CD3+ T-Cell Content of Short Term Ex Vivo Expanded UCB on a MSC Platform Predicts Human Engraftment in a NOD/SCID Model.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2303-2303
Author(s):  
M. Kozik ◽  
J. J. Banks ◽  
L. R. Fanning ◽  
M. R. Finney ◽  
Y. Huang ◽  
...  

Abstract Background. Umbilical cord blood (UCB), a source of hematopoeitic stem cells (HSC), is marked by delayed engraftment attributed to the limited cellular content of a single UCB unit. Cytokine-based ex vivo expansion of UCB is a way of increasing the number of cells available for allogeneic transplants, however, this strategy has not demonstrated improved engraftment in human clinical trials. Further studies have incorporated human mesenchymal stem cells (huMSC) which may provide signals that control the proliferation, survival, and differentiation of HSC. In attempt to reduce the occurrence and severity of GVHD following allogeneic transplants, strategies such as utilizing T-cell depleted grafts have been pursued, however, clinical trials using these grafts have shown decreased rates of donor engraftment, suggesting the requirement of accessory cells as well as HSC to achieve engraftment. Methods. UCB mononuclear cells (MNC) were cultured using cytokines (IL-3, IL-6, G-CSF, SCF, Flt-3L, EPO) with or without a feeder-layer of huMSC for 12 days. On day 12, viability, 4-color flow cytometry, and human engraftment potential were measured. Human engraftment potential was determined by injecting cells (without CD34+ selection) from each culture condition and non-cultured UCB MNC, via tail vein, into sublethal irradiated NOD/SCID mice. Mice were injected with unexpanded UCB MNC (n=23), UCB expanded in huMSC+cytokines (n=21) and UCB expanded in cytokines alone (n=10). 7–9 weeks following injection of human cells, bone marrow was harvested and analyzed for human content. Positive human engraftment was determined by a human %CD45+ of ≥ 0.4%. Results. An 8.77 fold expansion of UCB cultured in cytokines alone compared to a 7.14 fold expansion of UCB cultured in huMSC + cytokines was observed. Surface phenotyping of expanded UCB, and human cells emerging in the bone marrow of NOD/SCID mice following injection of cultured and non-cultured UCB are in Table 1. Unexpanded huMSC+cytokines Cytokines % CD3 44.0 (4.40M) 2.04 (10.1M) 1.52 (4.35M) % CD56 17.0 (1.70M) 7.07 (36.1M) 3.69 (13.8M) % CD34 3.41 (.341M) 2.25 (10.9M) 3.52 (12.3M) Bone marrow of NOD/SCID mice % CD45+ 2.57 3.58 2.38 % of CD45+ co-expressing CD3 11.0 9.25 18.7 % of CD45+ co-expressing CD19 33.5 16.6 19.4 % of CD45+ co-expressing CD56 10.1 8.04 1.60 Human engraftment was seen in 13 mice which received unexpanded UCB, 10 mice which received UCB expanded in huMSC+cytokines and only 3 mice which received UCB expanded in cytokines alone. Statistical analysis, using multivariable logistic regression to determine the factors that predict engraftment, revealed that the proportions of T and NK cells present in expanded UCB correlated with engraftment. A 10% increase in the proportion of CD45+ co-expressing CD3 was associated with a 1.79 fold increase in engraftment (p=0.016), whereas each 10% increase in the proportion of CD45+ co-expressing CD56 increased the odds of engrafting by 104% (p= 0.003). Conclusions. We observed an expansion of CD34 hematopoietic progenitors as well as a greater proportion of CD3+ cells, in expansion conditions incorporating huMSC. Additionally, we observed improved rates of engraftment in this expansion condition. Therefore, although the mechanism by which accessory cells including T and NK cells facilitate HSC engraftment is not known, we observed that the presence of accessory cells in addition to CD34 hematopoietic progenitors facilitated engraftment in NOD/SCID mice.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3666-3666
Author(s):  
Tarun K. Garg ◽  
Susann Szmania ◽  
Jumei Shi ◽  
Katie Stone ◽  
Amberly Moreno-Bost ◽  
...  

Abstract Immune-based therapies may improve outcome for multiple myeloma (MM) by eradicating chemo-resistant disease. Our recent trial utilizing IL2 activated, killer immunoglobulin-like receptor-ligand mismatched NK cell transfusions from haplo-identical donors yielded (n) CR in 50% of patients. Unfortunately, after NK cell therapy, 2/10 patients had progressive disease, and the median duration of response for the other 8/10 patients was only 105 days (range 58–593). This may have been due to an insufficient dose of alloreactive NK cells and early rejection. Furthermore, appropriate donors were identified for only 30% of otherwise eligible patients. We therefore investigated whether NK cells from MM patients could be expanded and activated to kill autologous MM. We then examined whether pre-treatment of MM cell targets with elotuzumab, a humanized antibody to the MM tumor antigen CS1, could further enhance NK cell-mediated lysis. PBMC from 5 MM patients were co-cultured for 14 days with irradiated K562 cells transfected with 4-1BBL and membrane bound IL15 in the presence of IL2 (300U/ml) as previously described (Imai et al, Blood2005;106:376–383). The degree of NK cell expansion, NK immunophenotype, and ability to kill MM (4 hour 51Cr release assays) were assessed. To determine the ability of ex vivo expanded NK cells to traffic to bone marrow, activated NK cells were injected into the tail vein of NK cell depleted NOD-SCID mice, which were then sacrificed after 48 hours. Flow cytometry for human CD45, CD3, and CD56 was performed on cells from blood, marrow and spleen. There was an average 64-fold expansion of NK cells (range: 8–200) after 2 weeks of co-culture with K562 transfectants. Expansion of T cells was not observed. The NK cell activating receptor NKG2D, and natural cytotoxicity receptors NKp30, NKp44, and NKp46 were up-regulated following the expansion. Expanded NK cells were able to kill autologous MM (E:T ratio 10:1, average 31%, range 22–41%), whereas resting NK cells did not. Pretreatment of autologous MM cells with elotuzumab increased the activated NK cell-mediated killing by 1.7-fold over target cells pretreated with an isotype control antibody. This level of killing was similar to that of the highly NK kill-sensitive cell line K562 (Figure). Autologous PHA blasts and CD34+ stem cells were not killed. Activated human NK cells were detectable in the bone marrow of NOD-SCID mice 48 hours after injection. Ex vivo activation of NK cells from MM patients with K562 transfectants can induce killing of autologous MM and produce large numbers of NK cells for potential therapy. The addition of elotuzumab to activated NK cell therapy enhances anti-MM effects by ADCC thus invoking an additional NK cell-mediated mechanism of MM killing. Importantly, ex vivo activated NK cells traffic to the bone marrow in mice. Autologous NK cell therapy eliminates the issues related to allo-donor availability and early NK cell rejection, and could provide an option for patients refractory to chemotherapy agents. Figure Figure


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2915-2915
Author(s):  
Xiao jun Huang ◽  
Daihong Liu ◽  
Kai Yan Liu ◽  
Lan ping Xu ◽  
Huan Chen ◽  
...  

Abstract Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only therapeutic option for many hematological malignancies. Many patients requiring allo-HSCT do not have a human leukocyte antigen (HLA)-matched donor. For those patients, unrelated bone marrow or cord blood is often used but with many limitations such as time-consuming search and deficient cell numbers. Recently, HLA mismatched family donors were used in allo-HSCT, but which are associated with higher rates of graft rejection and acute graft versus host disease (aGVHD) if T cells are not first depleted. We developed a new technique for HLA mismatched allogeneic HSCT using G-CSF primed bone marrow plus G-CSF-mobilized peripheral blood stem cells without ex vivo T cell depletion. In this study, 176 patients, including 88 with high-risk or advanced leukemia, were transplanted with cells from a HLA-haploidentical family donor with 1–3 mismatched loci. After conditioning, patients received G-CSF-primed bone marrow grafts that had not been depleted ex vivo of T cells, in combination with G-CSF-mobilized peripheral blood stem cells, as well as GVHD prophylaxis. The total infused nucleated cell count was 6x108 to 8x108/kg recipient weight. All patients achieved sustained, full donor-type engraftment. The incidence of grade II–IV aGVHD was 41.5% (73/176), including 21 patients with grade III–IV aGVHD. The development of aGVHD was not associated with the extent of HLA disparity. Chronic GVHD was observed in 81 of 120 evaluable patients (67.5%). Forty-six of the 81 patients had limited-stage disease and 35 had extensive cGVHD. Fifty-two patients died among whom 9 died of recurrent disease and 43 of transplant-related complications. One hundred and twenty-four of the 176 patients survived, and 112 remained disease free at the time of a median follow-up of 22 months (4 to 59 months). The 2-year probabilities of disease-free survival were 74.8% and 69.3% for standard- and high-risk patients, respectively. In summary, we have established a new regimen to use bone marrow from haploidentical family donors without ex vivo T cell depletion, in combination with G-PBSCs, as a source of stem cells even in cases of HLA mismatched transplantation. This method showed us good effect even similar as that of HLA-matched allo-HSCT. It will be very useful for patients with hematological malignancies especially much more patients in China who are the only child in the family.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1425-1425
Author(s):  
Prabal Banerjee ◽  
Michael D Lairmore ◽  
Juan Carlos Ramos ◽  
William J Harrington ◽  
Mark A Beilke ◽  
...  

Abstract Abstract 1425 Poster Board I-448 Human T-lymphotropic virus type-1 (HTLV-1) is the first human retrovirus linked to cancer and is the etiologic agent of adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4+ T cell malignancy. The molecular and genetic factors induced by HTLV-1 that initiate ATLL remain unclear, in part, due to the lack of an animal model which recapitulates leukemogenic events. In particular, early target cell infection and transformation events have not been identified or defined. Herein, we have created humanized NOD/SCID (HU-NOD/SCID) mice by inoculation of NOD/SCID mice with CD34+ hematopoietic progenitor and stem cells (CD34+ HP/HSCs) infected ex vivo with HTLV-1. HTLV-1-HU-NOD/SCID mice consistently developed CD4+ T cell lymphomas with characteristics similar to ATLL. Elevated proliferation of infected human stem cells (CD34+CD38−) in the bone marrow was observed in mice developing malignancies. Furthermore, examination of CD34+ HP/HSCs from HTLV-1-infected patients revealed proviral integrations suggesting a role of human bone marrow-derived stem cells in leukemogenesis. NOD/SCID mice reconstituted with CD34+ HP/HSCs transduced with a lentivirus vector (LV) expressing the HTLV-1 oncoprotein (Tax1) also developed CD4+ lymphomas. The recapitulation of a CD4+ T cell lymphoma in HTLV-1- and Tax1-HU-NOD/SCID mice suggest that hematopoietic stem cells serve as a viral reservoir in vivo and provide a cellular target for cell transformation in humans. This animal model of HTLV-1 induced ATLL will provide an important tool for the identification of molecular and cellular events that control the initiation and progression of the lymphoma and potential therapeutic targets to block tumor development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2002 ◽  
Vol 100 (13) ◽  
pp. 4391-4400 ◽  
Author(s):  
Wanda Piacibello ◽  
Stefania Bruno ◽  
Fiorella Sanavio ◽  
Sara Droetto ◽  
Monica Gunetti ◽  
...  

The ability of advanced-generation lentiviral vectors to transfer the green fluorescent protein (GFP) gene into human hematopoietic stem cells (HSCs) was studied in culture conditions that allowed expansion of transplantable human HSCs. Following 96 hours' exposure to flt3/flk2 ligand (FL), thrombopoietin (TPO), stem cell factor (SCF), and interleukin-6 (IL-6) and overnight incubation with vector particles, cord blood (CB) CD34+ cells were further cultured for up to 4 weeks. CD34+ cell expansion was similar for both transduced and control cells. Transduction efficiency of nonobese diabetic/severe combined immunodeficient (NOD/SCID) repopulating cells (SRCs) was assessed by transplants into NOD/SCID mice. Mice that received transplants of transduced week 1 and week 4 expanded cells showed higher levels of human engraftment than mice receiving transplants of transduced nonexpanded cells (with transplants of 1 × 105 CD34+ cells, the percentages of CD45+ cells were 20.5 ± 4.5 [week 1, expanded] and 27.2 ± 8.2 [week 4, expanded] vs 11.7 ± 2.5 [nonexpanded]; n = 5). The GFP+/CD45+ cell fraction was similar in all cases (12.5% ± 2.9% and 12.2% ± 2.7% vs 12.7% ± 2.1%). Engraftment was multilineage, with GFP+/lineage+ cells. Clonality analysis performed on the bone marrow of mice receiving transduced and week 4 expanded cells suggested that more than one integrant likely contributed to the engraftment of GFP-expressing cells. Serial transplantations were performed with transduced week 4 expanded CB cells. Secondary engraftment levels were 10.7% ± 4.3% (n = 12); 19.7% ± 6.2% of human cells were GFP+. In tertiary transplants the percentage of CD45+ cells was lower (4.3% ± 1.7%; n = 10); 14.8% ± 5.9% of human cells were GFP+, and human engraftment was multilineage. These results show that lentiviral vectors efficiently transduce HSCs, which can undergo expansion and maintain proliferation and self-renewal ability.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4950-4950
Author(s):  
Jin-Yeong Han ◽  
Rhee-Young Koh ◽  
Su-Yeong Seo ◽  
Joo-In Park ◽  
Hyuk-Chan Kwon ◽  
...  

Abstract Mesenchymal stem cells (MSC) are multipotent and believed to facilitate the engraftment of hematopoietic stem cells (HSC) when transplanted simultaneously in animal studies and recently even in human trials. In this study, we transfected culture-expanded MSC with GM-CSF, G-CSF, and CSF cytokine genes and then co-transplanted with HSC to further promote HSC engraftment. Mononuclear cells were harvested from the various sources and seeded in long-term culture for ex vivo MSC expansion. The phenotype and purity of MSC were assessed by flow cytometry. We transferred the above three cytokine genes into ex vivo expanded MSC, confirmed transfection by fluorescent microscope of GFP, and thereafter did co-transplantation with HSC. A total of 1x107 HSC plus MSC/uL were introduced to tail vein of SCID mice. After 3–7 weeks later, with venous blood from the eyeballs, homing and engraftment of human cells were determined by flow cytometry and fluorescence in situ hybridization (FISH) studies. The total nucleated cell count and the engraftment of CD45+/CD34+ cells and XX/XY-positive human cells significantly increased in co-transplanted mice and even higher with the cytokine gene-transfected MSC in the order of GM-CSF, SCF, and G-CSF transfections (P<0.05). These results suggest that MSC transfected with hematopoietic growth factor genes are capable of enhancing the hematopoietic engraftment. Now we are planning to deliver genes involved in homing and cell adhesions, e.g., CXCR4, VLA, or TPO into ex vivo expanded MSC and do co-transplantation with HSC to further increase the efficiency of stem cell transplantation.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1261-1261 ◽  
Author(s):  
Shuro Yoshida ◽  
Fumihiko Ishikawa ◽  
Masaki Yasukawa ◽  
Toshihiro Miyamoto ◽  
Goichi Yoshimoto ◽  
...  

Abstract Transplantation of human leukemic cells into severe combined immunodeficiency (SCID) mice has been used to analyze developmental mechanisms of human leukemogenesis. Previous models, however, were limited in efficient or long-term engraftment of leukemia initiating cells. Here we report a new SCID model that supports highly efficient long-term engraftment of primary human acute myelogenous leukemia (AML) cells. We have established a novel immune-compromised mouse by backcrossing a complete null mutation of the common cytokine receptor g chain onto NOD-scid mice (NOD/SCID/IL2rgnull mice), and reported that normal human cord blood-derived hematopoietic stem cells efficiently engrafted in newborn NOD/SCID/IL2rgnull mice as compared to NOD/SCID/b2mnull mice (Ishikawa et al, Blood in press). Injection of 5x106 total bone marrow mononuclear cells from primary AML patients (FAB subtypes: M1, M2, M3, M4 and M7) into sublethally-irradiated newborn NOD/SCID/IL2rgnull mice, however, did not result in efficient engraftment of AML cells, while predominant proliferation of human CD4+ and CD8+ T cells was seen. These human T cells expressed CD45RO, and levels of human IFN-g in sera of the recipients significantly elevated, suggesting that human T cells were activated and inhibited the engraftment of human AML cells in the xenogeneic setting. We thus transplanted AML cells after T cell depletion. Strikingly, transplantation of 4x106 T cell-depleted AML bone marrow cells into neonatal NOD/SCID/IL2rgnull mice resulted in the efficient AML engraftment, whose levels were significantly higher than those in transplantation of the same number of T cell-depleted AML cells into NOD/SCID/b2mnull newborns or NOD/SCID/IL2rgnull adults. We also transplanted 103–104 hCD34+hCD38− bone marrow cells purified from AML patients. These low-doses of hCD34+hCD38− cells also successfully engrafted, progressively giving rise to hCD34+hCD38+ and hCD34− leukemic cells over 16 weeks. hCD34+hCD38− cells purified from the bone marrow of primary NOD/SCID/IL2rgnull recipients again reconstituted AML in secondary recipients, indicating that this system supports self-renewal capacity of AML stem cells within the hCD34+hCD38− fraction. Thus, the NOD/SCID/IL2rgnull newborn system provides a powerful model to study human leukemogenesis as well as the interaction between human T cells and AML cells in vivo.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1359-1359
Author(s):  
Prabal Banerjee ◽  
Lindsey Crawford ◽  
Michelle Sieburg ◽  
Patrick Green ◽  
Mark A Beilke ◽  
...  

Abstract Human T-lymphotropic virus type-1 (HTLV-1) is a human retrovirus linked to cancer and is the etiologic agent of Adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4+/CD25+ T cell malignancy. The early molecular events induced by HTLV-1 infection as well as the role of various viral genes in the induction of leukemia remain unclear, predominantly due to the lack of an animal model that recapitulates ATLL development. HTLV-1 infection of humanized NOD/SCID mice (HTLV-1- HU-SCID) was achieved by inoculation of NOD/SCID mice with CD34+ hematopoietic progenitor cells and stem cells (CD34+ HP/HSCs) infected ex vivo with HTLV-1. HTLV-1-HU-NOD/SCIDmice consistently developed CD4+CD25+ T cell lymphomas with clinical characteristics associated with ATLL and infected mice showed hyperproliferation of infected human stem cells (CD34+CD38−) in the bone marrow. Inoculation of NOD/SCID mice withCD34+ HP/HSCs transduced with a lentivirus vector (LV) expressing the HTLV-1oncoprotein (Tax1) also developed CD4+CD25+ lymphomas. The HTLV-1 bZIP protein(HBZ), encoded by the minus strand of the HTLV-1 genome, is expressed in all ATLL cells and has been implicated in the maintenance of leukemogenesis. HBZ has previously been previously shown to interact with numerous cellular factors and can modulate Tax1 activity in vitro. To establish the role of HBZ in HTLV-1 replication and leukemogenesis in vivo, HU-SCID mice were infected with an infectious proviral clone lacking functional HBZ (HTLV-1ΔHBZ). HTLV-1ΔHBZ-infected HU-SCID mice developed lymphoproliferations with an immature preleukemic CD4−CD8−CD90+ phenotype starting at ~10 weeks post-reconstitution. In contrast wild type HTLV-1 infection reproducibly induces a mature CD4+CD25+ CD90− lymphoma. Lymphoma cells successfully engrafted naïve NOD/SCID mice when injected into the peritoneal cavity and these cells maintain the expression of viral proteins, gp46env and p19gag. HTLV-1 infection of CD34+ HP/HSCs and the recapitulation of a lymphoma similar to ATLL in HU-NOD/SCID mice suggest that hematopoietic stem cells provide a relevant cellular target and viral reservoir in vivo and that infection of these cells contribute to viral lymphomagenesis in humans. The HTLV-1-HU-SCID mouse model presents a compelling in vivo model to characterize molecular initiation and progression of events in the generation of ATL and to establish the role of HTLV-1 auxiliary proteins in viral pathogenesis.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 48-49
Author(s):  
Hans Jiro Jiro Becker ◽  
Masatoshi Sakurai ◽  
Satoshi Yamazaki

Background The application of gene editing in hematopoietic stem cells (HSCs) holds great promise for the treatment of genetic blood disorders such as severe combined immunodeficiency (SCID). However, one critical bottleneck is that edited HSCs cannot easily be expanded ex vivo without concomitant loss of self-renewal. This limitation excludes the possibility of growing functional HSCs from single cells, which would enable the selection of desired clones based on sequence verification of relevant on- and off-target modifications. We recently reported on a defined, serum-free, polymer-based culture protocol that selectively facilitates in vitro proliferation of murine HSCs [1]. In the current study, we aimed to expand functional, CRISPR/Cas9-edited HSCs from bulk cell populations as well as from cloned, single HSCs to generate grafts capable of hematopoietic reconstitution. We show that HSCs from the murine PrkdcSCID model, which harbors a point mutation in the Prkdc gene leading to B and T cell deficiency, can be edited and expanded to correct the immunodeficient phenotype after transplantation. Furthermore, we demonstrate that single, gene-edited HSCs can be cloned and expanded using our system to generate functional HSCs for SCT. Methods CD150+CD201+c-Kit+Lin- (CD150+CD201+KL) HSCs from C.B17/Icr-PrkdcSCID (SCID) mice were isolated and cultured in polymer-based medium supplemented with recombinant cytokines. Gene editing was performed with Cas9 protein and appropriate gRNAs delivered as ribonucleoprotein complexes (RNPs) via electroporation. HDR donors were supplied as single-strand oligonucleotides (ssODNs). Stem cell transplantations (SCTs) were carried out after lethal irradiation with 2.5 Gy. Results To demonstrate that edited HSCs can be expanded as a bulk population and efficiently engraft to correct a disease phenotype, SCID mouse-derived donor HSCs were subjected to Cas9-mediated gene editing at the Prkdc locus (Fig. 1a). Total cells and primitive CD201+CD150+cKit+Lin-(KL) cells expanded 70- and 10-fold, respectively, over seven days after which bulk populations were transplanted into SCID mice. Inference of CRISPR edits (ICE) analysis performed at the time of SCT indicated an HDR frequency of 29%±10%. The emergence of B and T cells in peripheral blood samples was observed from four weeks after transplantation (B220+: 21±7%, CD4+: 27±4%, CD8+: 4±1%; Fig 1b). We also confirmed the presence of B and T cells in the spleen and thymus of transplanted mice. Immunization experiments showed immunoglobulin titer levels equal to healthy control mice after challenge with a T-dependent antigen. We conclude that expansion and autologous SCT of edited HSCs restores a functional B and T cell compartment in SCID mice. We next inquired whether our system could be used to expand single, edited HSC clones. To this end, we sorted single, edited CD150+CD201+KL clones by flow cytometry and expanded them for two weeks. Genomic DNA was sampled from growing colonies and editing outcomes at the Prkdc locus were individually assessed to screen for corrected HSCs (Fig. 2a). After transplantation of the selected clones, B and T cells could be detected starting from 4 weeks and 8 weeks, respectively, in peripheral blood (Fig. 2b), suggesting that functional HSCs could be expanded from edited clones. Interestingly, we found that engraftment was associated with high expression of EPCR in the CD150+KL populations of single cell-derived HSC colonies. Conclusion We have shown that functional, Cas9-edited SCID and wildtype HSCs can be expanded in our defined culture system. Corrected SCID HSCs contributed to hematopoietic reconstitution of B and T lineages conferring restored immunity in vivo. Furthermore, we were able to generate transplantable HSCs from single edited clones. This approach has important applications in HSC gene editing and has potential to overcome marker-based selection strategies since individual clones can be interrogated and selected for targeted gene editing events prior to transplantation. Our expansion system will serve as a tool to further the development of targeted gene therapeutic strategies. [1] Wilkinson et al., Nature 571, 117-121 (2019) Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 931-931
Author(s):  
Bruno Fattizzo ◽  
Matteo Claudio Da Via' ◽  
Juri Alessandro Giannotta ◽  
Paola Bianchi ◽  
Luca Baldini ◽  
...  

Abstract Warm type autoimmune hemolytic anemia (wAIHA) is a rare disease characterized by variable severity and bone marrow (BM) compensation, unpredictable relapses and several complications (i.e. infections and thrombosis). Most therapies are aimed at restoring immune tolerance by targeting various immunologic mechanisms (autoantibody production, reticuloendothelial phagocytosis, and complement activation). BM composition during acute and chronic/relapsing disease phases is still under investigated, and preliminary studies showed that it may predict anemia severity and treatment response. We aimed at dissecting BM environment by single cell RNA sequencing (scRNA-seq) in patients with wAIHA. We selected 2 patients experiencing mild hemolytic reactivations handled with low dose steroids (M-AIHA) and 2 with severe relapses requiring high steroid doses and rituximab (S-AIHA). We focused on lymphoid/myeloid subpopulations and their gene expression and evaluated the association of output data with clinical features. We performed scRNA-seq for 17,989 single cells, detecting over 23,446 expressed genes per cell on average. Uniform manifold approximation and projection (UMAP), a method for nonlinear dimensionality reduction, showed the microenvironmental cell composition, including T-, B- and NK- lymphocytes and their subpopulations, plasma cells, CD14+ and CD16+ monocytes, hematopoietic stem cells, and myeloid precursors (Figure 1A). On the whole, BM microenvironment showed a high frequency of innate immunity effectors such as NK cells and monocytes (11% and 15% of total cells), likely reflecting the inflammatory state typical of autoimmune/autoinflammatory response. T-cell subpopulations were also highly represented. Specifically, more CD4+ memory than CD4+ naïve T-cells (58% vs 38%) were found, and T-regs represented a small fraction (4%). Also, CD8+ memory cells were more frequent than CD8+ naïve and CD8+ effectors (55% vs 24% vs 21%). Both CD8+ memory and effectors type 2 cells were higher than type 1 cells, indicating a likely participation of T cell compartment in disease phenotype. Finally, B cells were particularly underrepresented, probably due to recent therapy (steroids/rituximab). Figure 1B displays % of BM immune cells divided into M-AIHA and S-AIHA. S-AIHA patients showed higher CD14+ monocytes (57% vs 43%) and decreased NK cells (19% vs 81%) as compared to M-AIHA. Interestingly, within the latter compartment, the CD56 bright NKs were over-represented in S-AIHA (83% vs 17%), suggesting an attempt to negatively regulate activated lymphocytes. Moreover, S-AIHA showed a severe decrease of B cells as compared to M-AIHA, consistently with more recent rituximab treatment. Furthermore, we performed differential expression and gene set enrichment (GSEA) analysis within the different cell subset comparing M-AIHA and S-AIHA. Concerning T cells, we found differential expression of genes related to T-cell receptor, immunoglobulins and interferon alpha/gamma response. Regarding CD14+ monocytes, we observed a downregulation of pathways related to immunomodulatory/inflammatory cytokines, complement activation and apoptosis in S-AIHA versus M-AIHA. Finally, using the CopyKat tool, we found aneuploidies in myeloid cells, including stem cells, suggesting that the selective pressure from the immune environment may lead to accumulation of genetic lesions in chronic S-AIHA. This clonal evolution can possibly explain the clinical overlap with myeloid neoplasms. Overall, these preliminary data show for the first time that scRNA-seq technology is feasible in wAIHA patients and gives insights in the pathogenic role of bone marrow immunologic microenvironment. Additionally, BM composition appears to dynamically modify according to disease severity and treatment, potentially enabling tailored therapies. Figure 1 Figure 1. Disclosures Fattizzo: Kira: Speakers Bureau; Alexion: Speakers Bureau; Novartis: Speakers Bureau; Momenta: Honoraria, Speakers Bureau; Annexon: Consultancy; Apellis: Speakers Bureau; Amgen: Honoraria, Speakers Bureau. Bianchi: Agios pharmaceutics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Bolli: Celgene/BMS: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Honoraria; Amgen: Honoraria. Barcellini: Alexion Pharmaceuticals: Honoraria; Incyte: Membership on an entity's Board of Directors or advisory committees; Agios: Honoraria, Research Funding; Bioverativ: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document