lentiviral particle described herein is used to transduce a nucleic acid sequence (polynucleotide) encoding one or more chimeric antigen receptor (CARs) into a cell (e.g., a T lymphocyte). In some embodiments, the transduction of the lentiviral particle results in expression of one or more CARs in the transduced cells. [0301] CARs are artificial membrane-bound proteins that direct a T lymphocyte to an antigen and stimulate the T lymphocyte to kill cells displaying the antigen. See, e.g., Eshhar, U.S. Pat. No.7,741,465. Generally, CARs are genetically engineered receptors comprising an extracellular domain that binds to an antigen, e.g., an antigen on a cell, an optional linker, a transmembrane domain, and an intracellular (cytoplasmic) domain comprising a costimulatory domain and/or a signaling domain that transmits an activation signal to an immune cell. With a CAR, a single receptor can be programmed to both recognize a specific antigen and, when bound to that antigen, activate the immune cell to attack and destroy the cell bearing that antigen. When these antigens exist on tumor cells, an immune cell that expresses the CAR can target and kill the tumor cell. All other conditions being satisfied, when a CAR is expressed on the surface of, e.g., a T lymphocyte, and the extracellular domain of the CAR binds to an antigen, the intracellular signaling domain transmits a signal to the T lymphocyte to activate and/or proliferate, and, if the antigen is present on a cell surface, to kill the cell expressing the antigen. Because T lymphocytes may require two signals, a primary activation signal and a costimulatory signal, in order to maximally activate, CARs can comprise a stimulatory and a costimulatory domain such that binding of the antigen to the extracellular domain results in transmission of both a primary activation signal and a costimulatory signal. Illustrative CARs may be designed in a modular fashion, e.g. as described in (see, e.g., Guedan S, Calderon H, Posey AD, Maus MV, Molecular Therapy - Methods & Clinical Development.2019; 12: 145-156), incorporated by reference. [0302] In some embodiments, a lentiviral particle disclosed herein comprises a polynucleotide that encodes a CAR comprising an extracellular domain, optionally a hinge domain, a transmembrane domain, and an intracellular signaling domain. In some Attorney Ref. No.061479-506001WO embodiments, the intracellular signaling domain comprises a costimulatory domain and an activation domain. In some embodiments, the costimulatory and activation domains are a single domain, for example a single intracellular domain that provides both costimulation and activation signals to a cell. In other embodiments, the intracellular signaling domain comprises either a costimulatory domain or an activation domain. In some embodiments, the CAR comprises an extracellular domain, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a lentiviral particle disclosed herein encodes an extracellular domain, a CD28 hinge domain, a CD28 transmembrane domain, a CD28 co-stimulatory domain, and a CD3zeta signaling domain. In some embodiments, a lentiviral particle disclosed herein encodes an extracellular domain, an IgG4 hinge domain, a CD28 transmembrane domain, a 4-1BB co-stimulatory domain, and a CD3zeta signaling domain. In some embodiments, a lentiviral particle disclosed herein encodes a CAR comprising an extracellular domain, a CD8a hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. CAR Intracellular Domain [0303] In some embodiments, the intracellular domain of the CAR is or comprises an intracellular domain or motif of a protein that is expressed on the surface of T lymphocytes and triggers activation and/or proliferation of said T lymphocytes. In some embodiments, such a domain or motif is able to transmit a signal for activation of a T lymphocyte in response to antigen binding to the extracellular portion of the CAR. In some embodiments, this domain or motif comprises, or is, an ITAM (immunoreceptor tyrosine-based activation motif). ITAM-containing polypeptides suitable for CARs include, for example, the zeta 9:. MRKSW $9:.m% X[ ?I7B(MXW]KSWSWQ YX[]SXW\ ]RO[OXP) ?W \XVO OVLXNSVOW]\' ]RO SW][KMOUU^UK[ NXVKSW S\ K 9:.m SW][KMOUU^UK[ \SQWKUSWQ NXVKSW) ?W \XVO OVLXNSVOW]\' ]RO intracellular domain is from a lymphocyte receptor chain, a TCR/CD3 complex protein, an Fc receptor subunit or an IL-2 receptor subunit. In some embodiments, the intracellular \SQWKUSWQ NXVKSW XP 97G VKb LO ]RO \SQWKUSWQ NXVKSW\ XP PX[ OaKVYUO 9:.m' 9:.i' 9:--' CD79a, CD66d or CD39. “Intracellular signaling domain” refers to the part of a CAR polypeptide that participates in transducing the message of effective CAR binding to a target antigen into the interior of the immune effector cell to elicit effector cell function, e.g., activation, cytokine production, proliferation and cytotoxic activity, including the release of cytotoxic factors to the CAR-bound target cell, or other cellular responses elicited following antigen binding to the extracellular CAR domain. Attorney Ref. No.061479-506001WO [0304] In some embodiments, the intracellular domain of the CAR is the zeta CD3 chain (CD3zeta). [0305] In some embodiments, the lentiviral particle comprises a polypeptide comprising a CAR whose intracellular domain comprises a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 82. [0306] RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKP RRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYD ALHMQALPPR (SEQ ID NO: 82) [0307] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the intracellular domain of a CAR comprising a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 83. [0308] CGCGTGAAGTTCAGCCGGTCCGCCGATGCCCCTGCCTACCAGCAGGGC CAGAACCAGCTGTATAACGAGCTGAATCTGGGCCGGAGAGAGGAGTACGACG TGCTGGATAAGAGGAGGGGAAGGGACCCAGAGATGGGAGGCAAGCCTCGGA GAAAGAACCCACAGGAGGGCCTGTACAATGAGCTGCAGAAGGACAAGATGGC CGAGGCCTATTCTGAGATCGGCATGAAGGGAGAGAGGCGCCGGGGCAAGGGA CACGATGGCCTGTACCAGGGCCTGAGCACCGCCACAAAGGACACATATGATG CCCTGCACATGCAGGCCCTGCCACCTAGG (SEQ ID NO: 83) [0309] In some embodiments, the CAR additionally comprises one or more co-stimulatory domains or motifs, e.g., as part of the intracellular domain of the polypeptide. Co- stimulatory molecules may include cell surface molecules other than antigen receptors or Fc receptors that provide a second signal useful for efficient activation and function of T lymphocytes upon binding to antigen. The one or more co-stimulatory domains or motifs can, for example, be, or comprise, one or more of a co-stimulatory CD27 polypeptide sequence, a co-stimulatory CD28 polypeptide sequence, a co-stimulatory OX40 (CD134) polypeptide sequence, a co-stimulatory 4-1BB (CD137) polypeptide sequence, or a co- stimulatory inducible T-cell costimulatory (ICOS) polypeptide sequence, or other costimulatory domain or motif, or any combination thereof. In some embodiments, the one or more co-stimulatory domains are selected from the group consisting of intracellular domains of 4-1BB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 Attorney Ref. No.061479-506001WO (OX40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM, and ZAP70. [0310] In some embodiments, the co-stimulatory domain is an intracellular domain of 4- 1BB, CD28, or OX40. Illustrative CAR constructs comprising a CD28 signaling domain are disclosed in US Patent No. 7,446,190, incorporated by reference. Illustrative CAR constructs comprising a 4-1BB signaling domain are disclosed in US Patent No. 9,856,322 and US Patent No.8,399,964, each incorporated by reference. [0311] In some embodiments, the lentiviral particle encodes a CAR comprising an IgG4 linker operatively linked to a CD28 transmembrane domain operatively linked to a 4-1BB co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0312] In some embodiments, the lentiviral particle encodes a CAR comprising an IgG4 linker operatively linked to a CD8a transmembrane domain operatively linked to a 4-1BB co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0313] In some embodiments, the lentiviral particle encodes a CAR comprising an IgG4 linker operatively linked to a CD8a transmembrane domain operatively linked to a CD28 co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0314] In some embodiments, the lentiviral particle encodes a CAR comprising a CD8a linker operatively linked to a CD8a transmembrane domain operatively linked to a 4-1BB co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0315] In some embodiments, the lentiviral particle encodes a CAR comprising a CD8a linker operatively linked to a CD28 transmembrane domain operatively linked to a 4-1BB co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0316] In some embodiments, the lentiviral particle encodes a CAR comprising a CD28 linker operatively linked to a CD28 transmembrane domain operatively linked to a CD28 co-stimulatory domain operatively linked to a CD3zeta signaling domain. [0317] In some embodiments, the lentiviral particle comprises a polypeptide comprising a CAR whose intracellular domain comprises a co-stimulatory 4-1BB polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 84. [0318] KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO: 84) [0319] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the intracellular domain of a CAR comprising a co-stimulatory 4-1BB sequence that shares at Attorney Ref. No.061479-506001WO least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 85. [0320] CGCGTGAAGTTCAGCCGGTCCGCCGATGCCCCTGCCTACCAGCAGGGC CAGAACCAGCTGTATAACGAGCTGAATCTGGGCCGGAGAGAGGAGTACGACG TGCTGGATAAGAGGAGGGGAAGGGACCCAGAGATGGGAGGCAAGCCTCGGA GAAAGAACCCACAGGAGGGCCTGTACAATGAGCTGCAGAAGGACAAGATGGC CGAGGCCTATTCTGAGATCGGCATGAAGGGAGAGAGGCGCCGGGGCAAGGGA CACGATGGCCTGTACCAGGGCCTGAGCACCGCCACAAAGGACACATATGATG CCCTGCACATGCAGGCCCTGCCACCTAGG (SEQ ID NO: 85) [0321] In some embodiments, the lentiviral particle comprises a polypeptide comprising a CAR whose intracellular domain comprises an IgG4 linker operatively linked to a CD28 transmembrane domain operatively linked to a co-stimulatory 4-1BB polypeptide operatively linked to a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 86. [0322] ESKYGPPCPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKKLLYIF KQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNEL NLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMK GERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 86) [0323] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the intracellular domain of a CAR comprising an IgG4 linker operatively linked to a CD28 transmembrane domain operatively linked to a co-stimulatory 4-1BB polypeptide operatively linked to a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 87. [0324] GAGTCTAAGTATGGCCCTCCATGCCCCCCTTGTCCTATGTTCTGGGTGC TGGTGGTGGTGGGAGGCGTGCTGGCCTGTTACTCCCTGCTGGTGACCGTGGCC TTTATCATCTTCTGGGTGAAGCGCGGCCGGAAGAAGCTGCTGTATATCTTTAA GCAGCCCTTCATGAGACCTGTGCAGACCACACAGGAGGAGGACGGCTGCAGC TGTAGGTTTCCAGAGGAGGAGGAGGGAGGATGCGAGCTGCGCGTGAAGTTCT CTCGGAGCGCCGATGCCCCTGCCTACCAGCAGGGACAGAACCAGCTGTATAA CGAGCTGAATCTGGGCCGGAGAGAGGAGTACGACGTGCTGGATAAGAGGAGG GGAAGAGACCCAGAGATGGGAGGCAAGCCTCGGAGAAAGAACCCACAGGAG GGCCTGTACAATGAGCTGCAGAAGGACAAGATGGCCGAGGCCTATTCCGAGA TCGGCATGAAGGGAGAGAGGCGCCGGGGCAAGGGACACGATGGCCTGTACCA Attorney Ref. No.061479-506001WO GGGCCTGAGCACCGCCACAAAGGACACCTATGATGCCCTGCACATGCAGGCC CTGCCACCCAGG (SEQ ID NO: 87) [0325] In some embodiments, the lentiviral particle comprises a polypeptide comprising a CAR whose intracellular domain comprises an IgG4 linker operatively linked to a CD28 transmembrane domain operatively linked to a co-stimulatory 4-1BB polypeptide operatively linked to a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 88. [0326] ESKYGPPCPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKKLLYIF KQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNEL NLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMK GERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRGSGATNFSLLKQAGDVEE NPGP (SEQ ID NO: 88) [0327] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the intracellular domain of a CAR comprising an IgG4 linker operatively linked to a CD28 transmembrane domain operatively linked to a co-stimulatory 4-1BB polypeptide operatively linked to a CD3zeta domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 89. [0328] GAGTCCAAGTACGGCCCACCCTGCCCTCCATGTCCCATGTTTTGGGTGC TGGTGGTGGTGGGAGGCGTGCTGGCCTGTTATTCCCTGCTGGTGACCGTGGCC TTCATCATCTTTTGGGTGAAGCGCGGCCGGAAGAAGCTGCTGTACATCTTCAA GCAGCCCTTCATGAGACCCGTGCAGACCACACAGGAGGAGGACGGCTGCAGC TGTAGGTTCCCAGAGGAGGAGGAGGGAGGATGCGAGCTGAGGGTGAAGTTTT CCCGGTCTGCCGATGCCCCTGCCTATCAGCAGGGCCAGAATCAGCTGTACAAC GAGCTGAATCTGGGCAGGCGCGAGGAGTACGACGTGCTGGATAAGAGGAGAG GAAGGGACCCTGAGATGGGAGGCAAGCCAAGGCGCAAGAACCCTCAGGAGG GCCTGTATAATGAGCTGCAGAAGGACAAGATGGCCGAGGCCTACTCCGAGAT CGGCATGAAGGGAGAGCGGAGAAGGGGCAAGGGACACGATGGCCTGTATCA GGGCCTGAGCACCGCCACAAAGGACACCTACGATGCACTGCACATGCAGGCC CTGCCACCTAGAGGATCTGGAGCCACAAACTTCAGCCTGCTGAAGCAGGCCG GCGATGTGGAGGAGAATCCTGGACCA (SEQ ID NO: 89) [0329] In some embodiments, the intracellular domain can be further modified to encode a detectable, for example, a fluorescent, protein (e.g., green fluorescent protein) or any variants thereof. Attorney Ref. No.061479-506001WO CAR Transmembrane Region [0330] The transmembrane region can be any transmembrane region that can be incorporated into a functional CAR, e.g., a transmembrane region from a CD28, CD4, or a CD8 molecule. [0331] In some embodiments, the transmembrane domain of CAR may be the transmembrane domain of CD8, an alpha, beta or zeta chain of a T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1 BB (CD137), 4-1 BBL, GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRFI), CD160, CD19, IL-2R beta, IL-2R gamma, IL-7R a, ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRT AM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, NKG2D “in reverse orientation”, and/or NKG2C. In some embodiments, the transmembrane domain of the CAR may be the transmembrane domain of CD28. In some embodiments, the transmembrane domain of a CAR may be the transmembrane domain of 9:3' PX[ OaKVYUO' 9:3h) CAR Linker Region [0332] The optional linker or hinge of CAR positioned between the extracellular domain and the transmembrane domain may be a polypeptide of about 2 to over 100 amino acids in length. The linker can include or be composed of flexible residues such as glycine and serine so that the adjacent protein domains are free to move relative to one another. Longer linkers may be used, e.g., when it is desirable to ensure that two adjacent domains do not sterically interfere with one another. Longer linkers may also be advantageous when the target antigen is closer to the cell surface. [0333] In some embodiments, the linker is from a hinge region or portion of the hinge region of any immunoglobulin or other transmembrane protein. For example, the hinge region may be from IgG1, IgG2, IgG3, IgG4, PD1, CD8, or CD28, or a portion thereof. In some embodiments, the linker is from a portion of an immunoglobulin, for example IgG4. In some embodiments, the linker is a portion of an immunoglobulin, for example IgG1. In some Attorney Ref. No.061479-506001WO embodiments, the linker is a portion of the extracellular domain of CD28. In other embodiments, the linker is a portion of the extracellular domain of CD8. In other embodiments, the linker is a portion of the extracellular domain of PD1. [0334] In some embodiments, the linker is an IgG4 linker operably linked to a CD28 transmembrane domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 90. [0335] ESKYGPPCPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWV (SEQ ID NO: 90) [0336] In some embodiments, the linker is an IgG4 linker operably linked to a CD28 transmembrane domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 91. [0337] GAGTCTAAGTATGGCCCACCCTGCCCTCCATGTCCAATGTTCTGGGTGC TGGTGGTGGTGGGAGGCGTGCTGGCCTGTTACTCCCTGCTGGTGACCGTGGCC TTTATCATCTTCTGGGTG (SEQ ID NO: 91) CAR Extracellular Domain [0338] In some embodiments, the nucleic acid transduced into cells using the methods described herein comprises a sequence that encodes a polypeptide, wherein the extracellular domain of the polypeptide binds to an antigen of interest. In some embodiments, the extracellular domain comprises a receptor, or a portion of a receptor, that binds to said antigen. In some embodiments, the extracellular domain comprises, or is, an antibody or an antigen-binding portion thereof. In some embodiments, the extracellular domain comprises, or is, a single-chain Fv domain. The single-chain Fv domain can comprise, for example, a VL linked to VH by a flexible linker, wherein said VL and VH are from an antibody that binds said antigen. [0339] In some embodiments, the extracellular domain of CAR may contain any polypeptide that binds the desired antigen (e.g. prostate neoantigen or antigen expressed on a tumor of interest). The extracellular domain may comprise a scFv, a portion of an antibody or an alternative scaffold. CARs may also be engineered to bind two or more desired antigens that may be arranged in tandem and separated by linker sequences. For example, one or more domain antibodies, scFvs, llama VHH antibodies or other VH only antibody fragments may be organized in tandem via a linker to provide bispecificity or multispecificity to the CAR. [0340] The antigen to which the extracellular domain of the polypeptide binds can be any antigen of interest, e.g., can be an antigen on a tumor cell. The tumor cell may be, e.g., a Attorney Ref. No.061479-506001WO cell in a solid tumor, or a cell of a blood cancer. The antigen can be any antigen that is expressed on a cell of any tumor or cancer type, e.g., cells of a lymphoma, a lung cancer, a breast cancer, a prostate cancer, an adrenocortical carcinoma, a thyroid carcinoma, a nasopharyngeal carcinoma, a melanoma, e.g., a malignant melanoma, a skin carcinoma, a colorectal carcinoma, a desmoid tumor, a desmoplastic small round cell tumor, an endocrine tumor, an Ewing sarcoma, a peripheral primitive neuroectodermal tumor, a solid germ cell tumor, a hepatoblastoma, a neuroblastoma, a non-rhabdomyosarcoma soft tissue sarcoma, an osteosarcoma, a retinoblastoma, a rhabdomyosarcoma, a Wilms tumor, a glioblastoma, a myxoma, a fibroma, a lipoma, or the like. In some embodiments, said lymphoma can be chronic lymphocytic leukemia (small lymphocytic lymphoma), B-cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, Waldenström macroglobulinemia, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, extranodal marginal zone B cell lymphoma, MALT lymphoma, nodal marginal zone B cell lymphoma, follicular lymphoma, mantle cell lymphoma, diffuse large B cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, Burkitt's lymphoma, T lymphocyte prolymphocytic leukemia, T lymphocyte large granular lymphocytic leukemia, aggressive NK cell leukemia, adult T lymphocyte leukemia/lymphoma, extranodal NK/T lymphocyte lymphoma, nasal type, enteropathy-type T lymphocyte lymphoma, hepatosplenic T lymphocyte lymphoma, blastic NK cell lymphoma, mycosis fungoides, Sezary syndrome, primary cutaneous anaplastic large cell lymphoma, lymphomatoid papulosis, angioimmunoblastic T lymphocyte lymphoma, peripheral T lymphocyte lymphoma (unspecified), anaplastic large cell lymphoma, Hodgkin lymphoma, or a non-Hodgkin lymphoma. In some embodiments, in which the cancer is chronic lymphocytic leukemia (CLL), the B cells of the CLL have a normal karyotype. In some embodiments, in which the cancer is chronic lymphocytic leukemia (CLL), the B cells of the CLL carry a 17p deletion, an 11q deletion, a 12q trisomy, a 13q deletion or a p53 deletion. [0341] In some embodiments, the antigen is expressed on a B-cell malignancy cell, relapsed/refractory CD19-expressing malignancy cell, diffuse large B-cell lymphoma (DLBCL) cell, Burkitt’s type large B-cell lymphoma (B-LBL) cell, follicular lymphoma (FL) cell, chronic lymphocytic leukemia (CLL) cell, acute lymphocytic leukemia (ALL) cell, mantle cell lymphoma (MCL) cell, hematological malignancy cell, colon cancer cell, lung cancer cell, liver cancer cell, breast cancer cell, renal cancer cell, prostate cancer cell, Attorney Ref. No.061479-506001WO ovarian cancer cell, skin cancer cell, melanoma cell, bone cancer cell, brain cancer cell, squamous cell carcinoma cell, leukemia cell, myeloma cell, B cell lymphoma cell, kidney cancer cell, uterine cancer cell, adenocarcinoma cell, pancreatic cancer cell, chronic myelogenous leukemia cell, glioblastoma cell, neuroblastoma cell, medulloblastoma cell, or a sarcoma cell. [0342] In some embodiments, the antigen is a tumor-associated antigen (TAA) or a tumor- specific antigen (TSA). In some embodiments, without limitation, the tumor-associated antigen or tumor-specific antigen is B cell maturation antigen (BCMA), B cell Activating Factor (BAFF), GPRC5D, FCRL5/FCRH5, ROR1, L1-CAM, CD22, folate receptor, carboxy anhydrase IX (CAIX), claudin 18.2, FAP, mesothelin, IL-13Ra2, Lewis Y, CCNA1, WT-1, TACI, CD38, SLAMF7, CD138, DLL3, transmembrane 4 L six family member 1 (TM4SF1), epithelial cell adhesion molecule (EpCAM), PD-1, PD-L1, CTLA-4, AXL, ROR2, glypican-3 (GPC3), CD133, CD147, EGFR, MUC1, GD2, Her2, prostate stem cell antigen (PSCA), prostate-specific membrane antigen (PSMA) alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), EGFRvIII, cancer antigen-125 (CA-125), CA19- 9, calretinin, MUC-1, epithelial membrane protein (EMA), epithelial tumor antigen (ETA), tyrosinase, melanoma-associated antigen (MAGE), CD19, CD20, CD34, CD45, CD99, CD117, chromogranin, cytokeratin, desmin, glial fibrillary acidic protein (GFAP), gross cystic disease fluid protein (GCDFP-15), HMB-45 antigen, protein melan-A (melanoma antigen recognized by T lymphocytes; MART-1), myo-D1, muscle-specific actin (MSA), neurofilament, neuron-specific enolase (NSE), placental alkaline phosphatase, synaptophysis, thyroglobulin, thyroid transcription factor-1, vascular endothelial growth factor receptor (VEGFR), the dimeric form of the pyruvate kinase isoenzyme type M2 (tumor M2-PK), an abnormal ras protein, or an abnormal p53 protein. In some embodiments, the CAR comprises binding domains that target two or more antigens as disclosed herein, in any combination. For example: CD19 and CD3, BCMA and CD3, GPRC5D and CD3, FCRL5 and CD3, CD38 and CD3, CD19 and CD20, CD19 and CD22, BCMA and GPRC5D, or CD20 and CD22. In some embodiments, the CAR comprises binding domains that target two or more antigens on the same target protein, for example two epitopes in BCMA. [0343] In other embodiments, the CAR is a universal CAR and does not itself specifically target a tumor antigen. For example, the CAR could comprise a tag-specific scFv such that Attorney Ref. No.061479-506001WO an exogenous agent comprising the tag and a tumor-targeting domain could direct the universal CAR T cell to the target tumor. [0344] In some embodiments, the CAR is a second-generation CAR comprised of an anti- fluorescein scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. [0345] In some embodiments, the antigen is CD19. CAR T therapies targeting CD19 have been approved by the FDA and include Yescarta, Tecartus, Kymriah and Breyanzi. CARs targeting CD19 are described, for example, in US Publication No.20160152723, US Patent No.10,736,918, US Patent No.10,357,514, and US Patent No.7,446,190, each incorporated by reference. [0346] In some embodiments, a CAR comprises an extracellular domain comprising a FMC63 scFv binding domain for CD19 binding. In some embodiments, the CAR is a second-generation CAR comprised of the FMC63 mouse anti-human CD19 scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. In some embodiments, a CAR comprises a binding domain for CD19, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for CD19, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for CD19, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a FMC63 scFv binding domain for CD19 binding, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a FMC63 scFv binding domain for CD19 binding, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a FMC63 scFv binding domain for CD19 binding, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. [0347] In some embodiments, the lentiviral particle comprises a polypeptide comprising a CAR whose extracellular domain comprises a signal peptide that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 92. Attorney Ref. No.061479-506001WO [0348] MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 92) [0349] In some embodiments, the lentiviral particle comprises a polynucleotide encoding a 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ $9:,4 JA USWTON ]X K 9:,4 VH) that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 93. [0350] DIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHT SRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITG STSGSGKPGSGEGSTKGEVKLQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQ PPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYC AKHYYYGGSYAMDYWGQGTSVTVSS (SEQ ID NO: 93) [0351] The complementary determining regions (CDR) of this scFv are RASQDISKYLN, (CDR-L1; SEQ ID NO: 94), HTSRLHS (CDR-L2; SEQ ID NO: 95), QQGNTLPYT (CDR-L3; SEQ ID NO: 96), DYGV (CDR-H1; SEQ ID NO: 97), VIWGSETTYYNSALKS (CDR-H2; SEQ ID NO: 98), HYYYGGSYAMDY (CDR-H3; SEQ ID NO: 99). In some embodiments, the lentiviral particle comprises a polynucleotide OWMXNSWQ K 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ RK_SWQ ]RO\O 9:G\' `RO[OSW XY]SXWKUUb ]RO h9:,4 \M<_ \RK[O\ K] UOK\] 20#' K] UOK\] 3+#' K] UOK\] 30#' at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 93. [0352] In some embodiments, the lentiviral particle comprises a polynucleotide encoding K 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ RK_SWQ ]RO\O 9:G\' `RO[OSW XY]SXWKUUb ]RO h9:,4 \M<_ \RK[O\ K] UOK\] 20#' K] UOK\] 3+#' K] UOK\] 30#' K] UOK\] 4+#' K] least 95%, at least 99%, or 100% identity to SEQ ID NO: 93 or 100. [0353] MLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASLGDRVTISCRASQDISK YLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYF CQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVKLQESGPGLVAPSQSL SVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNS KSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQGTSVTVSS (SEQ ID NO: 100) [0354] In some embodiments, the lentiviral particle comprises a nucleic acid encoding a signal peptide for the extracellular domain of CAR that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 101. Attorney Ref. No.061479-506001WO [0355] ATGCTGCTGCTGGTGACCTCCCTGCTGCTGTGCGAGCTGCCTCACCCAG CCTTTCTGCTGATCCCC (SEQ ID NO: 101) [0356] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the Oa][KMOUU^UK[ NXVKSW XP K 97G MXVY[S\SWQ K h9:,4 \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 102. [0357] GACATCCAGATGACACAGACCACAAGCTCCCTGTCTGCCAGCCTGGGC GACAGAGTGACCATCTCCTGTAGGGCCTCTCAGGATATCAGCAAGTACCTGAA CTGGTATCAGCAGAAGCCAGATGGCACAGTGAAGCTGCTGATCTACCACACCT CCAGGCTGCACTCTGGAGTGCCAAGCCGGTTCTCCGGATCTGGAAGCGGCACC GACTATTCCCTGACAATCTCTAACCTGGAGCAGGAGGATATCGCCACATACTT TTGCCAGCAGGGCAATACCCTGCCATATACATTCGGCGGAGGAACCAAGCTG GAGATCACCGGATCCACATCTGGAAGCGGCAAGCCAGGAAGCGGAGAGGGAT CCACAAAGGGAGAGGTGAAGCTGCAGGAGAGCGGACCAGGACTGGTGGCAC CATCCCAGTCTCTGAGCGTGACCTGTACAGTGTCCGGCGTGTCTCTGCCTGACT ACGGCGTGTCCTGGATCAGGCAGCCACCTAGGAAGGGACTGGAGTGGCTGGG CGTGATCTGGGGCTCTGAGACCACATACTATAATTCTGCCCTGAAGAGCCGCC TGACCATCATCAAGGACAACTCCAAGTCTCAGGTGTTTCTGAAGATGAATAGC CTGCAGACCGACGATACAGCCATCTACTATTGCGCCAAGCACTACTATTACGG CGGCTCCTACGCCATGGATTATTGGGGCCAGGGCACCTCCGTGACAGTGTCTA GC (SEQ ID NO: 102) [0358] In some embodiments, the lentiviral particle comprises a polypeptide comprising a 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 103. [0359] MLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASLGDRVTISCRASQDISK YLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYF CQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVKLQESGPGLVAPSQSL SVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNS KSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQGTSVTVSS (SEQ ID NO: 103) [0360] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the Oa][KMOUU^UK[ NXVKSW XP K 97G MXVY[S\SWQ K h9:,4 \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] Attorney Ref. No.061479-506001WO 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 104. [0361] ATGCTGCTGCTGGTGACCAGCCTGCTGCTGTGCGAGCTGCCACACCCTG CCTTCCTGCTGATCCCAGATATCCAGATGACACAGACCACATCCTCTCTGTCCG CCTCTCTGGGCGACAGAGTGACCATCTCTTGTAGGGCCAGCCAGGATATCTCC AAGTACCTGAACTGGTATCAGCAGAAGCCTGACGGCACAGTGAAGCTGCTGA TCTACCACACCTCTAGGCTGCACAGCGGAGTGCCATCCCGGTTCAGCGGATCC GGATCTGGAACAGACTATTCTCTGACCATCAGCAACCTGGAGCAGGAGGATAT CGCCACATACTTTTGCCAGCAGGGCAATACCCTGCCATATACATTCGGCGGAG GAACCAAGCTGGAGATCACCGGAAGCACATCCGGATCTGGCAAGCCAGGATC CGGAGAGGGATCTACAAAGGGAGAGGTGAAGCTGCAGGAGAGCGGACCAGG ACTGGTGGCACCCAGCCAGTCCCTGTCTGTGACCTGTACAGTGTCTGGCGTGA GCCTGCCCGATTACGGCGTGTCCTGGATCAGACAGCCACCAAGGAAGGGACT GGAGTGGCTGGGCGTGATCTGGGGCTCTGAGACCACATACTATAATAGCGCCC TGAAGTCCCGGCTGACCATCATCAAGGACAACAGCAAGTCCCAGGTGTTTCTG AAGATGAATAGCCTGCAGACCGACGATACAGCCATCTACTATTGCGCCAAGC ACTACTATTACGGCGGCTCCTACGCCATGGATTATTGGGGCCAGGGCACCTCC GTGACAGTGAGCTCC (SEQ ID NO: 104) [0362] In some embodiments, the lentiviral particle comprises a polynucleotide encoding a 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 100. [0363] The complementary determining regions (CDR) of this scFv are RASQDISKYLN, (CDR-L1; SEQ ID NO: 94), HTSRLHS (CDR-L2; SEQ ID NO: 95), QQGNTLPYT (CDR-L3; SEQ ID NO: 96), DYGV (CDR-H1; SEQ ID NO: 97), VIWGSETTYYNSALKS (CDR-H2; SEQ ID NO: 98), HYYYGGSYAMDY (CDR-H3; SEQ ID NO: 99). In some embodiments, the lentiviral particle comprises a polynucleotide OWMXNSWQ K 97G `RX\O Oa][KMOUU^UK[ NXVKSW MXVY[S\O\ K h9:,4 \M<_ RK_SWQ ]RO\O 9:G\' `RO[OSW XY]SXWKUUb ]RO h9:,4 \M<_ \RK[O\ K] UOK\] 20#' K] UOK\] 3+#' K] UOK\] 30#' at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 100. [0364] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the Oa][KMOUU^UK[ NXVKSW XP K 97G MXVY[S\SWQ K h9:,4 \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] Attorney Ref. No.061479-506001WO 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 105. [0365] ATGCTGCTGCTGGTGACATCCCTGCTGCTGTGCGAGCTGCCACACCCAG CCTTCCTGCTGATCCCCGATATCCAGATGACCCAGACCACAAGCTCCCTGAGC GCCTCCCTGGGCGACAGGGTGACAATCTCTTGTCGGGCCAGCCAGGATATCTC CAAGTATCTGAATTGGTACCAGCAGAAGCCCGACGGCACCGTGAAGCTGCTG ATCTATCACACATCTAGACTGCACAGCGGCGTGCCTTCCAGGTTTTCTGGCAG CGGCTCCGGCACCGACTACTCTCTGACAATCAGCAACCTGGAGCAGGAGGAT ATCGCCACCTATTTCTGCCAGCAGGGCAATACCCTGCCTTACACATTTGGCGG CGGCACAAAGCTGGAGATCACCGGCTCTACAAGCGGATCCGGCAAGCCAGGA TCCGGAGAGGGATCTACCAAGGGAGAGGTGAAGCTGCAGGAGAGCGGACCTG GACTGGTGGCACCATCTCAGAGCCTGTCCGTGACCTGTACAGTGTCTGGCGTG AGCCTGCCAGATTATGGCGTGAGCTGGATCAGGCAGCCACCTAGGAAGGGAC TGGAGTGGCTGGGCGTGATCTGGGGCTCCGAGACCACATACTATAACAGCGCC CTGAAGTCCCGCCTGACCATCATCAAGGACAACTCTAAGAGCCAGGTGTTCCT GAAGATGAATTCCCTGCAGACCGACGATACAGCCATCTACTATTGCGCCAAGC ACTACTATTACGGCGGCTCTTATGCCATGGATTACTGGGGCCAGGGCACCAGC GTGACAGTGTCTAGC (SEQ ID NO: 105) [0366] In some embodiments, the CAR is a anti-FITC CAR and the ligand is composed of a fluorescein or fluorescein isothiocyanate (FITC) moiety conjugated to an agent that binds to a desired target cell (such as a cancer cell). Exemplary ligands are described in the section above. In some embodiments, the ligand is FITC-folate. [0367] In some embodiments, the CAR comprises an scFv domain. In some embodiments, the scFv domain comprises anti-fluorescein isothiocyanate (FITC) E2. In some embodiments, the scFv domain comprises a light chain variable domain (VL), a linker, and a heavy chain variable domain (VH). [0368] In some embodiments, the scFv VL comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ Attorney Ref. No.061479-506001WO ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL comprises the nucleotide sequence of SEQ ID NOs: 108 or 115. In some embodiments, the scFv VL consists of the nucleotide sequence of SEQ ID NOs: 108 or 115. [0369] In some embodiments, the scFv VL comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 109.In some embodiments, the scFv VL comprises the amino acid sequence of SEQ ID NO: 109. In some embodiments, the scFv VL consists the amino acid sequence of SEQ ID NO: 109. Attorney Ref. No.061479-506001WO [0370] In some embodiments, the scFv VH comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 112 or 117. In some embodiments, the scFv VH comprises the nucleotide sequence of SEQ ID NOs 112 or 117. In some embodiments, the scFv VH consists of the nucleotide sequence of SEQ ID NOs: 112 or 117. [0371] In some embodiments, the scFv VH comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 98% identical to the Attorney Ref. No.061479-506001WO amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH comprises the amino acid sequence of SEQ ID NO: 113. In some embodiments, the scFv VH consists the amino acid sequence of SEQ ID NO: 113. [0372] In some embodiments, the scFv linker comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker comprises the nucleotide sequence of SEQ ID NOs: 110 or 116. In some embodiments, the scFv linker consists the nucleotide sequence of SEQ ID NOs: 110 or 116. [0373] In some embodiments, the scFv linker comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ Attorney Ref. No.061479-506001WO ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 111.In some embodiments, the scFv linker comprises the amino acid sequence of SEQ ID NO: 111. In some embodiments, the scFv linker consists the amino acid sequence of SEQ ID NO: 111. [0374] In some embodiments, the scFv comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv comprises the nucleotide sequence of SEQ ID NOs: 106 or 114. In some embodiments, the scFv consists of the nucleotide sequence of SEQ ID NOs: 106 or 114. [0375] In some embodiments, the scFv comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of Attorney Ref. No.061479-506001WO SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv comprises the amino acid sequence of SEQ ID NO: 107. In some embodiments, the scFv consists of the amino acid sequence of SEQ ID NO: 107. [0376] In some embodiments, the viral particle comprises a nucleic acid encoding the Oa][KMOUU^UK[ NXVKSW XP K 97G MXVY[S\SWQ K h9:-+ \M<_ ]RK] \RK[O\ K] UOK\] 20#' K] UOK\] 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 174. In some embodiments, the viral particle comprises a nucleic acid encoding the Oa][KMOUU^UK[ NXVKSW XP K 97G MXVY[S\SWQ K h9:-+ \M<_ KWN MXVY[S\O\ KW KVSWX KMSN sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at UOK\] 44#' X[ ,++# SNOW]S]b ]X H;F ?: CD5 -,+) ?W \XVO OVLXNSVOW]\' ]RO h9:-+ \M<_ VL comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 176. In some OVLXNSVOW]\' ]RO h9:-+ \M<_ JA MXVY[S\O\ K W^MUOSM KMSN \OZ^OWMO K] UOK\] 3+#' 30#' 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ?: CD5 ,20) ?W \XVO OVLXNSVOW]\' ]RO h9:-+ \M<_ USWTO[ MXVY[S\O\ K W^MUOSM KMSN sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the KVSWX KMSN \OZ^OWMO XP H;F ?: CD5 ,22) ?W \XVO OVLXNSVOW]\' ]RO h9:-+ \M<_ J> comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 179. In some embodiments, the Attorney Ref. No.061479-506001WO h9:-+ \M<_ J> MXVY[S\O\ K W^MUOSM KMSN \OZ^OWMO K] UOK\] 3+#' 30#' 4+#' 40#' 41#' 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 178. [0377] In some embodiments, the viral particle comprises a nucleic acid encoding the extracellular domain of a CAR comprising a FLAG-tag that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 180. In some embodiments, the viral particle comprises a nucleic acid encoding the extracellular domain of a CAR comprising a FLAG-tag and the encoded amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 211. [0378] In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8a hinge and transmemebrane domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 181. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8a hinge and transmemebrane domain and the encoded amino acid sequence shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 212. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8a hinge domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 182. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8a transmemebrane domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 183. [0379] In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a 4-1BB co-stimulatory domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 184. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a 4-1BB co-stimulatory domain and the encoded amino acid sequence shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 213. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD3zeta signaling domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 185. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD3zeta signaling Attorney Ref. No.061479-506001WO domain and the encoded amino acid sequence shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 214. [0380] In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding an anti-CD20 CAR comprising a FLAG-tag that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 186. In some embodiments, the viral particle disclosed herein comprises a nucleic acid encoding an anti-CD20 CAR comprising a FLAG-tag and the encoded amino acid sequence shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 209. [0381] In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a modified IgG4 hinge domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 187, 188, or 189. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a PD1 hinge domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 190. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising an IgG1 hinge domain that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 191. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8 hinge domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 192. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD28 hinge domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 193. [0382] In some embodiments, the lentiviral particle comprises a nucleic acid encoding the extracellular domain of a CAR comprising an anti-CD19 scFv comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 194. In some embodiments, the lentiviral particle comprises a nucleic acid encoding the extracellular domain of a CAR comprising an anti-CD19 scFv comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: Attorney Ref. No.061479-506001WO ,40) ?W \XVO OVLXNSVOW]\' ]RO h9:,4 \M<_ JA MXVY[S\O\ KW KVSWX KMSN \OZ^OWMO K] UOK\] 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to ]RO KVSWX KMSN \OZ^OWMO XP H;F ?: CD5 -+1) ?W \XVO OVLXNSVOW]\' ]RO h9:-+ \M<_ \YKMO[ comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 207. In some embodiments, the h9:-+ \M<_ J> MXVY[S\O\ KW KVSWX KMSN \OZ^OWMO K] UOK\] 3+#' 30#' 4+#' 40#' 41#' 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 208. [0383] In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8 hinge domain comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 196. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD8 hinge domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 197. [0384] In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD28 transmembrane domain comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 198. In some embodiments, the lentiviral particle disclosed herein comprises a nucleic acid encoding a CAR comprising a CD28 transmembrane domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 199. [0385] In some embodiments, the lentiviral particle disclosed herein encodes a CAR comprising a 4-1BB co-stimulatory domain comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 200. In some embodiments, the lentiviral particle disclosed herein encodes a CAR comprising a 4-1BB co-stimulatory domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 201. In some embodiments, the lentiviral particle disclosed herein encodes a CAR comprising a CD3zeta signaling domain comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 202. In some embodiments, the lentiviral particle disclosed herein encodes a CAR comprising a CD3zeta Attorney Ref. No.061479-506001WO signaling domain comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 203. [0386] In some embodiments, the lentiviral particle comprises a nucleic acid encoding an anti-CD19 CAR comprising a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 204. In some embodiments, the lentiviral particle comprises a nucleic acid encoding an anti- CD19 CAR comprising an amino acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 205. Table 4B Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO Attorney Ref. No.061479-506001WO [0387] In some embodiments, the antigen is CD20. In some embodiments, a CAR comprises an extracellular domain comprising a scFv binding domain for CD20 binding. In some embodiments, the CAR is a second-generation CAR comprised of an anti-CD20 scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. In some embodiments, a CAR comprises a binding domain for CD20, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for CD20, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for CD20, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a Leu16 scFv binding domain for CD20 binding, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a Leu16 scFv binding domain for CD20 binding, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a Leu16 scFv binding domain for CD20 binding, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a 2B8 scFv binding domain for CD20 binding, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a 2B8 scFv binding domain for CD20 binding, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB Attorney Ref. No.061479-506001WO costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises an extracellular domain comprising a 2B8 scFv binding domain for CD20 binding, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. [0388] In some embodiments, the anti-fluorescein E2 scFv comprises a CDRL1, CDRL2, and CDRL3 having at least 80% amino acid identity, at least 90% amino acid identity or at least 95% amino acid identity to: TSNIGNNYVS (SEQ ID NO: 128), LMIYDVSKRPS (SEQ ID NO: 129), and AAWDDSLSEF (SEQ ID NO: 130), respectively, and CDRH1, CDRH2, and CDRH3 having at least 80% amino acid identity, at least 90% amino acid identity or at least 95% amino acid identity to: FTFGSFSMS (SEQ ID NO: 131), WVAGLSARSSLTHY (SEQ ID NO: 132), and RRSYDSSGYWGHFYSYMDV (SEQ ID NO: 133), respectively. [0389] In some embodiments, the CAR is a second-generation CAR comprised of the FMC63 mouse anti-human CD19 scFv linked to the CD28 costimulatory domain and the CD3zeta intracellular signaling domain. In some embodiments, the CAR is a second- generation CAR comprised of the FMC63 mouse anti-human CD19 scFv linked to a CD8 transmembrane domain, 4-1BB costimulatory domain, and the CD3zeta intracellular signaling domain. [0390] In some embodiments, the antigen is BCMA. CAR T therapies targeting BCMA have been approved by the FDA and include Abecma and Carvykti. CARs targeting BCMA are described, for example, in US Publication No. 2020/0246381; US Patent No. 10,918,665; US Publication No.2019/0161553; and US Publication No.2022/0033509 each of which is herein incorporated by reference. In some embodiments, a CAR comprises a binding domain for BCMA, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for BCMA, a CD8a hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for BCMA, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for BCMA, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. [0391] In some embodiments, the antigen is G protein-coupled receptor class C group 5 member D (GPRC5D). CARs targeting GPRC5D are described, for example, in US Attorney Ref. No.061479-506001WO Publication No.2018/0118803; US Publication No.2020/0231686, and US Publication No. 2021/10393689, each of which is herein incorporated by reference. In some embodiments, a CAR comprises a binding domain for GPRC5D, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for GPRC5D, a CD8a hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for GPRC5D, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for GPRC5D, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. [0392] In some embodiments, the antigen is Fc Receptor-like 5 (FcRL5). CARs targeting FcRL5 are described, for example, in US Publication No. US 2017/0275362, which is herein incorporated by reference. CARs may also comprise binding domains derived from known anti-FcRL5 antibodies including CD307e and REA391. In some embodiments, a CAR comprises a binding domain for FcRL5, a CD8a hinge, a CD8a transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for FcRL5, a CD8a hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for FcRL5, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for FcRL5, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. [0393] In some embodiments, the antigen is receptor tyrosine kinase-like orphan receptor 1 (ROR1). CARs targeting ROR1 are described, for example, in US Publication No. 2022/0096651, which is herein incorporated by reference. In some embodiments, a CAR comprises a binding domain for ROR1, a CD8a hinge, a CD8a transmembrane domain, a 4- 1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for ROR1, an IgG4 hinge, a CD28 transmembrane domain, a 4-1BB costimulatory domain, and a CD3zeta signaling domain. In some embodiments, a CAR comprises a binding domain for ROR1, a CD28 hinge, a CD28 transmembrane domain, a CD28 costimulatory domain, and CD3zeta signaling domain. Attorney Ref. No.061479-506001WO [0394] In some embodiments, the CAR is a second-generation CAR comprised an anti- BCMA scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. In some embodiments, the CAR is a second-generation CAR comprised an anti-GPRC5D scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. In some embodiments, the CAR is a second-generation CAR comprised an anti-ROR1 scFv linked to the 4-1BB costimulatory domain and the CD3zeta intracellular signaling domain. [0395] In some embodiments, the TAA or TSA is a cancer/testis (CT) antigen, e.g., BAGE, CAGE, CTAGE, FATE, GAGE, HCA661, HOM-TES-85, MAGEA, MAGEB, MAGEC, NA88, NY-ESO-1, NY-SAR-35, OY-TES-1, SPANXB1, SPA17, SSX, SYCP1, or TPTE. [0396] In some embodiments, the TAA or TSA is a carbohydrate or ganglioside, e.g., fuc- GM1, GM2 (oncofetal antigen-immunogenic-1; OFA-I-1); GD2 (OFA-I-2), GM3, GD3, and the like. [0397] In some embodiments, the TAA or TSA is alpha-actinin-4, Bage-1, BCR-ABL, Bcr- Abl fusion protein, beta-catenin, CA 125, CA 15-3 (CA 27.29\BCAA), CA 195, CA 242, CA-50, CAM43, Casp-8, cdc27, cdk4, cdkn2a, CEA, coa-1, dek-can fusion protein, EBNA, EF2, Epstein Barr virus antigens, ETV6-AML1 fusion protein, HLA-A2, HLA-All, hsp70- -' @?77D-+0' BK[]-' B^V(,' -' KWN .' WOX(E7E' VbX\SW MUK\\ ?' DH(4' YVU(G7Gh P^\SXW protein, PTPRK, K-ras, N-ras, triosephosphate isomerase, Gage 3,4,5,6,7, GnTV, Herv-K- mel, Lage-1, NA-88, NY-Eso-1/Lage-2, SP17, SSX-2, TRP2-Int2, gp100 (Pmel 17), tyrosinase, TRP-1, TRP-2, MAGE-1, MAGE-3, RAGE, GAGE-1, GAGE-2, p15(58), RAGE, SCP-1, Hom/Mel-40, PRAME, p53, H-Ras, HER-2/neu, E2A-PRL, H4-RET, IGH- IGK, MYL-RAR, human papillomavirus (HPV) antigens E6 and E7, TSP-180, MAGE-4, MAGE-5, MAGE-6, p185erbB2, p180erbB-3, c-met, nm-23H1, PSA, TAG-72-4, CA 19- 9, CA 72-4, CAM 17.1, NuMa, K-ras, 13-Catenin, Mum-1, p16, TAGE, PSMA, CT7, telomerase, 43-9F, 5T4, 791Tgp72, 13HCG, BCA225, BTAA, CD68\KP1, CO-029, FGF- 5, G250, Ga733 (EpCAM), HTgp-175, M344, MA-50, MG7-Ag, MOV18, NB\70K, NY- CO-1, RCAS1, SDCCAG16, TA-90, TAAL6, TAG72, TLP, TPS, CD19, CD20, CD22, CD27, CD30, CD70, CD123, CD133, B-cell maturation antigen, CS1, GPCR5, GD2 (ganglioside G2), EGFRvIII (epidermal growth factor variant III), sperm protein 17 (Sp17), mesothelin, PAP (prostatic acid phosphatase), prostein, TARP (T cell receptor gamma alternate reading frame protein), Trp-p8, STEAP1 (six-transmembrane epithelial antigen of the prostate 1), an abnormal ras protein, or an abnormal p53 protein. In some embodiments, Attorney Ref. No.061479-506001WO \KSN ]^VX[(K\\XMSK]ON KW]SQOW X[ ]^VX[(\YOMSPSM KW]SQOW S\ SW]OQ[SW h_k. $9:1,%' QKUKM]SW' K-Ras (V-Ki-ras2 Kirsten rat sarcoma viral oncogene), or Ral-B. Other tumor-associated and tumor-specific antigens may be used. [0398] Antibodies, and scFvs, that bind to TSAs and TAAs include antibodies and scFVs that are known in the art, as are nucleotide sequences that encode them. [0399] In some embodiments, the antigen is an antigen not considered to be a TSA or a TAA, but which is nevertheless associated with tumor cells, or damage caused by a tumor. In some embodiments, for example, the antigen is, e.g., a growth factor, cytokine or interleukin, e.g., a growth factor, cytokine, or interleukin associated with angiogenesis or vasculogenesis. Such growth factors, cytokines, or interleukins can include, e.g., vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), platelet growth factor (PDGF), hepatocyte growth factor (HGF), insulin-like growth factor (IGF), or interleukin-8 (IL-8). Tumors can also create a hypoxic environment local to the tumor. As such, in some embodiments, the antigen is a hypoxia-associated factor, e.g.' >?<(,h' >?<( ,k' >?<(-K' >?<(-k' >?<(.h' X[ >?<(.k) I^VX[\ MKW KU\X MK^\O UXMKUScON NKVKQO ]X WX[VKU tissue, causing the release of molecules such as damage associated molecular pattern molecules (DAMPs; also known as alarmins). In some embodiments, therefore, the antigen is a DAMP, e.g., a heat shock protein, chromatin-associated protein high mobility group box 1 (HMGB1), S100A8 (MRP8, calgranulin A), S100A9 (MRP14, calgranulin B), serum amyloid A (SAA), or can be a deoxyribonucleic acid, adenosine triphosphate, uric acid, or heparin sulfate. [0400] In some embodiments of the polypeptides described herein, the extracellular domain is joined to said transmembrane domain directly or by a linker, spacer or hinge polypeptide sequence, e.g., a sequence from CD28 or a sequence from CTLA4. [0401] In some embodiments, the extracellular domain that binds the desired antigen may be from antibodies or their antigen binding fragments generated using the technologies described herein. Universal CARs [0402] In some embodiments, the viral particle described herein comprises a nucleotide sequence encoding a universal CAR. Universal CARs allow for targeting to a cancer cell without the need to change the antigen specificity of the CAR. Universal CARs are Attorney Ref. No.061479-506001WO described, for example, in US Publication Nos. US 2016/0348073, US 2018/0085399, US 2019/0256597, and US 2014/0349402, each of which is herein incorporated by reference. [0403] In some embodiments, the viral particle described herein comprises a nucleotide sequence encoding a universal, modular, anti-tag chimeric antigen receptor (UniCAR). This system allows for retargeting of UniCAR engrafted immune cells against multiple antigens (see e.g., US Patent Publication US20170240612 A1 incorporated herein by reference in its entirety; Cartellieri et al., (2016) Blood Cancer Journal 6, e458 incorporated herein by reference in its entirety). [0404] In some embodiments, the viral particle described herein comprises a nucleotide sequence encoding a switchable CAR and/or CAR effector cell (CAR-EC) switches. In this system, the CAR -EC switches have a first region that is bound by a CAR on the CAR -EC and a second region that binds a cell surface molecule on a target cell, thereby stimulating an immune response from the CAR -EC that is cytotoxic to the bound target cell. In some embodiments, the CAR -EC switch may act as an “on-switch” for CAR -EC activity. Activity may be “turned off” by reducing or ceasing administration of the switch. These CAR-EC switches may be used with CAR-ECs disclosed herein, as well as existing CAR T-cells, for the treatment of a disease or condition, such as cancer, wherein the target cell is a malignant cell. Such treatment may be referred to herein as switchable immunotherapy (US Patent Publication US9624276 B2 incorporated herein by reference in its entirety). [0405] In some embodiments, the viral particle comprises a nucleotide sequence encoding a universal immune receptor (e.g., switchable CAR, sCAR) that binds a peptide neo-epitope (PNE). In some embodiments, the peptide neo-epitope (PNE) has been incorporated at defined different locations within an antibody targeting an antigen (antibody switch). Therefore, sCAR-T-cell specificity is redirected only against PNE, not occurring in the human proteome, thus allowing an orthogonal interaction between the sCAR-T-cell and the antibody switch. In this way, sCAR-T cells are strictly dependent on the presence of the antibody switch to become fully activated, thus excluding CAR T-cell off-target recognition of endogenous tissues or antigens in the absence of the antibody switch (Arcangeli et al., (2016) Transl Cancer Res 5(Suppl 2):S174-S177 incorporated herein by reference in its entirety). Other examples of switchable CARs is provided by US Patent Application US20160272718A1 incorporated herein by reference in its entirety. Attorney Ref. No.061479-506001WO [0406] As used herein, the term “tag” encompasses a universal immune receptor, a tag, a switch, or an Fc region of an immunoglobulin as described supra. In some embodiments, a viral particle comprises a nucleotide sequence encoding a CAR comprising a tag binding domain. In some embodiments, the CAR binds fluorescein isothiocyanate (FITC), streptavidin, biotin, dinitrophenol, peridinin chlorophyll protein complex, green fluorescent protein, phycoerythrin (PE), horse radish peroxidase, palmitoylation, nitrosylation, alkalanine phosphatase, glucose oxidase, or maltose binding protein. [0407] In some embodiments, the viral particle comprises a nucleotide sequence encoding a CAR to generate CAR cells to be used with a targeting small molecule. In some embodiments, the CAR targets a moiety that is not produced or expressed by cells of the subject being treated. This CAR system thus allows for focused targeting of the immune cells to target cells, such as cancer cells. The two-component CAR system has been previously described in e.g., US 2015/0320799; US 2019/0224237; and US 2020/0023009, each of which is herein incorporated by reference. [0408] In some embodiments, the targeting small molecule comprises a ligand of a tumor cell receptor. By administration of a targeting small molecule along with the CAR- expressing immune cell , the immune cell response can be targeted to only those cells expressing the tumor receptor, thereby reducing off-target toxicity, and the activation of immune cells can be more easily controlled due to the rapid clearance of the targeting small molecule. As an added advantage, the CAR-expressing immune cell can be used as a universal cytotoxic cell to target a wide variety of tumors without the need to prepare separate CAR constructs. The targeting small molecule recognized by the CAR may also remain constant. It is only the ligand portion of the targeting small molecule that needs to be altered to allow the system to target cancer cells of different identity. [0409] In some embodiments, a targeting small molecule comprises fluorescein linked to a ligand of a selected tumor cell receptor. In some embodiments, a targeting small molecule comprises FITC linked to a ligand of a selected tumor cell receptor. In some embodiments, the viral vector described herein encodes a CAR comprising an anti-fluorescein scFv. In some embodiments, the viral vector described herein encodes a CAR comprising an anti- FITC scFv. This CAR thus targets fluorescein or FITC instead of a tumor-associated antigen that might also be expressed by healthy, non-target cells. The two components are administered to a subject having cancer and the targeting small molecule is bound by the target tumor cells (through binding of the ligand portion of the molecule to cognate tumor Attorney Ref. No.061479-506001WO cell receptor). The FITC portion of the targeting small molecule is then recognized and bound by the anti-FITC CAR expressed by the T cells (second component). Upon binding, the anti-FITC CAR-expressing immune cells are activated and the tumor cell is killed. As will be apparent to the skilled artisan, the immune cells cannot kill cells without first binding to a tumor cell. As it will be further apparent, immune cells will not bind to non-target cells because the recognition region of the CAR will only recognize and bind FITC, which is not produced or expressed by cells of the subject. The targeting small molecule thus acts as a bridge between the immune cells and the target tumor cells. As long as the targeted moiety of the targeting small molecule is a moiety not found in the host, the activity of the immune cells can be limited to the target cells. Further, the activation of the CAR-expressing immune cells can be regulated by limiting the amount of targeting small molecule administered to a subject, for example, by manipulating infusion of the targeting small molecule if a side effect is detected. Illustrative anti-fluorescein and anti-FITC CARs are described in US Patent Application US20200405760A1 incorporated herein by reference in its entirety. [0410] In some embodiments, the targeting small molecule comprises 2,4-dinitrophenol (DNP), 2,4,6-trinitrophenol (TNP), biotin, digoxigenin, fluorescein, fluorescein isothiocyanate (FITC), NHS-fluorescein, pentafluorophenyl ester, tetrafluorophenyl ester, a knottin, a centyrin, a DARPin, an affibody, an affilin, an anticalin, an atrimer, an avimer, a bicicyclic peptide, an FN3 scaffold, a cys-knot, a fynomer, a Kunitz domain, or an Obody. In some embodiments, the viral particle comprises a nucleotide sequence encoding a CAR comprising an extracellular binding domain that binds 2,4-dinitrophenol (DNP), 2,4,6- trinitrophenol (TNP), biotin, digoxigenin, fluorescein, fluorescein isothiocyanate (FITC), NHS-fluorescein, pentafluorophenyl ester, tetrafluorophenyl ester, a knottin, a centyrin, a DARPin, an affibody, an affilin, an anticalin, an atrimer, an avimer, a bicicyclic peptide, an FN3 scaffold, a cys-knot, a fynomer, a Kunitz domain, or an Obody. [0411] In some embodiments, the CAR system utilizes conjugate molecules as the bridge between CAR-expressing cells and targeted cancer cells. The conjugate molecules are conjugates comprising a hapten and a cell-targeting moiety, such as any suitable tumor cell- specific ligand. Illustrative haptens that can be recognized and bound by CARs, include small molecular weight organic molecules such as DNP (2,4-dinitrophenol), TNP (2,4,6- trinitrophenol), biotin, and digoxigenin, along with fluorescein and derivatives thereof, including FITC (fluorescein isothiocyanate), NHS-fluorescein, and pentafluorophenyl ester (PFP) and tetrafluorophenyl ester (TFP) derivatives, a knottin, a centyrin, and a DARPin. Attorney Ref. No.061479-506001WO Suitable cell-targeting moiety that may themselves act as a hapten for a CAR include knottins (see Kolmar H. et al., The FEBS Journal. 2008.275(11):26684-90), centyrins, and DARPins (see Reichert, J.M. MAbs 2009.1(3):190–209). [0412] In some embodiments, the cell-targeting moiety is DUPA (DUPA-(99m) Tc), a ligand bound by PSMA-positive human prostate cancer cells with nanomolar affinity (KD = 14 nM; see Kularatne, S.A. et al., Mol Pharm. 2009. 6(3):780-9). In some embodiments, a DUPA derivative can be the ligand of the small molecule ligand linked to a targeting moiety, and DUPA derivatives are described in WO 2015/057852, incorporated herein by reference. [0413] In some embodiments, the cell-targeting moiety is CCK2R ligand, a ligand bound by CCK2R-positive cancer cells (e.g., cancers of the thyroid, lung, pancreas, ovary, brain, stomach, gastrointestinal stroma, and colon; see Wayua. C. et al., Molecular Pharmaceutics. 2013. ePublication). [0414] In some embodiments, the cell-targeting moiety is folate, folic acid, or an analogue thereof, a ligand bound by the folate receptor on cells of cancers that include cancers of the ovary, cervix, endometrium, lung, kidney, brain, breast, colon, and head and neck cancers; see Sega, E.I. et al., Cancer Metastasis Rev.2008.27(4):655-64). [0415] In some embodiments, the cell-targeting moiety is an NK-1R ligand. Receptors for NK-1R the ligand are found, for example, on cancers of the colon and pancreas. In some embodiments, the NK-1R ligand may be synthesized according the method disclosed in Int’l Patent Appl. No. PCT/US2015/044229, incorporated herein by reference. [0416] In some embodiments, the cell-targeting moiety may be a peptide ligand, for example, the ligand may be a peptide ligand that is the endogenous ligand for the NK1 receptor. In some embodiments, the small conjugate molecule ligand may be a regulatory peptide that belongs to the family of tachykinins which target tachykinin receptors. Such regulatory peptides include Substance P (SP), neurokinin A (substance K), and neurokinin B (neuromedin K), (see Hennig et al., International Journal of Cancer: 61, 786-792). [0417] In some embodiments, the cell-targeting moiety is a CAIX ligand. Receptors for the CAIX ligand found, for example, on renal, ovarian, vulvar, and breast cancers. The CAIX ligand may also be referred to herein as CA9. [0418] In some embodiments, the cell-targeting moiety is a ligand of gamma glutamyl transpeptidase. The transpeptidase is overexpressed, for example, in ovarian cancer, colon cancer, liver cancer, astrocytic gliomas, melanomas, and leukemias. Attorney Ref. No.061479-506001WO [0419] In some embodiments, the cell-targeting moiety is a CCK2R ligand. Receptors for the CCK2R ligand found on cancers of the thyroid, lung, pancreas, ovary, brain, stomach, gastrointestinal stroma, and colon, among others. [0420] In some embodiments, the cell-targeting moiety may have a mass of less than about 100,000 Daltons, less than about 90,000 Daltons, less than about 80,000 Daltons, less than about 70,000 Daltons, less than about 60,000 Daltons, less than about 50,000 Daltons, less than about 40,000 Daltons, less than about 30,000 Daltons, less than about 20,000 Daltons, less than about 10,000 Daltons, less than about 9000 Daltons, less than about 8,000 Daltons, less than about 7000 Daltons, less than about 6000 Daltons, less than about 5000 Daltons, less than about 4500 Daltons, less than about 4000 Daltons, less than about 3500 Daltons, less than about 3000 Daltons, less than about 2500 Daltons, less than about 2000 Daltons, less than about 1500 Daltons, less than about 1000 Daltons, or less than about 500 Daltons. In another embodiment, the small molecule ligand may have a mass of about 1 to about 100,000 Daltons, about 1 to about 90,000 Daltons, about 1 to about 80,000 Daltons, about 1 to about 70,000 Daltons, about 1 to about 60,000 Daltons, about 1 to about 50,000 Daltons, about 1 to about 40,000 Daltons, about 1 to about 30,000 Daltons, about 1 to about 20,000 Daltons, about 1 to about 10,000 Daltons, about 1 to about 9000 Daltons, about 1 to about 8,000 Daltons, about 1 to about 7000 Daltons, about 1 to about 6000 Daltons, about 1 to about 5000 Daltons, about 1 to about 4500 Daltons, about 1 to about 4000 Daltons, about 1 to about 3500 Daltons, about 1 to about 3000 Daltons, about 1 to about 2500 Daltons, about 1 to about 2000 Daltons, about 1 to about 1500 Daltons, about 1 to about 1000 Daltons, or about 1 to about 500 Daltons. [0421] In some embodiments, the linkage in a conjugate described herein can be a direct linkage (e.g., a reaction between the isothiocyanate group of FITC and a free amine group of a small molecule ligand) or the linkage can be through an intermediary linker. In some embodiments, if present, an intermediary linker can be any biocompatible linker, such as a divalent linker. In some embodiments, the divalent linker can comprise about 1 to about 30 carbon atoms. In another illustrative embodiment, the divalent linker can comprise about 2 to about 20 carbon atoms. In other embodiments, lower molecular weight divalent linkers (i.e., those having an approximate molecular weight of about 30 to about 300 Da) are employed. In another embodiment, linkers lengths that are suitable include, but are not limited to, linkers having 2, 3, 4, 5, 6, 7, 8, 9.10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37.38, 39 or 40, or more atoms. Attorney Ref. No.061479-506001WO [0422] In some embodiments, the hapten and the cell-targeting moiety can be directly conjugated through such means as reaction between the isothiocyanate group of FITC and free amine group of small ligands (e.g., folate, DUPA, and CCK2R ligand). However, the use of a linking domain to connect the two molecules may be helpful as it can provide flexibility and stability. Examples of suitable linking domains include: 1) polyethylene glycol (PEG); 2) polyproline; 3) hydrophilic amino acids; 4) sugars; 5) unnatural peptideoglycans; 6) polyvinylpyrrolidone; 7) pluronic F-127. Linker lengths that are suitable include, but are not limited to, linkers having 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40, or more atoms. [0423] In some embodiments, the linker may be a divalent linker that may include one or more spacers. [0424] Illustrative conjugates of the disclosure include the following molecules: FITC- (PEG) 12 -Folate, FITC-(PEG) 20 -Folate, FITC-(PEG) 108 -Folate, FITC-DUPA, FITC- (PEG) 12 -DUPA, FITC-CCK2R ligand, FITC-(PEG) 12 -CCK2R ligand, FITC-(PEG) 11 - NK1R ligand and FITC-(PEG)2-CA9. [0425] While the affinity at which the ligands and cancer cell receptors bind can vary, and in some cases, low affinity binding may be preferable (such as about 1 µM), the binding affinity of the ligands and cancer cell receptors may generally be at least about 100 µM, 1 nM, 10 nM, or 100 nM, at least about 1 pM or 10 pM, or at least about 100 pM. [0426] Examples of conjugates and methods of making them are provided in U.S. patent applications US 2017/0290900, US 2019/0091308, and US 2020/0023009, each of which are incorporated herein by reference in its entirety. Particles [0427] In some embodiments, the disclosure provides particles of various types, including but not limited to lentiviral particles (i.e., a virion), lipid nanoparticles (LNPs), lipoplexes, liposomes, and nanocarriers. The particle may include an adhesion molecule, a costimulatory molecule, an activation molecule, a combination thereof. Any of the adhesion molecule, costimulatory molecule, or activation may be included in a fusion molecule. The adhesion molecule, costimulatory molecule, activation molecule, or combination thereof may be included at a surface of the particle. The fusion molecule may be included at a surface of the particle. Attorney Ref. No.061479-506001WO [0428] The particle may be a lipid nanoparticle (LNP) or a poly(beta-amino) esters (PBAE) nanocarriers, both of which have been shown to transduce T cells when administered to a subject in vivo or contacted with T cells ex vivo. Using the compositions and methods described herein, transduction of T cells with LNPs and PBAE-based nanocarriers may be increased. [0429] In some embodiments, the particle is a viral particle. Methods for generating viral vectors from various virus types are known in the art. Exemplary types of viral particles that may be recombinantly engineered as delivery vehicles include retroviruses, lentivirus (e.g. HIV and its derivatives and SIV), adeno-associated virus, adenovirus, MMLV retrovirus, MSCV retrovirus, baculovirus, vesicular stomatitis virus, herpes simplex virus, and vaccinia virus. Examples include adeno-associated virus (AAV) particles used for gene therapy. In a preferred embodiment, the particle is a retroviral particle. In a particularly preferred embodiment, the particle is a lentiviral particle. [0430] Lentiviral particles may be made using packaging cell lines as described in WO 2016/139463 or by using a polycistronic vector as described in Int’l Pat. Pub. No. WO 2020/106992 A1. Each of the foregoing specifically describes methods for making lentiviral particles. Their disclosures are incorporated by reference herein. Numerous other methods for making viral particles, including lentiviral particles, may be useful. [0431] Retroviruses, a group that includes lentiviruses, are enveloped viruses. The fusion molecules described herein may be displayed on such enveloped viruses by expressing the fusion molecule, or its various components, in a host cell under the control of a suitable promotor (or promoters). Each component may include a signal sequence for secretion. At least one component should include a transmembrane region or anchor sequence (such as ]RO 9o]O[VSWKU \SQWKU \OZ^OWMO ]RK] NS[OM]\ ]RO K]]KMRVOW] XP K =E? KWMRX[%) IRO X]RO[ components may associate with the first component either during the secretion process or after secretion. In the case of single fusion proteins, only one transmembrane region or anchor sequence may be required, although those of skill in the art may envision and employ multiple transmembrane regions or anchor sequences. In some embodiments, the fusion molecule is a fusion protein comprising a C-terminal transmembrane region, expressed from a polynucleotide that encodes an N-terminal signal peptide. The signal peptide may be cleaved during expression of the protein at the cell surface of the producer cell, leaving a membrane-tethered fusion protein without the signal sequence. The lentiviral particle may Attorney Ref. No.061479-506001WO then be made when a virion buds from the surface of the producer cell, incorporating as its envelope portions of the cell membrane that include one or more copies of the fusion protein. [0432] Lentiviral particles generally package a vector genome and, incidentally or intentionally, may package other molecules present in the producer cell. The vector genome may be an artificial vector genome engineered to encode a heterologous protein or polynucleotide. [0433] Lentiviral particles may contain structural and/or functional genetic elements that are primarily from a virus. Lentiviral particles are characterized by the predominant source of genetic or structural material in the lentiviral particle. Thus, the term “retroviral particle” refers to a viral particle containing structural proteins and vector genome elements, primarily from a retrovirus. Likewise, the term “lentiviral particle” refers to a viral particle containing structural proteins and vector genome elements, primarily from a lentivirus. To package its vector genome, a lentiviral particle may generally require at least one copy of the long-terminal repeats (LTRs) that flank a native lentiviral vector genome, or functional variants thereof. [0434] In some embodiments, a viral particle comprises a viral glycoprotein. In some embodiments, the viral particle comprises a viral glycoprotein different from the native viral glycoprotein. When the viral glycoprotein is heterologous to the vector genome, the viral particle is termed a “pseudotyped” viral particle. For example, in some embodiments, the viral particle is derived from HIV, which typically includes the glycoprotein gp120. However, such HIV-based particles may be “pseudotyped” and, instead of expressing their native glycoprotein, express a glycoprotein from a different virus. For example, the viral glycoprotein may be a portion of RD114 or one of its variants, VSV-G, Gibbon-ape leukemia virus (GALV), the Amphotropic envelope glycoprotein, Measles envelope glycoprotein, or baboon retroviral envelope glycoprotein. In some embodiments, the viral envelope glycoprotein is a G protein from the Cocal strain (Cocal G), or a functional variant thereof. Illustrative viral glycoproteins include the VSV G protein, the Cocal G protein, and variants thereof. Illustrative viral glycoproteins may be expressed as a single protein or in multiple subunits or parts. The viral glycoprotein may serve as ligand for cell-surface receptors on a target cell, and thereby promote transduction or the target cell. The viral glycoprotein may be engineered to lack LDLR binding affinity—for example, by mutation at positions 47 (e.g., K47Q) and/or 354 (e.g., R354A). This may be termed a “blinded” viral glycoprotein. Illustrative envelope variants are provided in, e.g., US 2020/0216502 A1, Attorney Ref. No.061479-506001WO which is incorporated herein by reference in its entirety. Surprisingly, in some embodiments, the fusion molecules as described herein may permit use of a viral glycoprotein that does not, by itself, cause transduction of target cell. Without being bound by theory, it is believed that the fusion protein may serve as a ligand for cell-surface receptor while the viral glycoprotein retains a structural function, but not a function as a ligand for a cell-surface receptor. [0435] In some embodiments, the viral glycoprotein is a VSV-G glycoprotein that comprises a mutation at position 47. In some embodiments, the viral glycoprotein is a VSV- G glycoprotein that comprises a mutation at position 354. In some embodiments, the viral glycoprotein is a VSV-G glycoprotein that comprises a K47Q mutation. In some embodiments, the viral glycoprotein is a VSV-G glycoprotein that comprises a R354A mutation. In some embodiments, the viral glycoprotein is a VSV-G glycoprotein that comprises a K47Q and a R354A mutation. In some embodiments, the viral glycoprotein is a cocal glycoprotein that comprises a mutation at position 47. In some embodiments, the viral glycoprotein is a cocal glycoprotein that comprises a mutation at position 354. In some embodiments, the viral glycoprotein is a cocal glycoprotein that comprises a K47Q mutation. In some embodiments, the viral glycoprotein is a cocal glycoprotein that comprises a R354A mutation. In some embodiments, the viral glycoprotein is a cocal glycoprotein that comprises a K47Q and a R354A mutation. [0436] In some embodiments, the viral glycoprotein comprises a mutation at position 47. In some embodiments, the viral glycoprotein comprises a mutation at position 354. In some embodiments, the viral glycoprotein comprises a K47Q mutation. In some embodiments, the viral glycoprotein comprises a R354A mutation. In some embodiments, the viral glycoprotein comprises a K47Q and a R354A mutation. [0437] The Cocal G protein may have a polypeptide sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to the following sequence: [0438] NFLLLTFIVLPLCSHAKFSIVFPQSQKGNWKNVPSSYHYCPSSSDQNWHND LLGITMKVKMPKTHKAIQADGWMCHAAKWITTCDFRWYGPKYITHSIHSIQPTSE QCKESIKQTKQGTWMSPGFPPQNCGYATVTDSVAVVVQATPHHVLVDEYTGEWI DSQFPNGKCETEECETVHNSTVWYSDYKVTGLCDATLVDTEITFFSEDGKKESIG KPNTGYRSNYFAYEKGDKVCKMNYCKHAGVRLPSGVWFEFVDQDVYAAAKLP ECPVGATISAPTQTSVDVSLILDVERILDYSLCQETWSKIRSKQPVSPVDLSYLAPK Attorney Ref. No.061479-506001WO NPGTGPAFTIINGTLKYFETRYIRIDIDNPIISKMVGKISGSQTERELWTEWFPYEGV EIGPNGILKTPTGYKFPLFMIGHGMLDSDLHKTSQAEVFEHPHLAEAPKQLPEEET LFFGDTGISKNPVELIEGWFSSWKSTVVTFFFAIGVFILLYVVARIVIAVRYRYQGS NNKRIYNDIEMSRFRK (SEQ ID NO: 74). [0439] The Cocal G protein may have a polypeptide sequence having at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% sequence identity to the following sequence: [0440] MNFLLLTFIVLPLCSHAKFSIVFPQSQKGNWKNVPSSYHYCPSSSDQNWHN DLLGITMKVKMPKTHKAIQADGWMCHAAKWITTCDFRWYGPKYITHSIHSIQPTS EQCKESIKQTKQGTWMSPGFPPQNCGYATVTDSVAVVVQATPHHVLVDEYTGE WIDSQFPNGKCETEECETVHNSTVWYSDYKVTGLCDATLVDTEITFFSEDGKKESI GKPNTGYRSNYFAYEKGDKVCKMNYCKHAGVRLPSGVWFEFVDQDVYAAAKL PECPVGATISAPTQTSVDVSLILDVERILDYSLCQETWSKIRSKQPVSPVDLSYLAP KNPGTGPAFTIINGTLKYFETRYIRIDIDNPIISKMVGKISGSQTERELWTEWFPYEG VEIGPNGILKTPTGYKFPLFMIGHGMLDSDLHKTSQAEVFEHPHLAEAPKQLPEEE TLFFGDTGISKNPVELIEGWFSSWKSTVVTFFFAIGVFILLYVVARIVIAVRYRYQG SNNKRIYNDIEMSRFRK (SEQ ID NO: 247) [0441] Illustrative lentiviruses include but are not limited to: HIV (human immunodeficiency virus; including HIV type 1, and HIV type 2; visna-maedi virus (VMV) virus; the caprine arthritis-encephalitis virus (CAEV); equine infectious anemia virus (EIAV); feline immunodeficiency virus (FIV); bovine immune deficiency virus (BIV); and simian immunodeficiency virus (SIV). In some embodiments, the backbones are HIV-based vector backbones (i.e., HIV cis-acting sequence elements). Retroviral particles have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted, making the vector biologically safe. [0442] Illustrative lentiviral particles and methods for making them are described in Naldini et al. Science 272:263-7 (1996); Zufferey et al. J. Virol.72:9873-9880 (1998); Dull et al. J. Virol. 72:8463-8471 (1998); Miyoshi et al. J. Virol. 72:8150-57 (1998); U.S. Pat. No. 6,013,516; and U.S. Pat. No.5,994,136. [0443] Protocols for producing replication-defective recombinant viruses are provided in W095/14785, W096/22378, U.S. Pat. No.5,882,877, U.S. Pat. No.6,013,516, U.S. Pat. No. 4,861,719, U.S. Pat. No.5,278,056, and W094/19478. Attorney Ref. No.061479-506001WO [0444] Viral particles may be assessed in various ways, including, for example, measuring the vector copy number (VCN) or vector genomes (vg) in a sample of viral particle by quantitative polymerase chain reaction (qPCR) or digital droplet PCR (ddPCR), or testing to the viral particles on target cells to measure a “titer” of the virus in, e.g., infectious units per milliliter (IU/mL). For example, the titer may be assessed using a functional assay performed on the cultured tumor cell line HT1080 as described in Humbert et al. Molecular Therapy 24:1237–1246 (2016). When titer is assessed on a cultured cell line that is continually dividing, stimulation may be unnecessary, and hence the measured titer may be uninfluenced by surface engineering of the retroviral particle. Other methods for assessing the efficiency of retroviral vector systems are provided in Gaererts et al. BMC Biotechnol. 6:34 (2006). Payloads [0445] The particle may be used to deliver a payload. The term “payload” refers to any molecule or combination of molecules whose delivery to a target cell is desired. Various payloads may be delivered using the particles desired herein, including but not limited to small molecules, polynucleotide and proteins. When the selected target cell is a T cell, the particles of the disclosure may be used to deliver a therapeutic agent targeting T cells, to genetically modified T cells, or to deliver a polynucleotide encoding a protein of interest to the T cell. Similarly, particles disclosed herein may be used to delivery payloads to NK cells. [0446] The payload may be a polynucleotide, such as a polynucleotide whose sequence encodes a protein or a non-coding nucleic acid (e.g., shRNA, microRNA, or siRNA). The polynucleotide may be an RNA, such as a messenger RNA (mRNA) or the vector genome of an RNA virus. It may be a DNA, such as the vector genome of a DNA virus. [0447] The payload may be a polynucleotide comprising a polynucleotide encoding a chimeric antigen receptor (CAR). Illustrative CARs, and polynucleotide encoding them, are described herein. CARs useful in the present disclosure are also provided in in U.S. Pat. Nos.7,741,465; 9,856,322 and 8,399,964. [0448] In some embodiments, the CAR is a CAR that specifically binds CD19. CAR T therapies targeting CD19 have been approved by the FDA and include YESCARTA, TECARTUS, KYMRIAH AND BREYANZI. CARs targeting CD19 are described, for Attorney Ref. No.061479-506001WO example, in U.S. Pat. Pub. No. 20160152723; and U.S. Pat. Nos. 10,736,918; 10,357,514; and 7,446,190. [0449] The CAR may specifically binds BCMA. CAR T therapies targeting BCMA have been approved by the FDA and include ABECMA and CARVYKTI. CARs targeting BCMA are described, for example, in U.S. Pat. Pub. Nos.2020/0246381 and 2019/0161553; and U.S. Pat. Nos.10,918,665. [0450] The CAR may specifically bind G protein-coupled receptor class C group 5 member D (GPRC5D), as described, for example, in U.S. Pat. Pub. Nos. 2018/0118803 and 2021/10393689. [0451] The CAR may specifically bind Fc Receptor-like 5 (FcRL5), as described, for example, in U.S. Pat. Pub. No. US 2017/0275362. [0452] The CAR may specifically bind receptor tyrosine kinase-like orphan receptor 1 (ROR1), as described, for example, in U.S. Pat. Pub. No.2022/0096651. [0453] In some embodiments, the particle described herein comprises a polynucleotide having a polynucleotide sequence encoding a universal CAR. Universal CARs allow for targeting to a cancer cell without the need to change the antigen specificity of the CAR. Universal CARs are described, for example, in U.S. Pat. Pub. Nos. 2016/0348073, 2018/0085399, 2019/0256597, and 2014/0349402. Use of universal CARs are also described in U.S. Pat. Pub. Nos. 2015/0320799, 2019/0224237, US 2020/0023009. [0454] In some embodiments, the particle described herein comprises a polynucleotide having a polynucleotide sequence encoding a universal, modular, anti-tag chimeric antigen receptor (UniCAR) (U.S. Pat. Pub. No. 20170240612; Cartellieri et al., Blood Cancer J. 6:e458 (2016)) or a switchable CAR (U.S. Pat. No. 9,624,276 and U.S. Pat. Pub. No. 2016/0272718). [0455] The payload may comprise a polynucleotide whose sequence encodes small molecule-inducible cytokine receptor, such as a rapamycin-activated cell-surface receptor (RACR). Small molecule-inducible cytokine receptors are described in, e.g., U.S. Pat. Pub. No.2020/0123224. [0456] In some embodiments, the CAR is a CAR that specifically binds fluorescein and derivatives thereof, including fluorescein thiocynate and fluorescein conjugated to agent, (“anti-FITC CAR”). The agent may be a small molecule or protein that specifically binds to a desired target cell, such as a cancer cell. The particles disclosed herein, or cells made using them, may be used in comibination with a conjugate to treat disease, such as cancer. An Attorney Ref. No.061479-506001WO example agent is folate, which binds folate receptors (FR) on FR+ cancers; fluorescein conjugated to folate is termed “FITC-folate.” In some embodiments, anti-FITC CAR MXVY[S\O\ K USQKWN(LSWNSWQ NXVKSW' K 9:3h RSWQO' K ][KW\VOVL[KWO NXVKSW S\ Y[O\OW] (“TM”), a co-stimulation domain (e.g., 4-1BB), and an activation signaling domain (e.g., 9:.m%) [0457] An illustrative polynucleotide sequence encoding a universal CAR is SEQ ID NO: 75. [0458] An illustrative universal CAR amino acid sequence is SEQ ID NO: 14: MALPVTALLLPLALLLHAARPDVVMTQTPLSLPVSLGDQASISCRSSQSLVHSNGN TYLRWYLQKPGQSPKVLIYKVSNRVSGVPDRFSGSGSGTDFTLKINRVEAEDLGV YFCSQSTHVPWTFGGGTKLEIKSSADDAKKDAAKKDDAKKDDAKKDGGVKLDE TGGGLVQPGGAMKLSCVTSGFTFGHYWMNWVRQSPEKGLEWVAQFRNKPYNY ETYYSDSVKGRFTISRDDSKSSVYLQMNNLRVEDTGIYYCTGASYGMEYLGQGTS VTVSFVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFA CDIYIWAPLAGTCGVLLLSLVITLYCNHRNRFSVVKRGRKKLLYIFKQPFMRPVQT TQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYD VLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGH DGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 14) [0459] An illustrative polynucleotide insert for a particle is SEQ ID NO: 76. [0460] In some embodiments, the CAR may be encoded by a polynucleotide sequence that encodes a signal peptide to signal transport of the CAR in the cell. It is understood that typically the signal peptide is removed from the protein. [0461] An illustrative CAR amino acid sequence without a signal peptide may comprise SEQ ID NO: 77: DVVMTQTPLSLPVSLGDQASISCRSSQSLVHSNGNTYLRWYLQKPGQSPKVLIYK VSNRVSGVPDRFSGSGSGTDFTLKINRVEAEDLGVYFCSQSTHVPWTFGGGTKLEI KSSADDAKKDAAKKDDAKKDDAKKDGGVKLDETGGGLVQPGGAMKLSCVTSG FTFGHYWMNWVRQSPEKGLEWVAQFRNKPYNYETYYSDSVKGRFTISRDDSKSS VYLQMNNLRVEDTGIYYCTGASYGMEYLGQGTSVTVSFVPVFLPAKPTTTPAPRP PTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVI TLYCNHRNRFSVVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKN Attorney Ref. No.061479-506001WO PQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHM QALPPR (SEQ ID NO: 77) [0462] An illustrative CAR amino acid sequence signal peptide may comprise SEQ ID NO: 78: MALPVTALLLPLALLLHAARP (SEQ ID NO: 78) [0463] A further illustrative universal CAR amino acid sequence is SEQ ID NO: 79: [0464] LLLVTSLLLCELPHPAFLLIPSVLTQPSSVSAAPGQKVTISCSGSTSNIGNNY VSWYQQHPGKAPKLMIYDVSKRPSGVPDRFSGSKSGNSASLDISGLQSEDEADYY CAAWDDSLSEFLFGTGTKLTVLGSTSGSGKPGSGEGSTKGQVQLVESGGNLVQPG GSLRLSCAASGFTFGSFSMSWVRQAPGGGLEWVAGLSARSSLTHYADSVKGRFTI SRDNAKNSVYLQMNSLRVEDTAVYYCARRSYDSSGYWGHFYSYMDVWGQGTL VTVSSTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPL AGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGG CELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPR RKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDA LHMQALPPR [0465] A further illustrative polynucleotide sequence encoding a universal CAR is SEQ ID NO: 80. [0466] An illustrative polynucleotide insert for a particle is SEQ ID NO: 81. [0467] In various embodiments, the particle comprises a polynucleotide having a polynucleotide sequence according to one or more of SEQ ID NOs. 75-76 or 80-81, or polynucleotide sequence similar to it. The polynucleotide sequence may encode, and cells transduced by the particle may express, a CAR having a polypeptide sequence according to one or more of SEQ ID NOs. 14, 77, 79, or polynucleotide sequence similar to it. The polypeptide sequence may comprise a humanized immunoglobulin variable domain. [0468] As used herein, the term “similar” may refer to a polynucleotide or polypeptide sequence at least about 75%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or at least about 99.5% to a reference sequence. [0469] The payload may comprise a polynucleotide whose sequence encodes a gene-editing system (e.g., CRISPR-Cas, meganuclease, homing endonuclease, zinc finger enzyme). [0470] In some embodiments, the lentiviral particles of the present disclosure comprise a polynucleotide sequence encoding, in any order, on a polycistronic transcript: a promoter, a Attorney Ref. No.061479-506001WO therapeutic protein (e.g. CAR), optionally a cytosolic FRB domain or a portion thereof, and optionally a synthetic cytokine polypeptide (e.g. RACR). In some embodiments, the polycistronic transcript comprises a promoter and a CAR. Illustrative promoters include, without limitation, a cytomegalovirus (CMV) promoter, a CAG promoter, an SV40 promoter, an SV40/CD43 promoter, and a MND promoter. [0471] In some embodiments, the MND promoter comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 118. GAACAGAGAAACAGGAGAATATGGGCCAAACAGGATATCTGTGGTAAGCAGT TCCTGCCCCGGCTCAGGGCCAAGAACAGTTGGAACAGCAGAATATGGGCCAA ACAGGATATCTGTGGTAAGCAGTTCCTGCCCCGGCTCAGGGCCAAGAACAGAT GGTCCCCAGATGCGGTCCCGCCCTCAGCAGTTTCTAGAGAACCATCAGATGTT TCCAGGGTGCCCCAAGGACCTGAAATGACCCTGTGCCTTATTTGAACTAACCA ATCAGTTCGCTTCTCGCTTCTGTTCGCGCGCTTCTGCTCCCCGAGCTCTATATA AGCAGAGCTCGTTTAGTGAACCGTCAGATCGCTAGC (SEQ ID NO: 118) [0472] In some embodiments, the MND promoter comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 172. AATGAAAGACCCCACCTGTAGGTTTGGCAAGCTAGGATCAAGGTCAGGAACA GAGAGACAGCAGAATATGGGCCAAACAGGATATCTGTGGTAAGCAGTTCCTG CCCCGGCTCAGGGCCAAGAACAGTTGGAACAGCAGAATATGGGCCAAACAGG ATATCTGTGGTAAGCAGTTCCTGCCCCGGCTCAGGGCCAAGAACAGATGGTCC CCAGATGCGGTCCCGCCCTCAGCAGTTTCTAGAGAACCATCAGATGTTTCCAG GGTGCCCCAAGGACCTGAAATGACCCTGTGCCTTATTTGAACTAACCAATCAG TTCGCTTCTCGCTTCTGTTCGCGCGCTTCTGCTCCCCGAGCTCTATATAAGAGC CCACAACCCCTCACTCGGC (SEQ ID NO: 172) [0473] In some embodiments, the CSF2RA signal sequence comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 173. ATGCTGCTGCTGGTGACAAGCCTGCTGCTGTGCGAGCTGCCTCACCCAGCCTT TCTGCTGATCCCC (SEQ ID NO: 173) [0474] The disclosure provides a polynucleotide construct comprising a contiguous polynucleotide sequence encoding at least two synthetic receptors and methods for uses Attorney Ref. No.061479-506001WO thereof. In some embodiments, the polynucleotide construct is a polycistronic construct encoding a synthetic cytokine receptor, a synthetic chimeric antigen receptor (CAR), and a freely diffusible FRB, in which the cytokine receptor is responsive to rapamycin binding. Advantageously, FRB reduces the inhibitory effects of rapamycin on mTOR in cells engineered to express the polycistronic constructs provided herein. Expression of the freely diffusible FRB can promote consistent activation and proliferation of engineered cells. [0475] In some aspects, provided herein is a lentiviral vector comprising any one of the polycistronic constructs disclosed herein. In some aspects, provided herein is a cell comprising any of the lentiviral vectors disclosed herein. [0476] In some aspects, provided herein is a method of transducing a cell comprising contacting a target cell with any of the polycistronic constructs disclosed herein. [0477] In some aspects, provided herein is a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell. In some aspects, provided herein is a cell produced by any of the methods disclosed herein. [0478] In some aspects, provided herein is a method of administering to a subject any of the cells disclosed herein. In some aspects, provided herein is a method of administering to a subject any of the lentiviral vectors disclosed herein. [0479] All publications, including patent documents, scientific articles and databases, referred to in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication were individually incorporated by reference. If a definition set forth herein is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications that are herein incorporated by reference, the definition set forth herein prevails over the definition that is incorporated herein by reference. [0480] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described Polycistronic Constructs [0481] Provided herein are polycistronic constructs encoding one or more separate proteins. In some embodiments, the polycistronic constructs comprise one, two, three, or four expression cassettes each encoding a separate protein. In some embodiments, the polycistronic constructs comprise four expression cassettes each encoding a separate protein. In some embodiments, the expression cassettes are separated by cleavable linkers. Attorney Ref. No.061479-506001WO [0482] In some embodiments, the polycistronic constructs provided herein comprise a nucleotide sequence encoding an FRB. In some embodiments, the polycistronic constructs provided herein comprise a nucleotide sequence encoding a chimeric antigen receptor (CAR). In some embodiments, the polycistronic constructs provided herein comprise a nucleotide sequence encoding a synthetic cytokine polypeptide. In some embodiments, the synthetic cytokine polypeptide comprises a synthetic cytokine gamma chain polypeptide and a synthetic cytokine beta chain polypeptide. In some embodiments, the synthetic Mb]XTSWO QKVVK MRKSW MXVY[S\O\ SW]O[UO^TSW - [OMOY]X[ \^L^WS] l $?A(-G=%) ?W \XVO embodiments, the synthetic cytokine gamma chain further comprises FRB. In some OVLXNSVOW]\' ]RO \bW]RO]SM Mb]XTSWO LO]K MRKSW MXVY[S\O\ SW]O[UO^TSW - [OMOY]X[ \^L^WS] k (IL-2RB). In some embodiments, the synthetic cytokine gamma chain comprises further FKBP12. In other embodiments, the synthetic cytokine gamma chain comprises interleukin - [OMOY]X[ \^L^WS] l $?A(-G=%) ?W \XVO OVLXNSVOW]\' ]RO \bW]RO]SM Mb]XTSWO QKVVK MRKSW further comprises FKBP12. In some embodiments, the synthetic cytokine beta chain MXVY[S\O\ SW]O[UO^TSW - [OMOY]X[ \^L^WS] k $?A(-G8%) ?W \XVO OVLXNSVOW]\' ]RO \bW]RO]SM cytokine beta chain further comprises FRB. [0483] In some embodiments, the polycistronic construct provided herein comprises nucleotide sequences encoding an FRB, a synthetic cytokine polypeptide, and a CAR. [0484] In some embodiments, the polycistronic construct comprises a nucleotide sequence encoding FRB, a nucleotide sequence encoding a synthetic cytokine polypeptide, and a nucleotide sequence encoding a CAR. In some embodiments, the nucleotide sequence encoding the synthetic cytokine polypeptide comprises a first nucleotide sequence encoding FRB:IL-2RG and a second nucleotide sequence encoding FKBP12:IL-2RB. In some embodiments, the nucleotide sequence encoding the synthetic cytokine polypeptide comprises a first nucleotide sequence encoding FKBP12:IL-2RG and a second nucleotide sequence encoding FRB:IL-2RB. Cytosolic FRB [0485] In some embodiments, an expression cassette of the polycistronic construct encodes an FRB domain. The FRB domain is an approximately 270 base pair (bp) domain from the mTOR protein kinase. It may be expressed in the cytosol as a freely diffusible soluble protein. Attorney Ref. No.061479-506001WO [0486] In some embodiments, the first expression cassette in the polycistronic construct comprises a nucleotide sequence encoding an FRB. In some embodiments, when the FRB is expressed, it is a soluble, cytoplasmic protein (termed herein “free FRB”). [0487] In some of any embodiments, the method further including administering a non- physiological ligand to the subject. In some embodiments, the non-physiological ligand is able to bind to the synthetic cytokine receptor and induce gamma cytokine signaling in the cell. In some embodiments, the nonphysiological ligand includes rapamycin or a rapamycin analog. [0488] In some embodiments, the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 256, 257, or 258. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NOs: 256, 257, or 258. In some embodiments, the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NOs: 256, 257, or 258. [0489] In some embodiments, the FRB comprises an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs: 251, 252, or 260. In some embodiments, the FRB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs: 251, 252, or 260. In some embodiments, the FRB comprises the amino acid sequence of SEQ ID NOs: 251, 252, or 260. [0490] In some embodiments, synthetic cytokine receptor complex comprises a cytosolic polypeptide that binds to the ligand or a complex comprising the ligand. [0491] Advantageously, the cytosolic FRB confers resistance to the immunosuppressive effect of the non-physiological ligand (e.g., rapamycin or rapalog). Synthetic Cytokine Receptor [0492] In some embodiments, an expression cassette of the polycistronic construct encodes a synthetic cytokine receptor. The synthetic cytokine receptors of the present disclosure comprise a synthetic gamma chain and a synthetic beta chain, each comprising a dimerization domain. The dimerization domains controllable dimerize in the present of a non-physiological ligand, thereby activating signaling the synthetic cytokine receptor. [0493] The synthetic gamma chain polypeptide comprises a first dimerization domain, a first transmembrane domain, and an interleukin-2 receptor subunit gamma (IL-2RG) intracellular domain. The dimerization domain may be extracellular (N-terminal to the Attorney Ref. No.061479-506001WO transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2G intracellular domain. [0494] The synthetic beta chain polypeptide comprises a second dimerization domain, a second transmembrane domain, and an intracellular domain selected from an interleukin-2 receptor subunit beta (IL-2RB) intracellular domain, an interleukin-7 receptor subunit beta (IL-7RB) intracellular domain, or an interleukin-21 receptor subunit beta (IL-21RB) intracellular domain. The dimerization domain may be extracellular (N-terminal to the transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2RB or IL-7RB intracellular domain). [0495] In some embodiments, the polycistronic construct provided herein comprises one or more nucleotide sequences encoding a synthetic cytokine receptor. In some embodiments, the one or more nucleotide sequences correspond to one or more expression cassettes. In some embodiments, the polynucleotide construct provided herein comprises one expression cassette encoding IL-2RG and a second expression cassette encoding IL-2RB. [0496] In some embodiments, the synthetic gamma chain polypeptide is encoded by a nucleic acid sequence that encodes a signal peptide. In some embodiments, the synthetic beta chain polypeptide is encoded by a nucleic acid sequence that encodes a signal peptide. A skilled artisan is readily familiar with signal peptides that can provide a signal to transport a nascent protein in the cells. Any of a variety of signal peptides can be employed. [0497] In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 261, 262, or 263. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 100% identical to the nucleotide sequence of SEQ ID NOs: 261, 262, or 263. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide comprises the nucleotide sequence of SEQ ID NOs: 261, 262, or 263. [0498] In some embodiments, the synthetic cytokine gamma chain polypeptide comprises SW]O[UO^TSW - [OMOY]X[ \^L^WS] l $?A(-G=%) ?W \XVO OVLXNSVOW]\' ]RO ?A(-G= MXVY[S\O\ KW amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs: 264 or 265. In some embodiments, the IL-2RG comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs: 264 or 265. In some embodiments, the IL-2RG comprises the amino acid sequence of SEQ ID NOs: 264 or 265. Attorney Ref. No.061479-506001WO [0499] In some embodiments, the second expression cassette further comprises a nucleotide sequence encoding FRB. In some embodiments, the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 257. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NO: 257. In some embodiments, the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NO: 257. [0500] In some embodiments, the FRB comprises an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 252. In some embodiments, the FRB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 252. In some embodiments, the FRB comprises the amino acid sequence of SEQ ID NO: 252. [0501] In some embodiments, the second expression cassette is codon optimized. [0502] In some embodiments, the second expression cassette comprises a nucleotide sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 266. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 266. In some embodiments, the second expression cassette comprises the nucleotide sequence of SEQ ID NO: 266. [0503] In some embodiments, the second expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 267. In some embodiments, the second expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 267. In some embodiments, the second expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NO: 267. [0504] In some embodiments, the second expression cassette further comprises a nucleotide sequence encoding FKBP12. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 268 or 269. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 100% identical to the nucleotide sequence of SEQ ID NOs: 268 or 269. In some embodiments, the nucleotide Attorney Ref. No.061479-506001WO sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NOs: 268 or 269. [0505] In some embodiments, the FKBP12 comprises an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 253. In some embodiments, the FKBP12 comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 253. In some embodiments, the FKBP12 comprises the amino acid sequence of SEQ ID NO: 253. [0506] In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 270 or 271. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 100% identical to the nucleotide sequence of SEQ ID NOs: 270 or 271. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide comprises the nucleotide sequence of SEQ ID NOs: 270 or 271. [0507] In some embodiments, the synthetic cytokine beta chain polypeptide comprises SW]O[UO^TSW - [OMOY]X[ \^L^WS] k $?A(-G8%) [0508] In some embodiments, the IL-2RB comprises an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs: 272 or 273. In some embodiments, the IL-2RB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs: 272 or 273. In some embodiments, the IL-2RB comprises the amino acid sequence of SEQ ID NOs: 272 or 273. [0509] In some embodiments, the third expression cassette further comprises a nucleotide sequence encoding FKBP12. [0510] In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 274 In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 100% identical to the nucleotide sequence of SEQ ID NO: 274. In some embodiments, the nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NO: 274. [0511] In some embodiments, the FKBP12 comprises an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the Attorney Ref. No.061479-506001WO amino acid sequence of SEQ ID NO: 275. In some embodiments, the FKBP12 comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 275. In some embodiments, the FKBP12 comprises the amino acid sequence of SEQ ID NO: 275. [0512] In some embodiments, the third expression cassette is codon optimized. [0513] In some embodiments, the third expression cassette comprises a nucleotide sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 276. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 276. In some embodiments, the third expression cassette comprises the nucleotide sequence of SEQ ID NO: 276. [0514] In some embodiments, the third expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 277. In some embodiments, the third expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 277. In some embodiments, the third expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NO: 277. [0515] In some embodiments, the third expression cassette further comprises a nucleotide sequence encoding FRB. In some embodiments, the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 257. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NO: 275. In some embodiments, the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NO: 257. Intracellular Domain [0516] In some embodiments, the intracellular signaling domain of the first transmembrane receptor protein comprises an interleukin-2 receptor subunit gamma (IL-2RG) domain. [0517] In some embodiments, the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-2RB intracellular domain, and a second dimerization domain. [0518] In some embodiments, the synthetic beta chain comprises an interleukin-2 receptor subunit beta (IL-2RB) intracellular domain. IL-2RB is also known as IL-15RB or CD122. Attorney Ref. No.061479-506001WO Thus, when referred to herein, IL-2RB can also mean IL-15RB. That is, the terms are used interchangeably in the present disclosure. [0519] In some embodiments, the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-7RB intracellular domain, and a second dimerization domain. [0520] In some embodiments, the synthetic beta chain comprises an interleukin-7 receptor subunit beta (IL-7RB) intracellular domain. [0521] In some embodiments, the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-21RB intracellular domain, and a second dimerization domain. [0522] In some embodiments, the synthetic beta chain comprises an interleukin-21 receptor subunit beta (IL-21RB) intracellular domain. Dimerization Domain [0523] The dimerization domains may be heterodimerization domains, including but not limited to FK506-Binding Protein of size 12 kD (FKBP) and a FKBP12-rapamycin binding (FRB) domain, which dimerize in the presence of rapamycin or a rapalog. [0524] Alternatively, the first dimerization domain and the second dimerization domain may be a FK506-Binding Protein of size 12 kD (FKBP) and a calcineurin domain, which dimerize in the presence of FK506 or an analogue thereof. [0525] In some embodiments the dimerization domains are homodimerization domains selected from: i) FK506-Binding Protein of size 12 kD (FKBP); ii) cyclophiliA (CypA); or iii) gyrase B (CyrB); with the corresponding non-physiological ligands being, respectively i) FK1012, AP1510, AP1903, or AP20187; ii) cyclosporin-A (CsA); or iii) coumermycin or analogs thereof. [0526] In some embodiments, the first and second dimerization domains of the transmembrane receptor proteins are a FKBP domain and a cyclophilin domain. Attorney Ref. No.061479-506001WO [0527] In some embodiments, the first and second dimerization domains of the transmembrane receptor proteins are a FKBP domain and a bacterial dihydrofolate reductase (DHFR) domain. [0528] In some embodiments, the first and second dimerization domains of the transmembrane receptor proteins are a calcineurin domain and a cyclophilin domain. [0529] In some embodiments, the first and second dimerization domains of the transmembrane receptor proteins are PYR1-like 1 (PYL1) and abscisic acid insensitive 1 (ABI1). Transmembrane Domain [0530] The transmembrane domain is the sequence of the synthetic cytokine receptor that spans the membrane. The transmembrane domain may comprise a hydrophobic alpha helix. In some embodiments, the transmembrane domain is a human protein. [0531] In some embodiments, the TM domain and the intracellular signaling domain are from the same cytokine receptor. In some embodiments, the synthetic gamma chain polypeptide contains an IL-2RG TM domain and an IL-2RG intracellular domain. In some embodiments, the synthetic beta chain polypeptide contains an IL-2RB TM domain and an IL-2RB intracellular domain. In some embodiments, the synthetic beta chain polypeptide contains an IL-7RB TM domain and an IL-7RB intracellular domain. In some embodiments, the synthetic beta chain polypeptide contains an IL-21RB TM domain and an IL-21RB intracellular domain. [0532] In some embodiments, one or more additional contiguous amino acids of the ectodomain directly adjacent to the TM domain of the cytokine receptor also can be included as part of the polypeptide sequence of a chain of the synthetic cytokine receptor. In some embodiments, 1-20 contiguous amino acids of the ectodomain adjacent to the TM domain of the cytokine receptor is included as part of the polypeptide sequence of a chain of the synthetic cytokine receptor. The portion of the ectodomain may be a contiguous sequence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acids directly adjacent (e.g. N-terminal to) the TM sequence. [0533] In some embodiments, the synthetic cytokine receptor is able to be bound by the non-physiological ligand rapamycin or a rapamycin analog. In some embodiments, the synthetic cytokine receptor is responsive to the non-physiological ligand rapamycin or a rapamycin analog, in which binding of the non-physiological ligand to the dimerization Attorney Ref. No.061479-506001WO domains of the synthetic cytokine receptor induces cytokine receptor-mediated signaling in the cell, such as via the JAK/STAT pathway. Illustrative Polycistronic Constructs [0534] In some embodiments, the polycistronic construct comprises in 5’ to 3’ order a nucleotide sequence encoding FRB, a nucleotide sequence encoding a synthetic cytokine polypeptide, and a nucleotide sequence encoding a CAR. In some embodiments, the nucleotide sequence encoding the synthetic cytokine polypeptide comprises in 5’ to 3’ order a first nucleotide sequence encoding FRB operably linked to IL-2RG and a second nucleotide sequence encoding FKBP12 operably linked to IL-2RB. In some embodiments, the nucleotide sequence encoding the synthetic cytokine polypeptide comprises in 5’ to 3’ order a first nucleotide sequence encoding FKBP12 operably linked to IL-2RG and a second nucleotide sequence encoding sFRB operably linked to IL-2RB. [0535] In some embodiments, the lentiviral particles of the present disclosure comprise a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[ XW K YXUbMS\][XWSM ][KW\M[SY]5 (a) a MND promoter; (b) a CAR; (c) a cytosolic FRB domain or a portion thereof; (d) a RACR cell-surface receptor; and (e) a WPRE sequence [0536] In some embodiments, the lentiviral particles of the present disclosure comprise a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[5 (a) a CAR; (b) a cytosolic FRB domain or a portion thereof; and (c) a RACR cell-surface receptor. [0537] In some embodiments, the lentiviral particle comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 119. [0538] GAACAGAGAAACAGGAGAATATGGGCCAAACAGGATATCTGTGGTAA GCAGTTCCTGCCCCGGCTCAGGGCCAAGAACAGTTGGAACAGCAGAATATGG GCCAAACAGGATATCTGTGGTAAGCAGTTCCTGCCCCGGCTCAGGGCCAAGA ACAGATGGTCCCCAGATGCGGTCCCGCCCTCAGCAGTTTCTAGAGAACCATCA GATGTTTCCAGGGTGCCCCAAGGACCTGAAATGACCCTGTGCCTTATTTGAAC Attorney Ref. No.061479-506001WO TAACCAATCAGTTCGCTTCTCGCTTCTGTTCGCGCGCTTCTGCTCCCCGAGCTC TATATAAGCAGAGCTCGTTTAGTGAACCGTCAGATCGCTAGC (SEQ ID NO: 119) [0539] In some embodiments, the lentiviral particle comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 120. [0540] MLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASLGDRVTISCRASQDISK YLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYF CQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVKLQESGPGLVAPSQSL SVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRLTIIKDNS KSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQGTSVTVSSESKYGPP CPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKKLLYIFKQPFMRPVQTT QEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDV LDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHD GLYQGLSTATKDTYDALHMQALPPRGSGATNFSLLKQAGDVEENPGPEMWHEG LEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQAYGRDLMEAQE WCRKYMKSGNVKDLLQAWDLYYHVFRRISKGSGATNFSLLKQAGDVEENPGPM PLGLLWLGLALLGALHAQAGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKK FDSSRDRNKPFKFMLGKQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPG IIPPHATLVFDVELLKLGEGSNTSKENPFLFALEAVVISVGSMGLIISLLCVYFWLER TMPRIPTLKNLEDLVTEYHGNFSAWSGVSKGLAESLQPDYSERLCLVSEIPPKGGA LGEGPGASPCNQHSPYWAPPCYTLKPETGSGATNFSLLKQAGDVEENPGPMALPV TALLLPLALLLHAARPILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMM ERGPQTLKETSFNQAYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRI SKGKDTIPWLGHLLVGLSGAFGFIILVYLLINCRNTGPWLKKVLKCNTPDPSKFFS QLSSEHGGDVQKWLSSPFPSSSFSPGGLAPEISPLEVLERDKVTQLLLQQDKVPEPA SLSSNHSLTSCFTNQGYFFFHLPDALEIEACQVYFTYDPYSEEDPDEGVAGAPTGSS PQPLQPLSGEDDAYCTFPSRDDLLLFSPSLLGGPSPPSTAPGGSGAGEERMPPSLQE RVPRDWDPQPLGPPTPGVPDLVDFQPPPELVLREAGEEVPDAGPREGVSFPWSRPP GQGEFRALNARLPLNTDAYLSLQELQGQDPTHLV (SEQ ID NO: 120) [0541] In some embodiments, the lentiviral particle comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 121. Attorney Ref. No.061479-506001WO [0542] In some embodiments, the lentiviral particles of the present disclosure comprises a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[ XW K YXUbMS\][XWSM ][KW\M[SY]5 (a) a MND promoter; (b) a cytosolic FRB domain or a portion thereof; (c) a RACR cell-surface receptor; (d) a CAR; and (e) a WPRE sequence. [0543] In some embodiments, the lentiviral particles of the present disclosure comprises a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[5 (a) a cytosolic FRB domain or a portion thereof; (b) a RACR cell-surface receptor; and (c) a CAR. [0544] In some embodiments, the lentiviral particle comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 122. [0545] MEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQ AYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISKGSGATNFSLLKQ AGDVEENPGPMPLGLLWLGLALLGALHAQAGVQVETISPGDGRTFPKRGQTCVV HYTGMLEDGKKFDSSRDRNKPFKFMLGKQEVIRGWEEGVAQMSVGQRAKLTISP DYAYGATGHPGIIPPHATLVFDVELLKLGEGSNTSKENPFLFALEAVVISVGSMGLI ISLLCVYFWLERTMPRIPTLKNLEDLVTEYHGNFSAWSGVSKGLAESLQPDYSERL CLVSEIPPKGGALGEGPGASPCNQHSPYWAPPCYTLKPETGSGATNFSLLKQAGD VEENPGPMALPVTALLLPLALLLHAARPILWHEMWHEGLEEASRLYFGERNVKG MFEVLEPLHAMMERGPQTLKETSFNQAYGRDLMEAQEWCRKYMKSGNVKDLL QAWDLYYHVFRRISKGKDTIPWLGHLLVGLSGAFGFIILVYLLINCRNTGPWLKK VLKCNTPDPSKFFSQLSSEHGGDVQKWLSSPFPSSSFSPGGLAPEISPLEVLERDKV TQLLLQQDKVPEPASLSSNHSLTSCFTNQGYFFFHLPDALEIEACQVYFTYDPYSEE DPDEGVAGAPTGSSPQPLQPLSGEDDAYCTFPSRDDLLLFSPSLLGGPSPPSTAPGG SGAGEERMPPSLQERVPRDWDPQPLGPPTPGVPDLVDFQPPPELVLREAGEEVPD AGPREGVSFPWSRPPGQGEFRALNARLPLNTDAYLSLQELQGQDPTHLVGSGATN FSLLKQAGDVEENPGPMLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASLGDRV TISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTIS NLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVKLQES Attorney Ref. No.061479-506001WO GPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSA LKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQGTSV TVSSESKYGPPCPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKKLLYIF KQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNEL NLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMK GERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 122) [0546] In some embodiments, the lentiviral particle comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 123. [0547] In some embodiments, the lentiviral particles of the present disclosure comprise a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[ XW K YXUbMS\][XWSM ][KW\M[SY]5 (a) a MND promoter; (b) a cytosolic FRB domain or a portion thereof; (c) a CAR; $N% I=<(k :C NXVKSW X[ YX[]SXW ]RO[OXP6 KWN (e) a WPRE sequence. [0548] In some embodiments, the lentiviral particles of the present disclosure comprise a YXUbW^MUOX]SNO \OZ^OWMO OWMXNSWQ' SW 0j ]X .j X[NO[5 (a) a cytosolic FRB domain or a portion thereof; (b) a CAR; and $M% K I=<(k :C NXVKSW X[ YX[]SXW ]RO[OXP) [0549] In some embodiments, the lentiviral particle comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 124. [0550] MEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQ AYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISKGSGATNFSLLKQ AGDVEENPGPMLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASLGDRVTISCRA SQDISKYLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQE DIATYFCQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVKLQESGPGLV APSQSLSVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTYYNSALKSRL TIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQGTSVTVSSE SKYGPPCPPCPMFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRR Attorney Ref. No.061479-506001WO EEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRGSGATNFSLLKQAGDVEENPGPM GRGLLRGLWPLHIVLWTRIASTIPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDV RFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFI LEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPDLLLVIFQVTG ISLLPPLGVAISVIIIFYCYRVNRQQKRRR (SEQ ID NO: 124) [0551] In some embodiments, the lentiviral particle comprises a nucleic acid sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 125. [0552] (SEQ ID NO: 125) [0553] In some embodiments, the FRB domain comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 251. [0554] MEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQ AYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISK (SEQ ID NO: 251) [0555] In some embodiments, the IL-2 Receptor gamma domain comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 252. [0556] ILWHEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETS FNQAYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISK (SEQ ID NO: 252) [0557] In some embodiments, the IL-2 Receptor beta domain comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 253. [0558] GVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKFDSSRDRNKPFKFML GKQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLK L (SEQ ID NO: 253) [0559] In some embodiments, the Rapamycin-Activated Cell-Surface Receptor (RACR) and FRB domain complex comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 254. Attorney Ref. No.061479-506001WO [0560] MEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQ AYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISKASRRKRGSGEG RGSLLTCGDVEENPGPMPLPVTALLLPLALLLHAARPILWHEMWHEGLEEASRLY FGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQAYGRDLMEAQEWCRKYMK SGNVKDLLQAWDLYYHVFRRISKGSNTSKENPFLFALEAVVISVGSMGLIISLLCV YFWLERTMPRIPTLKNLEDLVTEYHGNFSAWSGVSKGLAESLQPDYSERLCLVSEI PPKGGALGEGPGASPCNQHSPYWAPPCYTLKPETGSGATNFSLLKQAGDVEENPG PMPLGLLWLGLALLGALHAQAGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDG KKFDSSRDRNKPFKFMLGKQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGH PGIIPPHATLVFDVELLKLGEGKDTIPWLGHLLVGLSGAFGFIILVYLLINCRNTGP WLKKVLKCNTPDPSKFFSQLSSEHGGDVQKWLSSPFPSSSFSPGGLAPEISPLEVLE RDKVTQLLLQQDKVPEPASLSSNHSLTSCFTNQGYFFFHLPDALEIEACQVYFTYD PYSEEDPDEGVAGAPTGSSPQPLQPLSGEDDAYCTFPSRDDLLLFSPSLLGGPSPPS TAPGGSGAGEERMPPSLQERVPRDWDPQPLGPPTPGVPDLVDFQPPPELVLREAG EEVPDAGPREGVSFPWSRPPGQGEFRALNARLPLNTDAYLSLQELQGQDPTHLV(S EQ ID NO: 254) [0561] In some embodiments, the Rapamycin-Activated Cell-Surface Receptor (RACR) and FRB domain complex and anti-CD19 CAR comprises a polypeptide sequence that shares at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 99%, or 100% identity to SEQ ID NO: 255. [0562] MEMWHEGLEEASRLYFGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQ AYGRDLMEAQEWCRKYMKSGNVKDLLQAWDLYYHVFRRISKASRRKRGSGEG RGSLLTCGDVEENPGPMPLPVTALLLPLALLLHAARPILWHEMWHEGLEEASRLY FGERNVKGMFEVLEPLHAMMERGPQTLKETSFNQAYGRDLMEAQEWCRKYMK SGNVKDLLQAWDLYYHVFRRISKGSNTSKENPFLFALEAVVISVGSMGLIISLLCV YFWLERTMPRIPTLKNLEDLVTEYHGNFSAWSGVSKGLAESLQPDYSERLCLVSEI PPKGGALGEGPGASPCNQHSPYWAPPCYTLKPETGSGATNFSLLKQAGDVEENPG PMPLGLLWLGLALLGALHAQAGVQVETISPGDGRTFPKRGQTCVVHYTGMLEDG KKFDSSRDRNKPFKFMLGKQEVIRGWEEGVAQMSVGQRAKLTISPDYAYGATGH PGIIPPHATLVFDVELLKLGEGKDTIPWLGHLLVGLSGAFGFIILVYLLINCRNTGP WLKKVLKCNTPDPSKFFSQLSSEHGGDVQKWLSSPFPSSSFSPGGLAPEISPLEVLE RDKVTQLLLQQDKVPEPASLSSNHSLTSCFTNQGYFFFHLPDALEIEACQVYFTYD PYSEEDPDEGVAGAPTGSSPQPLQPLSGEDDAYCTFPSRDDLLLFSPSLLGGPSPPS Attorney Ref. No.061479-506001WO TAPGGSGAGEERMPPSLQERVPRDWDPQPLGPPTPGVPDLVDFQPPPELVLREAG EEVPDAGPREGVSFPWSRPPGQGEFRALNARLPLNTDAYLSLQELQGQDPTHLVG SGATNFSLLKQAGDVEENPGPLLLVTSLLLCELPHPAFLLIPDIQMTQTTSSLSASL GDRVTISCRASQDISKYLNWYQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDY SLTISNLEQEDIATYFCQQGNTLPYTFGGGTKLEITGSTSGSGKPGSGEGSTKGEVK LQESGPGLVAPSQSLSVTCTVSGVSLPDYGVSWIRQPPRKGLEWLGVIWGSETTY YNSALKSRLTIIKDNSKSQVFLKMNSLQTDDTAIYYCAKHYYYGGSYAMDYWGQ GTSVTVSSGAVHTRGLDFACDFWVLVVVGGVLACYSLLVTVAFIIFWVKRGRKK LLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQ LYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYS EIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 255) Pharmaceutical Compositions and Kits [0563] In some embodiments, the disclosure provides a pharmaceutical composition comprising a particle according to the disclosure and a pharmaceutically acceptable carrier. [0564] In some embodiments, the disclosure provides a kit comprising the particle and instructions for use in transduction of target cells and/or treatment of a subject. The kit may include a pharmaceutically acceptable carrier and/or an injection device. The kit may further include suitable tubing for administering the particles. Formulations [0565] The formulations and compositions of the present disclosure may comprise a combination of any number of viral particles, and optionally one or more additional pharmaceutical agents (polypeptides, polynucleotides, compounds etc.) formulated in pharmaceutically acceptable or physiologically-acceptable compositions for administration to a cell, tissue, organ, or an animal, either alone, or in combination with one or more other modalities of therapy. In some embodiments, the one or more additional pharmaceutical agents further increases transduction efficiency of viral particles. [0566] In some embodiments, the formulations and compositions of the present disclosure may comprise a combination of any number of viral particles, and optionally one or more nanocarriers. Illustrative nanocarriers include, but are not limited to, micelles, polymers, liposomes, and lipid nanoparticles (LNPs). [0567] In some embodiments, the present disclosure provides compositions comprising a therapeutically-effective amount of a viral particle, as described herein, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. In some Attorney Ref. No.061479-506001WO embodiments, the composition further comprises other agents, such as, e.g., cytokines, growth factors, hormones, small molecules or various pharmaceutically active agents. [0568] In some embodiments, compositions and formulations of the viral particles used in accordance with the present disclosure may be prepared for storage by mixing a viral particle having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed. In some embodiments, one or more pharmaceutically acceptable surface-active agents (surfactant), buffers, isotonicity agents, salts, amino acids, sugars, stabilizers and/or antioxidant are used in the formulation. [0569] Suitable pharmaceutically acceptable surfactants comprise but are not limited to polyethylen-sorbitan-fatty acid esters, polyethylene-polypropylene glycols, polyoxyethylene-stearates and sodium dodecyl sulphates. Suitable buffers comprise but are not limited to histidine-buffers, citrate-buffers, succinate-buffers, acetate-buffers and phosphate-buffers. [0570] Isotonicity agents are used to provide an isotonic formulation. An isotonic formulation is liquid, or liquid reconstituted from a solid form, e.g. a lyophilized form and denotes a solution having the same tonicity as some other solution with which it is compared, such as physiologic salt solution and the blood serum. Suitable isotonicity agents comprise but are not limited to salts, including but not limited to sodium chloride (NaCl) or potassium chloride, sugars including but not limited to glucose, sucrose, trehalose or and any component from the group of amino acids, sugars, salts and combinations thereof. In some embodiments, isotonicity agents are generally used in a total amount of about 5 mM to about 350 mM. [0571] Non-limiting examples of salts include salts of any combinations of the cations sodium potassium, calcium or magnesium with anions chloride, phosphate, citrate, succinate, sulphate or mixtures thereof. Non-limiting examples of amino acids comprise arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine, tryptophan, methionine, serine, or proline. Non- limiting examples of sugars according to the invention include trehalose, sucrose, mannitol, sorbitol, lactose, glucose, mannose, maltose, galactose, fructose, sorbose, raffinose, glucosamine, N-methylglucosamine (also referred to as “meglumine”), galactosamine and Attorney Ref. No.061479-506001WO neuraminic acid and combinations thereof. Non-limiting examples of stabilizer includes amino acids and sugars as described above as well as commercially available cyclodextrins and dextrans of any useful kind and molecular weight. Non-limiting examples of antioxidants include excipients such as methionine, benzylalcohol or any other excipient used to minimize oxidation. [0572] The phrase “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human. The preparation of an aqueous composition that contains a protein as an active ingredient is well understood in the art. Typically, such compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared. The preparation can also be emulsified. [0573] As used herein, “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. Except insofar as any media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. [0574] As used herein “pharmaceutically acceptable carrier” includes solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible, including pharmaceutically acceptable cell culture media. In some embodiments, a composition comprising a carrier is suitable for parenteral administration, e.g., intravascular (intravenous or intra-arterial), intraperitoneal or intramuscular administration. Pharmaceutically acceptable carriers may include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. Except insofar as any media or agent is incompatible with the transduced cells, use thereof in the pharmaceutical compositions of the present disclosure is contemplated. [0575] The compositions may further comprise one or more polypeptides, polynucleotides, vector genomes comprising same, compounds that increase the transduction efficiency of vector genomes, formulated in pharmaceutically acceptable or physiologically-acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy. It will also be understood that, if desired, the Attorney Ref. No.061479-506001WO compositions of the present disclosure may be administered in combination with other agents as well, such as, e.g., cytokines, growth factors, hormones, small molecules or various pharmaceutically active agents. There is virtually no limit to other components that may also be included in the compositions, provided that the additional agents do not adversely affect the ability of the composition to deliver the intended therapy. [0576] The present disclosure also provides pharmaceutical compositions comprising an expression cassette or vector (e.g., therapeutic vector) disclosed herein and one or more pharmaceutically acceptable carriers, diluents or excipients. In some embodiments, the pharmaceutical composition comprises a lentiviral vector comprising an expression cassette disclosed herein, e.g., wherein the expression cassette comprises one or more polynucleotide sequences encoding one or more chimeric antigen receptor (CARs) and variants thereof. [0577] The pharmaceutical compositions that contain the expression cassette or vector genome may be in any form that is suitable for the selected mode of administration, for example, for intraventricular, intramyocardial, intracoronary, intravenous, intra-arterial, intra-renal, intraurethral, epidural, intrathecal, intraperitoneal, or intramuscular administration. The vector genome can be administered, as sole active agent, or in combination with other active agents, in a unit administration form, as a mixture with pharmaceutical supports, to animals and human beings. In some embodiments, the pharmaceutical composition comprises cells transduced ex vivo with any of the vector genomes according to the present disclosure. [0578] In some embodiments, the viral particle (e.g., lentiviral particle), or a pharmaceutical composition comprising that viral particle, is effective when administered systemically. For example, the viral vectors of the disclosure, in some cases, may be administered intravenously to subject (e.g., a primate, such as a non-human primate or a human). In some embodiments, the viral vectors of the disclosure are capable of inducing expression of CAR in various immune cells when administered systemically (e.g., in T-cells, dendritic cells, NK cells). [0579] In various embodiments, the pharmaceutical compositions contain vehicles (e.g., carriers, diluents and excipients) that are pharmaceutically acceptable for a formulation capable of being injected. Illustrative excipients include a poloxamer. Formulation buffers for viral vectors may contain salts to prevent aggregation and other excipients (e.g., poloxamer) to reduce stickiness of the viral particle. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium Attorney Ref. No.061479-506001WO or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. In some embodiments, the formulation is stable for storage and use when frozen (e.g., at less than 0 °C, about -60 °C, or about -72 °C). In some embodiments, the formulation is a cryopreserved solution. [0580] The pharmaceutical compositions of the present disclosure, formulation of pharmaceutically acceptable excipients and carrier solutions may be useful to those of skill in the art, such as for development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, intraperitoneal, and intramuscular administration and formulation. [0581] In certain circumstances, it may be desirable to deliver the compositions disclosed herein parenterally, intravenously, intramuscularly, or intraperitoneally, for example, in U.S. Pat. Nos. 5,543,158; 5,641,515 and 5,399,363 (each incorporated herein by reference in its entirety). Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. [0582] The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, incorporated herein by reference in its entirety). In all cases, the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Fluidity may be maintained, for example, by use of a coating, such as lecithin, by maintenance of a useful particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be facilitated by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In some embodiments, isotonic agents, for example, sugars or sodium chloride, are added. Prolonged Attorney Ref. No.061479-506001WO absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. [0583] For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if useful or necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In some embodiments, the solution intended for subcutaneous administration includes hyaluronidase. In this connection, a sterile aqueous medium that can be employed may be useful. One dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion (see, e.g., Remington: The Science and Practice of Pharmacy, 20th Edition. Baltimore, Md.: Lippincott Williams & Wilkins, 2005). Some variation in dosage may occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. In some embodiments, for human administration, preparations should meet sterility, pyrogenicity, and the general safety and purity standards set by the FDA Office of Biologics standards. [0584] In some embodiments, the present disclosure provides formulations or compositions suitable for the delivery of viral vector systems (i.e., viral-mediated transduction) including, but not limited to, retroviral (e.g., lentiviral) vectors. Method of Use In vitro or Ex Vivo [0585] The compositions described herein such as fusion proteins or particles described may be used in vitro or ex vivo. The lentiviral particles described may be used ex vivo, in a cell manufacturing process or at a bedside as described, e.g., in Int’l Pat. Pub. No. WO 2022/072885, Int’l Pat. Pub. No. 2019/217954, Int’l Pat. Pub. No. 2020/123649, and Int’l Pat. Pub. No. 2009/072003. In some embodiments, the disclosure provides an ex vivo method of transducing target cells, comprising contacting the target cells with the particle according to the present disclosure. In some embodiments, the particles described herein may be used to transduce cells that have not been previously activated. For example, the particles described herein may be useful for transducing cells that have not been previously contacted with cell activation beads or activation reagents (e.g. Dynabeads or other reagents comprising anti-CD3 and/or anti-CD28 antibodies or binding fragments thereof). Where a method herein describes use of a lentiviral particle, use of another particle is contemplated Attorney Ref. No.061479-506001WO where appropriate and feasible. Where a method herein describes use of a lentiviral particle, use of a composition or fusion molecule is also contemplated where appropriate and feasible. For example, a fusion molecule, contained on the surface of a lentiviral particle, or a pharmaceutical composition may be administered to or contacted with a cell such as an immune cell (e.g. T cell). [0586] Non-limiting examples of cells that can be the target of the lentiviral particle described herein include T lymphocytes, dendritic cells (DC), Treg cells, B cells, Natural Killer cells, and macrophages. Ex-Vivo Manufacturing [0587] In some aspects, the disclosure provides a method of delivering a nucleic acid to a cell ex vivo. In some embodiments, the disclosure provides a method of delivering a nucleic acid to an immune cell ex vivo. In some embodiments, the lentiviral particles of the disclosure activate and transduce an immune cell ex vivo. [0588] In some embodiments, the disclosure provides a method of delivering a nucleic acid to a cell in an ex vivo CAR T manufacturing process. Such methods typically involve the isolation of peripheral blood mononuclear cells (PBMCs) from a patient via leukapheresis. In some embodiments, such methods involve obtaining whole blood from a patient without isolation of PBMCs and forward processing the whole blood. The PBMCs may be washed and optionally further purified via one or more selection steps to isolate particular T cell populations of interest. In some aspects, these might include CD4+ and/or CD8+ T cells. The washed and/or purified cells may be optionally activated and then transduced using a lentiviral vector. The activation step may comprise contacting the cells with an exogenous activation agent such as anti-CD3 and anti-CD28 antibodies bound to a substrate or using unbound antibodies. Illustrative activation agents include anti-CD3 and anti-CD28- presenting beads and/or soluble polymers. Advantageously, the particles of the present disclosure may not require activation prior to transduction and so the activation step may be omitted. Transduction may be accomplished by contacting the patient’s PBMCs, isolated cells, or, in some cases, whole blood with the lentiviral particles described herein. After transduction, the cells may be optionally further washed and cultured until harvest. Methods of manufacturing engineered cell therapies, including CAR T cells (see e.g., Abou-el-Enein, M. et al. Blood Cancer Discov (2021), Vol 2(5): 408-422; Arcangeli, S. et al. Front. Immunol (19 Jun 2020), Vol. 11 (1217) 1-13; Ghassemi, S. et al. Nat Biomed Eng (Feb 2022), Vol 6(2): 118-128; Vormittag, P. et al. Curr Opin Biotechnol (Oct 2018), Vol. 54: Attorney Ref. No.061479-506001WO 164-181; each of which is herein incorporated by reference), may be useful. Illustrative methods of autologous CAR T manufacturing are disclosed in US Patent Publication Nos. 2019/0269727, 2016/0122782, 2021/0163893, and US 2017/0037369, each of which is incorporated herein in its entirety. [0589] In some embodiments, the disclosure provides a method of delivering a nucleic acid to a cell in an ex-vivo closed-loop manufacturing process. In some embodiments, an ex- vivo manufacturing process is an extracorporeal process. In exemplary embodiments, the lentiviral vectors disclosed herein permit delivery of a nucleic acid to a target cell during a closed-loop process. Exemplary methods of closed-loop and/or extracorporeal processes are disclosed in US Patent Publication No. 2021/0244871 and WO2022072885, each of which are incorporated herein in their entirety. In some embodiments, the lentiviral vectors as disclosed herein may be used to transduce cells ex vivo. For example, in exemplary closed- loop manufacturing processes, cells are obtained from a subject, washed, incubated and/or contacted with lentiviral particles, optionally washed again, and infused into the subject in a closed-loop system. In such embodiments, the lentiviral particles as disclosed herein are useful even without prior activation of the cells and are capable of binding to the cells in a short incubation and/or contacting step. In some embodiments, the incubation and/or contacting step is approximately or less than one hour. In some embodiments, the incubation and/or contacting step is approximately or less than one hour, approximately or less than two hours, approximately or less than three hours, approximately or less than four hours, or approximately or less than five hours. In some embodiments, the incubation and/or contacting step is less than 12 hours or less than 24 hours. In some embodiments, a nucleic acid is delivered to a cell by transduction with a lentiviral vector such that the nucleic acid enters the cell ex-vivo. In some embodiments, a nucleic acid is delivered to a cell by contacting the lentiviral vector to the surface of the cell. In such embodiments, the nucleic acid may enter the cell ex-vivo or in vivo after the cells (complexed with the lentiviral vector) are infused back into the subject. [0590] In some embodiments, provided herein are bedside systems and methods for performing cell-based therapies and treatments in a subject-connected, closed-loop continuous-flow manner, including cellular modifications and treatments, e.g., to produce chimeric antigen receptor T (CAR T) cells. In some embodiments of a system described herein, blood is removed from a subject, processed, customized, and returned to the subject in a closed-loop, continuous-flow manner. An arrangement of modules and units are used Attorney Ref. No.061479-506001WO sequentially for separation and collection of target cells from whole blood, employing for example, leukapheresis and/or other cell enrichment techniques, optionally including cell enrichment, purification and/or washing using an elutriation device, followed by one or more cell customization procedures, e.g., to generate CAR-T cells, optionally followed by cell enrichment, purification, fractionation, and/or washing, after which the processed and modified fraction comprising CAR-T cells are returned to the subject by means of an outlet conduit. One exemplary system is manufactured by Lupagen™ and is a closed-loop, continuous-flow system. Such systems and methods are disclosed in WO2019217964, which is incorporated herein by reference in its entirety. In some embodiments, the lentiviral vectors as disclosed herein eliminate the need for an ex-vivo activation step. In such embodiments, the isolated cells could be transduced directly after leukapheresis, washing, or selection. It is contemplated that the surface engineering described herein enables the lentiviral particles disclosed herein to activate and transduce cells in a single step. In such embodiments, the lentiviral particles disclosed herein may enable a short or truncated manufacturing process, reducing the time spent in ex-vivo manufacturing by eliminating one or more unit operations (e.g. activation prior to transduction) and/or reducing the amount of time that may be necessary in post-transduction cell culture. Without wishing to be bound by theory, in some embodiments the lentiviral vectors as described herein, in particular those particles comprising a fusion multidomain protein, bind to target cells with a higher avidity than lentiviral particles not comprising a fusion multidomain protein. In such embodiments, the fusion multidomain protein may allow the described lentiviral particles to bind target cells more tightly, reducing the incubation time for transduction and increasing transduction frequency and efficiency. In some embodiments, the time to effectively bind a lentiviral particle to a target cell may be one hour or less. [0591] It is contemplated that the present disclosure provides an ex vivo method of generating an engineered cell comprising contacting a target cell with a particle comprising a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule, a fusion molecule comprising an adhesion molecule linked to an activation molecule, or a fusion molecule comprising an adhesion molecule linked to a costimulatory molecule and an activation molecule wherein the contacting step is performed for approximately one hour, for approximately two hours, approximately three hours, approximately four hours, approximately five hours, approximately six hours, approximately 12 hours, approximately 24 hours, approximately 12-24 hours (inclusive of endpoints), or longer. This method may Attorney Ref. No.061479-506001WO require the contacting step to be performed in a closed-loop manufacturing or extracorporeal process as described herein. Alternatively, this method may require the contacting step to be performed in a traditional ex-vivo engineered cell manufacturing process. For example, in a perfusion incubator or a centrifuge (such as a Sepax or Rotea machine). Method of Use In Vivo [0592] In some embodiments, the lentiviral particles described herein transduce target cells in vivo. In some embodiments, the target cells are immune cells. In some embodiments, the immune cells are T cells. In some embodiments, the lentiviral particles described herein transduce T cells in vivo. In some embodiments, the lentiviral particles described herein transduce T cells in vivo generating CAR T cells. In some embodiments, the lentiviral particles described herein display a CD58-CD80-anti-CD3 scFv tri-fusion polypeptide and transduce T cells in vivo generating CAR T cells. Where a method herein describes use of a lentiviral particle, use of another particle is contemplated where feasible. [0593] In some embodiments, the viral particle is administered via a route selected from the group consisting of extracorporeal, parenteral, intravenous, intramuscular, subcutaneous, intratumoral, intraperitoneal, and intralymphatic. In some embodiments, the viral particle is administered multiple times. In some embodiments, the viral particle is administered by intralymphatic injection of the viral particle. In some embodiments, the viral particle is administered by intraperitoneal injection of the viral particle. In some embodiments, the viral particle is administered by intra-nodal injection – that is, the viral particle may be administered via injection into one or more lymph nodes. In some embodiments, the lymph nodes for administration are the inguinal lymph nodes. In some embodiments, the viral particle is administered by injection of the viral particle into tumor sites (i.e. intratumoral). In some embodiments, the viral particle is administered subcutaneously. In some embodiments, the viral particle is administered systemically. In some embodiments, the viral particle is administered intravenously. In some embodiments, the viral particle is administered intra-arterially. In some embodiments, the viral particle is a lentiviral particle. [0594] In some embodiments, the lentiviral particle is administered by intraperitoneal, subcutaneous, or intranodal injection. In some embodiments, the lentiviral particle is administered by intraperitoneal injection. In some embodiments, the lentiviral particle is administered by subcutaneous injection. In some embodiments, the lentiviral particle is administered by intranodal injection. Attorney Ref. No.061479-506001WO [0595] The present disclosure provides a method of treatment comprising administering a therapeutically effective dose of lentiviral particles to a subject in need thereof. In some embodiments, a therapeutically effective dose of the lentiviral particles described herein are administered. In some embodiments, a therapeutically effective dose comprises about 0.1×10 6 transducing units (TUs), about 0.2×10 6 TUs, about 0.3×10 6 TUs, about 0.4×10 6 TUs, about 0.5×10 6 TUs, about 0.6×10 6 TUs, about 0.7×10 6 TUs, about 0.8×10 6 TUs, about 0.9×10 6 TUs, about 1×10 6 TUs, about 1.2×10 6 TUs, about 1.4×10 6 TUs, about 1.6×10 6 TUs, about 1.8×10 6 TUs, about 0.1×10 6 TUs, about 0.1×10 6 TUs, about 0.1×10 6 TUs, about 0.1×10 6 TUs, about 2×10 6 TUs, about 2.5×10 6 TUs, about 3×10 6 TUs, about 4×10 6 TUs, about 5×10 6 TUs, about 6×10 6 TUs, about 7×10 6 TUs, about 8×10 6 TUs, about 9×10 6 TUs, about 1×10 7 TUs, about 2×10 7 TUs, about 3×10 7 TUs, about 4×10 7 TUs, about 5×10 7 TUs, about 6×10 7 TUs, about 7×10 7 TUs, about 8×10 7 TUs, about 9×10 7 TUs, about 1×10 8 TUs, about 2×10 8 TUs, about 3×10 8 TUs, about 4×10 8 TUs, about 5×10 8 TUs, about 6×10 8 TUs, about 7×10 8 TUs, about 8×10 8 TUs, about 9×10 8 TUs, about 1×10 9 TUs, or about 2×10 9 TUs. [0596] In some embodiments, the transduced immune cells comprising the polynucleotide of the present disclosure is administered to the subject. [0597] The disclosure provides a method of treating a malignancy in a subject, comprising administering to the subject the lentiviral particles or pharmaceutical composition of the disclosure. In some embodiments, the malignancy is a B-cell malignancy, a myeloma, or a solid tumor malignancy. The disclosure provides a method of treating diffuse large B-cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma in a subject, comprising administering to the subject the lentiviral particles or pharmaceutical composition of the disclosure. [0598] In some embodiments, the lentiviral particle is administered as a single injection. In some embodiments, the lentiviral particle is administered as at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 injections. Attorney Ref. No.061479-506001WO [0599] In some embodiments, the lentiviral particles disclosed herein may be used in a method to generate engineered cells in vivo. It has been observed that the lentiviral particles comprising surface engineering (e.g. a fusion protein as disclosed herein) may preferentially generate engineered central memory T cells (TCM). It has also been observed that administering lentiviral particles via one or more lymph nodes maycontribute to generation of a predominately TCM engineered cell phenotype. TCM may be characterized by expression of certain surface markers, for example, TCM may be CD62L+. TCM may also be CCR7+. TCM may also be characterized as CD45RA-, CD45RO+, and/or CD27+. In some embodiments, TCM are characterized as CCR7+, CD45RA-, CD45RO+, CD62L+, and CD27+. In some embodiments, TCM are characterized as CD45RA-, CCR7+. Without wishing to be bound by theory, it is contemplated that engineered TCM may persist for a longer time in vivo and may show improved effector function compared with engineered effector memory (TEM) or similar effector cell types. [0600] Thus, it is contemplated that the present disclosure further provides a method of generating predominately engineered TCM in vivo. Due to the presence of the fusion proteins on the surface of the engineered particles, similar observations may be found in the ex-vivo setting, so the present disclosure further provides a method of generating predominately engineered TCM ex vivo, using one or more of the methods disclosed here. For example, via extracorporeal delivery or traditional ex-vivo manufacturing. Method of Making [0601] In some embodiments, the disclosure provides a method of making a particle, comprising introducing a polynucleotide encoding a vector genome into a host cell comprising a polynucleotide encoding a fusion molecule (or fusion protein) as described herein. The fusion molecule (or fusion protein) and the vector genome are expressed by the host cell. The host cell packages the vector genome into a lentiviral particle comprising the fusion molecule (or fusion protein). [0602] In some embodiments, the disclosure provides an in vivo method of transducing target cells in a subject in need thereof, comprising administering to the subject a particle or pharmaceutical composition of the disclosure. The particle may be administered by intranodal, intravenous, or subcutaneous injection. [0603] Various disease or disorders may be treated using particles as disclosed herein, or pharmaceutical composition comprising them. The particles may be administered to a subject suffering from or at risk for a B-cell malignancy, relapsed/refractory malignancy, Attorney Ref. No.061479-506001WO diffuse large B-cell lymphoma (DLBCL), Burkitt’s type large B-cell lymphoma (B-LBL), follicular lymphoma (FL), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), mantle cell lymphoma (MCL), hematological malignancy, colon cancer, lung cancer, liver cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, skin cancer, melanoma, bone cancer, brain cancer, squamous cell carcinoma, leukemia, myeloma, B cell lymphoma, kidney cancer, uterine cancer, adenocarcinoma, pancreatic cancer, chronic myelogenous leukemia, glioblastoma, neuroblastoma, medulloblastoma, or sarcoma. [0604] Lentiviral particles of the present disclosure may enhance in vivo activity. Lentiviral particles of the present disclosure resist serum inactivation. Lentiviral particles of the present disclosure provide efficient targeting of activated T cells. Lentiviral particles of the present disclosure may require low physical particle per transducing unit compared to two component glycoproteins. Lentiviral particles of the present disclosure retain potential to transduce a broad range of non-T effector cells. Lentiviral particles of the present disclosure enhance particle to T cell binding. Lentiviral particles of the present disclosure enhance T cell activation. Lentiviral particles of the present disclosure enhance immune cell expansion. Lentiviral particles of the present disclosure enhance immune cell transduction. Lentiviral particles of the present disclosure enhance anti-tumor potency. Lentiviral particles of the present disclosure enhance immune cell persistence. [0605] Some embodiments include a method of making an adhesion molecule, a costimulatory molecule, an activation molecule, or a fusion molecule. The method may include transcribing or translating a nucleic acid (such as a DNA or RNA) that encodes a protein comprising the adhesion molecule, costimulatory molecule, activation molecule, or fusion molecule. Kits [0606] Disclosed herein, in some embodiments, are kits. In some embodiments, the kit includes an adhesion molecule. In some embodiments, the kit includes a costimulatory molecule. In some embodiments, the kit includes an activation molecule. In some embodiments, the kit includes a fusion molecule. In some embodiments, the kit includes a particle. In some embodiments, the kit includes a composition described herein. The kit may include instructions for use, such as instructions for use in a method herein. Attorney Ref. No.061479-506001WO EXAMPLES [0607] The following Examples describe how embodiments of the invention may be made, evaluated, and used. The Examples are intended to be illustrative and non-limiting. Example 1 [0608] This Example shows some impacts of incorporating a costimulatory molecule such as CD80, and/or an adhesion protein such as CD58, onto the surface of a lentiviral particle. A schematic of some such lentiviral particles is provided in FIG.1. Virus Production [0609] 1.2 × 10 6 293T cells were seeded into TC-treated 6-well plates in a total volume of 2.5 ml Complete DMEM media per well. 24 hours later, cells were transfected at room temperature. [0610] The following DNA was added to 500 µl serum free OptiMEM™ media: 2µg transfer plasmid, 1µg Gag/pol plasmid, 1µg REV plasmid, and 1µg envelope plasmid. 15 µl (15µg) PEI was then added to the media/DNA mix. The solution was then mixed well and incubated at room temperature for 20 minutes. The media/DNA/PEI mix was then added to 2.5 ml fresh Complete DMEM media. Seeding media in 293T-containing well was removed and replaced with fresh media containing the transfection reagents and placed in a 37° C humidified incubator. 48 hours later, the supernatant was collected and filtered through a 0.45µm PVDF filter. The virus-containing supernatant was concentrated using an Amicon-Ultra 15100K column and centrifuged at 3000xg for 30 minutes at 4° C. The virus was then stored at 4° C until use. 293T Transduction Titers [0611] 1 × 10 5 293T cells were seeded into TC-treated 12 well plates in 1ml Complete DMEM media. 24 hours later, empty wells were counted 3X to calculate titer. Then virus was added to wells in amounts of: 2ul, 1ul, 0.5ul, 0.2ul, 0.1ul, or 0.05ul virus per well. Virus was diluted 1:100 before adding to 293T cells. 3 days later, 293T cells were harvested for analysis by flow cytometry. Media was removed, and cells were washed in PBS. Cells were then washed in Trypsin and incubate for ~3-5 minutes in a 37° C incubator. Cells were resuspended in 1ml FACS buffer and ~100-200ul were added to a 96 well V bottom plate. Flow cytometry analysis was performed for mCherry expression. 293T Titer calculation [0612] TU/ml = (# of cells at time of transduction x %mCherry+ x 100)/(vector volume in ul x 1000) Attorney Ref. No.061479-506001WO [0613] Engineered particles packaging an anti-CD19 CAR containing either a CD3scFV alone or a CD3scFV+CD80, CD3scFV+CD58, or CD3scFV+CD80+CD58 were added to PBMCs from 2-3 donors. Example 2 [0614] This Example shows that incorporation of a costimulatory molecule and/or adhesion molecule on a lentiviral particle enhanced transduction of PBMCs by lentiviral particles as generated in Example 1. Virus Production [0615] All solutions used were the same as those described in Example 1. 28 × 10 6 293T cells were seeded into 16x T175 flasks (8x per vector) with 28e6293T cells each in a total volume of 25ml Complete DMEM media. 24 hours later, cells were transfected. Virus was produced as described in Example 1. All viruses included a Cocal envelope protein. [0616] List of Virus preps made for study: 1. CD3scfv only 2. CD3scfv+CD58 3. CD3scfv+CD80 PBMC Transduction and staining for flow cytometry [0617] 50 × 10 6 PBMCs were thawed, diluted to 2 × 10 6 cells/ml in complete media (e.g. RPMI or Optimem). IL-2 was added to a final concentration of 50IU/ml. [0618] 500µl(1e6 cells) were added to the wells of a Non TC-treated 48 well plate. Vector was added to the wells at MOI=10, 5, and 2 based on the SupT1 ddPCR titer and the plates were placed in 37° C incubator. [0619] After 3 days, vector was washed out and replaced with 500µl fresh RPMI media+IL- 2 (50IU/ml). Cells were mixed and 100-300µl were added to wells in a 96 well V-bottom plate for activation flow cytometry analysis. Cells were then washed with 200µl FACS buffer. The cell pellets were resuspended in 50-100µl PBS containing LiveDead Stain (1:1000) and incubate at 4° C for 20 min followed by another wash in 200µlFACS buffer. Cells were resuspended in 50µlof FACS buffer + surface stain cocktail, incubated for 30 min at 4° C, washed in 200µlFACS buffer. Results and Conclusions [0620] To assess if lentiviral particles with costimulatory molecules could better activate human T cells, the vector particles were added to human PBMCs at several MOI’s. 3 days later, the virus was removed and the cells were given fresh media and analyzed for the Attorney Ref. No.061479-506001WO activation marker CD25. CD3scfv+CD58 and CD3scfv+CD80 particles potently activated CD8 T cells compared to CD3scfv only (FIG. 2A and FIG. 2B). Furthermore, CD25 upregulation was dose-dependent (FIG.2A and FIG.2B). CD3scfv only lentiviral particles induced minimal levels of CD25 compared to the particles with CD80 or CD58 (FIG. 2A and FIG.2B). [0621] To examine transduction, 6 total days after vector addition, samples were analyzed for anti-CD19 CAR expression. Mirroring the CD25 expression on day 3, CD3scfv+CD58 and CD3scfv+CD80 particles were capable of transducing unstimulated PBMCs while CD3scfv only particles transduced unstimulated PBMCs to a lesser extent (FIGs. 2C-2F). Furthermore, transduction occurred in a dose-dependent manner for both CD3 and CD8 T cells (FIGs. 2C-2F). The data show that CD3scfv+CD58 and CD3scfv+CD80 particles efficiently activate and transduce unstimulated PBMCs in vitro compared to CD3scfv only. Importantly, the enhanced particles results in increased numbers of CAR+ T cells (FIGs. 2D-2F). [0622] To determine if adding costimulatory molecules to particles enhances Rapa- mediated expansion of CAR+ cells in vitro, the fold expansion of CD8 T cells was determined using CD3scfv+CD80 particles compared to CD3scfv only particles. PMBCs were cultures in either IL-2 only media or Rapamycin-only media. The addition of the costimulatory molecule did not affect the fold expansion when cultured with IL-2 only (FIG.2G) but the costimulatory molecule induced a dramatic expansion when cultured with Rapamycin-media (FIG. 2H). These results demonstrate that adding costimulatory molecules to particles enhanced Rapamycin-mediated expansion of CAR+ cells in vitro. [0623] This study demonstrated the ability of the CD3scfv + costimulatory molecules envelope construct to deliver payloads consisting of an anti-CD19 CAR to unstimulated PBMCs in vitro. The CD3scfv+CD58 and CD3scfv+CD80 particles induced activation of T cells as measured by CD25 expression and this activation correlated with transduction as measured by % of T cells expressing the anti-CD19 CAR and total CAR+ T cells. Furthermore, activation and transduction occurred in a dose-dependent manner. Costimulatory molecules also enhance Rapamycin-mediated expansion of CAR+ cells in vitro. This data further supports the use CD3scfv+CD58 and CD3scfv+CD80 particles to deliver CAR payloads to unstimulated PBMCs in vitro and in vivo. Attorney Ref. No.061479-506001WO Example 3 [0624] This Example shows that a combination of a costimulatory molecule (CD80 in this case), and an adhesion protein (CD58 in this case) further enhanced T cell activation and transduction. Particles having both molecules were generated. These particles were examined for their ability to activate and transduce unstimulated human PBMCs compared to particles only having anti-CD3scFv. Virus Production [0625] All solutions used were the same as those described in Example 1. 28 × 10 6 293T cells were seeded into 16x T175 flasks (8x per vector) with 28 × 10 6 293T cells each in a total volume of 25ml Complete DMEM media.24 hours later, cells were transfected. Virus was produced as described in Example 1. [0626] List of Virus preps made for study: 1. CD3scfv only 2. CD3scfv+CD58 3. CD3scfv+CD8 4. CD3scfv+CD80+CD58 PBMC Transduction and Analysis [0627] PBMCs were transduced and analyzed for expression as described in Example 2. [0628] Supernatant cytokine analysis was measured by Meso Scale Discovery (MSD) 3 days after transduction. [0629] Total K562.CD19, Raji, and Nalm6 tumor cells were tracked over time on an IncuCyte® for up to 15 days. Results and Conclusions [0630] To assess whether lentiviral particles with costimulatory molecules and adhesion molecules enhance T cell activation and transduction, the lentiviral particles were added to human PBMCs at several MOI’s. 3 days later, the virus was removed and the cells were given fresh media and analyzed for the activation marker CD25. CD3scfv+CD80+CD58 particles potently activated CD8 T cells compared to CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only (FIG. 3A and FIG. 3B). Furthermore, CD25 upregulation was dose- dependent and CD3scfv+CD80+CD58 particles activated CD8 T cells at a much lower dose (FIG. 3A and FIG. 3B). CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only lentiviral particles induced minimal levels of CD25 compared to the CD3scfv+CD80+CD58 particles (FIG. 3A and FIG. 3B). Attorney Ref. No.061479-506001WO [0631] To further characterize T cell activation, 3 total days after vector addition, samples were analyzed for cytokine expression. Similar to CD25 expression, CD3scfv+CD80 and CD3scfv+CD80+CD58 YK[]SMUO\ `O[O MKYKLUO XP SWN^MSWQ ?<C(l Y[XN^M]SXW ^W\]SV^UK]ON PBMCs at lower doses whereas CD3scfv+CD58 and CD3scfv only particles transduced unstimulated PBMCs to a lesser extent (FIG. 3C). Furthermore, CD3scfv+CD80+CD58 YK[]SMUO\ SWN^MON [XL^\] ?A(- KWN IC<(h `RO[OK\ 9:.\MP_&9:03, CD3scfv+CD80, and CD3scfv only did not (FIG. 3D and FIG.3E). The data show that CD3scfv+CD80+CD58 particles efficiently induce cytokine production in unstimulated PBMCs in vitro compared to CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only. [0632] To examine the role of CD80 and CD58 on transduction, 3 total days after vector addition, samples were analyzed for anti-CD19 CAR expression with CD3scfv+CD80 and CD3scfv+CD58 mixed particles (FIG. 3F and FIG. 3G) or CD3scfv+CD80+CD58 on the same particle (FIG. 3H and FIG. 3I ) compared to CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only. CD3scfv+CD80 and CD3scfv+CD58 mixed particles or CD3scfv+CD80+CD58 on the same particle were both capable of transducing unstimulated PBMCs to a greater extent than CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only (FIG. 3F, FIG. 3G, FIG. 3H and FIG. 3I). Furthermore, transduction occurred in a dose- dependent manner for both CD3 and CD8 T cells (FIG. 3F, FIG. 3G, FIG. 3H and FIG. 3I). The data show that both CD58 and CD80 either in mixed particles or on the same particle better activate and transduce unstimulated PBMCs in vitro compared to CD3scfv+CD58, CD3scfv+CD80, and CD3scfv only. [0633] To determine whether lentiviral particles with costimulatory and/or adhesion molecules have enhanced particle binding to T cells, the particles were cultured with PBMCs for 6 hours and then were analyzed for particle-associated molecules on T cells (Cocal, CD80, and CD58). Both CD3scfv+CD58 and CD3scfv+CD80+CD58 increased Cocal staining (FIG. 3J) and only CD3scfv+CD80+CD58 demonstrated high stating for CD80 (FIG. 3K) and CD58 (FIG. 3L). The data show that the combination of CD3scfv+CD80+CD58 enhances particle binding to T cells. [0634] To determine whether different T cell subtypes are generated by the lentiviral particles, PBMCs cultured with the lentiviral particles were profiled and gated on viable, CD3+ and CD8+. The cells were further analyzed by flow and principal component analysis was done based on parameters listed CCR7, CD45R, CD45RA, CD27, CD25, CAR+, total Attorney Ref. No.061479-506001WO cells, CD4, and CD8. The analysis revealed that 3 main clusters of differentiation are produced by the different particles (FIG.3M). [0635] Next, T cell subtypes generated by the particles was profiled. The cells were assessed using CD45RA and CCR7 markers 7 days post transduction at an MOI of 10. Naïve T cells are CD45RA+CCR7+, effector T cells (Teff) are CD45RA-CCR7-, central memory T cells (Tcm) are CD45RA-CCR7+, and terminally differentiated effector memory T cells (Temra) are CD45RA+CCR7-. In a first experiment, CD3scfv only particles produced a majority of Teff cells whereas CD3scfv+CD80 particles produced a majority of Tcm cells (FIG. 3N). In a second experiment, CD3scfv only particles produced both Teff and Tcm cells, CD3scfv+CD80 particles produced a majority of T cm cells, CD3scfv+CD58 particles produced a majority of T cm cells, and CD3scfv+CD80+CD58 produced a majority of T cm cells (FIG.3O). The data show that both the addition of CD80 and/or CD58 to the particle consistently produces a T cm cell phenotype. CD45RA-CCR7+ T cm cells are thought to have increased longevity and proliferative capacity and correlate with better antitumor responses in vivo. [0636] To assess the ant-tumor efficacy of CAR T cells generated using lentiviral particles expressing costimulatory and/or adhesion molecules, PBMCs were transduced and cultured with tumor cells. Specifically, particles comprising a nucleotide sequence encoding an anti- CD19 CAR were added to PBMCs at an MOI of 10, along with tumor cells (K562.CD19 or Raji cells) at PBMC:Tumor ratio of 5:1 and put directly on an Incucyte. Tumor cell killing was measured over time. The highest killing was observed with particles composed of at least CD80 in addition to CD3scfv (FIG. 4A and FIG. 4B). In a subsequent experiment, tumor cell killing was measured 7 days after transduction with an MOI of 10. The total number of CAR+ cells were calculated and incubated with either K562.CD19 or Raji cells at E:T ratios of 0.5 and 1, respectively. CAR T cells generated using a mixture of individual particles with CD80 or CD58. Similarly, the highest killing was observed with particles composed of at least CD80 in addition to CD3scfv, including CD80+CD58 (FIG. 4C and FIG. 4D). An additional experiment determined the effect for CAR T cells generated with a single lentiviral particle having both CD80 and CD58. Tumor cell killing was measured 7 days after transduction at an MOI 10. The total number of CAR+ cells were calculated and incubated with either K562.CD19 or Nalm6 cells at E:T ratios of 1:1, respectively. The CD80+CD58 dual particle provided the highest cytotoxic function (FIG.4E and FIG.4F). Attorney Ref. No.061479-506001WO [0637] This study demonstrated the CD3scfv+CD80+CD58 particles induced the highest differentiation of T cells and the highest cytokine production at the lowest MOI. CD3scfv+CD80+CD58 particles further had the highest T cell binding. Furthermore, this study demonstrated that CD3scfv+CD80+CD58 particles provided the highest cytolytic function in vitro. Example 4 [0638] This Example shows tumor control by in vivo transduction of T cells with a lentiviral particle with CD3scfv or CD3scfv+CD80. The lentiviral particle contains a polynucleotide encoding an anti-CD19 CAR. The lentiviral particle was delivered via intravenous injection into NSG MHCI/II KO mice. The mice used in the study were immune-compromised and contain engrafted human T cells and circulating human B cells. Study Design Virus preparation, animal strain, cell lines [0639] 11 female NSG MHCI/II KO mice (Jackson laboratory) were and housed following institutional guidelines (Fred Hutchinson Cancer Research Center). Study Protocol [0640] 11 female NSG MHCI/II KO mice were acclimated for one week after receipt. At day -7, blood from all mice was collected for flow cytometry analysis to quantify degree of humanization. Mice were randomized according to their total human CD3 levels into the treatment groups described in Table 5. Table 5: Study Treatment Groups Study Timeline [0641] At study Day 0 (SD0), 20 × 10 6 PBMCs were injected into the intraperitoneal cavity. Mice were then dosed with virus particles according to the table above, followed with a challenge of 5 × 10 5 luciferase+ Nalm6 tumor cells intravenously. Tumor burden was Attorney Ref. No.061479-506001WO measured for the duration of the study. At SD11, blood was collected and CAR T cells were measured. [0642] At SD75, survivor mice were rechallenged with 5 × 10 6 Nalm6 cells. Results and Conclusions [0643] On Day 11 of the study, blood was collected from both groups. The level of CAR T cells in the blood was higher in the CD3scfv+CD80 particle-treated group compared to the CD3scfv particle-treated group (FIG. 5A). The CD3scfv+CD80 particle-treated group was also able to decrease tumor burden during the initial challenge and subsequent rechallenge compared to the CD3scfv particle-treated group (FIG.5B and FIG.5C). [0644] In summary, when delivered intravenously, CD3scfv and CD3scfv+CD80 engineered lentivirus particles successfully transduced T cells in vivo. While both groups decreased tumor burden after initial challenge and subsequent rechallenge, particles with costimulatory molecule CD80 provided greater anti-tumor efficacy and anti-tumor immune response. Example 5 [0645] This Example shows transduction with engineered lentiviral particles as described herein to transduce T cells in a short incubation period. Without wishing to be bound by theory, this study provides proof of concept support that the engineered particles described may be useful in an extracorporeal intravenous system. [0646] PBMCs from 3 healthy donors were thawed and cultured with vector particles containing an anti-CD19 CAR-mCherry payload pseudotyped with either CD3scfv+cocal or CD3scfv+CD80+CD58+Cocal, generally as described in Example 2. After the indicated time point, cells were washed in serum-free media containing IL-2, human ab serum, HEPES, and glutamine. Cells were then plated in 1 ml serum-free media with IL-2 in a 24 well non-TC-treated plate. 3 days later cells were harvested and CD25 expression was measured by flow cytometry on viable T cells (FIG.6A). The remaining cells were washed and re-plated in 1ml fresh media containing IL-2. 4 days later (Day 7 after transduction) viable T cells were analyzed by flow cytometry for CAR surface expression (FIG. 6B). %CAR was measured by staining for anti-CD19 mAb and mCherry expression. [0647] As shown in FIGs. 6A-6B, vector particles comprising activation, costimulation, and adhesion molecules (e.g. CD3scFv+CD80+CD58 particles) efficiently transduced T cells after short incubation periods to a greater extent than particles comprising a CD3scFv Attorney Ref. No.061479-506001WO without costimulation and adhesion components. These results indicate that vector particles as described herein may enable T cell transduction during short incubation periods ex vivo, for example in a closed-loop and/or extracorporeal system. Example 6 [0648] This Example shows the transduction potential of lentiviral particles comprising a mutated (blinded) envelope protein. Envelope proteins, such as VSV-G or Cocal, can be mutated such that they cannot bind the LDL receptor. These modifications may enhance the specificity of lentiviral particles and reduce or eliminate off-target transduction. [0649] SupT1 cells were cultured with vector particles containing an anti-CD19 CAR- mCherry payload, generated generally as described in Example 2. Specifically, 0.02uL of concentrated particles were added to 3.75 x10 4 SupT1 cells and assessed for CAR expression 3 days later. The cells were cultured in the following conditions: Table 6 [0650] As shown in the top row of FIG.7A lentiviral particles comprising the blinded VSV- G mutant envelopes alone (without CD3scFv+CD80+CD58) exhibited greatly reduced transduction of SupT1 cells compared with a non-blinded VSV-G control. The bottom row depicted in FIG. 7A shows that the addition of activation, costimulation, and adhesion molecules in particles comprising blinded VSV-G mutant envelope proteins resulted in increased transduction. [0651] Wishing to confirm the results seen using a T cell line, the experiment was replicated using PBMCs. On day 0, PBMCs from 2 donors were thawed and 2 x 10^6 cells were placed in wells of a 24-well plate. Vector particles containing an anti-CD19 CAR-mCherry payload, generated generally as described in Example 2, were added to each cell-containing well according to the following chart. Attorney Ref. No.061479-506001WO Table 7 [0652] On day 3, the media was changed, and cells were re-plated with fresh media and samples were taken for assessment of transduction via flow cytometry. As shown in FIGs. 7B-7C, lentiviral particles comprising blinded VSV-G envelopes resulted in reduced transduction compared with the non-blinded VSV-G control in both CD4 (FIG. 7B) and CD8 (FIG. 7C) T cells. In addition, the addition of CD3scFv+CD80+CD58 to lentiviral particles resulted in increased transduction compared to lentiviral particles without CD3scFv+CD80+CD58. In addition, lentiviral particles comprising CD3scFv+CD80+CD58 without VSV-G also exhibited poor transduction. [0653] On day 5, additional samples were taken for assessment of transduction via flow cytometry. Expression of CAR on day 5 was similar to expression on day 3 (data not shown). [0654] The results of this study support the hypothesis that lentiviral particles comprising a blinded envelope protein and activation, costimulation, and adhesion molecules are capable of transducing primary T cells. Example 7 [0655] This Example shows expansion of non-transduced T cells after administration of a lentiviral particle with CD3scfv or CD3scfv+CD80+CD58. The lentiviral particle contains a polynucleotide encoding an anti-CD19 CAR. The lentiviral particle was delivered via intravenous injection into mice. Study Design [0656] Mice were acclimated for one week after receipt. At day -7, blood from all mice was collected for flow cytometry analysis to quantify degree of humanization. Mice were Attorney Ref. No.061479-506001WO randomized according to their total human CD3 levels into the treatment groups described in the table below. Table 8 Study Timeline [0657] At study Day 0 (SD0) mice were then dosed with virus particles according to the table above. At SD11, blood was collected, and CAR negative T cells were measured. Results and Conclusions [0658] On Day 11 of the study, blood was collected from both groups. The level of CAR negative T cells in the blood was higher in the CD3scfv+CD58+CD80 particle-treated group and was dose-dependent compared to the CD3scfv particle-treated group (FIG. 8). These results indicate, when delivered intravenously, CD3scfv+CD58+CD80 engineered lentivirus particles appears to activate and expand even non-transduced T cells in vivo. Without wishing to be bound by theory, this activation of non-transduced T cells may enable a lower dose of engineered lentiviral particles as non-transduced cells may exhibit anti- tumor activity. Example 8 [0659] The Examples shows that combination of a costimulatory molecules CD80, anti- CD3scFv, and an adhesion protein CD58 expressed as a single fusion polypeptide (FIG. 9A) further enhances T cell activation and transduction. Particles having the fusion polypeptide were generated. These particles were examined for their ability to activate and transduce unstimulated human PBMCs compared to particles expressing the three proteins separately and compared to particles expressing a CD80/CD58 fusion polypeptide (FIG. 9B) and expressing anti-CD3scFv separately. Virus Production [0660] All solutions used were the same as those described in Example 1. Virus was produced as described in Example 1. [0661] List of Virus preps made for study: Attorney Ref. No.061479-506001WO ,) h(9:.\MP_&9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO $"/43 ][SYUO P^\SXW% -) 9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO KWN h(9:.\MP_ OaY[O\\ON separately (#455 dual fusion) .) h(9:.\MP_&9:3+&9:03 OaY[O\\ON \OYK[K]OUb eHOYK[K]Of PBMC Transduction and Analysis [0662] Three healthy PBMC donors were incubated with the indicated lentiviral particles described above at a multiplicity if infection (MOI) of 10 in 0.9% Sodium Chloride buffer at a concentration of 20e6 cells/ml in a total volume of 100ul in a 96 well U-bottom plate. After 1 hour of incubation, the cells were washed, stained with antibodies, and analyzed by flow cytometry. The percentage of cells with bound Cocal and Cocal geometric mean fluorescence intensity (gMFI) are shown in FIG.10. Cells analyzed by flow cytometry were gated on viable, CD3+, CD14-, CD56-, CD8+, Cocal+ cells. Similar data was seen for CD4+ T cells (data not shown). Results and Conclusions [0663] To assess whether lentiviral particles with costimulatory molecules and adhesion VXUOM^UO\ h(9:.\MP_&9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO OWRKWMO I cell activation and transduction, the lentiviral particles were added to PBMCs from three healthy PBMC donors at several MOI’s and at 2E6 cells/ml in RPMI media. 3 days later, the virus was removed and the cells were washed, given fresh media, and analyzed for the activation marker CD25 by flow cytometry. Cell were gated on viable, CD3+, CD4+ or 9:3& MOUU\) h(9:.\MP_&9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO e"/43f particles potently activated CD4+ (FIG. 11A and FIG. 11C) and CD8+ (FIG. 11B and FIG. 11D) T cells. Furthermore, the triple fusion “#498” particles activated CD4+ (FIG. 11A and FIG. 11C) and CD8+ (FIG. 11B and FIG. 11D) T cells through displayed CD25 upregulation at a much lower dose as compared to “#455” the dual fusion and “Separate” lentiviral particles. [0664] To further characterize T cell activation, lentiviral particles were added to PBMCs from three healthy PBMC donors at several MOI’s and at 2E6 cells/ml in RPMI media. 3 days later, supernatant was harvested and cytokines were measured using V-PLEX™ Proinflammatory Panel 1 Human Kit. Similar to CD25 expression, the triple fusion “#498” particles were capable of inducing more T cell activation-associated cytokines including ?<C(l' ?A(-' KWN IC<(h K\ MXVYK[ON ]X e"/00f ]RO N^KU P^\SXW KWN eHOYK[K]Of YK[]SMUO\) >SQRO[ ?<C(l Y[XN^M]SXW SW ^W\]SV^UK]ON E8B9\ `K\ XL\O[_ON K] UX`O[ NX\O\ $FIG.12A). Attorney Ref. No.061479-506001WO Furthermore, the triple fusion “#498” YK[]SMUO\ SWN^MON [XL^\] ?A(- KWN IC<(h Y[XN^M]SXW as compared to “Fusion-455” and “Separate” particles (FIG.12B and FIG.12C). The data show that the triple fusion “#498” particles efficiently induce cytokine production in unstimulated PBMCs in vitro compared to “#455” the dual fusion and “Separate” particles. [0665] T cell subtypes generated by the particles were profiled. The cells were assessed using CCR7, CD27, CD28, and CD57 markers. Lentiviral particles were added to PBMCs from three healthy PBMC donors at several MOI’s and at 2E6 cells/ml in RPMI media. 7 days post transduction, cells were washed and CAR surface marker expression was analyzed by flow cytometry. Cells were gated on viable, CD3+, CD4+ or CD8+, CAR+ cells. [0666] Non-terminally differentiated memory T cells are CCR7+CD27+CD28+. Triple fusion-containing particles “#498” produced a greater percentage of CCR7+CD27+CD28+ memory-like CAR+ T cells as compared to dual fusion “#455” and “Separate” particles (FIG. 13A and FIG.13C). [0667] CCR7+CD27+CD28+ memory-like CAR+ T cells are thought to have increased longevity and proliferative capacity and correlate with better antitumor responses in vivo. Triple fusion particles “#498” produced a smaller percentage of senescence marker CD57 at an MOI of 2 as compared to dual fusion “#455” and “Separate” particles (FIG. 13B and FIG. 13D). Example 9 [0668] IRS\ ;aKVYUO \RX`\ I MOUU KM]S_K]SXW KWN ?<Cl Y[XN^M]SXW PXUUX`SWQ in vivo transduction of T cells by a lentiviral particle displaying #498 triple fusion polypeptide as compared to #455 dual fusion and “Separate” particles as described above. The lentiviral particle contains a polynucleotide encoding an anti-CD19 CAR. [0669] On study Day –4 NSG MHC I/II dKO mice were injected via tail vein injection with 2.5E5 Nalm6 cells expressing firefly luciferase (ffluc) (FIG.14A).3 days later (study Day –1), mice were imaged via bioluminescence imaging and randomized to study arms according to tumor burden (total flux), the same day all mice were humanized by injecting 20E6 human PBMCs intraperitoneally in 100 µl of 1X sterile PBS. The mice used in the study were immune-compromised and contain engrafted human T cells and circulating human B cells. [0670] The next day (study Day 0), mice were treated via intraperitoneal injection with different doses of lentiviral particles displaying: ,) h(9:.\MP_&9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO e"/43f ][SYUO P^\SXW6 Attorney Ref. No.061479-506001WO -) 9:3+&9:03 OaY[O\\ON K\ K \SWQUO P^\SXW YXUbYOY]SNO KWN h(9:.\MP_ OaY[O\\ON separately “#455” dual fusion; or .) h(9:.\MP_&9:3+&9:03 OaY[O\\ON \OYK[K]OUb eHOYK[K]Of) [0671] A control study arm treated mice with 1xPBS (Neg) via intraperitoneal injection. Mice were then weighted two times a week throughout the study to monitor body weight change and imaged weekly to monitor tumor burden. Mice were bleed on study Days 4, 11, 18, 25, and 32 to perform flow cytometry analysis. Study day 4 activation markers CD25 (FIG. 14B) and CD71 (FIG. 14C) on CD3+ T cells were analyzed. 4 days after lentiviral YK[]SMUO ][OK]VOW] $\]^Nb :Kb /% \O[^V `K\ MXUUOM]ON P[XV LUXXN KWN ?<Cl UO_OU\ SW ]RO serum were measured using V-PLEX™ proinflammatory panel 1 human kit (Mesoscale Discovery) (FIG.14D). [0672] Apheresis blood was washed on the Lupagen™ machine and incubated with lentiviral particles at an MOI of 2 for 1 hour in saline. The lentiviral particle contains a polynucleotide encoding an anti-CD19-mCherry transgene. [0673] The particle-bound cells were then washed of unbound particles to generate the “Final” material. Particle-bound cells were assessed by staining for Cocal on various cell populations (CD4+ T cells, CD8+ T cells, NK T cells, NK cells, CD56+ NK cells, monocytes, B cells, and other MFI) and analyzing by flow cytometery. Cocal geometric mean fluorescence intensity are shown (FIG. 15A-15C). The strongest binding was observed with particles displaying the triple fusion “#498” as compared to particles displaying the dual fusion “#455”. Example 10 [0674] This Examples shows in vivo antitumor responses to lentiviral particles displaying a costimulatory and adhesion molecule fusion protein using the Lupagen™ System. [0675] On study Day –4 NSG MHC I/II DKO mice were injected via tail vein injection with 2.5E5 Nalm6 cells expressing GFP/ firefly luciferase (ffluc). 3 days later (study Day –1), mice were imaged via Bioluminescence imaging and randomized to study arms according to tumor burden (total flux). On study Days 0 and 1, mice were injected with PBMCs from 2 different donors either after Lupagen™ wash or after incubation with lentiviral particles comprising “#455” dual fusion or triple fusion “#498” polypeptides in the lentiviral particle surface. On study Day 8 and every week through the study, mice were imaged via Bioluminescence imaging using the IVIS™ spectrum system to analyze tumor burden (total flux) (FIG. 16C and 16D). Serial weekly blood draws were collected to perform flow Attorney Ref. No.061479-506001WO cytometry analysis and assess CAR T cell expansion and persistence (FIG. 16A and 16B). Disease progression was monitored by Bioluminescence imaging once a week after d- Luciferin subcutaneous injection using the IVIS™ imaging system (FIG. 16E). Example 11 [0676] This Example shows screening of lentiviral particles displaying variations of CD58 and CD80 dual-fusion polypeptides and screening of lentiviral particles displaying variations of CD58, CD80, and anti-CD3 scFv tri-fusion polypeptides. [0677] Cryopreserved human PBMCs from normal donors were obtained from AllCells™. Human PBMCs were cultured in T cell growth (TCGM) media (RPMI1640 + 5% HuAB serum + 1x GlutaMax + HEPES). For lentiviral transduction, virus was added to the PBMC cells for 3 days. Stimulation and lentiviral infection were then terminated by washing and re-seeding PBMCs in fresh TCGM media. [0678] To analyze T cell activation, about 0.1×10 6 cells were pelleted after the 3-day production period following lentiviral transduction described above. Cells were then analyzed by flow cytometry as follows. Cells were resuspended in Fixable Viability Dye eFluor 780 in PBS for 10 minutes, then washed with Cell Staining Buffer. T cell activation was measured by detection of hCD25 marker using an anti-CD25-PE/Cy7 antibody diluted 1:100 in Cell Staining Buffer. [0679] To measure CAR expression levels and transduction efficiencies, about 0.1×10 6 cells were pelleted after a 7-day production period following lentiviral transduction. Cells were then analyzed by flow cytometry as follows. Cells were resuspended in Fixable Viability Dye eFluor 780 in PBS for 10 minutes, then washed with Cell Staining Buffer. FMC63 CAR surface expression was detected using an anti-ID-FITC antibody diluted 1:100 in Cell Staining Buffer. Cells were pelleted after a 20 minute incubation in the dark, followed by 2× wash with Cell Staining Buffer. All flow cytometric analysis was done on an Attune™ NxT Flow Cytometer and analyzed with FlowJo™. [0680] Day 7 transduced primary T cells expressing FMC63 CAR were counted, resuspended to a cell density equal to 0.4×10 6 CAR+ cells/ml in cell-assay media $GEB?,1/+ & ,+#<8H%) ,++ nU _XU^VO XP 97G& MOUU\ $/+'+++ 97G& MOUU\% `O[O KNNON ]X a flat-bottom 96 well plate containing 10,000 Nalm6 cells and incubated at 37° C for effector to target ratio of 4:1. CAR+ cells are serially diluted in cell-assay media prior to plating to achieve lower effector to target ratios. Killing of target cells was analyzed in an IncuCyte™ Live Cell Analysis System. Each well was imaged every 6 hours and the number of Nalm6 Attorney Ref. No.061479-506001WO cells was quantified to assess the kinetics of T cell cytotoxicity. After 24 hours, supernatant from each well was collected for cytokine measurements according to manufacturer's protocol. Nalm6 target cell lysis was tracked over >4 days. The Nalm6 target cell line was stably labeled with nuclear mKate2 by lentiviral transduction with IncuCyte™ NucLight Red Lentivirus Reagent. Results [0681] Healthy donor PBMCs were transduced with lentiviruses carrying a FMC63 CAR transgene and displaying various surface engineered dual-fusion proteins at MOI 2 and 5. Early activation is determined based on hCD25 staining on Day 3 (FIG. 17), and CAR expression level was measured by staining with an anti-FMC63 antibody conjugated to FITC (FIG. 18). CAR-T cells were challenged with Nalm6-NIR (FIG. 19A-19D) to compare killing kinetics and target-dependent cytokine production levels (FIG. 20). Pro- inflammatory cytokine concentrations in co-culture supernatant after 24 hours of assay setup, in combination of killing kinetics indicate that CAR-T cells comprising the #455 dual fusion construct and separately expressed anti-CD3 scFv performed the best in functional assay. [0682] With very low MOI transduction (MOI = 0.5 and 1), lentiviral particles produced using anti-CD3 scFv and dual-fusion plasmids promoted T cell activation on Day 3 (FIG. 21), enhanced transduction efficiency and increased CAR expression on Day 7 (FIG.22). [0683] Tri-fusion protein “#498” surface engineered particles were compared against dual- fusion “#455” particles in PBMC transduction using both high and low MOI (MOI=1 and 10). Lentiviral particles produced using tri-fusion versions #479, #496, and #498 enhanced early T cell activation in PBMCs (FIG. 23). #496 and #498 outperformed in transduction efficiency and CAR expression on Day 7 (FIG. 24). In this experiment, #498 had the most significant effect on CAR+ T cell expansion (FIG.25). Example 12 [0684] This Example shows T cell activation and transduction with lentiviral particles displaying a CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide. [0685] Human PBMCs from 3 normal donors were cultured in T cell growth (TCGM) media (RPMI1640 + 5% HuAB serum + 1x GlutaMax + HEPES). For lentiviral transduction, lentiviral particles were added to the PBMC cells. [0686] To analyze T cell activation, cells were pelleted after 3 days and then analyzed by flow cytometry. T cell activation was measured by detection of hCD25 marker using an Attorney Ref. No.061479-506001WO anti-CD25-PE/Cy7 antibody diluted 1:100 in Cell Staining Buffer. To measure CAR expression levels and transduction efficiencies, cells were pelleted after a 7-day production period following lentiviral transduction. Cells were then analyzed by flow cytometry. Anti- CD19 CAR surface expression was detected and all flow cytometric analysis was done on an Attune™ NxT Flow Cytometer and analyzed with FlowJo™. Day 7 transduced primary T cells expressing anti-CD19 CAR were counted, resuspended, and added to Nalm6 tumor cells. Killing of target Nalm6 cells was analyzed in an IncuCyte™ Live Cell Analysis System. Each well was imaged every 6 hours and the number of Nalm6 cells was quantified to assess the kinetics of T cell cytotoxicity. After 24 hours, supernatant from each well was collected for cytokine measurements according to manufacturer's protocol. [0687] Healthy donor PBMCs (from three donors) were contacted for less than one hour with lentiviruses carrying an anti-CD19 CAR transgene and displaying surface engineered tri-fusion proteins at MOI 2 (FIG. 28A). Consistent and efficient binding of T cells to engineered lentiviral particles was observed and measured by percentage of CD3+ T cells positively staining for Cocal (FIG. 28B). Selective T cell binding was observed in a Cocal staining peak shift for CD3+ T cells relative to CD3- T cells (FIG. 28C). Activation was determined based on hCD25 staining on Day 3 (FIG. 28D), and CAR expression level was measured (FIG. 28E). The engineered lentiviral particles demonstrated robust avidity and selectivity for T cell binding following short duration (< 1 hour) culture. Transduced PBMCs were cultured with Nalm6 tumor cells. Specifically, anti-CD19 CAR+ T cells were serial-stimulated with Nalm6 tumor cells every 2-3 days. Total Nalm6 tumor cells were measured over time using an IncuCyte® providing a measurement of tumor cell killing over time (FIG. 29). This assay measures the ability of the CAR T cells to expand and kill multiple tumor cells over time and showed that anti-CD19 CAR T cells generated with lentivirus particles displaying a CD58, CD80, and anti-CD3 scFv tri-fusion protein demonstrated serial killing in vitro. [0688] In a study of hematologic malignancy in a tumor xenograft model, on Day -4, 2.5 × 10 5 Nalm6 cells were intravenously injected into NSG MHCI/II KO mice. At study Day -1, 20 × 10 6 PBMCs were injected into the intraperitoneal cavity. At study Day 0, mice were dosed with virus particles displaying a CD58, CD80, and anti-CD3 scFv tri-fusion protein (FIG. 30A). [0689] Four days after lentiviral particle administration, cells were harvested and expression of activation markers CD25 (FIG. 30B) and CD71 (FIG. 30C% KWN Mb]XTSWO ?<C(l Attorney Ref. No.061479-506001WO production (FIG. 30D) were measured by flow cytometry on viable CD3+ T cells in the blood. CAR T cell expansion was analyzed at doses of 10 Million and 50 Million transducing units (TU). On Day 11, total anti-CD19 CAR+ T cells found in the blood were analyzed by flow cytometry for CAR surface expression (FIG. 30E). Tumor burden was assessed as total flux and measured for the duration of the study using an In vivo Imaging system (IVIS®) (FIG.30F). Example 13 [0690] This Example analyzed transduction of T cells by a lentiviral particle displaying either a dual-fusion (#455) construct or a triple fusion (#498) construct. FIGs. 32H-32I show total tumor burden (Total flux) over the course of 28 days of the study in the blood of mice injected with PBMCs from Donor 1 (FIG. 32H) or Donor 2 (FIG. 32I) after Lupagen™ incubation with untreated PBMC control, lentiviral particles displaying either a dual “#455” or triple “#498” fusion construct. The data show that extracorporeal incubation of PBMCs with the lentiviral particles described herein generates potent antitumor responses in vivo. Lentiviral particles displaying a CD58, CD80, and anti-CD3 scFv tri- fusion “#498” polypeptide showed enhanced antitumor activity in Donor 2 with a lower cell dose (Donor 2 - 15e6 cells were injected; Donor 1 - 25e6 cells were injected). [0691] On study Day 49 (rechallange Day 0) the mice were injected via tail vein injection with an additional 2.5E5 Nalm6 cells expressing firefly luciferase (ffluc) to assess clearance of tumor re-challenge (FIG. 33A). Tumor burden was assessed as total flux and measured for the duration of the rechallange study for Donor 1 (D1) and Donor 2 (D2) using an In vivo Imaging system (IVIS®) (FIG. 33C). FIG. 33B shows the tumor burden in NSG MHCI/II KO mice after administration of T cells produced via extracorporeal incubation of PBMCs from Donor 1 (D1) or Donor 2 (D2) incubated with lentiviral particles displaying either a dual fusion “#455” or triple fusion “#498”construct following tumor cell rechallenge at Day 49.Lentiviral particles displaying a CD58, CD80, and anti-CD3 scFv tri-fusion “#498” polypeptide generated anti-CD19 CAR T cells, which showed persistence following primary tumor clearance and protection against tumor rechallenge in vivo. Example 14 [0692] This Example shows T cell transduction and activation with viral particles displaying a CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide. [0693] Human PBMCs from 3 normal donors were cultured in T cell growth (TCGM) media (RPMI1640 + 5% HuAB serum + 1x GlutaMax + HEPES). For lentiviral transduction, viral particles were added to the PBMC cells. Attorney Ref. No.061479-506001WO [0694] Healthy donor PBMCs (from three donors) were contacted for one hour with engineered lentiviruses carrying an anti-FITC CAR transgene and displaying multi-domain fusion (MDF) surface proteins (FIG.34A). Consistent and efficient binding of T cells to the engineered lentiviral particles was observed and measured by percentage of CD3+ T cells positively staining for Cocal (FIG.34B). Lentiviral particles displaying MDF (multidomain fusion) proteins showed a significant increase in particle-bound cells after just one hour of incubation. Selective T cell binding was observed in inreased % Cocal (FIG. 34C) and geometric mean fluorescent intensity (gMFI) Cocal (FIG.34D) for CD4+ and CD8+ T cells relative to B cells, monocytes, and NK clls. The lentiviruses engineered to display MDF surface proteins showed selective binding to T cells over NK cells, B cells, or monocytes in a dose dependent manner. Activation was determined by %CD25 staining on Day 3 (FIG.34F) and % CAR expression level on Day 7 post-PBMC transduction (FIG.34G). The engineered lentiviral particles demonstrated robust transduction and activation of T cells in vitro in a dose-dependent manner. Example 15 [0695] Example 15 describes the efficacy of lymph node administration of engineered viral particles in non-human primates. T cell transduction and activation with viral particles surface engineered to display a CD58 and CD80 di-fusion polypeptide and anti-CD3 scFv showed in vivo CAR T cell generation. The viral particles were engineered to display an anti-CD3 scFv that binds NHP CD3 and a payload comprising a human-specific anti-CD20 CAR which cross-reacts with NHP CD20 (FIG.35A). Table 9 Abbreviations [0696] The objectives of the study included analyzing the ability of engineered viral particles to transduce T cells and generate functional CAR T cells in vivo in a large animal model. Another objective of the study was to assess lymph node injection as a viable route of administration for Attorney Ref. No.061479-506001WO engineered viral particles. Another objective of the study was to analyze the function of the generated CAR T cells on B cell depletion as well the persistence of the CAR T cells and/or B cell depletion. [0697] In the present NHP study, the subjects of the study were Pig-tailed macaques (Macaca nemestrina) that were either male or female, a minimum size of 3.5-5 kg, no age restriction (other than to ensure sufficient size). Three animals were studied. A summary of the animals treated is shown in FIG.35C. [0698] Engineered viral particles were generated with surface molecules that are cross-reactive with pig-tailed macaque (Human CD58-Human CD80 fusion polypeptide and an NHP-specific anti-CD3 scFv) (FIG. 35A) and !CD20-CAR and low-affinity nerve growth factor receptor (LNGFR) molecule payload (FIG.35B). [0699] The particles were dosed in transducing units per kg (TU/kg) as determined by ddPCR titration on SupT1 cells. FIG.36 shows the study design and timeline for the study. [0700] Vector formulations were maintained at -80°C until the day of animal treatment. Vector was kept on ice for transport to the animal facility and equilibrated at room temperature for approximately 5-15 minutes prior to lymph node injection in macaques using a single-use sterile needle and 1 ml syringe. Vector was injected within 2 hours of preparation. [0701] Animals were individually housed (i) during the initial pre-study period in which the animals were acclimated to the jacket and tether and (ii) for at least 4 weeks following engineered viral particle injection. Animals had ad libitum access to water and food was only restricted prior to sedation or anesthesia. Study Procedure [0702] Blood was drawn pre-study for isolation of (i) PBMCs, (ii) serum, and (iii) gDNA. Serum and gDNA were isolated and stored at -80 ºC for later analyses. PBMCs were cryopreserved, analyzed by a pre-injection flow cytometry panel and transduced with the same engineered viral particles that were injected in vivo. [0703] In addition, in the two-week period prior to lymph node injection, up to two blood draws were taken to determine baseline values for the assays below. [0704] Day 0: - Prophylactic diphenhydramine (IV injection, one dose) - Surgical placement of central catheter (if on tether) - Surgical placement of telemetry device to monitor body temperature - Optional: blood draw - Ultrasound-guided Lymph Node injection of engineered viral particles [0705] Animals with a central intravenous catheter implanted received a continuous IV antibiotic infusion via the catheter to prevent septicemia. Attorney Ref. No.061479-506001WO [0706] Animal Monitoring and Blood Collection: Blood was drawn throughout the study with biweekly or triweekly blood draws planned for the first 3-4 weeks of the experiment and weekly thereafter. The number of draws will depend on assay results and the clinical condition of each animal. Blood draw schedule may be adjusted dependent on maximum blood draw allowance of 10ml/kg body weight every two weeks. [0707] Animals were observed daily for general health, appetite, activity level, responsiveness, and fecal production. Body temperature was measured continually using a telemetry device. Animals were monitored for clinical signs of cytokine release syndrome (CRS) and immune effector cell- associated neurotoxicity syndrome (ICANS) based on an established set of criteria. Clinical signs of CRS or ICANS were confirmed by CBCs, serum chemistries, serum IL-6, serum ferritin and serum CRP levels. Animals treated with engineered viral particle product in the present study did not display any clinical signs of CRS. [0708] Study Endpoint: The animals were monitored and have blood drawn periodically following injection of engineered viral particles. Analytical Assays: [0709] A. Cell Surface Flow Cytometry [0710] B. Serum IL-6 [0711] C. Serum ferritin [0712] D. Serum CRP Safety Efficacy Criteria: [0713] Safety and Efficacy was assessed in each animal using a combination of analytics comparing results pre- and post-injection. A. Depletion of B Cells (Flow Cytometry). The fraction of starting CD20+ cells were plotted longitudinally starting with pre-injection samples and extending throughout the study. B. Clinical Symptoms of CRS. The serum concentration of IL-6, ferritin, and CRP were plotted longitudinally starting with pre-injection samples and extending throughout the study. This study is a pilot however, despite the small study size, meaningful trends in regards to B cell depletion in vivo were detected. Safety Criteria: [0714] Safety was assessed using a combination of analytics. A. CRS Monitoring a. Body temperature b. CBCs c. Serum IL-6 Attorney Ref. No.061479-506001WO d. Serum ferritin e. Serum CRP f. Serum chemistries (kidney (creatinine, BUN, total protein, electrolytes) and liver (AST, ALT, ALP, total bilirubin, albumin) function or damage) B. Clinical and Physical Assessments – including general health, appetite, activity level, responsiveness, hydration, and fecal production C. Necropsy and Histopathology on a subset of animals a. Gross findings b. Histopathology of tissue sections c. qPCR of tissue sections d. RNA-ISH of tissue positive by qPCR. Results [0715] Depletion of B Cells was analyzed throughout the study in the fraction of starting CD20+ cells. In two of the three animals studied, B cell reduction was observed throughout the course of the study (FIG. 37). In vivo transduction with the engineered viral particles was well-tolerated in the animals as seen in minor increases in inflammatory markers following injection (FIG.38). The safety data confirm none of the animals showed clinical symptoms of Cytokine release syndrome (CRS). Thus, this study supports a finding that a viral particle comprising a dual-fusion polypeptide along with a separately expressed anti-CD3 targeting protein can effectively transduce and generate functional CAR T cells in vivo via an intranodal pathway administration. Example 16 [0716] Example 16 describes in vivo T cell transduction and activation with viral particles surface engineered to display a CD58, CD80, and anti-CD3 scFv tri-fusion polypeptide. The methods depict successful in vivo CAR T cell generation. This non-human primate (NHP) study was conducted in M. nemestrina and showed generation of anti-CD20 CAR T cells and was well tolerated in all animals. The viral particles were engineered to display an anti-CD3 scFv that binds NHP CD3 and a payload comprising a human-specific anti-CD20 CAR which cross-reacts with NHP CD20 (FIG. 39A). Study Design and study methods are in large part the same as those described in Example 15. [0717] The objectives of the study included analyzing the ability of engineered viral particles to transduce T cells and generate functional CAR T cells in a large animal model. Another objective of the study was to assess lymph node injection as a viable route of administration for engineered viral particles. [0718] In the preliminary experiment described in Example 14 above, healthy donor PBMCs were contacted for one hour with engineered lentiviruses displaying CD58-anti-CD3 scFv-CD80 multi- domain fusion (MDF) surface proteins (FIG. 34A). Lentiviral particles displaying MDF Attorney Ref. No.061479-506001WO (multidomain fusion) proteins showed a significant increase in particle-bound cells after just one hour of incubation (FIG.34B). Selective T cell binding was observed in increased % Cocal (FIG. 34C) and geometric mean fluorescent intensity (gMFI) Cocal (FIG. 34D) for CD4+ and CD8+ T cells relative to B cells, monocytes, and NK clls. Engineered lentiviruses displaying MDF surface proteins 7 days after the PBMCs were contacted with the engineered particles displaying CD58, CD80, and anti-CD3 scFv MDF polypeptide showed an increased yield of CAR T cells (FIG.34H and FIG.34I). [0719] In the present NHP study, the subjects of the study were Pig-tailed macaques (Macaca nemestrina) that were either male of female, a minimum size of 3.5-5 kg, no age restriction (other than to ensure sufficient size). Six animals were studied: 4 test article treated + 2 control (PBS or OVY]b YK[]SMUO% ][OK]ON) ESQ]KSUON VKMKZ^O\ K[O `OUU \^S]ON PX[ UOW]S_S[KU \]^NSO\ N^O ]X IG?B0h KUUOUO ]RK] YO[VS]\ OPPSMSOW] ][KW\N^M]SXW) IG?B0h SW [RO\^\ KWN MbWXVXUQ^\ VKMKZ^O\ [O\][SM]\ retroviral transduction. A summary of the aminals treated is shown in FIG.41B. [0720] Engineered viral particles were generated with surface molecules that are cross-reactive with pig-tailed macaque (Human CD58-NHP-specific anti-CD3 scFv-Human CD80 multi-domain fusion (MDF) polypeptide) (FIG.39A) KWN h9:-+(97G(<UKQ YKbUXKN $FIG.39B). The particles showed efficient transduction of pig-tailed macaque PBMCs in vitro at MOI=0.2 (FIG.40). [0721] The particles were dosed in transducing units per kg (TU/kg) as determined by ddPCR ]S][K]SXW XW H^YI, MOUU\) IRO YKbUXKN `K\ NO\SQWON ]X OaY[O\\ K <UKQ(]KQQON h9:-+(97G \SWMO ]RO[O S\ WX K_KSUKLUO KW]SLXNb ]RK] [OMXQWScO\ ]RO h9:-+(97G) [0722] FIG.41A shows the study design and timeline for the study. [0723] Vector formulations were maintained at -80°C until the day of animal treatment. Vector was kept on ice for transport to the animal facility and equilibrated at room temperature for approximately 5-15 minutes prior to lymph node injection in macaques using a single-use sterile needle and 1 ml syringe. Vector was injected within 2 hours of preparation. Control animals had an equivalent volume of vehicle (PBS) or equivalent number of surface engineered particles that did not contain a viral payload (empty particle) injected into the lymph node. [0724] Animals were individually housed (i) during the initial pre-study period in which the animals were acclimated to the jacket and tether and (ii) for at least 4 weeks following engineered viral particle injection. Animals had ad libitum access to water and food was only restricted prior to sedation or anesthesia. Study Procedure [0725] Blood was drawn pre-study for isolation of (i) PBMCs, (ii) serum, and (iii) gDNA. Serum and gDNA were isolated and stored at -80 ºC for later analyses. PBMCs were cryopreserved, analyzed by a pre-injection flow cytometry panel and transduced with the same engineered viral particles that were injected in vivo. Attorney Ref. No.061479-506001WO [0726] Criteria for exclusion from study include: (i) inability of engineered viral particles to activate T cells, (ii) inability of engineered viral particles to transduce T cells, and (iii) inability of CAR expressing T cells to kill target cells and produce cytokines. [0727] Pre-Study Blood Draws: In the two-week period prior to lymph node injection, up to two blood draws were taken to determine baseline values for the assays below. [0728] Day 0: - Prophylactic diphenhydramine (IV injection, one dose) - Surgical placement of central catheter (if on tether) - Surgical placement of telemetry device to monitor body temperature - Optional: blood draw - Ultrasound-guided LN injection of engineered viral particles [0729] Animals with a central intravenous catheter implanted received a continuous IV antibiotic infusion via the catheter to prevent septicemia. [0730] Animal Monitoring and Blood Collection: Blood was drawn throughout the study with biweekly or triweekly blood draws planned for the first 3-4 weeks of the experiment and weekly thereafter. The number of draws will depend on assay results and the clinical condition of each animal. Blood draw schedule may be adjusted dependent on maximum blood draw allowance of 10ml/kg body weight every two weeks. [0731] Animals were observed daily for general health, appetite, activity level, responsiveness, and fecal production. Body temperature was measured continually using a telemetry device. Animals were monitored for clinical signs of cytokine release syndrome (CRS) and immune effector cell- associated neurotoxicity syndrome (ICANS) based on an established set of criteria. Clinical signs of CRS or ICANS will be confirmed by CBCs, serum chemistries, serum IL-6, serum ferritin and serum CRP levels. Animals treated with engineered viral particle product in the present study did not display any clinical signs of CRS. [0732] Study Endpoint: The animals were monitored and have blood drawn periodically following injection of engineered viral particles. Analytical Assays: [0733] A. Cell Surface Flow Cytometry a. NHP PBMC Screening Panel Table 10 Attorney Ref. No.061479-506001WO B. qPCR Assay on gDNA extracted from peripheral blood to detect transduced cells C. MSD Assay to measure cytokines using serum isolated from peripheral blood D. CBC E. Serum chemistries F. Serum IL-6 G. Serum ferritin H. Serum CRP Efficacy Criteria: [0734] Efficacy was assessed in each animal using a combination of analytics comparing results pre- and post-injection. a. Depletion of B Cells (Flow Cytometry). The number of B cells per ql present were plotted longitudinally starting with pre-injection samples and extending throughout the study. b. Cytokine Release (MSD Assay). The serum concentration of each cytokine were plotted longitudinally starting with pre-injection samples and extending throughout the study. c. Detection of CAR T Cells in Peripheral Blood (Flow Cytometry, qPCR). The number of Flag-expressing T cells (Flow Cytometry) present were plotted longitudinally starting with pre-injection samples and extending throughout the study. [0735] With a total of 4 animals treated significant trends in regards to CAR T cell generation and B cell depletion in vivo were detected. Safety Criteria: [0736] Safety will be assessed using a combination of analytics. a. CRS Monitoring i. Body temperature ii. CBCs iii. Serum IL-6 Attorney Ref. No.061479-506001WO iv. Serum ferritin v. Serum CRP vi. Serum chemistries (kidney (creatinine, BUN, total protein, electrolytes) and liver (AST, ALT, ALP, total bilirubin, albumin) function or damage) [0737] Clinical and Physical Assessments: including general health, appetite, activity level, responsiveness, hydration, and fecal production [0738] Necropsy and Histopathology on a subset of animals: a. Gross findings b. Histopathology of tissue sections c. qPCR of tissue sections d. RNA-ISH of tissue positive by qPCR. Results [0739] Intranodal injection of engineered viral particles generated functional CAR T cells in Animal #1 (FIG.42). Flow cytometry analysis showed a recurring expansion of the CAR T cells as well as persistence and the emergence of a population of memory CAR T cells at Day 35 (FIG.42). CAR T cell kinetics were analyzed through flow cytometry analysis of cell staining with the activation marker CD25 (FIG. 43). CAR T cell activation peaked on Day 7 and Day 49 and such kinetics are consistent with antigen-engagement and B cell eradication. The presence of CD25_CAR T cells at Day 49 indicates antigen-driven expansion from a persistent memory T cell population. The generation of functional CAR T cells was further evidenced by sustained B cell aplasia for approximately 70 days (FIG.44). The depletion of B cells coincides with the increase in CAR+ T cells (FIG.45). In vivo transduction with the engineered viral particles was well-tolerated in Animal #1. FIG. 46A shows a timeline of observed clinical symptoms. Increase in inflammatory markers and the onset of clinical symptoms of mild CRS (FIG.46B peaks) and neurotoxicity (FIG.46B first peak) coincided with CAR T cell expansion (FIG.46B). Animal #1 responded rapidly to invention with a single dose of each medication. In summary, the data show that intranodal administration of engineered viral particles was well-tolerated in Animal #1. The engineered viral particles show potent in vivo CAR T cell generating activity as demonstrated by flow cytometric detection of CAR T cells and sustained B cell depletion. [0740] A dose de-escalation study was performed with Animal #2 wherein the animal was dosed with a half-log lower dose (FIG.47). Animal #2 did not show symptoms of CRS: no fever, decreased appetite, or decreased activity. [0741] Animal #3 showed symptoms of mild CRS: fever, decreased appetite, and decreased activity on Day 3. Animal #3 responded rapidly to invention with Tocilizumab and Anakinra. Expansion of CAR+ T cells on Day 7 appeared concomitantly with B cell aplasia (FIG. 48). B cell aplasia was complete and persistent rhgouh at least day 50 of the study. Attorney Ref. No.061479-506001WO [0742] Animal #4 did not show symptoms of CRS or ICAN. Animal #4 was prophylactically treated with Anakinra between Day 7 and Day 10 (FIG. 49). Similar to Animals #1 and #3, Animal #4 exhibited peak expansion of CAR+ T cells around Day 10 with corresponding complete and persistent B cell aplasia through at least Day 50 of the study. [0743] In summary, viral particles displaying multi-domain fusion (MDF) protein surface engineering showed potent in vivo anti-CD20 CAR T cell generation. Consistent generation of anti- CD20 CAR T cells that drive persistent B cell aplasia was observed in three out of three animals at the full dose (Animals 1, 3, and 4 of this Example). Typically, circulating CAR T cells are not detectable by industry standard flow cytometry. However, surprisingly, substantial circulating CAR- T cells generated by the viral particles described were also detectable by an industry standard flow cytometry assay. Further, none of the animals in the studies underwent lymphodepletion or supportive cytokine treatment prior to administration. The engineered viral particles were well- tolerated and toxicity associated with particle administration was either mild or not observed during peak CAR T expansion in most animals studied. [0744] In conclusion, these findings demonstrate in vivo CAR T cell generation was achieved with CAR+ T cell expansion peaking around Day 10 of the study. The generation of functional CAR T cells was further evidenced B cell aplasia which coincided with the increase in CAR+ T cells beginning at Day 7 of the study. Thus, this study supports a finding that a viral particle comprising a tri-fusion polypeptide comprising binding domains from CD58 and CD80, and an anti-CD3 binding domain, can effectively transduce and generate functional CAR T cells in vivo via an intranodal route of administration. Example 17. In Vivo Engineered CAR-T Cells for Oncology: Nonclinical Safety and Biodistribution [0745] This Example demonstrates the biodistribution and preliminary safety of the engineered particles of the present disclosure when delivered via either intranodal (IN) or intravenous (IV) routes of administration (ROAs). [0746] In vivo biodistribution was assessed in canines and humanized mice (FIG. 52A- 52B). FIG. 52A: CD34-NCG mice were treated IV or IP (a surrogate ROA as IN is not feasible in this model) with viral particles encoding a CAR and RACR payload with CD58+CD3 scFv+CD80 trifusion polypeptide and Cocal glycoprotein surface engineering. There were no adverse safety events during the in-life portion and mice were sacrificed at 1 week to assess biodistribution. FIG.52B: Canines were treated with a maximal IN dose of viral particles encoding an eGFP payload, or a 10-fold higher dose IV. Animals were sacrificed at 1 week or 8 weeks to assess biodistribution. There were no test article related Attorney Ref. No.061479-506001WO adverse findings after a full non-GLP tox assessment (in life, histopathology, clin chem & hematology). [0747] Viral particle transduction was dose-dependent and highest in spleen and liver following IP or IV dosing of CD34-NCG mice (FIG. 53A-53B). Viral particle biodistribution was measured using a qPCR assay performed on genomic DNA extracted from tissues 1 week post IP or IV administration of engineered viral particle or vehicle control. N=6-7; bars are group median and dots individual animals. In FIG. 53A, for each tissue type, the left bar represents vehicle data, the middle bar represents low dose intraperitoneal data, and the right bar represents high dose intraperitoneal data. In FIG.53B, for each tissue type, the left bar represents vehicle data, and the right bar represents high dose intraperitoneal intravenous data. [0748] Viral particle transduction events in CD34-NCG mice were predominantly detected in immune cells (hCD3+ or mCD68+). A multiplex RNA ISH (RNAscope) assay was performed to determine transduced cell type in qPCR positive tissues. Across all tissues, transduction was predominantly detected in mouse CD68+ phagocytic cells. In the spleen, transduction of human T cells was also detected but dependent on surface engineering with the trifusion protein. [0749] Quantifiable viral particle transduction was observed in lymphoid tissues following IN or IV administration to canines (FIG. 54). The figure depicts transduction 1 week after viral particle IN or IV administration as measured using a qPCR assay on genomic DNA extracted from tissues. Samples below LOD are depicted as “1” for graphing; samples below LLOQ depicted at LOD. N=2-3; bars are group median and dots individual animals. Following IN or IV administration of viral particles to canines, transduction of immune cells was obversed. [0750] Nonclinical studies in two species demonstrated a favorable viral particle safety and biodistribution profiles following multiple ROAs. Administration of engineered viral particles to CD34-NCG mice by IP (surrogate IN) or IV injection showed transduction of immune cells. Human T cell transduction improved with trifusion surface engineering. Following IN or IV administration to canines at high doses, quantifiable transduction was observed in lymphoid tissues. No observable in-life toxicity was observed following high viral particle doses in humanized mice or beagles. These findings support the usefulness of the engineered viral particles disclosed herein and related embodiments herein to generate Attorney Ref. No.061479-506001WO CAR T cells in vivo, which may be useful for generation of CAR T cells without the need for ex vivo cell therapy manufacturing or lymphodepletion. Example 18. Functional Study of Engineered Particles in Lentiviral Vector Transduction [0751] Engineered lentiviral particles comprising two different fusion proteins were evaluated. [0752] The lentiviral particles comprise two variations of a CD58-CD3 scFv-CD80 trifusion polypeptide and Cocal glycoprotein surface engineering. Version 1 of the fusion polypeptide comprised, in order, a CD58 binding domain, an anti-CD3 scFv, and a CD80 full length polypeptide. Version 2 comprised, in order, an anti-CD3 scFv, a CD58 binding domain, and the full length CD80 polypeptide.

Attorney Ref. No.061479-506001WO Table 11 [0753] As shown in Figs. 55A-55B, both versions of the fusion polypeptide effectively transduced target cells and resulted in CAR-T positive cells (FIG.55A-55B). Example 19 [0754] This Example shows the incorporation of a costimulatory molecule on a lentiviral particle enhances transduction of PBMCs by lentiviral particles as generated in Example 1. Virus Production [0755] All solutions used were the same as those described in Example 1. 293T cells were seeded into T175 flasks with Complete DMEM media.24 hours later, cells were transfected. Virus was produced as described in Example 1. All viruses included a Cocal envelope protein. [0756] List of Virus preps made for study (viruses contained an anti-CD19 CAR payload): 1. CD3scfv only 2. CD3scfv, CD80, and CD58 proteins separately expressed (“Tri protein”) PBMC Transduction and staining for flow cytometry [0757] 50 × 10 6 PBMCs were thawed, diluted to 2 × 10 6 cells/ml in complete media (e.g. RPMI or Optimem). IL-2 was added to a final concentration of 50IU/ml. [0758] 500µl (1e6 cells) were added to the wells of a Non-TC-treated 48 well plate. Vector was added to the wells at MOI=10, 5, and 2 based on the SupT1 ddPCR titer and the plates were placed in 37° C incubator. [0759] After 3 days, vector was washed out and replaced with 500µl fresh RPMI media+IL- 2 (50 IU/ml). Cells were mixed and 100-300µl were added to wells in a 96 well V-bottom plate for activation flow cytometry analysis. Cells were then washed with 200µlFACS buffer. The cell pellets were resuspended in 50-100µl PBS containing LiveDead Stain (1:1000) and incubate at 4° C for 20 min followed by another wash in 200µlFACS buffer. Cells were resuspended in 50µlof FACS buffer + surface stain cocktail, incubated for 30 min at 4° C, washed in 200µlFACS buffer. Attorney Ref. No.061479-506001WO Results and Conclusions [0760] To assess if lentiviral particles with separately expressed costimulatory and adhesion molecules can better activate human T cells, the vector particles were added to human PBMCs at several MOI’s.3 days later, the virus was removed and the cells were given fresh media and analyzed for the activation marker CD25. Tri protein particles potently activated CD8 T cells compared to CD3scfv only (FIG. 56A). Furthermore, CD25 upregulation was dose-dependent (FIG. 56A). CD3scfv only lentiviral particles induced minimal levels of CD25 compared to the Tri protein particles (FIG. 56A). To determine whether lentiviral particles with costimulatory and/or adhesion molecules have enhanced particle binding to T cells, the particles were cultured with PBMCs for 6 hours and then were analyzed for particle-associated molecules on T cells (Cocal). The separate expression of CD58, CD80, and an anti-CD3 scFv increased Cocal staining (FIG. 56B) [0761] To examine transduction, 7 total days after transduction, samples were analyzed for anti-CD19 CAR expression. Tri protein particles were capable of transducing unstimulated PBMCs while CD3scfv only particles transduced unstimulated PBMCs to a lesser extent (FIG.56C). Furthermore, transduction occurred in a dose-dependent manner for both CD4+ and CD8+ T cells (FIG.56C). The data show that Tri protein particles efficiently activate and transduce unstimulated PBMCs in vitro compared to CD3scfv only. Importantly, the enhanced particles result in increased numbers of CAR+ T cells (FIG. 56C, right panels: Total CAR+ cells). [0762] To further characterize T cell activation, 3 total days after vector addition, samples were analyzed for cytokine expression. Tri protein particles were capable of inducing IFN- l Y[XN^M]SXW SW ^W\]SV^UK]ON E8B9\ K] UX`O[ NX\O\ `RO[OK\ 9:.\MP_ XWUb YK[]SMUO\ transduced unstimulated PBMCs to a lesser extent (FIG. 56D). Furthermore, Tri protein YK[]SMUO\ SWN^MON [XL^\] ?A(- KWN IC<(h `RO[OK\ 9:.\MP_ XWUb NSN WX] $FIG. 56D). The data show that Tri protein particles efficiently induce cytokine production in unstimulated PBMCs in vitro compared to CD3scfv only. [0763] Transduced PBMCs were then cultured with Nalm6 tumor cells. Specifically, anti- CD19 CAR+ T cells were serial-stimulated with Nalm6 tumor cells every 2-3 days. Total Nalm6 tumor cells were measured over time using an IncuCyte® providing a measurement of tumor cell killing over time (FIG. 56E). This assay measures the ability of the CAR T cells to expand and kill multiple tumor cells over time and showed that anti-CD19 CAR T Attorney Ref. No.061479-506001WO cells generated with lentiviral particles displaying “Tri protein” demonstrated serial killing in vitro as compared to particles displaying CD3scfv only. [0764] To determine whether lentiviral particles with costimulatory and/or adhesion molecules have enhanced particle binding to T cells, the particles were cultured with PBMCs for 6 hours and then were analyzed for particle-associated molecules on T cells (Cocal, anti-CD3scFv, CD80, and CD58). Both Tri protein and Fusion particles demonstrated high stating for anti-CD3scFv, CD80, and CD58, and only Fusion particles demonstrated high staining for CD3scfv+CD80+CD58 (data not shown). The data show that the fusion of CD58v+CD3 scFv+CD80 enhances particle binding to T cells. Samples were then analyzed for cytokine expression. Tri protein particles were capable of inducing IFN- l Y[XN^M]SXW SW ^W\]SV^UK]ON E8B9\ `RO[OK\ 9:.\MP_ XWUb YK[]SMUO\ ][KW\N^MON unstimulated PBMCs to a lesser extent (FIG. 56F). Furthermore, Tri protein particles SWN^MON [XL^\] ?A(- KWN IC<(h Y[XN^M]SXW `RO[OK\ 9:.\MP_ XWUb NSN WX] $FIG.56F). The data show that Tri protein surface engineered particles efficiently induce cytokine production in unstimulated PBMCs as compared to CD3scfv only. [0765] To determine whether different T cell subtypes are generated by the lentiviral particles, PBMCs cultured with the lentiviral particles were profiled and gated on viable, CD4+ and CD8+. The cells were further analyzed by flow cytometry and analysis was done based on the parameters CCR7+ and CD27+ (FIG. 56G). Tri protein particles showed an increased population of CCR7+CD27+ T cells as compared to CD3 scFv only. CCR7+CD27+CD28+ memory-like CAR+ T cells are thought to have increased longevity and proliferative capacity and correlate with better antitumor responses in vivo. Example 20 [0766] IRS\ ;aKVYUO \RX`\ I MOUU KM]S_K]SXW KWN ?<Cl Y[XN^M]SXW PXUUX`SWQ in vivo transduction of T cells by a lentiviral particle displaying CD58, CD80, and an anti-CD3 scFv separately expressed as compared to CD3scfv only. The lentiviral particles contained a polynucleotide encoding an anti-CD19 CAR. [0767] On study Day –4 NSG MHC I/II dKO mice were injected via tail vein injection with 2.5E5 Nalm6 cells expressing firefly luciferase (ffluc) (FIG.57A).3 days later (study Day –1), mice were imaged via bioluminescence imaging and randomized to study arms according to tumor burden (total flux), the same day all mice were humanized by injecting 20E6 human PBMCs intraperitoneally in 100 µl of 1X sterile PBS. The mice used in the Attorney Ref. No.061479-506001WO study were immune-compromised and contain engrafted human T cells and circulating human B cells. [0768] The next day (study Day 0), mice were treated via intraperitoneal injection with different doses of lentiviral particles displaying: ,) 9:03' 9:3+' KWN KW h(9:. \M<_ \OYK[K]OUb OaY[O\\ON $eI[S Y[X]OSWf%6 -) h(9:.\MP_ XWUb) [0769] A control study arm treated mice with 1xPBS (Neg) via intraperitoneal injection. Mice were then weighted two times a week throughout the study to monitor body weight change and imaged weekly to monitor tumor burden. Mice were bleed on study Days 4, 11, 18, 25, and 32 to perform flow cytometry analysis. Study day 4 activation markers CD25 (FIG. 57B) and CD71 on T cells were analyzed. A blood draw on Day 11 was collected to perform flow cytometry analysis and assess CAR T cell expansion and persistence (FIG. 57C, top panel) and CAR expression level was measured by staining with an anti-FMC63 antibody (FIG. 57C, bottom panel). On study Day 6 and every week through the study, mice were imaged via Bioluminescence imaging using the IVIS™ spectrum system to analyze tumor burden (total flux) (FIG.57D). Example 21 [0770] This Example shows the incorporation of a costimulatory molecule and an adhesion molecule on a lentiviral particle enhances transduction of PBMCs by lentiviral particles as generated in Example 1. Virus Production [0771] All solutions used were the same as those described in Example 1. 28 × 10 6 293T cells were seeded into 16x T175 flasks (8x per vector) with 28e6293T cells each in a total volume of 25ml Complete DMEM media. 24 hours later, cells were transfected. Virus was produced as described in Example 1. All viruses included a Cocal envelope protein and an anti-CD19 CAR payload. [0772] List of Virus preps made for study: 1. CD3scfv, CD80, and CD58 separately expressed (“Tri protein”) 2. CD58+CD3scFv+CD80 expressed as a fusion protein (“Fusion”) Results and Conclusions [0773] To assess if lentiviral particles with costimulatory molecules can better activate human T cells, the vector particles were added to human PBMCs at several MOI’s. CD58+CD3 scFv+CD80 fusion particles potently activated CD4+ and CD8+ T cells Attorney Ref. No.061479-506001WO compared to the Tri protein particles (FIG. 58B). Furthermore, CD25 upregulation was dose-dependent (FIG.58B). To determine whether lentiviral particles with a singular fusion protein comprising costimulatory and/or adhesion molecules have enhanced particle binding to T cells, the particles were cultured with PBMCs and then were analyzed for particle- associated molecules on T cells (Cocal). The fusion particles resulted in increased Cocal staining (FIG.58A). [0774] To examine transduction, 7 total days after transduction, samples were analyzed for anti-CD19 CAR expression. CD58+CD3 scFv+CD80 fusion particles were capable of transducing unstimulated PBMCs at lower doses while Tri protein particles transduced unstimulated PBMCs to a lesser extent (FIG. 58C). Furthermore, transduction occurred in a dose-dependent manner for both CD4+ and CD8+ T cells (FIG.58C). The data show that CD58+CD3 scFv+CD80 fusion particles efficiently activate and transduce unstimulated PBMCs in vitro compared to Tri protein particles. [0775] To further characterize T cell activation, 3 total days after vector addition, samples were analyzed for cytokine expression. CD58+CD3 scFv+CD80 fusion particles induced [XL^\] ?A(- KWN IC<(h Y[XN^M]SXW K\ MXVYK[ON ]X I[S Y[X]OSW YK[]SMUO\ $FIG. 58D). The data show that CD58+CD3 scFv+CD80 fusion particles efficiently induce cytokine production in unstimulated PBMCs in vitro compared to Tri protein displaying particles. [0776] Transduced PBMCs were then cultured with Nalm6 tumor cells. Specifically, anti- CD19 CAR+ T cells were serial-stimulated with Nalm6 tumor cells every 2-3 days. Total Nalm6 tumor cells were measured over time using an IncuCyte® providing a measurement of tumor cell killing over time. This showed that CAR T cells generated with lentiviral particles displaying a CD58+CD3 scFv+CD80 fusion protein demonstrated improved serial killing in vitro as compared to Tri protein displaying particles. In particular, CAR T cells generated with the fusion particles (Fusion) were able to continously control tumor cells for at least 35 days. CAR T cells generated with the separately expressed proteins (Tri protein) exhibited slow tumor outgrowth beginning around day 15. [0777] To determine whether different T cell subtypes are generated by the lentiviral particles, PBMCs cultured with the lentiviral particles were profiled and gated on viable, CD4+ and CD8+. The cells were further analyzed by flow cytometry based on parameters CCR7+ and CD27+. At most MOIs tested, both the tri protein and fusion particles were capable of generating high levels of CCR7+ and CD27+ CAR+ cells. Attorney Ref. No.061479-506001WO Example 22 [0778] IRS\ ;aKVYUO \RX`\ I MOUU KM]S_K]SXW KWN ?<Cl Y[XN^M]SXW PXUUX`SWQ in vivo transduction of T cells by a lentiviral particle displaying the CD58+CD3 scFv+CD80 fusion as compared to particles comprising CD58, CD3 scFv, and CD80 separately expressed. The lentiviral particle contains a polynucleotide encoding an anti-CD19 CAR. [0779] On study Day –4 NSG MHC I/II dKO mice were injected via tail vein injection with 2.5E5 Nalm6 cells expressing firefly luciferase (ffluc). 3 days later (study Day –1), mice were imaged via bioluminescence imaging and randomized to study arms according to tumor burden (total flux), the same day all mice were humanized by injecting 20E6 human PBMCs intraperitoneally in 100 µl of 1X sterile PBS. The mice used in the study were immune-compromised and contain engrafted human T cells and circulating human B cells. [0780] The next day (study Day 0), mice were treated via intraperitoneal injection with different doses of lentiviral particles displaying: 1. CD3scfv, CD80, and CD58 separately expressed (“Tri protein”) 2. CD58+CD3scFv+CD80 expressed as a fusion protein (“Fusion”) [0781] A control study arm treated mice with 1xPBS (Vehicle) via intraperitoneal injection. Mice were then weighted two times a week throughout the study to monitor body weight change and imaged weekly to monitor tumor burden. Mice were bleed on study Days 4, 11, 18, 25, and 32 to perform flow cytometry analysis. Study day 4 activation markers CD25 (FIG. 59A) and CD71 on T cells were analyzed. A blood draw on Day 11 was collected to perform flow cytometry analysis and assess CAR T cell expansion and persistence (FIG. 59B, top panel) and CAR expression level was measured by staining with an anti-FMC63 antibody (FIG. 59B, bottom panel). On study Day 6 and every week through the study, mice were imaged via Bioluminescence imaging using the IVIS™ spectrum system to analyze tumor burden (total flux) (FIG. 59C). As shown in FIG. 59C, tumor growth was better controlled in both of the fusion particle cohorts, with the higher dose exhibiting more robust tumr control. Overall % survival of the mice was analyzed over the course of the study. Lenitviral particles displaying CD58+CD3 scFv+CD80 fusion particles transduced at 50E6 TU showed increased overall survival in mice as compared to lentiviral Tri protein displaying particles. * * * Attorney Ref. No.061479-506001WO [0782] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. Specifically, features described in one section may be combined with features in any other section of the description. [0783] Unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. [0784] The singular forms “a”, “an”, and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. [0785] As used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (or). [0786] All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, controls. However, mention of any reference, article, publication, patent, patent publication, and patent application cited herein is not, and should not be taken as an acknowledgment, or any form of suggestion, that they constitute valid prior art or form part of the common general knowledge in any country in the world. [0787] While illustrative embodiments have been described and depicted, it will be appreciated that various changes can be made to these illustrative embodiments without departing from the spirit and scope of the invention.