Artificial Antigen-Presenting Cells Permit Selective In Vitro Generation of CMV-Specific T-Cells of Desired HLA Allelic Restriction for Adoptive Immunotherapy in Recipients of HLA Disparate Allografts.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1298-1298 ◽  
Author(s):  
Deepa Trivedi ◽  
Wouter J.W. Kollen ◽  
Lorna Barnett ◽  
Michel Sadelain ◽  
Richard J. O’Reilly

Abstract Cytomegalovirus (CMV) infections remain a major cause of morbidity and mortality following allogeneic hematopoietic cell transplantation. For recipients of HLA haplotype disparate grafts, the risk of CMV infection is especially high and extends late into the post-transplant period. Early clinical trials indicate that adoptive transfer of ex vivo generated CMV-specific T-cells from the donor can be effective in the treatment and prevention of such infections. However, because seropositive donor T-cells sensitized with autologous infected or antigen loaded APCs regularly exhibit a repertoire of CMV-specific T-cells restricted by one or two immunodominant HLA alleles, such T-cells will only be effective if these immunodominant alleles are shared by the infected host. For example, among donors inheriting HLA-B*0702, CD8+ T-cell responses to CMV-pp65 peptides are usually, and often exclusively, restricted by this allele. To address this limitation, we tested whether artificial antigen-presenting cells, consisting of murine 3T3 cells transduced to express human B7.1, LFA1, ICAM1, β 2M and HLA-A*0201 alpha chain (Papanicolaou et al., Blood 2003), could be used to generate CMV-specific T-cells restricted by this allele in a series of normal donors inheriting HLA-A*A0201 and HLA-B*0702. Accordingly, T-cells were sensitized in vitro with either autologous monocyte-derived DCs or AAPCs expressing HLA-A*A0201, each loaded with a pool of 138 overlapping pentadecapeptides spanning the CMV-pp65 protein. Thereafter, specificity of responding T-cells was identified by mapping epitopes using an intersecting matrix of peptide subpools and measuring T-cells producing IFN-γ following secondary restimulation. HLA restriction was identified by analysis of T-cell cytotoxic responses against panels of EBV BLCL sharing single HLA alleles with the donor and loaded with the targeted peptide. Findings were confirmed by quantitation of T-cell binding tetramers containing targeted epitopes bound to either HLA-A*0201 or B*0702. In each of the donors, sensitization with DCs loaded with the peptide pool resulted in the generation of CMV-pp65-specific T-cells specific for epitopes predominantly or exclusively presented by HLA-B*0702. In contrast, sensitization with peptide pool loaded AAPC led to the generation of high numbers of T-cells recognizing separate CMV-pp65 epitopes in the context of HLA-A*A0201. These HLA-A*0201-restricted T-cells were also able to lyse peptide-loaded BLCLs or PBMCs expressing HLA-A*0201. Taken together, these studies demonstrate that AAPCs expressing single HLA alleles can be used to generate fully functional T-cells specific for epitopes presented by subdominant HLA alleles. This strategy may thus permit generation of virus-specific T-cells of desired HLA-restriction for adoptive immunotherapy in HLA-disparate transplant recipients.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4086-4086
Author(s):  
Ekaterina Doubrovina ◽  
Aisha Hasan ◽  
Banu Oflaz-Sozmen ◽  
Annamalai Selvakumar ◽  
Michel W Sadelain ◽  
...  

Abstract Abstract 4086 Poster Board III-1021 We have recently established a panel of artificial antigen-presenting cells (AAPCs) each expressing a single HLA class I allele (such as A0201, A0301, A2402, B0702, C0401) and costimulatory molecules (B7.1, ICAMI, LFA3, b2-microglobulin). These cells were capable of eliciting T cells responses specific for immunogenic epitopes of the cytomegalovirus protein CMVpp65 presented by single HLA allele expressed by each AAPC (J.Immunol, 2009;183(4):2837-50). The initial studies with HLA-A0201 expressing AAPCs had suggested that they could sensitize the T cells against single immunogenic A0201-binding peptides derived from antigens such as S100 and telomerase. However, there are no data regarding the potentials of AAPCs expressing other HLA alleles to present epitopes of endogenous self antigens and stimulate specific T cells. We have recently defined a series of immunogenic epitopes for WT1 that can be presented by each of the HLA alleles represented in this panel, by an epitope mapping analysis of WT1 specific T cells sensitized in vitro with autologous dendritic cells(DC) loaded with the pool of overlapping 15-mer peptides spanning the sequence of WT1. In the present study we asked whether the established panel of AAPCs expressing different HLA alleles when loaded with the human WT1 peptide pool, could be used to generate T cells directed against this self antigen, whether the peptides recognized by these T cells would be identical to the peptides defined to be immunogenic when presented in the context of the same HLA alleles after stimulation with the autologous DC loaded with the same WT1 pool and further whether AAPCs could also be used to elicit responses against subdominant epitopes presented by an HLA allele expressed on AAPCs that were not elicited among T cells sensitized with peptide pool loaded autologous DC. The pool of 141 synthetic pentadecapeptides each overlapping the next by 11aa loaded on the autologous DC elicited responses in 80% of 14 normal individuals tested. Epitope mapping permitted identification immunodominant WT1 peptide sequences eliciting responses and their restricting HLA allele(s) as determined in a Cr51 release assay against the panel of WT1 peptide loaded EBV transformed B cells matching one of the HLA alleles of the T cell genotype.. AAPCs expressing single HLA allele shared by the same T cells and loaded with the same WT1 total peptide pool elicited responses in each normal individual. In each case the T cells recognized one of the same epitopes defined to be previously immunogenic in man when presented in the context of the HLA allele expressed by the AAPCs. At the same time, single peptide epitopes that were defined to be presented by, for example, HLA A0201 alleles after stimulation with WT1 total pool loaded DCs did not elicit WT1 specific T cells responses if they were loaded on AAPCs expressing another HLA allele such as B0702. In comparisons of T cells sensitized with autologous DCs versus AAPCs loaded with WT1 peptides pool we also found that while AAPCs expressing the HLA allele presenting the immunodominant epitope recognized by T cells sensitized with pool loaded autologous DC also elicited strong responses, AAPCs expressing HLA alleles shared by the responding T cells could also elicit responses against immunogenic but subdominant epitopes that were not generated after sensitization of the same T cells with WT1 peptide pool loaded autologous DCs. Thus, these studies suggest that the panel of murine derived AAPCs can effectively present self antigens, such as WT1, and permits in vitro sensitization and propagation of tumor reactive WT1 peptide specific T cells of desired HLA restriction for the adoptive immunotherapy of WT1+ malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2589-2589 ◽  
Author(s):  
Aisha Hasan ◽  
Wouter J. Kollen ◽  
A. Selvakumar ◽  
D. Trivedi ◽  
M. Sadelain ◽  
...  

Abstract Adoptive transfer of antigen specific T cells can be effective in treating viral infections complicating allogeneic hematopoietic stem cell transplant (HSCT) recipients. However, in practice, generation of T cells is often limited by insufficient supply of autologous antigen presenting cells; therapeutic activity in HLA disparate patients may also be impaired if the immuno- dominant T cells generated are restricted by HLA alleles not shared by the host. AAPCs have theoretical advantages for T cell therapies in terms of sustained supply and capacity to selectively stimulate T cells restricted by HLA alleles shared by donor and host. However, to date, only AAPC systems expressing HLA A*0201 have been characterized. Accordingly, we established a panel of AAPC consisting of NIH 3T3 mouse fibroblast cells, each transduced to express β2- microglobulin and a prevalent HLA class-I allele, specifically HLA A*0201, A*0301, A*2402, B*0702, B*0801 or C*0401, as well as the human co-stimulatory molecules B7.1, LFA-3 and ICAM-1. Novel promotor sequences were introduced to secure stable high expression of the allele on the AAPCs. Sensitization of T cells from seropositive donors with AAPCs expressing each of these alleles (4-8 donors/allele), either loaded with overlapping 15-mer peptides spanning the CMVpp65 sequence or transduced to express the CMV pp65 protein, resulted in 12-35 fold expansions of CD8 + T cells exhibiting CMV pp65 epitope-specific, HLA restricted activity, as quantitated by peptide -HLA tetramer binding, epitope specific production of interferon gamma, and cytotoxic activity against peptide loaded or CMV infected targets. Although both peptide pool loaded and transduced AAPCs induce CMV pp65 epitope specific T cells, yields were higher when transduced AAPCs were employed. In studies of T-cells from 5 donors when sensitized with either peptide pool loaded autologous dendritic cells (DC) or HLA sharing AAPCs, sensitization with DC selectively induced T-cells specific for 1-2 immunodominant CMV pp65 epitopes. In contrast, while sensitization with a panel of peptide loaded or transduced AAPCs expressing shared HLA alleles elicited responses to the same dominant epitopes, we could also regularly generate comparable cytotoxic T cell responses to subdominant epitopes which were either not produced or only present at low frequencies in T cells sensitized with autologous DC. Thus, this panel of AAPCs stably expressing a series of HLA alleles which, in aggregate, are detected in 70% of the patients referred for HSCT, can be employed for rapid generation of CMV-pp65 specific T cells of desired HLA restriction for adoptive therapy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2918-2918
Author(s):  
Aisha Hasan ◽  
Ekatarina Doubrovina ◽  
Deepa Trivedi ◽  
Nancy A. Kernan ◽  
Susan Prockop ◽  
...  

Abstract T-cell depleted hematopoietic stem cell transplants (HSCT) from HLA haplotypedisparate related donors are being increasingly used for the treatment of patients lacking an HLA-compatible related or unrelated donor. Acute and chronic graft versus host disease can be prevented without added prophylaxis if techniques achieving extensive T-cell depletion are employed. Improved conditioning regimens and transplants providing high doses of CD34+ cells have also insured consistent engraftment. However, these patients have a higher and more prolonged risk of serious viral infections than recipients of matched unmodified or T-cell depleted grafts. Adoptive immunotherapy with donor-derived pathogen-specific T-cells can restore effective resistance in these patients. However, T-cells sensitized and propagated in vitro usually respond to only 1–2 immunodominant epitopes from any antigeneic protein presented by different donor HLA alleles. We questioned how often T-cells generated from an HLA-haplotype disparate donor would be restricted by donor-unique, rather than shared HLA alleles since such T-cells would be inactive against virus-infected cells of a haplotype disparate host. Accordingly, we tested 8 cytomegalovirus (CMV) seropositive HLA haplotype disparate parent or sibling donors, as well as 4 patients, each transplanted from one of these donors 1 or more years previously. In each case, T-cells were sensitized with autologous DCs loaded with a pool of overlapping 15-mers spanning the sequence of CMVpp65, an immunodominant protein of cytomegalovirus, for 21–28 days. Using a matrix of peptide subpools, we then mapped the epitopes eliciting IFNγ+ T-cell responses and then determined their HLA-restriction by assaying their specific cytotoxicity against a panel of EBV B-cell lines loaded with the immunogenic epitope each sharing a single HLA allele with the donor. Of the 8 donors tested, 6 responded to CMV pp65 epitopes presented by HLA alleles shared with the haplotype matched recipient, while 2 donors responded exclusively to epitopes presented by donor unique HLA alleles. Recipients of transplants from the latter two donors experienced prolonged CMV viremia, one with CMV chorioretinitis, requiring months of antiviral treatment. CMV-specific T-cells isolated from each of the four transplanted patients over 1 year post transplant were specific for epitopes presented by HLA alleles shared by their donors. In the patients transplanted from donors whose CMV-pp65 specific T-cells were restricted by donor-unique HLA alleles, the epitope targeted and HLA restriction of the engrafted T-cell differed from that of the transplant donors own T-cells, suggesting that these T-cells may have developed from donor precursors differentiating in the host thymus. Thus, in a significant proportion of cases (2/8 in this study), virus-specific T-cells generated in vitro from HLA haplotype disparate donors may be restricted by donor-unique HLA alleles and, as a consequence, inactive against virus-infected host cells. For such patients, techniques insuring selective generation of T-cells specific for epitopes presented by shared alleles should be employed. Our findings may also in part explain the prolonged susceptibility of these patients to viral infection. Since immunodominant T-cells reactive against the same epitopes are also those detected at highest frequency in the donor’s blood, they would likely be the only virus-reactive T-cells transferred in a T-cell depleted graft. As a consequence, effective CMV-specific responses might be delayed until new T cells are generated from precursors developing in the host thymus.


2021 ◽  
Vol 12 ◽  
Author(s):  
Manoj Patidar ◽  
Naveen Yadav ◽  
Sarat K. Dalai

IL-15 is one of the important biologics considered for vaccine adjuvant and treatment of cancer. However, a short half-life and poor bioavailability limit its therapeutic potential. Herein, we have structured IL-15 into a chimeric protein to improve its half-life enabling greater bioavailability for longer periods. We have covalently linked IL-15 with IgG2 base to make the IL-15 a stable chimeric protein, which also increased its serum half-life by 40 fold. The dimeric structure of this kind of IgG based biologics has greater stability, resistance to proteolytic cleavage, and less frequent dosing schedule with minimum dosage for achieving the desired response compared to that of their monomeric forms. The structured chimeric IL-15 naturally forms a dimer, and retains its affinity for binding to its receptor, IL-15Rβ. Moreover, with the focused action of the structured chimeric IL-15, antigen-presenting cells (APC) would transpresent chimeric IL-15 along with antigen to the T cell, that will help the generation of quantitatively and qualitatively better antigen-specific memory T cells. In vitro and in vivo studies demonstrate the biological activity of chimeric IL-15 with respect to its ability to induce IL-15 signaling and modulating CD8+ T cell response in favor of memory generation. Thus, a longer half-life, dimeric nature, and anticipated focused transpresentation by APCs to the T cells will make chimeric IL-15 a super-agonist for memory CD8+ T cell responses.


2014 ◽  
Vol 2 (S3) ◽  
Author(s):  
Karlo Perica ◽  
Joan Bieler ◽  
Christian Schuetz ◽  
Juan Varela ◽  
Mathias Oelke ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4886-4886
Author(s):  
Hanwen Mao ◽  
Wenli Liu ◽  
Zhe Gen ◽  
Wei Huang ◽  
Yicheng Zhang ◽  
...  

Abstract The antigen-specific cytotoxic T lymphocyte activated by antigen presenting cell is widely used in cell immunotherapy recently. CML28, which was screened from chronic myelogenous leukemia (CML) patients, was reported to be a specific tumour antigen and over-expressed on CML cells and acute leukemia cells. Therefore, CML28 could be a potential target for leukemia treatment. Dendritic cells (DC) are the most important antigen present cells, but it is hard to isolate and culture DCs for clinical use, which hampers the specific cell immunotherapy. Our investigation aimed to study the cytotoxic effects of CML28 specific T cells activated by artificial antigen presenting cells, on acute leukemia cells in vitro. Artificial antigen presenting cells were prepared by connecting CML28 to magnetic superbead that containing HLA-A2-Ig and B7-1 molecule. Mononuclear cells were isolated from the bone marrow or peripheral blood of healthy donors with positive HLA-A2. The artificial antigen-presenting cells were co-cultured with isolated mononuclear cells for four weeks. The activation and proliferation of CML28-specific T cells were measured by dimmer binding technique using flow cytometry. The cytotoxic effects of CML28-specific T cells on leukemia cells, which were isolated from leukemia patient, were evaluated by lactate dehydrogenase (LDH) releasing assay. Increased proportion of CML28-specific T cells was observed in artificial antigen-presenting group than in control group (29.27±3.54% vs 2.95±0.66%, p<0.05). For cytotoxic effects assay, significant higher killing efficiency was seen in artificial antigen-presenting group (41.47±4.23%vs3.56±0.71%, when the effector: target ration is 40:1, p<0.01). Therefore, we concluded that the artificial antigen presenting cells could mimic antigen presenting cells to induce specific T cell activation and proliferation, and cytotoxic effects on target cells, indicating that artificial antigen presenting cell-induced cytotocix T cells could be an option for leukemia treatment.


2021 ◽  
Vol 12 ◽  
Author(s):  
Sophia Giang ◽  
David A. Horwitz ◽  
Sean Bickerton ◽  
Antonio La Cava

Artificial antigen-presenting cells (aAPCs) are synthetic versions of naturally occurring antigen-presenting cells (APCs) that, similar to natural APCs, promote efficient T effector cell responses in vitro. This report describes a method to produce acellular tolerogenic aAPCs made of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) and encapsulating IL-2 and TGF-β for a paracrine release to T cells. We document that these aAPCs can induce both human CD4+ and CD8+ T cells to become FoxP3+ T regulatory cells (Tregs). The aAPC NP-expanded human Tregs are functional in vitro and can modulate systemic autoimmunity in vivo in humanized NSG mice. These findings establish a proof-of-concept to use PLGA NPs as aAPCs for the induction of human Tregs in vitro and in vivo, highlighting the immunotherapeutic potential of this targeted approach to repair IL-2 and/or TGF-β defects documented in certain autoimmune diseases such as systemic lupus erythematosus.


2019 ◽  
Author(s):  
Fatemeh S. Majedi ◽  
Mohammad Mahdi Hasani-Sadrabadi ◽  
Timothy J. Thauland ◽  
Song Li ◽  
Louis-S. Bouchard ◽  
...  

AbstractActivation of T cells by antigen presenting cells allows them to proliferate, produce cytokines, and kill infected or cancerous cells. We and others have shown that T cell receptors receive and in fact require mechanical forces from their own movements and the movements of antigen presenting cells. Emulation of T cell activation in vitro allows for the massive expansion of T cells necessary for clinical applications. In this paper, we studied the impact of augmenting novel artificial antigen presenting cells of various sizes and antigenic signal strength with mechanical, oscillatory movement. We showed that dynamic culture roughly doubles signal strength as compared to conventional, static culture. We demonstrated that tuning the strength of signal to a “sweet spot” allows for robust expansion of induced regulatory T cells, which is impeded by approaches that simply maximize activation.


Sign in / Sign up

Export Citation Format

Share Document