Treatment with Tyrosine Kinase Inhibitors May Impair the Potential Curative Effect of Allogeneic Stem Cell Transplantation.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 857-857
Author(s):  
Inge Jedema ◽  
Linda van Dreunen ◽  
Roelof Willemze ◽  
J.H. Frederik Falkenburg

Abstract Abstract 857 Tyrosine kinase inhibitors (TKI) like imatinib and dasatinib are the current treatment of choice for patients with chronic myeloid leukemia (CML). Although most patients enter a complete remission during treatment, cure of the disease is usually not achieved since recurrence of the disease is seen in the majority of patients upon discontinuation of the treatment, indicating that the leukemic stem cell is not efficiently targeted. Furthermore, in accelerated phase and blast crisis of CML TKI treatment only results in temporary control of the disease. In these situations allogeneic stem cell transplantation (allo-SCT) and application of donor T cells may be the only curative treatment. Besides the direct anti-leukemic effect of allo-SCT, alloreactive T cells recognizing CML (progenitor) cells, and the formation of immunological memory may lead to effective lifelong immune surveillance. Therefore, we investigated whether the leukemic cells persisting during TKI treatment are susceptible targets for the anti-leukemic effect mediated by donor T cells after allo-SCT and whether continuous TKI treatment may have an additive effect during the immunological intervention. To investigate the anti-leukemic effect of the two strategies, CD34+ positive CML cells were isolated from bone marrow, and labeled with the fluorescent dyes CFSE or PKH to allow monitoring of single cell proliferation. CML cells were exposed to imatinib (1-100μM) or dasatinib (0.01-50nM), and/or to CD8+ alloreactive cytotoxic T lymphocyte (CTL) clones in the presence of proliferation-inducing cytokines. The number, phenotype, and proliferative status of the CML cells persisting after single and combined interventions were measured by quantitative flowcytometric analysis. In the absence of therapeutic interventions the majority of CD34+ CML cells entered proliferation. However, a small population of CD34+ CML stem cells residing in the non-dividing peak could be identified despite the addition of cytokines. Addition of imatinib or dasatinib resulted in efficient dose-dependent induction of cell death of the leukemic cells (99% lysis by 25μM imatinib or 10nM dasatinib). However, the population of quiescent CD34+ CML stem cells was not affected. Moreover, the number of cells present in the non-dividing population increased 2-fold compared to the non-treated controls at the highest TKI concentrations, indicating additional growth arrest of a population of proliferating CML precursor cells. We next tested the capacity of different HLA-A2-restricted CD8+ CTL clones to kill non-treated or imatinib or dasatinib treated CML cells. Whereas the proliferating CD34+ CML precursors were efficiently lysed, the population of quiescent stem cells was capable of withstanding CTL exposure. Detailed phenotypic analysis revealed significant downregulation of HLA-A2 and the adhesion molecules CD49d and CD58 on these quiescent cells, probably resulting in the impaired ability of these target cells to form a high avidity interaction with the T cells. The increased population of non-dividing cells as a result of the TKI pretreatment showed similar resistance to T cell induced cell death, indicating that TKI treatment may even diminish the anti-leukemic effect of allo-SCT. In the absence of therapeutic control, as mimicked by the removal of T cells and TKI from the cultures, outgrowth of the leukemic cells re-occurred, illustrating their capacity to give rise to a relapse of the disease. Next, we analyzed the effect of TKI treatment on T cell survival and functionality. Whereas resting primary T cells were insensitive to TKI treatment, T cells activated by either polyclonal stimulation with anti-CD3/CD28 beads or stimulation with allogeneic stimulator cells died after TKI exposure at similar concentrations as the leukemic cells. In conclusion, TKI treatment results in selection of a population of quiescent leukemic stem cells showing cross-resistance to CTL-induced cell death, most likely due to their inability to form a high avidity interaction. Moreover, T cells actively participating in the anti-leukemic immune response after allo-SCT are suppressed by TKI. These data indicate that continuous TKI treatment may potentially hamper the curative effect of allo-SCT. Disclosures: No relevant conflicts of interest to declare.

2006 ◽  
Vol 6 ◽  
pp. 246-253 ◽  
Author(s):  
Elizabeth Hexner

Much attention has focused on the immune recovery of donor T cells following hematopoietic stem cell transplantation (HSCT). Termed immune reconstitution, a better understanding of the dynamics of the functional recovery of immune cells following HSCT has important implications both for fighting infections and, in the allogeneic setting, for providing antitumor activity while controlling graft-vs.-host disease (GVHD). The immune cells involved in immune reconstitution include antigen-presenting cells, B lymphocytes, natural killer cells, and, in particular, T lymphocytes, the immune cell that will be the subject of this review. In addition, T cells can play an important role in the process of engraftment of hematopoietic stem cells. The evidence for a T cell tropic effect on hematopoietic engraftment is both direct and indirect, and comes from the clinic as well as the research lab. Animal models have provided useful clues, but the molecular mechanisms that govern the interaction between donor stem cells, donor T cells, the host immune system, and the stem cell niche remain obscure. This review will describe the current published clinical and basic evidence related to T cells and stem cell engraftment, and will identify future directions for translational research in this area.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3173-3173
Author(s):  
Yukio Kondo ◽  
Xingmin Feng ◽  
Yoshihisa Kumano ◽  
Shinji Nakao

Abstract Aberrantly expressed self-antigens in leukemic cells serve as leukemia-associated-antigens (LAAs) of leukemia reactive T cells. Although such self-antigen-derived LAAs potentially induce high avidity CTLs in patients with leukemia, these CTLs usually do not persist due to apoptosis upon encountering leukemic cells. In allogeneic stem cell transplant (allo-SCT) recipients with leukemia, residual leukemic cell may sensitize donor-derived T cells by LAA in vivo and induce high avidity CTLs specific to leukemic cells. Cyclin-dependent kinase 2 (CDK2) is a cell cycle regulator protein that is aberrantly expressed in AML, MDS, ALL and MCL cells. We previously reported that CDK2-derived nonamer peptides (CDK2158: TYTHEVVTL, CDK2167: WYRQPEILL, CDK2178: KYYSTAVDI) avidly bound to HLA-A24 molecule to elicit each peptide-specific CTL from peripheral blood mononuclear cells (PBMCs) of healthy individuals (Blood. 106 (11): a3103. 2005). When we generated CDK2158-specific CD8+ T cells from an HLA identical sibling donor of a patient with AML by stimulating donor PBMCs with CDK2158-coated HLA-A24-transfected T2, CDK2158-specific CD8+ T cells preferentially killed the recipient AML cells which aberrantly expressed CDK2 proteins. The percentages of specific lysis in the 51Cr-release assay at an E/T ratio of 40 were 36.8% for AML cells and 24.8% for autologous PBMCs. To determine if CDK2-specific CD8+ T cells are present in PBMCs from allo-SCT patients, we studied 14 patients possessing HLA-A24 [6 patients with AML (2 AML-M0, 3 AML-M2, 1 AML with multilinage dysplasia), 1 with MDS, 1 with CML, 2 with ALL, 2 with MCL and 2 with RCC] using CDK2158/A24 and CDK2178/A24 multimers. Cryopreserved PBMCs obtained before and 4–73 months after allo-SCT were assayed for multimer staining. The source of graft was unrelated BM in 4, related BM in 2, related PBMC in 3 and CB in 5 patients. All CB and 1 BM graft were HLA mismatched. Seven patients were in complete remission (CR) at the time of SCT while 7 were in non-CR. Ten patients remained in CR 4–79 months after SCT. Small populations (0.13–0.77 % of lymphocyte) of CDK2158 and CDK2178-specific CTL were detectable in 5 patients (3 with AML-M2, 1 with CML and 1 with ALL) 10–73 months after SCT. All of the 5 remained in CR 11–79 months after SCT. Only one of them had active cGvHD at sampling. On the other hand, among 7 patients who did not show an increase in the proportion of neither CDK2158 nor CDK2178-specific CTLs, 2 patients with AML-M0 relapsed 4 and 5 months after SCT, respectively. Both of them had active cGvHD at sampling. CDK2-specific CTLs were also undetectable in 2 patients with RCC after allo-SCT. These data provide evidence for the first time that CDK2-specific CTLs can be induced from donor-derived T cells without peptide immunization possibly due to in vivo sentitization of donor T cells by residual leukemic cells. Vaccination with CDK2-derived peptides after allo-SCT may therefore be useful in inducing CDK2-specific CTLs and thereby preventing relapse of leukemia. Fig. Appearance of CDK2-CTLs after allo SCT in patients with leukemia in remission. Fig. Appearance of CDK2-CTLs after allo SCT in patients with leukemia in remission.


2019 ◽  
Vol 4 (42) ◽  
pp. eaaz6821
Author(s):  
Kai Kretzschmar ◽  
Hans Clevers

IFN-γ produced by T cells directly induces intestinal stem cell death upon inflammation-induced intestinal injury (see the related Research Article byTakashimaet al.).


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 594-594 ◽  
Author(s):  
D. Polchert ◽  
J. Sobinsky ◽  
M. Kidd ◽  
A. Moadsiri ◽  
E. Reina ◽  
...  

Abstract Mesecnhymal stem cells have been observed to inhibit graft versus host disease clinically, however the timing of infusion of these cells has not been well characterized. In previous studies, we have observed MSC to rescue lethally irradiated hosts that had received sub-optimal numbers of stem cells, permit the reduction of host conditioning while establishing equal or better levels of engraftment than the combination of intensive host conditioning and untreated HSC grafts, and enable xenogeneic engraftment (rat→ mouse) suggesting that administration of MSC in combination with an allogeneic transplant significantly alters host immune responses to enhance engraftment.. These findings could only be observed if MSC were given on the same day as the bone marrow stem cells. The purpose of this study was to determine to what extent MSC might affect donor immune responses involved in GVHD and to determine the optimal timing of these effects, in order to optimize the maximal beneficial effects of allogeneic stem cell grafts engineered with MSC. Since GVHD, mediated by donor T cells, requires host antigen presentation for initiation, we tested whether the effect of MSC occurred before or after this interaction. We used an established GVHD model in which 20x 10^6 Balb/c bone marrow cells in combination with 15 x 10^6 Balb/c splenocytes were administered to lethally irradiated B6 recipients to test whether MSC (1.0 x 105) could inhibit initiation of GVHD and to what extent these cells could mitigate or abrogate ongoing GVHD. In control animals, we observed donor T cell expansion to occur in the absence of B6 host T cells with corresponding destructive effects resulting in 100% lethality by day 48. Four experimental groups (n=10 per group) were used to test MSC administration at 4 time points: 1) on day 0 following co-culture with the graft to test whether cell contact between MSC and GVHD-producing splenocytes is necessary, 2) on day 2 to test whether donor T cell exposure to host antigen is required, 3) on day 20, to test the magnitude of effect of MSC on ongoing GVHD, and 4) on day 30 in which GVHD is severe and usually irreversible. Mice were weighed twice weekly and monitored daily for survival and clinical evidence of GVHD (ruffled fur, cachexia, alopecia, and diarrhea). When compared to survival of control animals, no statistically significant effect was observed when MSC were given with the stem cell grafts on day 0. Strikingly, survival was significantly increased to 60% when given on day 2 (p=0.01, log rank test), to 50% when given on day 20 (p=0.005), and to 40% for day 30 treated animals (p=0.009). Following MSC infusion, those animals that developed signs of GVHD such as ruffled fur and alopecia had dramatic improvement of these physical findings with most surviving animals experiencing a complete reversal to normal appearing fur. The observation that no effect occurred with MSC administered at the time of bone marrow transplantation suggests that the mechanism of effect requires host antigen presentation. We conclude that optimal timing for the infusion of donor specific MSC to abrogate GVHD begins after donor T cells have encountered host antigen and can be equally effective during early, late, and severe GVHD. Clinical strategies involving the use of allogeneic stem cell grafts engineered with MSC are likely to be more powerful in overcoming GVHD if the MSC infusion is administered in a delayed fashion.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2710-2710
Author(s):  
Mary T Scott ◽  
Koorosh Korfi ◽  
Paolo Gallipoli ◽  
Peter Saffrey ◽  
Heather Jorgensen ◽  
...  

Abstract Chronic myeloid leukaemia (CML) is a hematological malignancy resulting from the transformation of a primitive hematopoietic progenitor by the fusion oncogene BCR-ABL, a constitutively active tyrosine kinase. In recent years major advances have been made in the treatment of CML with the development of tyrosine kinase inhibitors (TKIs), resulting in high rates of remission in CML chronic phase (CP) patients. However, relapse is driven by quiescent and self-renewing BCR-ABL+ CML stem cells (LSCs) that are resistant to TKIs. Consequently, identification of novel proteins or pathways which can be drug-targeted to eliminate the LSCs is a primary goal of current CML research. Through comparative analysis between CML and non-leukemic samples, we show that components of the repressive Polycomb group (PcG) complex PRC2 are significantly misregulated in CML samples. By performing genome-wide mRNA and epigenetic screens, we demonstrate that this has led to as many as 3-fold more gene repression events in CML cells being associated with gains in the histone modification H3K27me3. This misregulation results in different biological pathways being targeted by PRC2 than those found in non-leukemic samples. We demonstrate that the majority of this misregulation is present in the LSCs. EZH2 is a key component of the PRC2 complex, responsible for laying down the H3K27me3 mark. To determine the effect of inhibition of the complex on LSC survival we have utilised an inhibitor of EZH2, CPI-625. In the absence and presence of TKI, treatment of CP CML CD34+ cells (n=3) with CPI-625 resulted in decreased cell viability (p<0.001 and p<0.05, -/+ TKI respectively) and increased apoptosis (p<0.05 without TKI) in a dose dependent manner. Significantly, there was also a decrease in the number of cells in the undivided, quiescent ‘TKI resistant’ population relative to controls (p<0.01 and p<0.05 -/+ TKI respectively). This was accompanied by an increase in apoptosis (p<0.05 without TKI). Moreover, treatment with CPI-625 resulted in decreasing Colony Forming Cell (CFC) numbers, both in the absence (p<0.05) and presence (p<0.01) of TKI relative to controls. Similar results were seen with treatment of the more primitive CD34+38- cells. Importantly, these effects were not observed in non-leukemic cells. These results demonstrate that CPI-625 is capable of selective targeting of the LSC population. Our data strongly points to changes in H3K27me3 gene targets in CML as a feature related to misregulation of the PRC2 complex. We have demonstrated that targeting of this complex may have efficacy in the treatment of CML, including eradication of the drug resistant LSCs. Disclosures: Holyoake: Novartis: Membership on an entity’s Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity’s Board of Directors or advisory committees; Ariad: Membership on an entity’s Board of Directors or advisory committees.


2021 ◽  
Vol 22 (15) ◽  
pp. 7946
Author(s):  
Chang Youn Lee ◽  
Seahyoung Lee ◽  
Seongtae Jeong ◽  
Jiyun Lee ◽  
Hyang-Hee Seo ◽  
...  

The acute demise of stem cells following transplantation significantly compromises the efficacy of stem cell-based cell therapeutics for infarcted hearts. As the stem cells transplanted into the damaged heart are readily exposed to the hostile environment, it can be assumed that the acute death of the transplanted stem cells is also inflicted by the same environmental cues that caused massive death of the host cardiac cells. Pyroptosis, a highly inflammatory form of programmed cell death, has been added to the list of important cell death mechanisms in the damaged heart. However, unlike the well-established cell death mechanisms such as necrosis or apoptosis, the exact role and significance of pyroptosis in the acute death of transplanted stem cells have not been explored in depth. In the present study, we found that M1 macrophages mediate the pyroptosis in the ischemia/reperfusion (I/R) injured hearts and identified miRNA-762 as an important regulator of interleukin 1b production and subsequent pyroptosis. Delivery of exogenous miRNA-762 prior to transplantation significantly increased the post-transplant survival of stem cells and also significantly ameliorated cardiac fibrosis and heart functions following I/R injury. Our data strongly suggest that suppressing pyroptosis can be an effective adjuvant strategy to enhance the efficacy of stem cell-based therapeutics for diseased hearts.


Cells ◽  
2021 ◽  
Vol 10 (2) ◽  
pp. 225
Author(s):  
Claire Racaud-Sultan ◽  
Nathalie Vergnolle

In adult stem cells, Glycogen Synthase Kinase 3β (GSK3β) is at the crossroad of signaling pathways controlling survival, proliferation, adhesion and differentiation. The microenvironment plays a key role in the regulation of these cell functions and we have demonstrated that the GSK3β activity is strongly dependent on the engagement of integrins and protease-activated receptors (PARs). Downstream of the integrin α5β1 or PAR2 activation, a molecular complex is organized around the scaffolding proteins RACK1 and β-arrestin-2 respectively, containing the phosphatase PP2A responsible for GSK3β activation. As a consequence, a quiescent stem cell phenotype is established with high capacities to face apoptotic and metabolic stresses. A protective role of GSK3β has been found for hematopoietic and intestinal stem cells. Latters survived to de-adhesion through PAR2 activation, whereas formers were protected from cytotoxicity through α5β1 engagement. However, a prolonged activation of GSK3β promoted a defect in epithelial regeneration and a resistance to chemotherapy of leukemic cells, paving the way to chronic inflammatory diseases and to cancer resurgence, respectively. In both cases, a sexual dimorphism was measured in GSK3β-dependent cellular functions. GSK3β activity is a key marker for inflammatory and cancer diseases allowing adjusted therapy to sex, age and metabolic status of patients.


2021 ◽  
Vol 12 ◽  
Author(s):  
Motoko Koyama ◽  
Geoffrey R. Hill

Allogeneic stem cell transplantation (alloSCT) is a curative therapy for hematopoietic malignancies. The therapeutic effect relies on donor T cells and NK cells to recognize and eliminate malignant cells, known as the graft-versus-leukemia (GVL) effect. However, off target immune pathology, known as graft-versus-host disease (GVHD) remains a major complication of alloSCT that limits the broad application of this therapy. The presentation of recipient-origin alloantigen to donor T cells is the primary process initiating GVHD and GVL. Therefore, the understanding of spatial and temporal characteristics of alloantigen presentation is pivotal to attempts to separate beneficial GVL effects from detrimental GVHD. In this review, we discuss mouse models and the tools therein, that permit the quantification of alloantigen presentation after alloSCT.


1999 ◽  
Vol 189 (4) ◽  
pp. 693-700 ◽  
Author(s):  
Taila Mattern ◽  
Gundolf Girroleit ◽  
Hans-Dieter Flad ◽  
Ernst T. Rietschel ◽  
Artur J. Ulmer

CD34+ hematopoietic stem cells, which circulate in peripheral blood with very low frequency, exert essential accessory function during lipopolysaccharide (LPS)-induced human T lymphocyte activation, resulting in interferon γ production and proliferation. In contrast, stimulation of T cells by “conventional” recall antigens is not controlled by blood stem cells. These conclusions are based on the observation that depletion of CD34+ blood stem cells results in a loss of LPS-induced T cell stimulation as well as reduced expression of CD80 antigen on monocytes. The addition of CD34-enriched blood stem cells resulted in a recovery of reactivity of T cells and monocytes to LPS. Blood stem cells could be replaced by the hematopoietic stem cell line KG-1a. These findings may be of relevance for high risk patients treated with stem cells or stem cell recruiting compounds and for patients suffering from endotoxin-mediated diseases.


2019 ◽  
Vol 4 (42) ◽  
pp. eaay8556 ◽  
Author(s):  
S. Takashima ◽  
M. L. Martin ◽  
S. A. Jansen ◽  
Y. Fu ◽  
J. Bos ◽  
...  

Despite the importance of intestinal stem cells (ISCs) for epithelial maintenance, there is limited understanding of how immune-mediated damage affects ISCs and their niche. We found that stem cell compartment injury is a shared feature of both alloreactive and autoreactive intestinal immunopathology, reducing ISCs and impairing their recovery in T cell–mediated injury models. Although imaging revealed few T cells near the stem cell compartment in healthy mice, donor T cells infiltrating the intestinal mucosa after allogeneic bone marrow transplantation (BMT) primarily localized to the crypt region lamina propria. Further modeling with ex vivo epithelial cultures indicated ISC depletion and impaired human as well as murine organoid survival upon coculture with activated T cells, and screening of effector pathways identified interferon-γ (IFNγ) as a principal mediator of ISC compartment damage. IFNγ induced JAK1- and STAT1-dependent toxicity, initiating a proapoptotic gene expression program and stem cell death. BMT with IFNγ–deficient donor T cells, with recipients lacking the IFNγ receptor (IFNγR) specifically in the intestinal epithelium, and with pharmacologic inhibition of JAK signaling all resulted in protection of the stem cell compartment. In addition, epithelial cultures with Paneth cell–deficient organoids, IFNγR-deficient Paneth cells, IFNγR–deficient ISCs, and purified stem cell colonies all indicated direct targeting of the ISCs that was not dependent on injury to the Paneth cell niche. Dysregulated T cell activation and IFNγ production are thus potent mediators of ISC injury, and blockade of JAK/STAT signaling within target tissue stem cells can prevent this T cell–mediated pathology.


Sign in / Sign up

Export Citation Format

Share Document