Construction of Lentiviruses Pseudotyped with Sindbis E2-Single Chain Antibody (SCA) Fusions or Membrane-Anchored SCAs for Targeted Therapies in Lymphoma.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3571-3571
Author(s):  
Quang Trong Luong ◽  
Kouki Morizono ◽  
Irvin SY Chen ◽  
Sven De Vos

Abstract Abstract 3571 Poster Board III-508 Previously, we demonstrated specific targeting of CD20+ and CD30+ cells can be achieved in vitro using vectors of Morizono et al. [Cell Cycle 2005; Nature Medicine 2005]. These lentiviruses expressed modified Sindbis E2 proteins that contain the Fc receptor binding domain (ZZ) of protein A. The lentiviruses could be specifically and effectively directed to target cells by conjugation of an antibody to the modified ZZ Sindbis viral envelope (m168). CD30+ HL cell lines and CD20+ non-Hodgkin lymphoma (NHL) cell lines were used. Anti-CD30-labeled viruses (m168-αCD30) specifically transduced CD30+ HL cells while avoiding CD30− NHL cells; in addition, transduction of CD20+ NHL cells was achieved using m168-αCD20 viruses (unpublished data). However, these viruses were unsuitable for in vivo studies because of the unstable conjugation of antibody and envelope protein, and the ambiguity of the ZZ domains to bind Fc receptors. We developed two targeting approaches. For the first approach, we constructed vectors containing single chain antibodies (SCA) directly fused to the Sindbis E2 spike glycoprotein. Approximately 100 different construct-variants have been produced so far, that contain SCA-E2 fusions with a variety of flexible linkers to allow for adequate folding and expression of the fusion proteins. These constructs contain either anti-CD30-E2 (T405 or T215) or anti-CD20-E2 sequences. The second approach was to construct membrane-anchored SCA vectors that can be expressed on the envelope of the lentiviruses. We produced in excess of 30 variants containing anti-CD30 or anti-CD20 sequences with transmembrane domains of CD4 or VSV-G, or a Glycosylphosphoinositol (GPI)-anchor. Potentially, these constructs are more stable and might allow an in vivo targeting approach. We have tested these constructs for expression of SCA-E2 or membrane-anchored SCA by Western analyses. The expression of SCA-E2 fusion proteins was dependent on the length of the linker used with greater expression observed with longer linkers. The linker length should contain a minimum of 20 amino acids either side of the cloning/fusion site to permit adequate expression of SCA-E2 fusion proteins. SCA-GPI-anchor proteins, on the other hand, are not fused to the Sindbis E2 protein but contain similar length flexible linkers to permit proper folding of the SCA and GPI proteins. We tested whether the viruses produced with these modified envelope proteins or membrane-anchored proteins were able to infect CD30 or CD20 expressing target cells. We achieved up to 40% infection of 293-T CD20+ cells with anti-CD20-E2 constructs but only about 10% infection of 293-T CD30+ cells using T405-E2 (anti-CD30) constructs. In contrast, we found that T405-GPI (anti-CD30) constructs were able to infect 60% of target cells while anti-CD20-GPI infected less than 10% of target cells. Additionally, we found that T215-E2 (anti-CD30) pseudotyped viruses were not able to infect CD30+ target cells. We conclude from our data (1) that while single chain antibody sequences were the functional components that determine specificity of pseudotyped lentiviruses, not all single chain antibodies will be functional in these systems; and (2) the success of the different anchoring techniques seems to depend on the single chain antibody and the target antigen. These vectors will now be subjected to in vivo targeting. Disclosures: No relevant conflicts of interest to declare.

2003 ◽  
Vol 77 (24) ◽  
pp. 13396-13398 ◽  
Author(s):  
Esteban Veiga ◽  
Víctor de Lorenzo ◽  
Luis Angel Fernández

ABSTRACT We report here that fusions of single-chain antibodies (scFvs) to the autotransporter β domain of the IgA protease of Neisseria gonorrhoeae are instrumental in locating virus-neutralizing activity on the cell surface of Escherichia coli. E. coli cells displaying scFvs against the transmissible gastroenteritis coronavirus on their surface blocked in vivo the access of the infectious agent to cultured epithelial cells. This result raises prospects for antiviral strategies aimed at hindering the entry into target cells by bacteria that naturally colonize the same intestinal niches.


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
Toshiki Ochi ◽  
Masaki Maruta ◽  
Kazushi Tanimoto ◽  
Fumitake Kondo ◽  
Toshihiro Yamamoto ◽  
...  

AbstractCancer immunotherapy using T cells redirected with chimeric antigen receptor (CAR) has shown a lot of promise. We have established a single-chain antibody (scFv) generation system in which scFv library-expressing CAR-T cells can be screened appropriately based on their antitumor functions. A variable region library containing the variable and J regions of the human immunoglobulin light or heavy chain was fused with the variable region of a heavy or light chain encoded by an existing tumor-specific antibody to generate a new scFv library. Then, scFv library-expressing CAR-T cells were generated and stimulated with target cells to concentrate the antigen-specific population. Using this system, target-specific recognition of CAR-T cells appeared to be finely tuned by selecting a new variable region. Importantly, we have demonstrated that the newly optimized scFv-expressing CAR-T cells had better proliferation capacity and durable phenotypes, enabling superior reactivity against advanced tumors in vivo in comparison with the original CAR-T cells. Therefore, the optimization of an scFv is needed to maximize the in vivo antitumor functions of CAR-T cells. This system may allow us to adjust an immunological synapse formed by an scFv expressed by CAR-T cells and a target antigen, representing an ideal form of CAR-T-cell immunotherapy.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A665-A665
Author(s):  
Emily Hsiue ◽  
Katharine Wright ◽  
Jacqueline Douglass ◽  
Michael Hwang ◽  
Brian Mog ◽  
...  

BackgroundTP53 is the most commonly mutated cancer driver gene but drugs that target TP53 are not yet available. A peptide derived from the most common p53 mutation R175H (HMTEVVRHC) can be presented by a common human leukocyte antigen (HLA-A*02:01) after proteasomal degradation.1 We aimed to develop T cell receptor (TCR)-mimic antibody targeting this shared neoantigen.MethodsWe constructed a single-chain variable fragment (scFv) phage display library that presents scFvs at an estimated diversity of 3.6e10. Mutant peptide-HLA (pHLA)-specific scFvs were enriched through five rounds of positive and negative selections. Mutant pHLA-specific scFv clones were converted into bispecific T cell retargeting antibodies in the single-chain diabody (scDb) format by linking with the anti-CD3 scFv UCHT1.2 These scDbs were tested in T cell co-culturs in the presence of target pulsed cells or target cells that either overexpress the p53 neoantigen or present the p53 neoantigen at endogenous levels. In vivo efficacy was assessed by administering scDb in NSG xenograft mouse model. The structural basis of the binding specificity was evaluated by X-ray crystallography.ResultsWe identified an scFv, termed clone H2, that specifically binds p53 R175H/HLA-A*02:01 pHLA and not its wild-type counterpart at a Kd of 86 nM (figure 1A). H2-scDb induced T cell cytokine release and cytotoxicity in the presence of 1) HLA-A*02:01-expressing cells pulsed with the p53R175H peptide, 2) cells transfected with HLA-A*02:01 and p53 R175H, and 3) cancer cell lines KMS26, KLE, and TYK-nu that express endogenous HLA-A*02:01/p53 R175H (figure 1B-E). T cell activation was diminished when TP53 was knocked out from these cancer cell lines using CRISPR (figure 1E). When administered to NOD scid gamma (NSG) mice systemically engrafted with the KMS-26 cell line, H2-scDb significantly suppressed tumor growth (figure 1 F, G). The structure of p53 R175H/HLA-A2 bound to the H2-Fab fragment shows that four complementarity-determining region loops of the H2 antibody formed a cage-like configuration around the C-terminus of the p53 R175H peptide, trapping the mutant histidine (His175) and the adjacent arginine (Arg174) residues in a stable interaction, which provides the structural basis for the specificity (figure 2).ConclusionsWe have developed a TCR-mimic bispecific T cell engager H2-scDb that recognized the shared neoantigen HLA-A*02:01/p53 R175H pHLA complex with exquisite specificity. It effectively activated T cells and lysed tumor cells both in vitro and in vivo. This approach could in theory be used to target cancers containing mutations that are difficult to target in conventional ways.AcknowledgementsWe thank Surojit Sur, Nicolas Wyhs, Ashley Cook, Marco Dal Molin, Richard L. Blosser, Lilian Dasko-Vincent, Christopher Thoburn, Jianhong Cao, and José Rodríguez Molina for assistance with this study.Trial RegistrationNAEthics ApprovalNAConsentNAReferencesLo W, Parkhurst M, Robbins PF, Tran E, Lu YC, Jia L, Gartner JJ, Pasetto A, Deniger D, Malekzadeh P, Shelton TE, Prickett T, Ray S, Kivitz S, Paria BC, Kriley I, Schrump DS, Rosenberg SA. Immunologic recognition of a shared p53 mutated neoantigen in a patient with metastatic colorectal cancer. Cancer Immunol. Res 2019;7:534–543.Stork R, Campigna E, Robert B, Muller D, Kontermann RE. Biodistribution of a bispecific single-chain diabody and its half-life extended derivatives. J Biol Chem 2009;284:25612–25619


2015 ◽  
Vol 21 ◽  
pp. 2110-2115 ◽  
Author(s):  
Fei Chen ◽  
Chuming Fan ◽  
Xuezhong Gu ◽  
Haixi Zhang ◽  
Qian Liu ◽  
...  

1997 ◽  
Vol 328 (2) ◽  
pp. 669-675 ◽  
Author(s):  
L. Tamara DOERING ◽  
Randy SCHEKMAN

The yeast mating pheromone precursor prepro-alpha factor was fused to C-terminal signals for glycosyl-phosphatidylinositol (GPI) anchor attachment, based on the sequence of the Saccharomyces cerevisiae protein Gas1p. Maturation of fusion proteins expressed in vivo required the presence of both a functional GPI attachment site and the synthesis of GPI precursors. Constructs were translated in vitro for use in cell-free studies of glycolipid attachment. The radiolabelled polypeptides were post-translationally translocated into yeast microsomes, where at least one third of the molecules received a GPI anchor. This approach offers distinct advantages over anchor attachment reactions that require co-translational translocation of secretory peptide substrates.


2001 ◽  
Vol 75 (24) ◽  
pp. 12028-12038 ◽  
Author(s):  
Benhur Lee ◽  
George Leslie ◽  
Elizabeth Soilleux ◽  
Una O'Doherty ◽  
Sarah Baik ◽  
...  

ABSTRACT DC-SIGN is a C-type lectin expressed on dendritic cells and restricted macrophage populations in vivo that binds gp120 and acts intrans to enable efficient infection of T cells by human immunodeficiency virus type 1 (HIV-1). We report here that DC-SIGN, when expressed in cis with CD4 and coreceptors, allowed more efficient infection by both HIV and simian immunodeficiency virus (SIV) strains, although the extent varied from 2- to 40-fold, depending on the virus strain. Expression of DC-SIGN on target cells did not alleviate the requirement for CD4 or coreceptor for viral entry. Stable expression of DC-SIGN on multiple lymphoid lines enabled more efficient entry and replication of R5X4 and X4 viruses. Thus, 10- and 100-fold less 89.6 (R5/X4) and NL4–3 (X4), respectively, were required to achieve productive replication in DC-SIGN-transduced Jurkat cells when compared to the parental cell line. In addition, DC-SIGN expression on T-cell lines that express very low levels of CCR5 enabled entry and replication of R5 viruses in a CCR5-dependent manner, a property not exhibited by the parental cell lines. Therefore, DC-SIGN expression can boost virus infection in cis and can expand viral tropism without affecting coreceptor preference. In addition, coexpression of DC-SIGN enabled some viruses to use alternate coreceptors like STRL33 to infect cells, whereas in its absence, infection was not observed. Immunohistochemical and confocal microscopy data indicated that DC-SIGN was coexpressed and colocalized with CD4 and CCR5 on alveolar macrophages, underscoring the physiological significance of these cis enhancement effects.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 12-12 ◽  
Author(s):  
Nikhil Hebbar ◽  
Rebecca Epperly ◽  
Abishek Vaidya ◽  
Sujuan Huang ◽  
Cheng Cheng ◽  
...  

Finding the ideal immunotherapy target for AML has proven challenging and is limited by overlapping expression of antigens on hematopoietic progenitor cells (HPCs) and AML blasts. Intracellular Glucose-regulated-protein 78 (GRP78) is a key UPR regulator, which normally resides in the endoplasmic reticulum (ER). GRP78 is overexpressed and translocated to the cell surface in a broad range of solid tumors and hematological malignancies in response to elevated ER stress, making it an attractive target for immune-based therapies with T cells expressing chimeric antigen receptors (CARs). The goal of this project was to determine the expression of GRP78 on pediatric AML samples, generate GRP78-CAR T cells, and evaluate their effector function against AML blasts in vitro and in vivo. To demonstrate overexpression of GRP78 in AML, we performed gene expression analysis by RNAseq on a cohort of cord blood CD34+ cell samples (N=5) and 74 primary AML samples. Primary AML samples included RUNX1-RUNX1T1 (N=7), CBFB-MYH11(N=17), KMT2A rearrangement (N=28) and NUP98 (N=22). Analysis showed increased GRP78 expression in AML samples, especially in KMT2A- and NUP98-rearranged AML. To demonstrate surface expression of GRP78, we performed flow cytometry of AML (Kg1a, MOLLM13, THP-1, MV4-11) cell lines as well as 11 primary AML samples and 5 PDX samples; non transduced (NT) T cells served as control. All AML samples, including cell lines, primary AML blasts, and PDX samples, showed increased expression of GRP78 on their cell surface in comparison to NT T cells We then designed a retroviral vector encoding a GRP78-CAR using a GRP78-specific peptide as an antigen recognition domain, and generated GRP78-CAR T cells by retroviral transduction of primary human T cells. Median transduction efficiency was 82% (± 5-8%, N=6), and immunophenotypic analysis showed a predominance of naïve and terminal effector memory subsets on day 7 after transduction (N=5). To determine the antigen specificity of GRP78-CAR T cells, we performed coculture assays in vitro with cell surface GRP78+ (AML cell lines: MOLM13, MV-4-11, and THP-1 and 3 AML PDX samples) or cell surface GRP78- (NT T cells) targets. T cells expressing CARs specific for HER2-, CD19-, or a non-functional GRP78 (DGRP78)-CAR served as negative controls. GRP78-CAR T cells secreted significant amounts of IFNg and IL-2 only in the presence of GRP78+ target cells (N=3, p<0.005); while control CAR T cells did not. GRP78-CAR T cells only killed GRP78+ target cells in standard cytotoxicity assays confirming specificity. To test the effects of GRP78-CAR T cells on normal bone marrow derived HPCs, we performed standard colony forming unit (CFU) assays post exposure to GRP78-CAR or NT T cells (effector to target (E:T) ratio 1:1 and 5:1) and determined the number of BFU-E, CFU-E, CFU-GM, and CFU-GEMM. No significant differences between GRP78-CAR and NT T cells were observed except for CFU-Es at an E:T ratio of 5:1 that was not confirmed for BFU-Es. Finally, we evaluated the antitumor activity of GRP78-CAR T cells in an in vivo xenograft AML model (MOLM13). Tumor growth was monitored by serial bioluminescence imaging. A single intravenous dose of GRP78-CAR T cells induced tumor regression, which resulted in a significant (p<0.001) survival advantage in comparison to mice that had received control CAR T cells. In conclusion, GRP78 is expressed on the cell surface of AML. GRP78-CAR T cells have potent anti-AML activity in vitro and in vivo and do not target normal HPCs. Thus, our cell therapy approach warrants further active exploration and has the potential to improve outcomes for patients with AML. Disclosures Hebbar: St. Jude: Patents & Royalties. Epperly:St. Jude: Patents & Royalties. Vaidya:St. Jude: Patents & Royalties. Gottschalk:TESSA Therapeutics: Other: research collaboration; Inmatics and Tidal: Membership on an entity's Board of Directors or advisory committees; Merck and ViraCyte: Consultancy; Patents and patent applications in the fields of T-cell & Gene therapy for cancer: Patents & Royalties. Velasquez:St. Jude: Patents & Royalties; Rally! Foundation: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 477-477
Author(s):  
Erica Dander ◽  
Giuseppina Li Pira ◽  
Ettore Biagi ◽  
Fabrizio Manca ◽  
Andrea Biondi ◽  
...  

Abstract BACKGROUND: Reactivation of latent CMV in immunocompromised recipients of allogeneic stem cell transplantation remains a major cause of morbidity and mortality. Reconstitution of immunity by CMV specific immunotherapy is an attractive alternative to drugs currently used, which show high toxicity and are sometimes ineffective. It has been demonstrated that CD4 helper T-cell function is crucial for the persistence of in vivo transferred CD8 CMV-specific CTL. Based on this finding, we have explored the feasibility of generating both anti-CMV CD4 and anti-CMV CD8 T-cell lines. METHODS: Dendritic Cells (DC) were generated from donor peripheral blood (PB) monocytes after a 7-day culture in the presence of GM-CSF plus IL-4 and matured with TNF-α, IFN-α, IFN-γ, IL1-β, POLI I:C. Matured-DC were then pulsed with a pool of 50 peptides spanning pp65 and IE1 proteins which are recognised by both CD4 and CD8 T lymphocytes. Donor T cells were stimulated three times at a T cell/DC ratio of 1:6 on day 0, +7 and +14 with mature peptide pulsed-DC. At the end of the culture the specificity of generated T cells was determined as percentage of pentamer-positive cells and intracellular IFN-γ production after incubation with peptide pulsed-DC. Cultured T cells were also analysed for their ability to proliferate in response to peptide pulsed-target cells, to kill them in a standard citotoxicity assay and to migrate in response to inflammatory (CXCL9, CCL3 and CCL5) and constitutive (CXCL12) chemokines. RESULTS: CMV-specific T cell lines were generated from five CMV seropositive donors. In four cases CD4 and CD8 CMV-specific T cell lines were expanded successfully. Cultured T cells expressed CD8 (mean= 70%, range 60–81%) and CD4 (mean= 20%, range 15–28%) and showed a CD45RA- CCR7- Effector Memory phenothype (mean=26%, range 19–30%) or a CD45RA+ CCR7- T Effector Memory RA-Positive phenothype (mean=67%, range 59–77%). An enriched CMV-specific T cell population was observed after staining with pentamers (7–45% pentamer-positive T cells). Furthermore, 90% of CD8+ and 40% of CD4+ T cells expressed high levels of intracytoplasmatic perforin and granzyme. In 4/5 cases tested, cutured T cells showed a cytolitic activity against CD8-peptide pulsed target cells (average lysis=50%, range 40–55%) and to a lesser extent against CD4-peptide pulsed target cells (average lysis=35%, range 30–40%). In addition, cultured T lymphocytes were able to proliferate and to produce intracytoplasmic IFN-γ (average production=50%, range 35–60%) after exposure to peptide-pulsed DC. Finally, Cultured T cells strongly migrated in response to chemokines (CXCL9, CCL3 and CCL5) involved in the recruitment of effector cells during viral infection. DISCUSSION: In conclusion, a great advantage of this method is represented by the possibility to generate anti-CMV CD4+ T cells, which could support in vivo the persistence of re-infused CMV-specific CTL. Moreover, the possibility of generating peptides under GMP conditions would facilitate the translation of this approach into clinical intervention.


Sign in / Sign up

Export Citation Format

Share Document