feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 3894-3894
    Abstract: Background: Vaccines against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been approved rapidly. However, pivotal studies have been conducted in healthy volunteers, while recipients of allogeneic hematopoietic cell transplantations (allo-HCT) may have different dynamics and patterns of response to the vaccine and data in this cohort is lacking. Methods: Here, we examined longitudinal antibody (AB) titers to SARS-CoV-2 vaccination with BNT162b (Comirnaty ®) or mRNA-1273 (Moderna Covid-19 Vaccine ®) in allo-HCT recipients who had undergone allo-HCT & gt;3months (m) ago and in healthy controls (hospital employers). Serial AB titers (prior to (T0); 1m after 1 st dose (T1); 1m (T2), 3m (T3), 6m (T4) post 2 nd dose) were measured with an in-house developed multiplex Antibody CORonavirus Assay (ABCORA) that measures SARS-CoV-2 IgG, IgA, and IgM reactivities against RBD (receptor binding domain), S1 (subunit 1 of the spike protein), S2 (subunit 2 of the spike protein) and N (nucleoprotein), thereby allowing to differentiate immunity after vaccination versus immunity after infection. As neutralization activity correlates well with S1 AB binding, the potency of the AB response was defined as the sum of S1 IgG, IgA and IgM reactivities (cumulative S1 (cS1)). Based on computational methods high neutralization potency was predicted above a cS1 threshold of 17. Results: We enrolled 114 allo-HCT patients (median age 57y (range 18y-74y)) between March 9th 2021 and May 31st 2021 at the University Hospital Zurich, Switzerland. Currently, AB responses at T1, T2, and T3 are available for 99, 95 and 89 patients, respectively. Patients were grouped into those (A) 3-6m post-HCT (T1: n=25 at, T2: n=23, T3: n=20); (B) 6-12m post-HCT (T1: n=13, T2: n=13, T3: n=12); and (C) & gt;12m post-HCT (T1: n=61, T2: n=59, T3: n=57). In addition, AB responses are available for healthy controls (median age 35y (range 23y-64y)) (T1: n=75, T2: n=69, T3: n=48). There were 10 patients and 5 healthy subjects with a reported or detected SARS-CoV-2 infection. There was a statistically significant difference of cS1 AB levels between the 4 groups at T1, T2, and T3 (ANOVA p-values (p) & lt;0.001, respectively, Fig 1) with the lowest AB response in group A (cS1 median value 0.957 at T1, 5.22 at T2, 1.90 at T3) and B (cS1 median value 0.973 at T1, 4.76 at T2, 11.9 at T3) compared to group C (cS1 median value 6.21 at T1, 199 at T2, 76.4 at T3) and healthy controls (cS1 median value 54.9 at T1, 228 at T2, 91.1 at T3). Using a multivariate linear regression analysis adjusted on age and gender, we found that patients in groups A and B had significantly lower cS1 levels than groups C and healthy subjects (p & lt;0.001, p & lt;0.001, p=0.034 of healthy versus groups A, B, C respectively at T2, and p & lt;0.001, p=0.004, p=0.12 at T3), and that preinfected patients had higher cS1 levels at T2 and T3 respectively (p=0.003 and 0.006). The dynamics of the AB response were more diverse in allo-HCT recipients. In a multivariate linear regression analysis (Fig 2) assessing factors associated with humoral immune responses in allo-HCT recipients, we found consistently lower cS1 responses in patients early post-HCT (group A+B (p=0.002)) and higher cS1 levels in those who had been preinfected with SARS-CoV-2 (p=0.012). Patients under immunosuppressive treatment (IST) and those who had relapsed disease post-HCT showed significantly lower cS1 immune responses (p=0.028 and 0.005, respectively). The presence of moderate or severe chronic GVHD was not a statistically significant factor influencing AB levels. This may be explained by (i) the heterogeneity of the condition of chronic GVHD and low patient numbers; (ii) the late time point & gt;12m post-HCT with generally higher AB levels. Consistent with other reports age & gt;65y was also associated with lower cS1 responses (p=0.03). Conclusion: Allo-HCT recipients early post-transplant, those of older age, and those given IST displayed insufficient AB titers to the vaccine. Such knowledge is of critical importance to transplant recipients and their physicians to guide treatment decisions regarding re-vaccination, and social behavior during this pandemic. Monitoring AB development in all allo-HCT recipients and vulnerable patients with other immunocompromising conditions may be crucial to determine those at increased risk for infection and for the timing of booster vaccines. Figure 1 Figure 1. Disclosures Manz: CDR-Life Inc: Consultancy, Current holder of stock options in a privately-held company; University of Zurich: Patents & Royalties: CD117xCD3 TEA.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 75-75
    Abstract: Abstract 75 While non-myeloablative conditioning significantly reduces morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT), the risk of graft rejection increases, because persistent host cells mediate host-vs-graft reactivity. Here we studied the dynamics of engraftment and hematopoietic reconstitution after non-myeloablative radiation (XRT) in a minor antigen-mismatched model with high barriers to engraftment. BALB.K mice received purified hematopoietic stem cells (HSC: cKit+Thy1.1loSca1+Lin−) +/− TC from AKR/J donors. Recipients of pure HSC or HSC+CD8 TC regularly achieved stable mixed chimerism; however, if grafts contained CD4+CD25− TC (CD4con), recipients failed to engraft. This lack of engraftment was associated with BM hypocellularity (median 3.8 vs 13.3 x10^6 cells/2 legs in HSC+CD4con vs HSC recipients, respectively; p=0.0003), and lymphopenia ( 〈 5% vs 〉 40%) at 2 weeks (w) post-HCT. In addition, 2w post-HCT the BM of mice given HSC+CD4con contained increased proportions of CD4 TC which expressed high levels of IFNγ (median 45% of donor vs. 13% of host CD4 TC; p 〈 .001) that exceeded the levels of IFNγ present their spleens or in the BM of control groups (HSC only, wild type [WT], or XRT 〈 10%). A primary determinant of naïve TC immunoreactivity is antigen presentation by dendritic cells (DC). Plasmacytoid DC (CD11c+B220+), which have tolerogenic activity, dominated the BM DC pool in mice given pure HSC at 1–2w post-HCT, but were lacking in recipients of HSC+CD4con. In this latter group, primarily myeloid DC (CD11c+Mac1+) were present that strongly expressed MHCII, CD40, and CD80. This inflammatory profile of myeloid DC was more pronounced in the BM than in the spleens of HSC+CD4con recipients, and IL-12 secretion was measurable in the myeloid DC of these recipients, even without the external endotoxin stimulation which is usually required to detect cytokine production. The IL-12/IFNγ axis is reported to be important in autoimmune pathologies and acute graft rejection of solid organs. To elucidate the mechanism by which IFNγ producing donor CD4 TC suppress engraftment and lymphopoiesis in our system, the progenitor pool in the BM of HSC+/−CD4con recipients was analyzed at 1–2w post-HCT. As expected, the BM recipients of purified HSC had all maturation stages of stem and progenitor cells (HSC: cKit+Sca1+Lin−Flt3−CD34−CD150+ [LT=long-term] /CD150− [ST=short term]; multipotent progenitors, MPP: cKit+Sca1+Lin−Flt3+/−CD34+/−). In contrast, mice given HSC+CD4con lacked MPP, while their population of ST ( 〉 LT) HSC was enlarged. These HSC were not apoptotic (as assessed by annexin V staining), but were cell cycle arrested (assessed by DNA content analysis). To study the functionality of these HSC outside the inflammatory environment, LT+ST HSC from HSC+CD4con recipients were FACS purified at 2w post-HCT and infused into secondary Rag2γc-/- recipients. These adoptively transferred HSC and promptly sustained multilineage hematopoiesis derived from the primary BALB.k host. Our results suggest that interactions between naïve donor CD4 TC and immunocompetent residual host cells, such as DC, create a pro-inflammatory environment involving the IL-12/IFNγ axis. We evaluated several other MHC-identical strain combinations which demonstrated lower levels of cytokine production with improved hematopoiesis compared to the AKR into BALK.K combination. These differences imply that impaired hematopoiesis is due to activation of donor CD4 TC in response to disparate minor alloantigens, and not due to non-specific inflammation. Such immune reactions appear to prevent engraftment of donor HSC and also inhibit the HSC expansion required to restore hematopoiesis early post-HCT. We conclude that HCT with pure HSC can result in superior immune reconstitution and donor chimerism after non-myeloablative conditioning. Pure HSC are immunologically anergic, and do not trigger inflammation in the BM microenvironment. As it is generally believed that TC augment donor HSC engraftment, our results are of broad significance because they reveal critical interactions between donor and host populations after non-myeloablative conditioning. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3545-3545
    Abstract: Abstract 3545 Poster Board III-482 Standing in the line of first defense, the liver is a critical immunocompetent organ. It is armed with lymphocytes, including T cells (TC), natural killer (NK) cells, NK T cells, and a variety of antigen-presenting cells (APC), such as dendritic cells and resident macrophages (Mph), called Kupffer cells. Because it is exposed to large amounts of toxins and antigens, both destructive and harmless, liver immunity must provide immunogenic and tolerogenic mechanisms. Moreover, as the organ of fetal blood production the liver can, if required, resume its hematopoietic function. Here, we studied the role of the liver as a hematopoietic and lymphatic organ after hematopoietic cell transplantation (HCT). Lethally irradiated BALB.K and BALB.B mice were given MHC-matched, FACS purified hematopoietic stem cells (HSC; cKit+Sca1+Thy1.1loLin-) from AKR/J and C57BL/6 donors, respectively, alone or supplemented with 10∧7 splenocytes (SP) for GVHD induction. Mononuclear cells (MNC) were Ficoll-separated from flushed livers 1 to 6 weeks (w) post transplant (pTX) and FACS analyzed. In recipients of TC-containing grafts, the liver was a major target organ of acute graft-vs-host disease (GVHD) with prominent donor lymphocyte expansion causing destruction of the hepatic portal morphology. Rare HSC-derived cells were observed in the livers. In contrast, mice given purified HSC showed no clinical or histological signs of GVHD, yet early pTX a high proportion of donor HSC-derived MNC was observed within the livers, comprising ∼75% of the MNC at 2w. Phenotype analysis revealed that these HSC-derived MNC were primarily NK cells (DX5+CD122+) or Mph (Mac1+F4/80+). In fact, amongst all nucleated cells, NK cells represented 〉 10% and were mixed donor/host type. Interestingly, the Mph were all donor derived. This observation of over-representation by cells of innate immunity (including NK cells and Mph) in livers of recipients of HSC alone led us to hypothesize that these cells might exert protective functions against increased amounts of pathogens and toxins entering the circulation from irradiation-damaged intestines. Thus, to suppress donor Mph reconstitution pTX, silica was injected intraperitoneally on d-1, and every 3d thereafter. All recipients of HSC alone recovered rapidly after irradiation (d5-7), while at this time point recipients of HSC plus silica showed severe weight loss, hunched posture, ruffled fur, diarrhea, with 〈 50% (7/15) survival. These survivors clinically stabilized around d12, suggesting that the intestines recovered from injury. To test if the presence of the HSC derived NK cells and APC could contribute to host protection from GVHD, a lethal dose of SP (10∧7) was injected simultaneously with HSC, or with a delay of 7d or 9d. All mice given SP on d0 died within 9d and 3/5 of those receiving SP on d7 died by d12. However, all mice given SP on d9 recovered fully and showed no signs of GVHD, despite the lymphopenic host environment that usually promotes homeostatic expansion of mature donor TC. In conclusion, the role of the liver as an immunologically active organ after ‘conventional’ HCT is often masked by donor TC expansion with subsequent GVHD. Here, we provide evidence that if grafts are devoid of mature lymphoid cells, innate immunity recovers rapidly, and in fact exceeds unmanipulated controls. Donor Mph may protect the host from pathogens and endotoxemia. Moreover, they may neutralize activated donor TC and thereby mediate tolerance between donor and host. Likewise, the elevated proportion of donor and host NK cells, which is lacking in GVHD affected mice, suggest another beneficial mechanism of protection, as NK cells have been reported to be capable of reducing GVHD. Immunohistochemical studies for a better quantitative assessment of resident immune cells in the liver pTX are underway. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 1896-1896
    Abstract: Abstract 1896 The curative potential of MHC-matched allogeneic bone marrow transplantation (BMT) is in part due to immunologic graft-versus-tumor (GvT) reactions mediated by donor T cells that recognize host minor histocompatibility antigens. Immunization with leukemia-associated antigens, such as Wilm's Tumor 1 (WT1) peptides, induces a T cell population that is tumor antigen specific. We determined whether BMT combined with immunotherapy using WT1 peptide vaccination of donors induced more potent anti-tumor activity when combined with allotransplantation. WT1 peptide vaccinations of healthy syngeneic or allogeneic donor mice with a 9-mer WT1 peptide (amino acids 126–134, the WT1 9-mer which has the highest binding affinity for H-2Db) and Incomplete Freund's Adjuvant induced CD8+ T cells that were specifically reactive to WT1-expressing FBL3 leukemia cells. We found that compared to vaccination with IFA alone, four weekly WT1 vaccinations induced an increased percentage of WT1-tetramer+CD8 T-cells (0.15% vs. 1%) in the peripheral blood 28 days following the first vaccination (Figure A *p 〈 .001). CD8 T-cells producing IFN-γ+ after co-culture with tumor cells were similarly increased (0.11% vs. 13.6%) at this timepoint (Figure B *p 〈 .001). They were CD44hi suggesting a memory phenotype, specifically reactive to WT1-expressing tumor (FBL3 and not H11), and increased in a vaccination dose-dependent fashion (Figure A and B). Four weekly WT1 vaccinations prevented tumor growth in donors following intravenous leukemia challenge. In contrast, in tumor-bearing mice, WT1 vaccinations failed to induce WT1-tetramer+ or IFN-γ+ CD8 T-cells and were ineffective as a therapeutic vaccine based on intensity of bioluminescence from luciferase-labeled FBL3 leukemia and mortality. BMT from WT1 vaccinated MHC-matched donors including LP/J and C3H.SW, but not C57BL/6 syngeneic donors, into C57BL/6 recipient tumor-bearing mice was effective as a therapeutic maneuver and resulted in eradication of luciferase-labeled FBL3 leukemia and survival of 70–90% of mice. Interestingly, the transfer of total CD8+ T cells from immunized donors was more effective than the transfer of WT1-tetramer+CD8+ T cells, likely as a result of alloreactive and tumor-antigen reactive T cells contained with the donor total CD8+ T cells. Total and tetramer+CD8+ T cells required CD4+ T cell help for maximal anti-tumor activity, which was equivalent in efficacy from immunized or unimmunized CD4+ T cell donors. Total CD4+ T cells, alone, from immunized donors provided no anti-tumor activity. The infused donor LP/J or C3H.SW CD8+ T cells collected from cured C57BL/6 recipients, were highly reactive against WT1-expressing FBL3 leukemia cells (14% IFN-γ+) compared to non-WT1-expressing H11 leukemia cells (5% IFN-γ+). The circulating, WT1-tetramer+CD8+ T cell population expanded in cured recipients, peaking at 3.5% on day 50 and contracting through day 100 post-BMT to 0.56%. These findings show that peptide vaccination of donor mice with a tumor antigen dramatically enhances GvT activity and is synergistic with allogeneic BMT. This novel and broadly applicable approach, using leukemia-associated antigen immunization to enhance GvT by creating an “educated” donor T cell graft for allogeneic transplantation of patients with acute myeloid leukemia and myelodysplastic syndrome, is currently being translated to a Phase 1 clinical trial at our institution. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3732-3732
    Abstract: Abstract 3732 Infections due to impaired immune function are a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Adoptively transferred donor (do) T cells (TC) are thought to provide protective immunity, as TC-depleted grafts are associated with increased viral infections post HCT (pTX). TC-replete grafts, however, require immunosuppression to prevent GVHD, and can damage lymphoid organs causing additional immune dysfunction. Using an MHC-matched, minor antigen (miAg) mismatched GVHD model, we compared immune responses to murine cytomegalovirus (MCMV) in lethally irradiated BALB.B recipients infused with purified hematopoietic stem cells (HSC; cKit+Sca1+Thy1.1loLin−) or HSC+TC (subsets) from congenic C57BL/6 (B6) donors which could be distinguished based upon CD45 alleles. Hosts were infected with sublethal doses of MCMV at 2 or 8 weeks (w) pTX. 2w after infection, lymphoid organs were harvested to assess the immune function of: 1) transferred doTC from uninfected and pre-immunized donors, 2) HSC-derived doTC, and 3) residual host TC. Anti-MCMV CD8 TC were measured by M45 tetramer staining and an MCMV ELISPOT assay was used to measure IFNg production of FACS-separated doTC-, doHSC-, and host-derived spleen populations when exposed to the viral peptide. Mice given TC-replete allografts and infected at 2w pTX mounted significantly lower responses than control congenic- or HSC only recipients. In the latter group reactivity originated from residual host cells. Transplantation of TC from immunized donors did not improve the response of doTC transplanted into miAg-mismatched hosts. However, when HSC were supplemented with memory (CD62L+/ & minus;CD44+) CD8 TC from immunized donors, recipients of memory, but not of naive CD8 TC (CD62L+CD44−), mounted strong anti-MCMV responses in both assays. At 8w pTX recipients of pure HSC were mixed TC chimeras, while mice given HSC+TC were full donor chimeras with the majority of TC originating from transferred doTC. When mice were infected 8w pTX, anti-MCMV reactivity of CD8 TC was significantly greater in recipients of pure HSC, as compared to weak responses seen in recipients of HSC+TC. In recipients of pure HSC, both donor and host populations demonstrated strong reactivity, while mice given HSC+TC had weak responses of both doTC-, and doHSC-derived TC (Fig.1). When recipients of HSC alone or HSC + total TC (ToTC), naive CD8, memory CD8 or M45 tetramer+ CD8 TC were infected with a lethal dose of MCMV at 2w pTX, 50% of ToTC and 20% of HSC recipients died before day 40 pTX, while all mice given any of the CD8 subsets were protected from infection. 6w post MCMV infection organs of survivors were analyzed. Hepatic CD8 cells contained a median of 〈 0.05% M45 tetramer reactive cells in ToTC-, 〈 0.5% in HSC only- and naive CD8-, and 0.65% in memory CD8 recipients (p=0.03 compared to ToTC group). Strikingly, in mice given as few as 7,000 M45 tetramer+ cells, the proportion of M45-reactive cells reached a median 8.7% in the liver, and 4.3% in the spleen. In mice given ToTC, 〉 90% of TC were doTC-derived with an effector memory phenotype. In contrast, recipients of naive, memory, or tetramer-sorted CD8 cells had TC originating from expanded doTC, doHSC, or residual host. Nascent doHSC-derived lymphopoiesis sustained a robust pool of naive TC - a sign of healthy immune reconstitution. Thus, our key findings are: 1) mature doTC do not maintain their full anti-viral potential when transplanted into miAg-disparate mice, even when obtained from pre-immunized donors; 2) residual host cells contribute substantially to protective immunity early pTX, but are eradicated by conventional TC-replete grafts; 3) HSC-derived TC arising in a healthy host are superior to those that develop and undergo selection in a GVHD-affected lymphoid system; and 4) low numbers of selected cell subsets, such as tetramer-sorted CMV-specific CD8 TC, expand dramatically in a lymphopenic environment and provide functional protection against infections. Our results challenge the conventional assumption that doTC in a hematopoietic allograft are required for optimal regeneration of the immune system. Rather, our studies suggest that long-term lymphoid function will greatly benefit from rigorous TC-depletion of the graft, and avoidance of GVHD. Moreover, co-transfer of small numbers of highly selected TC clones can provide effective antigen-specific protection. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 654-654
    Abstract: The bone marrow (BM) is a complex microsystem to support lifelong blood production. At steady-state most hematopoietic stem cells (HSC) are quiescent. However, in situations of increased demand, their activation is triggered by an array of signals, such as cytokines. Following hematopoietic cell transplantation (HCT) in the phase of hematopoietic reconstitution maximal blood production is needed. In an HCT donor HSC are given together with immune cells in the belief that T cells support HSC engraftment and regeneration of the blood system. Yet, states of immune mediated BM insufficiency, hypoplasia, and cytopenias are often observed. Moreover, with increased use of reduced intensity conditioning (RIC) engraftment failure has reemerged as a serious problem. Here, we studied the effects of distinct T cell subsets on hematopoietic reconstitution following HCT; specifically, we examined how conventional CD4+CD25- T cells (CD4conv) vs regulatory T cells (CD4+CD25+, Treg) modify the BM environment and influence donor-HSC activity and engraftment. We used minor-mismatched mouse models, non-myeloablative total body irradiation (TBI) conditioning and transplantations of purified HSC (KTLS; cKit+Thy1.1loLin–Sca-1+) plus selected T cell subsets. Recipients of HSC, HSC+Treg or HSC+CD8+ - but not HSC+CD4conv- demonstrated prompt donor engraftment with mixed chimerism in all lineages. Transplantation of HSC+Treg resulted in significantly faster lymphocyte recovery and higher levels of donor chimerism compared with recipients of HSC, HSC+CD8+ or HSC+CD4conv. Particularly B-cell regeneration was markedly higher in HSC+Treg-recipients compared with all other groups. In contrast, (B-) lymphopoiesis was severely impaired in recipients of HSC+CD4conv; when lymphocytes recovered eventually they were of host type. Moreover, in BM and spleens of HSC+CD4conv recipients pronounced hypocellularity was observed. This suppression of hematopoiesis was due to IFNg secretion of donor CD4conv cells, which were activated by dendritic cells via IL-12. High cytokine levels (of both IL-12 and IFNg) were only detectable in the BM (and not the spleen) of HSC+CD4conv recipients, where they resulted in an arrest of early hematopoiesis at the stage of short-term HSC and decreased cell-cycle activity within the progenitor compartment. As a consequence more mature multipotent progenitors were lacking. The key role of IFNg in halting hematopoietic maturation was confirmed by using CD4conv cells from IFNg-/- mice, which had no suppressive effects on BM cellularity and maturation of blood cells; rather, recipients of HSC+IFNg-/-CD4conv cells had equivalent cell numbers and subset distributions as mice given HSC alone. We hypothesized that differences of hematopoietic regeneration and donor engraftment relate to cell cycle activity of HSC in presence of CD4conv vs Tregs. To study the influence of these CD4-subsets on HSC cycling in more detail FACS-purified Treg vs CD4conv cells were infused into congenic mice following low-dose TBI-stimulation. In fact, on d+8 post-infusion HSC in the BM of Treg-recipients had increased cell-cycling activity in long-term-HSC and multipotent-progenitor fractions compared with mice given CD4conv cells or radiation only. These data lead us to speculate that Tregs promote, directly or indirectly, HSC proliferation; in the context of an allogeneic HCT this increased cycling of host HSC my open-up the niche space required to allow donor HSC to engraft. In contrast, CD4convinhibited HSC cycling activity, resulting in BM hypoplasia and cytopenias; at the same time donor HSC engraftment was impaired due to HSC-niche occupation by quiescent host HSC. Our findings underscore the critical role of T cells in regulating hematopoiesis under physiologic conditions and even more following allogeneic HCT. While donor T cells are generally believed to improve regeneration of the blood post-HCT and to be required to overcome host barriers, our data suggest Treg facilitate engraftment and hematopoiesis by increasing HSC cycling-activity and thereby making marrow sites available. CD4conv appear to have the opposite effect, resulting in decreased HSC proliferation and maturation - thus occupation of HSC niches. Our studies are of particular relevance to allogeneic HCT settings using RIC, where host HSC persist and grafts can be rejected by residual host immune cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3413-3413
    Abstract: Hematopoietic cell transplant (HCT) recipients are at increased risk for infections. Staphylococcus aureus (SA) colonizes 20-50% of healthy individuals and is a risk factor for subsequent invasive SA infections. Colonization rates in patients undergoing allogeneic HCT and clinical relevance for the time of aplasia and severely reduced immune function following HCT are not known. Only some retrospective data on methicillin-resistant SA infection rates are available. In this study, we prospectively assessed the prevalence of SA colonization in 110 consecutive patients before and during allo-HCT in a single-center observational study from June 2013 to January 2016. All patients undergoing allo-HCT were screened for nasal, pharyngeal and inguinal SA colonization weekly beginning at the time of admission to the transplant unit until neutrophil recovery. After swabs for the initial SA screening were taken all patients were put on oral gentamicin and vancomycin for gut decontamination until neutrophils had recovered. Quantitative stool analyses were performed weekly. In case of fever or increased of inflammatory laboratory parameters (C-reactive protein) blood cultures were drawn. In our cohort we found a SA prevalence of 14.5% (16/110 patients) at the time of admission to the transplant unit. All SA strains detected were sensitive to methicillin. Most patients colonized with SA in the nose (13/16), while pharyngeal and inguinal colonization was found less frequently (n=5 and n=6, respectively). Patients aged 60-67 years (n=14) showed the highest SA carrier rate (5/14, 36%, RR=1.36, p=0.02). There was no correlation between SA colonization and sex, underlying disease or chemotherapeutic pretreatment. Prior systemic antibiotic treatments using SA effective drugs within six months before admission to the transplant unit did not have relevant impact on SA prevalence at the time of screening. Within the group of the 16 SA-positive patients there were 2 patients (12.5%) who had received oral antibiotic gut decontamination (vancomycin and /or gentamicin) within twelve weeks prior to admission (during induction chemotherapy). In the SA negative group a similar proportion of patients had received oral gut decontamination (12/94; 12.8%). Despite the severe immunosuppression and skin and mucosal lesions incl. indwelling catheters no systemic SA infections (including bacteremia) were found during hospitalization in any of the HCT patients. All SA positive patients became SA negative within three weeks. These observations imply that decolonization is achieved by the consistent oral gut decontamination that all patients received in conjunction with the antibacterial soaps used. In conclusion, the SA colonization prevalence in our cohort of patients undergoing allogeneic HCT was 14.5% which is lower than described previously in the literature. Of note, our cohort did not comprise patients with MRSA. Here, we demonstrate in a prospective study that oral gut decontamination with vancomycin and gentamicin in addition to strict hygiene measures resulted in eradication of SA colonization in all 16 colonized patients within three weeks. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 4030-4030
    Abstract: Abstract 4030 Impaired immune function is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Adoptively transferred donor T cells (TC) mediate graft-versus-host disease (GVHD), and make the use of pharmacological immunosuppression necessary, yet are believed to protect against infections post-HCT (pTX). In an MHC-matched, minor antigen mismatched mouse model, we studied the impact of allogeneic doTC on GVH-related damage of lymphoid organs, their recovery and immune function pTX. Lethally irradiated BALB.B mice received purified hematopoietic stem cells (HSC; cKit+Sca1+Thy1.1loLin−) +/− TC from C57BL/6 (B6) donors. Recipient lymphoid organs, marrow and livers were analyzed at multiple time points. Mice given HSC+TC promptly became full donor chimeras in all lineages and developed moderate GVHD. HSC recipients retained some residual host cells, and had no signs of GVHD. By tetramer analysis and ELISPOT assay, we showed that in pure HSC recipients early pTX residual host TC protected against murine cytomegalovirus infection. Once a nascent donor HSC-derived TC pool was established, these TC reacted robustly against the virus. In contrast, transferred mature allogeneic TC proliferated and expanded in HSC+TC recipients, but did not respond or protect against the virus. To better understand why MHC-matched mature donor TC did not react against this pathogen, even when derived from pre-immunized donors, we studied the lymphatic microenvironment, hypothesizing that only highly organized interactions between cellular and humoral immunity, antigen-presenting and innate immune cells provide full immune function. Comparing tissues of mice given pure HSC +/− TC we found that: (i) lethal radiation caused marked reduction in lymphoid organ cellularity at 2 wks pTX in all transplanted groups. While the cellularity increased rapidly in HSC recipients, severe hypocellularity persisted in HSC+TC recipients, was associated with significantly decreased organ size (lymph nodes [LN] and thymuses) and marked disruption of the histological architecture; (ii) TC phenotypes present in reconstituted mice comprised a mixture of naïve (CD62L+CD44−), central memory (CM; CD62L+CD44+) and effector memory (EM; CD62L−CD44+) cells in HSC recipients, but were largely EM and rarely naïve TC in HSC+TC recipients; (iii) at ∼3 wks pTX donor B cells (BC) dominated lymphoid tissues in HSC recipients, but were absent in HSC+ToTC recipients; (iv) NK cells regenerated promptly and steadily in HSC recipients, constituting up to 20% of all lymphoid cells in the liver at 3 wks pTX, while in HSC+TC recipients they incessantly decreased to near-complete absence; (v) in mesenteric (m) LN of HSC-, but not HSC+TC- recipients, CD4+CD8+CD3− TC were observed, derived from donor HSC. This latter population resembled immature thymic TC, suggesting that peripheral lymphoid organs participate in lymphoid reconstitution in adult recipients; (vi) at 2 wks pTX we noted only in HSC, but not HSC+ToTC recipients persisting and regenerating innate lymphoid cells (ILC). No ‘classical' lymphoid tissue inducer cells, as described in embryogenesis of lymphoid organs, but cells with features of ILC were identified. Donor HSC-derived NKp46+RORgt+CD3− NK-like cells and IL-17A-secreting residual host CD4 TC were present, especially in livers and mLN. Cells that secreted IL-17A only and IL-17A+IFNγ were identified. IL-17A was not detected in recipients of HSC+ToTC, rather their donor TC were characterized by high IFNγ levels. Our studies show that, paradoxically, pure HSC grafts have advantages over TC-replete grafts with regard to immune function pTX. We hypothesize this superiority may be due to the presence of ILC that participate in the repair of lymphoid organs, and the whole spectrum of immune cells, including TC, BC, innate and immature cells emerging in HSC recipients. An intact microenvironment appears to be critical for optimal activation of TC to respond to pathogens. Moreover, our studies imply that donor TC cannot exert their protective immune function, when rapidly expanding alloreactive TC prevent the development of other lymphoid and innate immune cell, and disrupt the lymphoid tissue architecture. Controlling the negative effects of mature donor TC remains a challenge, and requires better understanding of the complexity of functional immune reconstitution pTX. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 3528-3528
    Abstract: Abstract 3528 Poster Board III-465 In the last decade the field of allogeneic hematopoietic cell transplantation has made significant strides in the reduction of morbidity and mortality using non-myeloablative conditioning. However, with reduced conditioning intensity the risk of allograft rejection has increased due to the persistence of host cells that mediate host-vs-graft alloreactivity or that occupy niches in the host hematopoietic microenvironment. Here we studied hematopoietic reconstitution and chimerism after non-myeloablative total body irradiation (TBI: 4 Gy [sublethal]; 7 Gy [lethal] ) or total lymphoid irradiation (TLI: 17×240 cGy) in a MHC-matched mouse model with known high resistance to engraftment. BALB.K (H-2K) hosts received FACS-purified hematopoietic stem cells (HSC: cKIT+Thy1.1loSca1+Lin-) alone or supplemented with T cells (TC) from AKR/J (H-2K) donors. Peripheral blood (PB), bone marrow (BM), and spleen (SP) were analyzed 2 and 4 weeks (w) post transplant (pTX), and PB chimerism was followed beyond day (d) 100. Mice conditioned with lethal TBI survived, when grafts contained HSC only, and became mixed chimeric for TC, but converted to primarily donor type for B cells and the myeloid lineage. When grafts were supplemented with TC, chimerism converted to full donor type, but hosts developed fulminant acute GVHD. In contrast, sublethal TBI resulted in only mild symptoms of GVHD and mice recovered rapidly. Recipients of HSC alone or HSC+CD8 TC became mixed chimeras in all lineages, even long-term ( 〉 100d). However, mice given grafts of either HSC with total TC, or HSC+CD4 TC remained host type for T and B cells, and achieved only low levels of donor engraftment in the myeloid lineage. These findings suggested that the addition of peripheral donor CD4 TC resulted in powerful lymphoid suppression as well as retardation of myeloid engraftment. Indeed at 2w pTX recipients of HSC+CD4 TC displayed severe lymphopenia ( 〈 5% of live cells [vs 〉 40% in recipients of HSC alone]) and B cell reconstitution was the most severely affected (6% vs 75% of lymphocytes). Graft suppression was also evidenced by absolute cytopenia as mice given HSC+CD4 TC vs HSC alone, had reduced cellularity in BM (median 3.8×10∧6 [n=7] vs 13.3 ×10∧6 cells [n=4]; p=0.0003) and spleen (median 4.4×10∧6 vs 14×10∧6 cells; p=0.03). Suppression of hematopoiesis by CD4 TC grafts was accompanied by an expansion of donor and host CD4 TC, each comprising ∼6% of BM cells as compared with 〈 1% in groups that received pure HSC or were only irradiated sublethally. PMA-stimulated donor, but not host CD4 TC secreted high levels of IFNγ (30-50%) in the BM. This IFNγ expression far exceeded the levels of IFNγ measured in CD4 TC of the spleen from treated (HSC+CD4) mice as well as in CD4 TC of BM or spleen from transplanted, irradiated or unmanipulated control mice (3-15%). The proportion of NK cells in the BM was also substantially increased in recipients of HSC+CD4 TC vs recipients of HSC only or sublethal TBI controls (median 14% vs 〈 1%), as were the absolute NK cell numbers (6.3×10∧5 vs 〈 0.4 ×10∧5 cells). NK cells were derived from both donor and host in recipients of HSC, but solely of host type in recipients of HSC+CD4 TC. Findings similar to those observed in the 4 Gy TBI-treated mice given donor CD4 TC with their graft were noted when mice underwent non-myeloablative TLI for conditioning: while recipients of HSC alone were mixed chimeras in their lymphoid and myeloid lineages [n=4], mice that received HSC supplemented with splenocytes or total TC failed to engraft with T and B cells, had marginal myeloid engraftment [n=12] , but had increased proportions of host NK cells in the PB. In conclusion: Our data show in the setting of non-myeloablative conditioning transplantation of purified HSC alone or with CD8 TC results in superior immune reconstitution and donor chimerism as compared to grafts of HSC+CD4 or HSC+total TC. These results are of broad significance as it is generally believed that TC augment donor HSC engraftment. The way in which CD4 TC retard engraftment appears to be by activation of inflammation within the BM and expansion of host lymphoid populations that may mediate resistance. IFNγ, which is a known regulator of innate and acquired immune responses, may be central in activating host CD4 TC and enhancing NK cell mediated rejection of the graft and/or suppression of donor hematopoiesis. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 5425-5425
    Abstract: Graft-vs-host disease (GVHD) remains a major complication of allogeneic hematopoietic cell transplantation (HCT). Acute GVHD results from activated donor T cells that infiltrate and damage target organs, producing an inflammatory state. In contrast, the pathophysiology of chronic graft-vs-host disease (cGVHD) remains poorly understood. cGVHD can follow acute GVHD or emerge de novo ( 〉 d+100). The clinical picture varies and manifestations can resemble autoimmune disorders. Because IL-17 has emerged as a principal cytokine involved in autoimmunity, Th17 cells have attracted much attention in the transplant community. While IFNg-producing Th1 cells appear to drive acute GVHD, the role of Th17 cells in the pathophysiology of GVHD has not been fully clarified. Here, we used an established minor-antigen disparate mouse model of acute and chronic GVHD to examine the emergence of IL-17+CD4+ Th17 cells post-HCT. Lethally irradiated BALB.B mice received pure hematopoietic stem cells (HSC; cKIT+Thy1.1loSca1+Lin-) or HSC plus splenic T cells from C57BL/6 donors (HSC: GFP; TC: CD45.1+). At several time points lymphoid and GVHD target organs were analyzed for donor T cell infiltration and T cell IL-17 expression. In this model recipients of HSC + T cells developed acute GVHD with intestinal involvement (diarrhea, weight loss) and a mortality of ∼30%, while mice given pure HSC remained healthy. Survivors stabilized around d45, but developed clinically evident chronic GVHD after 6-12 mo manifested by sclerodermatous skin excoriations and liver fibrosis/cirrhosis. Donor T cell infiltration of tissues (spleen, lymph nodes (LN), liver, intestines) was high at 2 and 4 wks post-HCT, but there was no detectable IL-17 production by CD4 cells during acute GVHD. The degree of donor T cell infiltration decreased (as acute GVHD improved) in these tissues. However, at 2 mo post-HCT higher percentages CD4+IL-17+ cells were observed, first in intestines and mesenteric LN, followed by liver and skin. At all time points post-HCT proportions of Th17 cells were higher in HCT recipients (of HSC +/- T cells) as compared to normal wild-type (WT) tissues. To summarize, our key findings are: (i) In our model acute GVHD was driven by adoptively transferred mature (CD4+) T cells that acquired a Th1 phenotype, whereas IL-17 producing donor cells were not detectable during this period. IFNg and T-bet are negative regulators of RORgT, the master regulator of Th17. Thus, this observation is consistent with the idea that in the presence of donor Th1 cells the development of Th17 cells is suppressed. (ii) The effect of Th1-related suppression of Th17 persisted beyond the acute phase: recipients of T-cell replete grafts that survived acute GVHD but later developed chronic GVHD did not demonstrate increased CD4+IL-17+ cells. In these mice, organ-infiltrating donor T cells were primarily adoptively transferred T cells, supporting the postulation that no plasticity exists between committed Th1 and Th17 cells. (iii) Signs of chronic GVHD were observed in animals that had not suffered from severe acute GVHD. In particular, in groups without acute GVHD we observed CD4+IL-17+ cells starting at 2 mo, peaking around 6 mo and which stabilized 〉 1 yr post-HCT. In spleen and peripheral LN of these mice only low levels of CD4+IL-17+ cells were detectable, but their proportion was high in GVHD target organs (liver, intestines, skin). The susceptibility of organs appeared to change post-HCT with high proportions of CD4+IL-17+ cells in the intestines at 2 mo post-HCT that decreased over time. In contrast, CD4+IL-17+ cells in the liver increased later in the time course (Figures). (iv) A centrally important observation was that CD4+IL-17+ cells primarily originated from donor HSC, even in recipients of mature donor T cells. Likewise, recipients of pure HSC showed increasing proportions of Th17 cells over time, and could also manifest signs of cGVHD. From our model we conclude that IL-17 does not contribute to acute inflammatory GVHD. However, IL-17 can be involved in an alternative pathophysiologic mode of chronic GVHD development in the absence of acute inflammation. Since CD4+IL-17+ cells derive from donor HSC and undergo maturation in the host this form of GVHD is delayed, and the emergence and activity of these cells appears to constitute a true autoimmune phenomenon. Our novel hypothesis may explain parts of the complex and obscure pathophysiology of chronic GVHD. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages