Use of Engineered Exosomes Expressing HLA and Costimulatory Molecules to Generate Antigen-specific CD8+ T Cells for Adoptive Cell Therapy

2017 ◽  
Vol 40 (3) ◽  
pp. 83-93 ◽  
Author(s):  
Sueon Kim ◽  
Hyun-Jung Sohn ◽  
Hyun-Joo Lee ◽  
Dae-Hee Sohn ◽  
Seung-Joo Hyun ◽  
...  
2013 ◽  
Vol 62 (10) ◽  
pp. 1563-1573 ◽  
Author(s):  
Anna Casati ◽  
Azam Varghaei-Nahvi ◽  
Steven Alexander Feldman ◽  
Mario Assenmacher ◽  
Steven Aaron Rosenberg ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 354-354
Author(s):  
Marcus O. Butler ◽  
Sascha Ansén ◽  
Makito Tanaka ◽  
Osamu Imataki ◽  
Alla Berezovskaya ◽  
...  

Abstract Abstract 354 Adoptive cell therapy utilizes unique mechanisms of action to prevent the development of infections in immunocompromised patients and treat chemotherapy resistant malignancies. In adoptive cell therapy, the major effector cells appear to be CD8+ T cells, since they are armed with antigen-specific effector functions, i.e. cytotoxicity and cytokine secretion. However, the roles of antigen-specific CD4+ T cells in T cell immunity are also critical. In immunocompromised patients adoptively transferred with CMV-specific CD8+ T cells, long-term in vivo persistence was achieved only when CMV-specific CD4+ T cells were also present in vivo. Recently, adoptive transfer of a NY-ESO-1 specific CD4+ T cell clone was reported to induce a complete response in a patient with metastatic melanoma. These results suggest that adoptive cell therapy for infectious diseases and cancer can be improved by infusing both antigen-specific CD4+ helper T cells as well as CD8+ CTL. Unfortunately, however, few versatile systems are available for producing large numbers of antigen-specific human CD4+ T cells for the purpose of adoptive therapy. K562 is a human erythroleukemic cell line, which lacks the expression of HLA class I and II, invariant chain (Ii), and HLA-DM, but does express adhesion molecules such as ICAM-1 and LFA-3. Given this unique immunologic phenotype, K562 has served as a useful tool in clinical cancer immunotherapy trials. Previously, we reported the generation of a K562-based artificial APC (aAPC), which expresses HLA-A2, CD80, and CD83. aAPC/A2 can uniquely support the priming and prolonged expansion of large numbers of antigen-specific CD8+ CTL which display a central/effector memory phenotype, possess potent effector function, and can be maintained in vitro without any feeder cells or cloning. aAPC/A2 is equipped with constitutive proteasome and inducible immunoproteasome machinery and can naturally process and present CD8+ T cell peptides via transduced A2 molecules. We have successfully generated clinical grade aAPC/A2 under cGMP conditions and conducted a clinical trial where patient with advanced melanoma are infused with large numbers of MART1-specific CTL generated ex vivo using aAPC/A2, IL-2 and IL-15. Based on our experience with aAPC/A2 and CD8+ T cells, we have generated a series of novel aAPC (aAPC/DR1, DR3, DR4, DR7, DR10, DR11, DR13, and DR15) to stimulate HLA-DR-restricted antigen-specific CD4+ T cells. K562 has been engineered to express HLA-DRα and β chains as a single HLA allele in conjunction with Ii, HLA-DMα and β chains, CD80 and CD83. CD83 delivers a CD80-dependent T cell stimulatory signal that allows T cells to be long-lived. Following the transduction of Ii, CLIP (class II invariant chain-associated peptide) appeared on the cell surface of aAPC. Furthermore, CLIP expression on aAPC was almost completely abrogated by the introduction of HLA-DM. This result is in accordance with previous studies showing that HLA-DM catalyzes the removal of CLIP from DR thus enabling exogenous peptides to bind to empty DR molecules in late endosomes. In addition to its endogenous pinocytic activity, aAPC was made capable of Fcγ receptor-mediated endocytosis by transduction of CD64. Comparison of naïve aAPC and CD64-transduced aAPC confirmed that Fcγ receptor-mediated endocytosis is more efficient than pinocytosis to take up soluble protein and process and present DR-restricted peptides to CD4+ T cells. Using these standardized and renewable aAPC, we determined novel viral protein-derived DR-restricted CD4+ T cell epitopes and expanded large numbers of viral antigen-specific CD4+ T cells without growing bystander Foxp3+ regulatory T cells. Without any feeder cells or cloning, expansion of CD4+ T cells using aAPC and low dose IL-2 and IL-15 was sustainable up to 150 days. Immunophenotypic analysis using HLA-DR tetramers and specific mAbs revealed that expanded CD4+ T cells were CD45RA−, CD45RO+, CD62L+-, demonstrating a central/effector memory phenotype. Furthermore, intracellular cytokine analysis showed that expanded DR-restricted viral-specific CD4+ T cells secreted IL-2 and IFN-γ but much less IL-4, displaying a Th1-biased phenotype. Taken all together, these results suggest that K562-based aAPC may serve as a translatable platform to generate both antigen-specific CD4+ helper T cells and CD8+ CTL. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A106-A106
Author(s):  
David Langan ◽  
Jourdain Lemaster ◽  
Lauren Suarez ◽  
Pratima Kunwar ◽  
Sojung Kim ◽  
...  

BackgroundNexImmune is developing highly differentiated immunotherapies to target, activate and expand tumor antigen-specific T cells using the proprietary Artificial Immune Modulation (AIM™) nanotechnology platform. The AIM nanoparticle (AIM-np) technology functions as synthetic dendritic cells capable of directing a specific T cell-mediated immune response. By mimicking natural T cell biology, NexImmune’s cellular therapy product candidates (AIM ACT) are designed to combine the attributes of cellular precision, potency, and persistence with reduced potential for undesired toxicities. Human papilloma virus (HPV) is responsible for >45,000 cancers yearly in the United States, according to the CDC. From 2013–2017 an estimate 79% of cervical, vulva, penis, vaginal, anus, and oropharyngeal cancers were attributed to HPV and of these about 80% were associated with high-risk HPV types 16 and 18. Although multivalent vaccines against high-risk HPV infections exist, significant clinical challenges remain. A limited vaccination rate means many remain vulnerable and vaccination does not treat pre-existing HPV infections or malignancies.MethodsTherefore, NexImmune is employing its AIM-np technology to generate an adoptive cell therapy (ACT) using its proprietary enrichment and expansion (E+E) ex vivo process to expand clinically relevant numbers of CD8+ T cells that recognize the HPV16 and HPV18 oncogenic antigens (i.e., E6 and E7) expressed by malignant cells of head and neck, cervical, and anal cancers. Using the Immune Epitope Database and Analysis Resource (IEDB), 44 HLA-A2 restricted peptides were identified as potential immunogenic targets for preclinical screening. Using PBMCs from healthy donor-derived apheresis material, different combinations of these peptides were used in the E+E process to expand HPV-cancer specific CD8+ T cells.ResultsAfter multiple E+E experiments were concluded, 5 peptides were identified that consistently elicited the strongest T cell responses. Furthermore, these CD8+ T cells were predominantly from the central memory (CD62L+CD45RA-) and effector memory (CD62L-CD45RA-) phenotype (sum total 82.18 ± 8.29 [Mean ± SEM]) suggesting their in vivo functionality and persistence will combine anti-tumor activity with long-term immunologic memory.ConclusionsA similar E+E screening is being conducted with PBMCs isolated from HPV+ cancer patients. A comparison of the CD8+ T cell responses from healthy donor and cancer patient cells will provide critical preclinical data to support a planned FIH trial for HPV-associated malignancies. The current study demonstrates the ability for high-throughput peptide screening to identify clinically relevant peptide cocktails capable of expanding multi-antigen tumor-specific CD8+ T cell populations within 2 weeks.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1338-1338
Author(s):  
Sueon Kim ◽  
Hyun-Jung Sohn ◽  
Hyun-Joo Lee ◽  
Dae-Hee Sohn ◽  
Seung-Joo Hyun ◽  
...  

Abstract S.K and H.-J.S. contributed equally for this works Dendritic cell-derived exosome (DEX) has been known as an efficient stimulator of T cells. However, the production of sufficient DEX remains a barrier to broad utility for immunotherapy. In this study, we engineered K562 cells expressing triple-co-stimulatory signals (CD80, 4-1BBL, and CD83) with HLA-A2 as an AAPC. Specifically, CD137L (4-1BBL) is an ideal signaling molecule for long-term propagation of CD8+ T cells, and the addition of other co-stimulatory molecules, such as CD80 and CD83, is used to support the expansion of naive T cell subsets. DC-derived exosomes display immunologically important molecules such as HLA and co-stimulatory molecules. Likewise, CoEX-A2 expressed high levels of HLA-A2, CD80, CD83, and CD137L (41BBL) and mediate strong, antigen-specific CD8+ T lymphocyte activation. The stimulation of freshly isolated peripheral CD8+ T cells with the appropriate antigen specificity observed here was likely made possible by the use of the sensitive ELISPOT assay. Viral or tumor protein-pulsed exosomes can directly stimulate CD8+ T cell proliferation and differentiation into CTLs in vitro. In addition, exosomes can be taken up by both CD8+ T cells and K562 cells. Meanwhile, K562 cells that have taken up exosomes can also stimulate CD8+ T cells, which may be due to the higher levels of HLA-A2, CD80, CD83, and 41BBL expression observed on exosomes. Therefore, the CD8+ T cell antigen-specific expansion observed in our cultures is likely the result of coated CoEX-A2s working directly or in a cross-dressed manner. The results suggest that these novel exosomes may provide a crucial source to generate antigen-specific CD8+ T cells for adoptive cell therapy against viral infection and tumors. Disclosures No relevant conflicts of interest to declare.


Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 2018
Author(s):  
Eun M. Yu ◽  
Eunjung Cho ◽  
Rohit Singh ◽  
Seon-Hee Kim ◽  
Chungyong Han ◽  
...  

Adoptive cell therapy (ACT) using tumor-reactive T cells is a promising form of immunotherapy to specifically target cancer. However, the survival and functional maintenance of adoptively transferred T cells remains a challenge, ultimately limiting their efficacy. Here, we evaluated the use of recombinant IL7-Fc in ACT. In a lymphopenic murine melanoma model, IL7-Fc treatment led to the enhanced inhibition of tumor growth with an increased number of adoptively transferred CD8+ T cells in tumor tissue and tumor-draining lymph nodes. Additionally, IL7-Fc further enhanced anti-tumor responses that were induced by recombinant human IL2 in the same mouse model. In contrast, in an immunocompetent murine melanoma model, IL7-Fc dampened the anti-tumor immunity. Further, IL7-Fc decreased the proliferation of adoptively transferred and immune-activated tumor-reactive CD8+ T cells in immunocompetent mice by inducing the massive expansion of endogenous T cells, thereby limiting the space for adoptively transferred T cells. Our data suggest that IL7-Fc is principally beneficial for enhancing the efficacy of tumor-reactive T-cells in lymphopenic conditions for the ACT.


2013 ◽  
Vol 57 (1-3) ◽  
pp. 23-33 ◽  
Author(s):  
C. Marcela Díaz-Montero ◽  
Abdel-Aziz Zidan ◽  
Maria F. Pallin ◽  
Vasileios Anagnostopoulos ◽  
Mohamed L. Salem ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A822-A822
Author(s):  
Sri Krishna ◽  
Frank Lowery ◽  
Amy Copeland ◽  
Stephanie Goff ◽  
Grégoire Altan-Bonnet ◽  
...  

BackgroundAdoptive T cell therapy (ACT) utilizing ex vivo-expanded autologous tumor infiltrating lymphocytes (TILs) can result in complete regression of human cancers.1 Successful immunotherapy is influenced by several tumor-intrinsic factors.2 3 Recently, T cell-intrinsic factors have been associated with immunotherapy response in murine and human studies.4 5 Analyses of tumor-reactive TILs have concluded that anti-tumor neoantigen-specific TILs are enriched in subsets defined by the expression of PD-1 or CD39.6 7 Thus, there is a lack of consensus regarding the tumor-reactive TIL subset that is directly responsible for successful immunotherapies such as ICB and ACT. In this study, we attempted to define the fitness landscape of TIL-enriched infusion products to specifically understand its phenotypic impact on human immunotherapy responses.MethodsWe compared the phenotypic differences that could distinguish bulk ACT infusion products (I.P.) administered to patients who had complete response to therapy (complete responders, CRs, N = 24) from those whose disease progressed following ACT (non-responders, NRs, N = 30) by high dimensional single cell protein and RNA analysis of the I.P. We further analyzed the phenotypic states of anti-tumor neoantigen specific TILs from patient I.P (N = 26) by flow cytometry and single cell transcriptomics.ResultsWe identified two CD8+ TIL populations associated with clinical outcomes: a memory-progenitor CD39-negative stem-like TIL (CD39-CD69-) in the I.P. associated with complete cancer regression (overall survival, P < 0.0001, HR = 0.217, 95% CI 0.101 to 0.463) and TIL persistence, and a terminally differentiated CD39-positive TIL (CD39+CD69+) population associated with poor TIL persistence post-treatment. Although the majority (>65%) of neoantigen-reactive TILs in both responders and non-responders to ACT were found in the differentiated CD39+ state, CR infusion products also contained a pool of CD39- stem-like neoantigen-specific TILs (median = 8.8%) that was lacking in NR infusion products (median = 23.6%, P = 1.86 x 10-5). Tumor-reactive stem-like T cells were capable of self-renewal, expansion, and persistence, and mediated superior anti-tumor response in vivo.ConclusionsOur results support the hypothesis that responders to ACT received infusion products containing a pool of stem-like neoantigen-specific TILs that are able to undergo prolific expansion, give rise to differentiated subsets, and mediate long-term tumor control and T cell persistence, in line with recent murine ICB studies mediated by TCF+ progenitor T cells.4 5 Our data also suggest that TIL subsets mediating ACT-response (stem-like CD39-) might be distinct from TIL subsets enriched for anti-tumor-reactivity (terminally differentiated CD39+) in human TIL.6 7AcknowledgementsWe thank Don White for curating the melanoma patient cohort, and J. Panopoulos (Flowjo) for helpful discussions on high-dimensional analysis, and NCI Surgery Branch members for helpful insights and suggestions. S. Krishna acknowledges funding support from NCI Director’s Innovation Award from the National Cancer Institute.Trial RegistrationNAEthics ApprovalThe study was approved by NCI’s IRB ethics board.ReferencesGoff SL, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol 2016;34:2389–2397.Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–2199.McGranahan N, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351:1463–1469.Sade-Feldman M, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 2019;176:404.Miller BC, et al. Subsets of exhausted CD8 T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol 2019;20:326–336.Simoni Y, et al. Bystander CD8 T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018;557:575–579.Gros A, et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J Clin Invest 2014;124:2246–2259.


2010 ◽  
Vol 60 (3) ◽  
pp. 349-360 ◽  
Author(s):  
Huanfa Yi ◽  
Xiaofei Yu ◽  
Chunqing Guo ◽  
Masoud H. Manjili ◽  
Elizabeth A. Repasky ◽  
...  

2018 ◽  
Vol 507 (1-4) ◽  
pp. 59-66 ◽  
Author(s):  
Yan Zheng ◽  
Ning Gao ◽  
Yu-Long Fu ◽  
Bing-Yong Zhang ◽  
Xiu-Ling Li ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document