Additional file 3 Visualization of the imputation process. a, c Heatmap of SF and OV lab test data before imputation. b, d Heatmap of SF and OV lab test data after imputation. Black tiles refer to missing entries. Abbreviations: NK, Natural killer cells, Th, T-helper lymphocyte. Ts, T-suppressor lymphocyte. CRP, C reactive protein. PCT, procalcitonin. IFN-γ, interferon-γ. TNF-α, tumor necrosis factor α. IL-1β, interleukin 1β. IL-2R, interleukin 2 receptor. IL-4, interleukin 4. IL-6, interleukin 6. IL-8, interleukin 8. IL-10, interleukin 10. C-IGM, SARS-COV-2 specific antibody IgM. C-IGG, SARS-COV-2 specific antibody IgG. SF, Sino-French New City Campus of Tongji Hospital. OV, Optical Valley Campus of Tongji Hospital.
Male infertility induced by heat stress has been attracting more and more attention. Heat stress not only causes apoptosis of spermatocytes but also has adverse effects on Sertoli cells, further damaging spermatogenesis. Lycium barbarum polysaccharide (LBP) is the main bioactive component of Lycium barbarum, which has a protective effect on male reproduction, but its mechanism is still unclear. In this study, our results proved that LBP blocked the inhibitory effect on the proliferation activity of Sertoli cells after heat stress, reversed the dedifferentiation of Sertoli cells induced by heat stress, and ameliorated the structural integrity of the blood-testis barrier. In addition, it increased the expression of the androgen receptor and activated Akt signaling pathway to resist heat-stress-induced injury of Sertoli cells.
Patients with Burkitt lymphoma who are refractory to initial therapy or who relapse after undergoing intensive chemotherapy and autologous stem cell transplantation (ASCT) usually have a poor prognosis. While there has been considerable progress in the use of chimeric antigen receptor-modified (CAR) T cell immunotherapy for the treatment of relapsed and refractory (r/r) malignancies, explicit data on adult patients with r/r Burkitt lymphoma are limited. We conducted two single-arm clinical trials to evaluate the clinical efficacy and toxicity of CD19/CD22 CAR T cell immunotherapy both alone (trial A) and in combination with ASCT (trial B) in adult patients with r/r Burkitt lymphoma. In total, 28 adult patients with r/r Burkitt lymphoma were enrolled [trial A (n = 15) and trial B (n = 13)]. The median doses of CD22 and CD19 CAR T cell infusions were 4.1 × 10 6 /kg and 4.0 × 10 6 /kg, respectively. Subsequently, after CAR T cell infusion, overall and complete responses were observed in 19 (67.9%) and 16 (57.1%) patients, respectively. The cumulative incidence rates of grade 2–4 cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome were 39.3% (11/28) and 10.7% (3/28), respectively. After a median follow-up duration of 12.5 months, 16 patients (5 in trial A and 11 in trial B) survived. Both the estimated 1-year progression-free and overall survival rates were 55.6%. Our preliminary results indicated that salvage therapy with CD19/CD22 CAR T cell infusion alone and that in combination with ASCT are effective in treating some adult patients with r/r Burkitt lymphoma.
Dear Editor, In this study, we reported the clinical and genetic characteristics of 10 Epstein-Barr virus (EBV)-triggered late-onset primary hemophagocytic lymphohistiocytosis (HLH) patients. As HLH is a rare and devastating disorder characterized by uncontrolled immune activation resulting from the impaired function of natural killer and cytotoxic T cells.1 And HLH can be further classified as either primary or secondary based on the predisposing genetic deficiency.2-4 Although primary HLH usually arises in infants and children, the late-onset primary HLH is increasingly reported in the literature.5 It has been suggested that synergetic effects of the atypical heterozygous HLH-associated mutations and environmental triggers, including EBV infection, were associated with the pathogenesis of late-onset primary HLH.6 However, the role of EBV-infected cell subpopulation in primary HLH is unknown. Therefore, we retrospectively investigated 10 patients with EBV-triggered late-onset primary HLH (occurring at over 12 years of age) from June 2013 and June 2018 in our department. According to the Diagnostic Guidelines for HLH, all of the cases met at least five of eight criteria and thereby were diagnosed as HLH, and detailed clinical parameters are listed in Table 1. Pathological studies of bone marrow, lymph node, and pleural fluids samples demonstrated that abnormal T lymphocytes, which were characterized by CD2bri+CD4–CD5–CD7dim+CD8+CD45RA–CD45RO+Ki67str+, existed in the bone marrow of case 1 and 2. In contrast, abnormal NK cell phenotype, which was marked by CD7dim+CD8–CD11bdim/–CD16dim/+CD56dim/+/str+CD57–, existed in the bone marrow for cases 7, 9, 10, and pleural effusion of case 4. No malignant clone was found in any of these patients. Despite all the patients were treated with regimens based on HLH-2004 protocol and supportive therapy, seven of 10 patients died of disease progression or complications, and the median survival time was 3.4 months. Three patients who received allogeneic hematopoietic stem cell transplantation (allo-HSCT) from unrelated donors acquired longer survival time compared with patients who did not receive allo-HSCT (Figure 1A). HLH-2004; BR RCHOP; HLH-2004; P-Gemox HLH-2004 Allo-HSCT (A) Kaplan-Meier survival curves of late-onset primary HLH patients who received or not received allo-HSCT. p values calculated by the Gehan-Breslow-Wilcoxon test. (B) Target cell identification by sorting-PCR in the late-onset primary HLH patients. EBV-DNA copies quantification of different cell types (PBMCs, T, B, and NK) by qualitative PCR demonstrated that EBV primarily infected NK cells in 100% (10/10) of them, and concomitant infection in T cells in 10% of patients (1/10). (C) The pedigrees of the 10 families affected by hemophagocytic lymphohistiocytosis were included in the present study. Squares, males; circles, females; slash, deceased; half-filled, heterozygous; darkened, homozygous; arrows, probands of the families. The affected genes and amino acid substitution caused by mutations in each family are indicated below the corresponding pedigree. (D) The expression of perforin protein on NK cells of case 2, case3, case 4, case 5, case 7, case 9, and case 10 was decreased, while relative regular perforin expression on CD56+CD3–NK cells was observed in case 1, case 6, and case 8. Numbers indicate the percentage of perforin expression. (E) The degranulation of rIL-2 stimulated NK cells (CD56+CD3–) was determined by the expression of CD107a. The results showed that stimulated degranulation of NK cells was significantly decreased in case 1, case 4, and case 6; however, relatively healthy stimulated degranulation of NK cells was observed in case 5, case 7, case 9, and case 10. Numbers indicate the percentage of CD107a expression Abbreviation: PBMCs, peripheral blood mononuclear cells. Firstly, to investigate the cell subpopulation pattern of EBV infection, immunobead sorting followed by quantitative PCR (sorting-PCR) and fluorescence in situ hybridization (sorting-FISH) assay was performed. Peripheral blood mononuclear cells (PBMCs) were isolated and fractionated into CD3+, CD19+, and CD56+ cells using an immunobead method, purities of which were confirmed by flow cytometry to be 97%–99% for B and T cells and 91%–95% for NK cells.7 Then purified cells were analyzed by quantitative PCR assay and fluorescence in situ hybridization with the EBER probe (green) as previously described.7 The results demonstrated that T cells, B cells, and NK cells were all shown to be infected with EBV, while EBV significantly infected NK cells in nine patients (cases 1–4, 6–10). EBV predominantly infected both NK cells and T cells in one patient (case 5) (Figure 1B). Sorting-FISH was performed in two cases (cases 8 and 10). EBV was found predominantly in NK cells, consistent with the results of sorting-PCR. Secondly, to understand the pathogenesis of EBV-triggered late-onset primary HLH, we carried out next-generation sequencing (NGS) using a custom design for the HLH panel under the Ion AmpliSeq Ready-to-Use custom designer platform following the guide (https://www.ampliseq.com/protected/dashboard.action), and detailed NGS procedure has been reported in previous literature.8 As described in Table 2, a total of five cases had one single gene mutation at heterozygous status (cases 1, 5, 6, 9, and 10), two cases had gene mutations in two primary HLH-related genes at heterozygous state (cases 2 and 4). Case 3 had a compound heterozygous mutation in PRF1 (this case had been reported in the previous article8), case 7 had a compound heterozygous mutation in UNC13D, while case 8 had two sites of heterozygous mutations in UNC13D, which were descended from her father. Most of the mutations were predicted to be damaging by software SIFT (Sorting Intolerant From Tolerant). All the details of 10 pedigrees are shown in Figure 1C and Table 2. UNC13D UNC13D G863D A397T 0.001 - 0.000 (Damaging) 0.262 (Tolerated) UNC13D UNC13D G863D I410L 0.001 0.003 0.000 (Damaging) 1.000 (Tolerated) UNC13D UNC13D G863D I410L Thirdly, to evaluate the impact of genetic mutations on their NK cell function, NK-cell activity assay, NK-cell degranulation assay, and perforin staining assay were subsequently performed on whole blood samples from these pedigrees. NK-cell activity assay was performed as previously described.8 Perforin staining assay was performed by staining PBMC samples for NK cell markers before fixation and permeabilization. Then anti-perforin or phycoerythrin-conjugated mouse immunoglobulin G2b (BD Pharmingen) was stained. NK cells were first gated and subsequently analyzed for the expression of the PRF1 protein. As for the NK-degranulation assay, PBMCs were incubated with or without rIL-2 to investigate activated or resting NK-cell degranulation, respectively. For analysis, lymphocytes were gated based on forward scatter, then CD3–CD56+ NK cells were gated and assessed for surface expression of CD107a. Results demonstrated that reduced NK-cell activity was observed in all EBV-triggered late-onset primary HLH patients (Table 1), while NK-cell activity defect was found in four patients, which was consistent with previous research.9 Substantially reduced perforin expression on CD3–CD56+ NK cells was detected in seven patients (Figure 1D). One case had abnormal resting NK-cell degranulation, and four cases had defective resting NK-cell degranulation. Abnormal activated NK-cell degranulation was recorded in three cases (Figure 1E). Nine cases had either reduced perforin expression or decreased activated NK-cell degranulation. Based on the above findings, our study demonstrated that, unlike sporadic HLH, EBV-triggered late-onset primary HLH usually has one or two pathologic mutations of primary HLH-associated genes, reduced perforin expression, NK activity, or degranulation. Most patients have high load EBV infection, and the target cells of EBV are usually NK cells, sometimes with T cells, which was typical for FHL (familial hemophagocytic lymphohistiocytosis) patients.10 Through the pedigree investigation, we also noticed that although some family members of the patient had the same mutations, they did not develop the disease, possibly attributing to their lack of EBV infection or other "second-hit" factors such as additional genetic mutations. Therefore, genetic mutations, as well as EBV-infected lymphocyte subtypes, may collectively be involved in the pathogenesis of late-onset primary HLH. HLH gene sequencing and pedigree investigation, combined with EBV-infected cell type identification, are valuable in the differential diagnosis of late-onset primary HLH. Timely allo-HSCT is recommended to improve the poor prognosis of late-onset primary HLH. The authors would like to acknowledge laboratory colleagues for data mining and analysis of next-generation sequencing, and we also acknowledge all the patients and their family members who participated in the study. The authors declare that they have no conflict of interest. All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by Liang Huang, Yi Xiao, Jinniu Deng, Miao Zheng, Hui Luo, and Lijun Jiang. The first draft of the manuscript was written by Lili Gao with the help of Li Yang and revised by Chunrui Li, Min Xiao, and Jianfeng Zhou. All authors commented on previous versions of the manuscript and agreed on the final manuscript.
Although dramatic clinical success has been achieved in acute promyelocytic leukemia (APL), the success of differentiating agents has not been reproduced in non-APL leukemia. A key barrier to the clinical success of arsenic is that it is not potent enough to achieve a clinical benefit at physiologically tolerable concentrations by targeting the leukemia cell differentiation pathway alone. We explored a novel combination approach to enhance the eradication of leukemia stem cells (LSCs) by arsenic in non-APL leukemia. In the present study, phosphatidylinositol 3-kinase /AKT/mammalian target of rapamycin (mTOR) phosphorylation was strengthened after As 2 S 2 exposure in leukemia cell lines and stem/progenitor cells, but not in cord blood mononuclear cells (CBMCs). propidium iodide-103, the dual PI3K/mTOR inhibitor, effectively inhibited the transient activation of the PI3K/AKT/mTOR pathway by As 2 S 2 . The synergistic killing and differentiation induction effects on non-APL leukemia cells were examined both in vitro and in vivo . Eradication of non-APL LSCs was determined using the nonobese diabetic/severe combined immunodeficiency mouse model. We found that a combined As 2 S 2 /PI-103 treatment synergized strongly to kill non-APL leukemia cells and promote their differentiation in vitro . Furthermore, the combined As 2 S 2 /PI-103 treatment effectively reduced leukemia cell repopulation and eradicated non-APL LSCs partially via induction of differentiation while sparing normal hematopoietic stem cells. Taken together, these findings suggest that induction of the PI3K/AKT/mTOR pathway could provide a protective response to offset the antitumor efficacy of As 2 S 2 . Targeting the PI3K/AKT/mTOR pathway in combination with As 2 S 2 could be exploited as a novel strategy to enhance the differentiation and killing of non-APL LSCs.
8025 Background: CT103A, which is designed with a fully human BCMA-specific CAR structure, has shown sustained efficacy and durable safety in heavily pretreated relapsed and refractory multiple myeloma patients. Here, we report updated efficacy and safety data of its phase 1b/2 study (FUMANBA-1) with longer duration of follow-up. Methods: FUMANBA-1 is conducted in 14 centers in China. This study enrolled RRMM patients who received ≥ 3 lines of prior therapies containing at least a proteasome inhibitor and an immunomodulatory agent and were refractory to their last line of treatment. Patients who have progressed on previous BCMA CAR-T cell therapy were also included. All patients received a single infusion of CT103A at the dose of 1.0 x 10 6 CAR-T cells/Kg. The objective is to evaluate the efficacy, safety and PK/PD of CT103A. Results: As of the September 9 th , 2022, 103 patients [53.4% male; median age 58.0 years (range 39-70)] with RRMM received CT103A (17 in phase 1b; 86 in phase 2) with a median follow-up of 13.8 months (range 0.4 to 27.2). The treated patients had received a median of 4 (range 3-23) lines of prior therapy. 101 patients were evaluable for efficacy assessment. The median time to first response was 16 days (range 11-179). A 96% ORR was observed, with 74.3% ≥ CR. Median DOR and median PFS have still not reached. The 12-month PFS rate was 78.8% (95% CI: 68.6–85.97). For patients without prior BCMA CAR-T therapy (N = 89), ORR was 98.9% with 78.7% ≥ CR. For patients without prior BCMA CAR-T therapy and received CT103A for more than 6 months as of the cutoff date (N = 81), ORR was 98.8% with 80.2% ≥ CR. For patients with prior BCMA CAR-T cell therapy, 4/5 (80%) of those who achieved sCR still sustained sCR over 18 months post infusion. Of the 101 patients, 95% achieved MRD-negativity with a median time to MRD-negative of 14 days (range 13-185), and all patients with CR/sCR were MRD-negative. Furthermore, 82.4% (95%CI 70.90-89.72%) achieved sustained MRD negativity over 12 months. Since the previous publication in 64 th ASH meeting, no new events of CRS and ICANS occurred. The most common ≥ grade 3 treatment-related AEs were still hematologic. The expansion of CT103A reached the median peak level of 87570.6 copies/μg gDNA at a median of 12 days. CT103A was still above lower limit of qualification (100 copies/μg gDNA) in 50.0% (28/56) patients at 12 months and 40.0% (4/10) patients at 24 months after infusion. In addition, only 20 of 103 patients (19.4%) with evaluable samples were detected to be positive for the anti-drug antibody. Conclusions: At a longer median follow-up of 13.8 months, CT103A achieved deep and long-lasting responses in heavily pre-treated patients with MM. Furthermore, patients with prior BCMA CAR-T cell therapy who achieved sCR had sustained sCR over 18 months. CT103A demonstrated a favorable safety profile with no new risk observed with longer follow-up. Clinical trial information: NCT05066646 .