Adoptive Transfer of CMV-Specific Donor T Cells Following Allogeneic Transplantation Leads to Rapid and Safe Systemic Reconstitution.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 44-44
Author(s):  
Karl S Peggs ◽  
Kirsty Thomson ◽  
Edward Samuel ◽  
Gemma Dyer ◽  
Julie Armoogum ◽  
...  

Abstract Abstract 44 Reactivation of CMV remains a significant problem following allogeneic hematopoietic stem cell transplantation. Antiviral drug therapy is effective but toxic, and resistant strains of CMV are increasingly being reported. Virus-specific T lymphocytes are necessary for the control of viral reactivation. Adoptive transfer of donor derived CMV-specific T cells has been reported previously but most methods to produce such cells have involved several weeks of in vitro culture or have produced a therapeutic product restricted to CD8 T cells. The current method involves a short incubation of donor peripheral blood mononuclear cells with either CMV-pp65 protein (20 hours) or a pool of peptides from pp65 (6 hours) with subsequent isolation of interferon-gamma secreting cells by CliniMACS using IFNψ capture microbeads (Miltenyi Biotec). This technique permits rapid isolation of an enriched IFNψ secreting T cell product, manufactured to clinical grade, which is then cryopreserved in dosed aliquots for subsequent infusion. Here we report the outcome of a single arm phase I/II in which CMV-T cells given pre-emptively at first detection (qPCR) of CMV DNA in peripheral blood, or at day +40-60 as prophylaxis. CMV replication was monitored by weekly PCR and reconstitution of CMV-specific T cells by pentamer labelling and/or IFNψ secretion assay. Conventional antiviral drug therapy was instituted if the viral load rose above institutional threshold. 30 recipients of T cell depleted low intensity transplants from HLA-matched CMV-seropositive related donors were enrolled between 2006 and 2008. Donors underwent a second, short apheresis procedure approximately 15 days after collection of the mobilised HPC-A for the collection of CMV-T cells. 26 clinical-grade products were produced to full cGMP standards; four donors were unsuitable or withdrew. The mean yield of cells following enrichment was 41.7% with a median purity of 43.9% (range 1.4-81.8). Adequate CMV-T cells were isolated from all donors. Both pp65 and peptide stimulated products contained both CD4 and CD8 reactive T cells. Median dose of CMV-specific CD4 T cells was 2840/kg and of CMV-specific CD8 was 630/kg. Eighteen patients received a single dose of 1×10^4 CD3+/kg; 13 were CMV seropositive; 11 were treated pre-emptively and 7 prophylactically. 83% had received T cell deplete regimens. Within 2 weeks of infusion in vivo expansion of CMV-T cells was observed in 17 of 18 patients. One patient required 4 weeks to generate detectable CMV-T cell in his peripheral blood. TCR-BV usage of the CMV-T cells post infusion matched that of the cells which had been infused. The 7 patients who had cells infused prophylactically all showed expansions of CMV-T cells in the absence of detectable viral DNA in peripheral blood. Subsequent low level CMV-reactivation was seen in one of these and was associated with rapid CMV-T cell expansion with clearance of virus without anti-viral drug therapy. One developed subsequent extensive chronic GvHD and required antiviral treatment for multiple reactivation episodes following introduction of steroids. Of the 11 patients treated pre-emptively, 9 received antiviral therapy for the initial reactivation, although in 7 patients this was required for only 7-15 days. (compared to a median of 21 days in historical controls). Three patients had a further CMV reactivation event. One followed prednisolone therapy for acute grade II GvHD. The second was the patient who had shown poor T cell expansion post infusion and had required prolonged anti-viral therapy (33 days) for the initial CMV reactivation. The third patient received no treatment and cleared virus following a further in vivo expansion of CMV-reactive T cells, suggesting the presence of a functional memory population. GVHD incidence and severity was no worse than seen in comparable historical controls. 3 patients suffered grade 2-3 acute GvHD. 3/17 evaluable patients developed extensive chronic GvHD (2 were recipients of T replete grafts). 16/18 patients were alive at the end of the 6 month monitoring period and CMV-reactive T cells were detectable in all 16. CMV-specific donor T cells can be readily produced to cGMP compliance which can be safely infused and lead to early immune reconstitution in at-risk patients. Cells expand in response to subsequent CMV-reactivation and patients appear to require fewer anti-viral treatment episodes which is being tested in an ongoing phase III trial. Disclosures: Lowdell: Cell Medica Ltd: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 589-589 ◽  
Author(s):  
Kirsty Thomson ◽  
Moran Julie ◽  
Kwok Pang ◽  
Ed Samuel ◽  
Michael Desborough ◽  
...  

Abstract Reactivation of CMV is a significant cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation. Available antiviral drug therapy is effective but toxic, and contributes to the negative impact of CMV infection in affected patients. Virus-specific T lymphocytes are necessary for the control of viral reactivation, and therapeutic strategies using adoptive transfer of donor derived CMV-specific T cells are being explored. Most methods to produce donor-derived antigen-specific T cells involve several weeks of in vitro culture. The current method involves a 20hr incubation of donor-derived PBMCs with CMV-pp65 protein with subsequent isolation of T cells secreting interferon-gamma (IFNγ) by CliniMACS using IFNγ capture microbeads (Miltenyi Biotec). This technique permits rapid isolation of an enriched IFNγ secreting T cell product, manufactured to clinical grade, which is then cryopreserved in dosed aliquots for subsequent infusion. A single arm phase I study is currently underway, with CMV-specific T cells given pre-emptively at first detection (by quantitative PCR) of CMV DNA in peripheral blood, or at day +40–50 as prophylaxis. Cells are infused at an initial dose of 1x104 CD3+/kg recipient weight, and CMV monitored by weekly PCR. Antiviral drug therapy is instituted if the viral load subsequently rises above threshold, according to institutional guidelines. Patients have received at least one dose of CMV-T cells and all are alive and well. Follow-up data are available currently on 7. These 7 patients had all received T cell deplete regimens carrying a CMV reactivation risk of >85%. The mean yield of cells following enrichment was 15.6 x106 (range 5.2–26.7) of which 82.3% were CD3+, 22.0% were CD4+/IFNγ+ and 10.6% were CD8+/IFNγ+, giving a total mean CMV-reactive cell yield of 3.4 x106 per donor. Following infusion, in vivo expansion of CMV-reactive T cells was observed in all patients. CMV-reactive cells represented a mean of 9.0% of CD4+ cells and 7.3% of CD8+ cells by 2–4 weeks post-infusion; the result of in vivo expansions of CMV-reactive cells of 700- to 5000-fold by week 4 post infusion. Two patients had CMV-specific cells infused prophylactically at day 40–50 and expansions of CMV-reactive T cells were seen in both despite consistent absence of detectable viral DNA in peripheral blood. Of the remaining 5 patients, all received antiviral therapy for the initial reactivation, although in 4 patients this was required for a significantly shorter period than in historical controls (11–14 days). In the final patient, more prolonged treatment was required (33 days) and a second reactivation also required drug therapy- in this patient, the expansion of CMV reactive T cells was substantially delayed compared to the other 6. Two other patients had a further CMV reactivation event. One followed prednisolone therapy for acute grade II graft-versus-host disease (in keeping with expected rates of acute GVHD following the primary allograft procedure) and required antiviral drug treatment. The third patient received no treatment and cleared virus following a further in vivo expansion of CMV-reactive T cells, suggesting the presence of functional memory cells. CMV-reactive T cells have been detectable up to the final 6 month sampling date. This technique provides a rapid source of clinical-grade CMV-reactive CD4+ and CD8+ T cells which early evidence suggests provide effective antiviral immunity.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 223-223
Author(s):  
Ulrike Gerdemann ◽  
Usha L Katari ◽  
Jacqueline Keirnan ◽  
John A. Craddock ◽  
Janet Salinas ◽  
...  

Abstract Abstract 223 We have previously demonstrated that small numbers of ex vivo-expanded, trivirus-specific T cells targeting Epstein Barr virus (EBV), cytomegalovirus (CMV), and Adenovirus (Adv) are safe, proliferate in vivo and protect human subjects against all 3 viruses following HSCT. However, broader implementation is limited by the need for infectious virus (EBV) to establish an EBV-transformed B lymphoblastoid cell line (EBV-LCL), for clinical grade adenoviral vector, and by prolonged (6wks for EBV-LCL and 6wks for T cells) and complex manufacture. Moreover, competition between viral antigens limits extension to additional viruses. We now evaluate whether it is possible to make clinically effective T cell lines using methods that exclude all viral components and utilize simplified manufacturing technology. With NHLBI-Production Assistance for Cellular Therapies (PACT) support, 29 clinical-grade rCTL lines have been generated. From an initial 15×106 PBMCs, we prepared a median of 214±88 × 106 T cells (range 100–420×106) over 9–11 days by using dendritic cells (DCs) nucleofected with DNA plasmids encoding immunogenic EBV (LMP2, EBNA1 and BZLF1), Adv (Hexon and Penton), and CMV (pp65 and IE1) antigens, and expanding them with IL4+7 in gas permeable (G-Rex) devices. The rCTL lines were polyclonal, comprising both CD4+ (33±3%) and CD8+ (60.5±3%) cells, that expressed the activation and memory markers CD45RO+ CD62L+ (64.3±26.6%) and CD45RO+ CD62L- (17.4±14%). Twenty lines generated from donors that were seropositive for all three viruses demonstrated activity against all 3 targets - CMV (IE1: 359±100; pp65: 637±177 SFC/2×105), EBV (LMP2: 217±60, EBNA1: 67±19 and BZLF1: 111±31) and Adv (Hexon: 265±74, Penton: 191±53) - while 9 lines generated from donors who were CMV seronegative demonstrated activity exclusively against EBV (LMP2: 197±70, EBNA1: 145±51 and BZLF1: 239±84) and Adv (Hexon: 271±96, Penton: 254±90). None of the lines reacted against recipient PHA blasts (median spontaneous Cr51 release of 0% at a 20:1 effector to target cell ratio). To date we have administered these lines to 10 recipients of allogeneic HSCT. Five patients received dose level (DL) 1 (5×106/m2), 2 received DL2 (1×107/m2) and 3 had DL3 (2×107/m2) of this phase I/II study. Three patients were infused as treatment for CMV, 2 for Adv, 2 for EBV, 1 for EBV+Adv, and 2 for CMV+Adv. Our major anticipated concern was that these once stimulated, unselected rCTLs might cause GvHD in vivo, but that was not the case. One patient developed a skin rash 2 weeks after rCTLs but no other toxicity related to the infused cells was observed. Eight of the 10 treated patients including one patient with a biopsy-proven EBV lymphoma and the 3 patients with double reactivations had complete responses to rCTL therapy with a return of viral load to normal and resolution of all other symptoms. Response was associated with an increase in the frequency of virus-specific T cells detected in the peripheral blood against the infecting virus. For CMV there was an increase from a median of 0.5 to 96 and 1 to 277 SFC/4×105 IE1 and pp65-specific T cells respectively 3–6wks post-infusion; for Adv an increase from a mean of 0.5 to 137 and 0.5 to 99 SFC/4×105 Hexon and Penton-specific cells 2wks post-infusion, respectively, and for EBV an increase from 2.75 to 227, 1.5 to 39, and 1 to 188.5 SFC/4×105 EBNA1, LMP2, and BZLF1-specific T cells 2–4wks post-infusion, respectively. Two patients failed to respond. The first had a 3 year history of persistent CMV colitis despite high circulating CMV-specific precursors (297 IE1-specific and 193 pp65-specific T cells/4×105 PBMCs). Post rCTL we saw no increase in T cell precursor levels and no clinical improvement. The second was treated for an elevated EBV viral load but also had high pre-existing EBV-specific T cell precursors (60, 23, and 240 SFC/4×105 EBNA1, LMP2, and BZLF1-specific T cells). Again, post-rCTL we did not detect an increase in EBV-specific precursors and no response. Thus, infusion of rCTLs has been safe and in 8/10 patients was associated with the appearance of virus-reactive T cells directed against the infecting virus in peripheral blood and subsequent virus clearance. rCTLs have the potential to increase the availability of cell products for HSCT recipients and we are currently extending this platform to additional viruses, thereby broadening the spectrum of pathogens that can be targeted by adoptive transfer of a single T cell line. Disclosures: Off Label Use: IND cell therapy product.


Blood ◽  
2008 ◽  
Vol 112 (6) ◽  
pp. 2232-2241 ◽  
Author(s):  
Jeff K. Davies ◽  
John G. Gribben ◽  
Lisa L. Brennan ◽  
Dongin Yuk ◽  
Lee M. Nadler ◽  
...  

AbstractWe report the outcomes of 24 patients with high-risk hematologic malignancies or bone marrow failure (BMF) who received haploidentical bone marrow transplantation (BMT) after ex vivo induction of alloantigen-specific anergy in donor T cells by allostimulation in the presence of costimulatory blockade. Ninety-five percent of evaluable patients engrafted and achieved full donor chimerism. Despite receiving a median T-cell dose of 29 ×106/kg, only 5 of 21 evaluable patients developed grade C (n = 4) or D (n = 1) acute graft-versus-host disease (GVHD), with only one attributable death. Twelve patients died from treatment-related mortality (TRM). Patients reconstituted T-cell subsets and immunoglobulin levels rapidly with evidence of in vivo expansion of pathogen-specific T cells in the early posttransplantation period. Five patients reactivated cytomegalovirus (CMV), only one of whom required extended antiviral treatment. No deaths were attributable to CMV or other viral infections. Only 1 of 12 evaluable patients developed chronic GVHD. Eight patients survive disease-free with normal performance scores (median follow-up, 7 years). Thus, despite significant early TRM, ex vivo alloanergization can support administration of large numbers of haploidentical donor T cells, resulting in rapid immune reconstitution with very few viral infections. Surviving patients have excellent performance status and a low rate of chronic GVHD.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ditte E. Jæhger ◽  
Mie L. Hübbe ◽  
Martin K. Kræmer ◽  
Gael Clergeaud ◽  
André V. Olsen ◽  
...  

AbstractAdoptive T-cell transfer (ACT) offers a curative therapeutic option for subsets of melanoma and hematological cancer patients. To increase response rates and broaden the applicability of ACT, it is necessary to improve the post-infusion performance of the transferred T cells. The design of improved treatment strategies includes transfer of cells with a less differentiated phenotype. Such T cell subsets have high proliferative potential but require stimulatory signals in vivo to differentiate into tumor-reactive effector T cells. Thus, combination strategies are needed to support the therapeutic implementation of less differentiated T cells. Here we show that systemic delivery of tumor-associated antigens (TAAs) facilitates in vivo priming and expansion of previously non-activated T cells and enhance the cytotoxicity of activated T cells. To achieve this in vivo priming, we use flexible delivery vehicles of TAAs and a TLR7/8 agonist. Contrasting subcutaneous delivery systems, these vehicles accumulate TAAs in the spleen, thereby achieving close proximity to both cross-presenting dendritic cells and transferred T cells, resulting in robust T-cell expansion and anti-tumor reactivity. This TAA delivery platform offers a strategy to safely potentiate the post-infusion performance of T cells using low doses of antigen and TLR7/8 agonist, and thereby enhance the effect of ACT.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5603-5603 ◽  
Author(s):  
Cherie Tracy Ng ◽  
Jeanette Ampudia ◽  
Robert J. Soiffer ◽  
Jerome Ritz ◽  
Stephen Connelly

Background: CD6 is a co-stimulatory receptor, predominantly expressed on T cells, that binds to activated leukocyte cell adhesion molecule (ALCAM), a ligand expressed on antigen presentation cells and various epithelial and endothelial tissues. The CD6-ALCAM pathway plays an integral role in modulating T cell activation, proliferation, differentiation and trafficking and is central to inflammation. While effector T cell (Teff) are CD6hi and upregulate expression upon activation, regulatory T cells (Treg) remain CD6lo/-, making this an attractive target to modulate Teff activity while preserving Treg activity. Early studies by Soiffer and colleagues demonstrated using T12, an anti-CD6 monoclonal antibody (mAb) that ex-vivo depletion of CD6+ donor cells prior to transplantation decreased the incidence of both acute and chronic GVHD, highlighting the importance of CD6+ cells in GVHD pathogenesis and validating it as a therapeutic target. However, it remains to be shown whether modulating the CD6-ALCAM pathway in vivo can attenuate GVHD. We investigated the use of itolizumab, a humanized anti-CD6 mAb that has demonstrated clinical efficacy in other autoimmune diseases, as both a preventive and therapeutic treatment for GVHD, using a humanized xenograft mouse model. Methods: Humanized xenograft mice were generated by intravenous transfer of 2x10^7 human PBMCs into 6-8 weeks old NOD/SCID IL2rγ-null (NSG). To investigate the ability of itolizumab to prevent GVHD, mice were dosed with either 60μg or 300μg of itolizumab, 150μg of abatacept (CTLA4-Ig), or vehicle, starting one day prior to PBMC transplantation. To investigate the therapeutic effect of itolizumab, mice were dosed with either 150μg of itolizumab or vehicle, starting at Day 5 post-PBMC transfer, when transplanted T cells are already activated. All treatments were administered IP every other day. Weight and disease scores were monitored throughout the study. At Days 18 and 35, peripheral blood was evaluated by flow cytometry to examine T cell prevalence, and tissues were collected for histological examination of pathology and T cell infiltration. Results: When administered as prevention (Day -1), treatment with either 60μg or 300μg of itolizumab significantly decreased mortality compared to the vehicle control (100% vs. 10%); this decrease was similar to the positive control group treated with abatacept (Figure 1). At 60μg, itolizumab-treated mice demonstrated significant reductions in the prevalence of human T cells in peripheral blood vs. vehicle-treated mice at Day 18 (<0.2% vs. 74.5%; p < 0.001). The reduction in peripheral T cells was accompanied by reductions in tissue-infiltrating T cells in lung (85-fold) and gut (9.5-fold), as well as reductions in disease scores and weight loss. When administered therapeutically, treatment with itolizumab was associated with a survival rate of 50% compared to 10% in the control group (Figure 2). Similarly, peripheral T cell prevalence (34.3% vs. 65.1%; p < 0.001), weight loss, and disease scores were inhibited by itolizumab compared to vehicle control mice. Conclusions: These data suggest that systemic treatment with itolizumab can modulate pathogenic Teff cell activity, establishing this antibody as a potential therapeutic for patents with GvHD. A phase I/II study using itolizumab as first line treatment in combination with steroids for patients with aGVHD is currently ongoing (NCT03763318). Disclosures Ng: Equillium: Employment, Equity Ownership. Ampudia:Equillium: Employment. Soiffer:Mana therapeutic: Consultancy; Kiadis: Other: supervisory board; Gilead, Mana therapeutic, Cugene, Jazz: Consultancy; Juno, kiadis: Membership on an entity's Board of Directors or advisory committees, Other: DSMB; Cugene: Consultancy; Jazz: Consultancy. Ritz:Equillium: Research Funding; Merck: Research Funding; Avrobio: Consultancy; TScan Therapeutics: Consultancy; Talaris Therapeutics: Consultancy; Draper Labs: Consultancy; LifeVault Bio: Consultancy; Celgene: Consultancy; Aleta Biotherapeutics: Consultancy; Kite Pharma: Research Funding. Connelly:Equillium: Employment, Equity Ownership.


Blood ◽  
1987 ◽  
Vol 70 (5) ◽  
pp. 1595-1603 ◽  
Author(s):  
K Welte ◽  
CA Keever ◽  
J Levick ◽  
MA Bonilla ◽  
VJ Merluzzi ◽  
...  

Abstract The ability of peripheral blood mononuclear cells (PBMC) to produce and respond to interleukin-2 (IL-2) was evaluated in 50 recipients of HLA- identical bone marrow (BM) depleted of mature T cells by soybean agglutination and E rosetting (SBA-E-BM). In contrast to our previous findings in recipients of unfractionated marrow, during weeks 3 to 7 post-SBA-E-BM transplantation (BMT), PBMC from the majority of patients spontaneously released IL-2 into the culture medium. This IL-2 was not produced by Leu-11+ natural killer cells, which were found to be predominant in the circulation at this time, but by T11+, T3+, Ia antigen-bearing T cells. The IL-2 production could be enhanced by coculture with host PBMC frozen before transplant but not by stimulation with mitogenic amounts of OKT3 antibody, thus suggesting an in vivo activation of donor T cells or their precursors by host tissue. Spontaneous IL-2 production was inversely proportional to the number of circulating peripheral blood lymphocytes and ceased after 7 to 8 weeks post-SBA-E-BMT in most of the patients. In patients whose cells had ceased to produce IL-2 spontaneously or never produced this cytokine, neither coculture with host cells nor stimulation with OKT3 antibody thereafter induced IL-2 release through the first year posttransplant. Proliferative responses to exogenous IL-2 after stimulation with OKT3 antibody remained abnormal for up to 6 months post-SBA-E-BMT, unlike the responses of PBMC from recipients of conventional BM, which responded normally by 1 month post-BMT. However, the upregulation of IL- 2 receptor expression by exogenous IL-2 was found to be comparable to normal controls when tested as early as 3 weeks post-SBA-E-BMT. Therefore, the immunologic recovery of proliferative responses to IL-2 and the appearance of cells regulating in vivo activation of T cells appear to be more delayed in patients receiving T cell-depleted BMT. Similar to patients receiving conventional BMT, however, the ability to produce IL-2 after mitogenic stimulation remains depressed for up to 1 year after transplantation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 593-593
Author(s):  
Andreas Beilhack ◽  
Stephan Schulz ◽  
Jeanette Baker ◽  
Georg F. Beilhack ◽  
Courtney B. Wieland ◽  
...  

Abstract To study the complex pathophysiology of aGvHD in allogeneic hematopoietic cell transplantation (HCT) we transplanted transgenic luciferase expressing T cell populations into lethally irradiated HCT recipients (murine MHC major mismatch model, H-2q into H-2d). Tracking of light emitting donor T cells in living animals and detailed studies by multi color immunofluorescence microscopy (IFM) and FACS revealed the tight links of spatial and temporal evolution in this complex immune process. Donor derived T cells migrate to T cell areas in lymphoid tissues within a period of 12 hours. In the initial periods donor CD4+ T cells appear first with CD8+ T cell infiltration at later time points. Donor T cells start proliferating in lymphatic tissues on day 2 after transfer, as observed by BrdU stainings. Although alloreactive T cells are similarly activated in all lymphoid organs, they only up-regulate gut homing molecules after more than 5 cell divisions (CFSE proliferation analysis by FACS) in certain lymphoid organs (Peyer’s patches, mesenteric LN and spleen). Abruptly on day 4 after HCT, T cells migrate into intestinal sites. These findings strongly suggested, that specific priming sites are required for alloreactive T cells to induce a distinct type of tissue tropism in GvHD. In contrast to previous reports peformed without host conditioning, depletion of certain lymphoid organs (e.g. Peyer’s patches) before HCT or antibody blocking experiments did not control aGVHD. BLI showed, that anti-L-selectin or anti-MAdCAM-1 antibody treatment alone or in combination was effective in blocking donor T cell migration to lymph nodes and Peyer’s patches, while redirecting these cells to liver and spleen. Subsequently cells proliferated predominantly in the spleen until day 3 after HCT. Surprisingly we observed a full picture of gut infiltration on day 4 and skin involvement on day 5–6, similar in dynamics and strength to the aGvHD isotype control group. These findings demonstrated, that other lymphoid organs can functionally compensate for inducing gut and skin homing of alloreactive T cells. Of importance, we demonstrated that T cells that lacked homing molecules for secondary lymphoid organs had alloreactive properties in vitro, yet did not cause aGVHD in vivo. In summary, the activation of alloreactive T cells in specific sites throughout the body is complex and involves the acquisition of homing molecule expression. Transplantation of T cells with defined homing properties therefore, appears to be a promising alternative in conferring protective immunity early after HCT without the risk of aGvHD.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 573-573
Author(s):  
Jian-Ming Li ◽  
Cynthia Giver ◽  
Doug McMillan ◽  
Wayne Harris ◽  
David L. Jaye ◽  
...  

Abstract Introduction: Impaired or inappropriate immune reconstitution after allogeneic bone marrow transplantation (BMT) can lead to infection, graft-versus-host disease (GvHD) and leukemia relapse. We have previously reported that BM contains two populations of dendritic cell (DC) subsets, CD11b+ DC and CD11b− DC, and that CD11b depleted donor BM promoted increased donor T-cell chimerism and increased graft-versus-leukemia (GvL) activity in C57BL/6 → B10BR transplants [BBMT, 2004, 10: 540]. To explore the mechanism by which CD11b-depletion improved allo-reactivity, we performed allogeneic hematopoietic cell transplants using defined populations of donor stem cells, DCs, and T-cells in a MHC mis-matched BMT model. Methods: We transplanted FACS purified populations of 50,000 GFP+ CD11b- DC or CD11b+ DC in combination with 5,000 FACS purified Lin- Sca-1+ c-kit+ hematopoietic stem cells (HSC) and 300,000 or 1,000,000 congenic spleen T-cells from C57BL/6 donors into C57BL/6[H-2Kb], B10BR[H-2Kk] and PL/J[H-2Ku] recipients. Proliferation of CFSE stained donor T-cells was measured at 72 hours post-transplant. FACS cytometric bead array and intracellular cytokine staining measured serum and intracellular cytokines in donor T-cells. Results: The initial proliferation and Ki-67 expression of CFSE labeled donor T-cells in allogeneic recipients were much higher than in syngeneic recipients (homeostatic proliferation). Confocal microscopy showed co-localization of donor DC subsets with donor T-cells in the recipient spleens at 3 and 10 days post-transplant. In the allogeneic transplant settings, donor T-cells co-transplanted with CD11b- DC showed increased IFN-γ synthesis at 3 and 10 days post-transplant compared to donor T-cells co-transplanted with HSC plus CD11b+ DC or HSC alone. Increased proliferation of donor T-cells led to increased donor T-cell chimerism at day 10, 30, 60, and day105 post-transplant among recipients of CD11b- DC compared to recipients of HSC alone or HSC plus CD11b+ DC (Figure 1). Transplantation of spleen T-cells and CD11b- DC did not increase GvHD, but was associated with full donor chimerism. In contrast, transplantation of allogeneic CD11b+ DC led to persistence and expansion of residual host T-cells (Figure 2), increased numbers of donor CD4+CD25++Foxp3+ T-cells, and higher serum level of IL-10 supporting early post-transplant expansion of donor T regulatory cells (Treg). Conclusions: Donor CD11b- DC promoted immune reconstitution by polarizing donor T-cells to Th1 immune responses associated with increased IFN-γ synthesis and donor T-cell proliferation, while donor CD11b+ DC suppressed immune reconstitution by inhibiting donor T-cell allogeneic immune responses. These data support a novel paradigm for the regulation of post-transplant immunity and suggest clinical methods to test the hypothesis that manipulation of the DC content of a hematopoietic cell allograft regulates post transplant immunity in the clinical setting. Figure 1. Donor Spleen Derived T-cells in Peripheral Blood [* p<0.05, v.s. recipients of HSC plus CD11b(+)DC and spleen T-cells] Figure 1. Donor Spleen Derived T-cells in Peripheral Blood [* p<0.05, v.s. recipients of HSC plus CD11b(+)DC and spleen T-cells] Figure 2. Host Derived T-cells in Peripheral Blood [* p<0.05, v.s. recipients of HSC plus CD11b(-)DC and spleen T-cells] Figure 2. Host Derived T-cells in Peripheral Blood [* p<0.05, v.s. recipients of HSC plus CD11b(-)DC and spleen T-cells]


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3662-3662
Author(s):  
Robert M. Dean ◽  
Daniel H. Fowler ◽  
Nancy M. Hardy ◽  
Jeanne Odom ◽  
Kathleen Castro ◽  
...  

Abstract Allogeneic hematopoietic stem cells (HSC) generally engraft rapidly and completely after myeloablative conditioning. However, with reduced-intensity conditioning (RIC), mixed chimerism and graft failure are more common. Host immune status and HSC number are factors known to affect engraftment after reduced-intensity stem cell transplantation (RIST). In addition, donor T cells within the allograft may also influencethe kinetics of donor engraftment after RIST. To evaluate this, we performed a controlled comparison of engraftment outcomes among 3 groups undergoing RIST, varying by ex vivo T cell depletion (TCD) or in vivo depletion of activated T cells with methotrexate (MTX) to prevent graft-versus-host disease (GVHD). Group I (n = 50) received T cell replete (TCR) peripheral blood stem cells (PBSC) with cyclosporine (CSA) alone for GVHD prophylaxis. Group II (n = 17) received ex vivo TCD PBSC (positive/negative selection with T cell add-back to uniform dose of 1 x 105 CD3+ cells/kg) with CSA alone for GVHD prophylaxis. Group III (n = 31) received TCR PBSC with CSA plus MTX (5 mg/m2 IV x 4 doses) for GVHD prophylaxis. The 3 groups were similarly immunosuppressed from prior therapy before RIST (median absolute lymphocyte counts 330/μL, 260/μL, and 307/μL for Groups I, II, and III, respectively), and received an identical RIC regimen (fludarabine/cyclophosphamide) plus comparable numbers of filgrastim-mobilized PBSC from HLA-matched sibling donors (median 7.9 x 106, 7.6 x 106, and 6.8 x 106 CD34+ cells/kg, respectively; median 3.6 x 108, 1.0 x 105, and 3.2 x 108 CD3+ cells/kg, respectively). Hematopoietic recovery was slowest in Group III, consistent with the myelosuppressive effects of MTX (Table). A greater proportion of patients in Group I achieved complete donor chimerism (≥ 95%) by day +28 than in Groups II or III (P &lt; 0.025), and at day +100, mixed donor chimerism persisted more often in Groups II and III than in Group I patients (P &lt; 0.01). Correspondingly, early (&lt; day +42) occurrence of grade 3–4 acute GVHD, before initiation of planned sequential donor lymphocyte infusions (DLI) in Group II, was more frequent in Group I than in either Groups II or III (p=0.08). Table: Hematopoietic Recovery, Engraftment, and GVHD Group Days to ANC &gt; 500, median (range) Days to plt &gt; 100, median (range) Donor chimerism ≥ 95% Early acute GVHD, grades 3–4 Day +28 Day +100 I 9 (7–13) 15.5 (12-42) 37/44 (84%) 36/38 (95%) 9/50 (18%) II 9 (7–10) 17.5 (11–40) 8/17 (47%) 9/14 (65%) 0/17 (0%) III 14 (7–21) 21.5 (12–85) 23/31 (74%) 21/31 (68%) 2/31 (6%) Thus, the deletion of T cells by either ex vivo TCD or in vivo MTX administration measurably alters the kinetics and degree of donor T cell engraftment after RIST. These observations provide evidence that donor T cells are an independent factor affecting engraftment of allogeneic HSC after RIST by compensating for incomplete host immune ablation. These data also support the hypothesis that a graft-versus-host effect plays a significant role in engraftment after RIST. Manipulation of donor T cells through graft engineering techniques may be a useful strategy to enhance engraftment in the setting of RIST.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2262-2262 ◽  
Author(s):  
Guenther Koehne ◽  
Ekaterina Doubrovina ◽  
Aisha Hasan ◽  
Juliet Barker ◽  
H.R. Castro-Malaspina ◽  
...  

Abstract Abstract 2262 Poster Board II-239 We have developed a novel alternative method of generating donor-derived T-cell lines specific for CMV-pp65 peptides for use in adoptive immunotherapy. In this approach, 138 pentadecapeptides (15-mers), with 11 amino acid overlaps spanning the entire 561 amino acid sequence of the CMV protein pp65 were synthesized and manufactured under conditions ensuring their purity and sterility for application in clinical trials. A pool of the 138 pentadecapeptides is loaded onto monocyte-derived dendritic cells derived from seropositive donors and the same donor's T cells are sensitized under GMP conditions. In our preclinical studies, we have been able to generate CMV-pp65 specific T-cell lines from each seropositive donor tested, irrespective of HLA genotype. During the culture period of 21-28 days, populations of T cells specific for CMV-pp65 selectively expanded 200-300 fold while T cells reactive against major or minor alloantigens were depleted. Currently, 9 patients (pts) with persistent/refractory CMV antigenemia, one pt with CMV pneumonia diagnosed four weeks after treatment, have been treated using cells manufactured by this method on a clinical trial approved by the FDA. One pt was treated on a single patient IND prior to final FDA approval; 8 pts were enrolled onto the clinical trial - 3 pts at a T cell dose of 5×105/kg; 3 pts at dose level II of 1×106/kg and 2 pts at dose level III of 2×106/kg. We report here the results of the first 7 pts, (the 2 latter pts are too early in treatment to be included at time of abstract admission). CMV specific CTLs were generated from HLA-identical unrelated donors (3 pts) or from HLA-identical siblings (6 pts) for 2 pts who underwent non-myeloablative conventional and 7 pts who received myeloablative T-cell depleted allogeneic transplants. Pts were eligible if they had persistent CMV antigenemia despite treatment with antiviral drugs or had toxicities precluding further treatment with antiviral agents. Prior to infusion, the T cells were tested to be CMV specific by cytotoxicity, intracellular Interferon gamma (IFN-g) production and MHC-tetramer staining (if available). Cells were also assayed to establish lack of alloreactivity, microbiological sterility and low endotoxin levels. The cytotoxic T cells demonstrated cytolytic activity against peptide-loaded autologous PHA blasts, but exhibited no cytotoxicity against non-pulsed HLA-matched or pulsed HLA-mismatched target cells. The HLA-restriction and the specific pp65-derived epitopes of the CMV-specific T-cells were characterized prior to the infusion. CMV-specific frequencies of the CD8+ cells measured by intracellular IFN-g or MHC tetramers ranged from 2 -70%. Post infusion, an increase in the absolute lymphocyte count correlated with an increase in CMV-specific T-cell frequencies to levels as high as 12% of CD8+ cells. These persisted for at least 7 months (10% of CD8+ cells) following the infusion. Notably, the same pp65-derived epitopes and their HLA-restrictions, which characterized the pre-infusion CTLs were detected in the pt specimens post infusion. Freshly isolated T cells from the blood of HLA-A*0201 positive pts obtained 3 months post infusion showed significant lysis of CMV infected and peptide-pulsed MRC-5 fibroblasts, while non infected/pulsed fibroblasts and pulsed HLA-mismatched targets were not lysed. Three of the treated pts exhibited an HLA-A*0201 as well as HLA-B*0701 allele. In all three pts, epitope-specific T cells for the HLA-A*0201 restricted NLVPMVATV peptide and the B*0701 restricted RPHERNGFTV peptide were detected and monitored in pre and post infusion T-cell populations. All 7 pts who were treated for persistent CMV antigenemia tolerated treatment well. None developed early signs or symptoms of GvHD at the dose levels tested. Six of the 7 pts cleared CMV viremia by 2-4 weeks following the T-cell infusions. One of the pts died six weeks after the CTL infusion of respiratory failure despite clearing CMV from blood and bronchial aspirates. The one pt who remained viremic following the CTL infusion continued on oral Valgancyclovir and subsequently became CMV antigen negative. The clearance of CMV antigenemia was preceded by an initial spike of CMV antigenemia and/or CMV PCR in all pts. These results from the first 7 pts on this trial indicate that donor T cells sensitized with this pool of synthetic overlapping CMV pp65 15-mers are safe and clear CMV viremia resistant to standard therapy. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document