2664. Whole-genome sequencing reveals complex genomic features underlying anti-CD19 CAR T-cell treatment failures in lymphoma.
作者: Michael D Jain.;Bachisio Ziccheddu.;Caroline A Coughlin.;Rawan Faramand.;Anthony J Griswold.;Kayla M Reid.;Meghan Menges.;Yonghong Zhang.;Ling Cen.;Xuefeng Wang.;Mohammad Hussaini.;Ola Landgren.;Marco L Davila.;Jonathan H Schatz.;Frederick L Locke.;Francesco Maura.
来源: Blood. 2022年140卷5期491-503页
CD19-directed chimeric antigen receptor (CAR-19) T cells are groundbreaking immunotherapies approved for use against large B-cell lymphomas. Although host inflammatory and tumor microenvironmental markers associate with efficacy and resistance, the tumor-intrinsic alterations underlying these phenomena remain undefined. CD19 mutations associate with resistance but are uncommon, and most patients with relapsed disease retain expression of the wild-type receptor, implicating other genomic mechanisms. We therefore leveraged the comprehensive resolution of whole-genome sequencing to assess 51 tumor samples from 49 patients with CAR-19-treated large B-cell lymphoma. We found that the pretreatment presence of complex structural variants, APOBEC mutational signatures, and genomic damage from reactive oxygen species predict CAR-19 resistance. In addition, the recurrent 3p21.31 chromosomal deletion containing the RHOA tumor suppressor was strongly enriched in patients for whom CAR T-cell therapy failed. Pretreatment reduced expression or monoallelic loss of CD19 did not affect responses, suggesting CAR-19 therapy success and resistance are related to multiple mechanisms. Our study showed that tumor-intrinsic genomic alterations are key among the complex interplay of factors that underlie CAR-19 efficacy and resistance for large B-cell lymphomas.
2665. Subclonal evolution of CLL driver mutations is associated with relapse in ibrutinib- and acalabrutinib-treated patients.
作者: Gage S Black.;Xiaomeng Huang.;Yi Qiao.;Szabolcs Tarapcsak.;Kerry A Rogers.;Shrilekha Misra.;John C Byrd.;Gabor T Marth.;Deborah M Stephens.;Jennifer A Woyach.
来源: Blood. 2022年140卷4期401-405页 2666. Enhanced BNT162b2 vaccine-induced cellular immunity in anti-CD19 CAR T cell-treated patients.
作者: Bernice Ling Zhi Oh.;Nicole Tan.;Ruklanthi de Alwis.;Kamini Kunasegaran.;Zhiwei Chen.;Michelle Poon.;Esther Chan.;Jenny G H Low.;Allen Eng Juh Yeoh.;Antonio Bertoletti.;Nina Le Bert.
来源: Blood. 2022年140卷2期156-160页
Patients receiving CD19 CAR T-cell therapy for relapsed/refractory lymphoma experience prolonged and profound B-cell aplasia and hypogammaglobulinemia, placing them at a higher risk for severe COVID-19. Independently, Oh et al and Atanackovic et al demonstrate that despite attenuated humoral response to mRNA-based vaccines, patients demonstrate normal or heightened functional T-cell responses, including antiviral T-cell activity against SARS-CoV-2 variants including Omicron. Collectively, these data reinforce the importance of COVID-19 vaccination following CD19 CAR T-cell therapy, despite long-term B-cell aplasia.
2667. Procoagulant platelet sentinels prevent inflammatory bleeding through GPIIBIIIA and GPVI.
作者: Rainer Kaiser.;Raphael Escaig.;Jan Kranich.;Marie-Louise Hoffknecht.;Afra Anjum.;Vivien Polewka.;Magdalena Mader.;Wenbo Hu.;Larissa Belz.;Christoph Gold.;Anna Titova.;Michael Lorenz.;Kami Pekayvaz.;Stefan Kääb.;Florian Gaertner.;Konstantin Stark.;Thomas Brocker.;Steffen Massberg.;Leo Nicolai.
来源: Blood. 2022年140卷2期121-139页
Impairment of vascular integrity is a hallmark of inflammatory diseases. We recently reported that single immune-responsive platelets migrate and reposition themselves to sites of vascular injury to prevent bleeding. However, it remains unclear how single platelets preserve vascular integrity once encountering endothelial breaches. Here we demonstrate by intravital microscopy combined with genetic mouse models that procoagulant activation (PA) of single platelets and subsequent recruitment of the coagulation cascade are crucial for the prevention of inflammatory bleeding. Using a novel lactadherin-based compound, we detect phosphatidylserine (PS)-positive procoagulant platelets in the inflamed vasculature. We identify exposed collagen as the central trigger arresting platelets and initiating subsequent PA in a CypD- and TMEM16F-dependent manner both in vivo and in vitro. Platelet PA promotes binding of the prothrombinase complex to the platelet membrane, greatly enhancing thrombin activity and resulting in fibrin formation. PA of migrating platelets is initiated by costimulation via integrin αIIbβ3 (GPIIBIIIA)/Gα13-mediated outside-in signaling and glycoprotein VI signaling, leading to an above-threshold intracellular calcium release. This effectively targets the coagulation cascade to breaches of vascular integrity identified by patrolling platelets. Platelet-specific genetic loss of either CypD or TMEM16F as well as combined blockade of platelet GPIIBIIIA and glycoprotein VI reduce platelet PA in vivo and aggravate pulmonary inflammatory hemorrhage. Our findings illustrate a novel role of procoagulant platelets in the prevention of inflammatory bleeding and provide evidence that PA of patrolling platelet sentinels effectively targets and confines activation of coagulation to breaches of vascular integrity.
2668. Targeted autophagy disruption reveals the central role of macrophage iron metabolism in systemic iron homeostasis.
Iron homeostasis depends on both intracellular control through iron-responsive proteins and the systemic level of iron through hepcidin-ferroportin axis. Indeed, the hormone hepcidin downregulates the ferroportin iron exporter to control iron recycling from macrophages and iron uptake from enterocytes. Here, we focused on the role of autophagy in macrophage iron metabolism and systemic iron homeostasis. Mice deficient for autophagy in macrophages (LysM-Atg5-/-) mimicked a primary iron overload phenotype, resulting in high ferroportin expression in both macrophages and enterocytes that correlated with marked parenchymal iron overload. Furthermore, LysM-Atg5-/- mice exhibited increased hematopoietic activity with no sign of anemia but correlating with rather high plasma iron level. Compared with wild-type cells, bone marrow-derived macrophages from LysM-Atg5-/- mice had significantly increased ferroportin expression and decreased iron content, confirming high iron export. In erythrophagocytic macrophages, autophagy regulates hemosiderin storage mechanisms as well as degradation of ferroportin and subsequently its plasma membrane localization and iron export; furthermore, ferroportin colocalization with hepcidin indicates hepcidin autocrine activity. Relatively high hepatic hepcidin expression and decreased hepcidin level in the spleen of LysM-Atg5-/- mice, correlating with low hemosiderin iron storage, as well as in erythrophagocytic Atg5-/- macrophages were evidenced. Therefore, our results highlight the critical role of autophagy in macrophages for iron trafficking and systemic iron homeostasis. We propose that in macrophages, autophagy restricts ferroportin level and iron export, resulting in hepcidin expression with an autocrine-paracrine effect that plays a role in the regulation of ferroportin expression in duodenal enterocytes.
2669. Risk of vaso-occlusive episode after exposure to corticosteroids in patients with sickle cell disease.
作者: Ondine Walter.;Pierre Cougoul.;Julien Maquet.;Pablo Bartolucci.;Maryse Lapeyre-Mestre.;Margaux Lafaurie.;Guillaume Moulis.
来源: Blood. 2022年139卷26期3771-3777页
Vaso-occlusive episodes (VOEs) are a major concern in patients with sickle cell disease (SCD). Exposure to systemic corticosteroids has been suspected to increase the occurrence of VOEs in case reports or series. No comparative study has been conducted to investigate this risk, which is still debated. Several clinical trials demonstrated the effectiveness of corticosteroids for the treatment of VOEs, but with increased rates of readmission. The aim of the study was to assess the risk of hospitalization for VOE associated with exposure to systemic corticosteroids in patients with SCD. We used a case-case-time-control design in a nationwide population-based cohort built in the French national health insurance database between 2010 and 2018. The population included all patients with SCD with at least 1 hospitalization for VOE. Corticosteroids were identified using out-of-hospital dispensing data. The outcome was the first hospitalization for VOE. The case-case-time-control design induces self-adjustment for time-invariant confounders, including genotype. Analyses were adjusted for time-dependent confounders (infections, red blood transfusions) and stratified by exposure to hydroxyurea. Overall, 5151 patients were included in the main analysis. Corticosteroid exposure was significantly associated with the occurrence of hospitalizations for VOEs: adjusted odds ratio, 3.8; 95% confidence interval [CI], 2.4-5.6). In patients exposed to hydroxyurea, the adjusted odds ratio was 2.6 (95% CI, 1.1-6.4); it was 4.0 (95% CI, 2.5-6.3) in unexposed patients. These results were consistent in children and adults. In conclusion, systemic corticosteroids were associated to an increased risk of hospitalization for VOEs and should be limited in patients with SCD.
2670. Inflammation and myeloid malignancy: quenching the flame.
Chronic inflammation with aging ("inflammaging") plays a prominent role in the pathogenesis of myeloid malignancies. Aberrant inflammatory activity affects many different cells in the marrow, including normal blood and stromal marrow elements and leukemic cells, in unique and distinct ways. Inflammation can promote selective clonal expansion through differential immune-mediated suppression of normal hematopoietic cells and malignant clones. We review these complex roles, how they can be understood by separating cell-intrinsic from extrinsic effects, and how this informs future clinical trials.
2671. NfκB signaling dynamics and their target genes differ between mouse blood cell types and induce distinct cell behavior.
作者: Tobias Kull.;Arne Wehling.;Martin Etzrodt.;Markus Auler.;Philip Dettinger.;Nicola Aceto.;Timm Schroeder.
来源: Blood. 2022年140卷2期99-111页
Cells can use signaling pathway activity over time (ie, dynamics) to control cell fates. However, little is known about the potential existence and function of signaling dynamics in primary hematopoietic stem and progenitor cells (HSPCs). Here, we use time-lapse imaging and tracking of single murine HSPCs from green fluorescent protein-p65/H2BmCherry reporter mice to quantify their nuclear factor κB (NfκB) activity dynamics in response to tumor necrosis factor α and interleukin 1β. We find response dynamics to be heterogeneous between individual cells, with cell type-specific dynamics distributions. Transcriptome sequencing of single cells physically isolated after live dynamics quantification shows activation of different target gene programs in cells with different dynamics. Finally, artificial induction of oscillatory NfκB activity causes changes in granulocyte/monocyte progenitor behavior. Thus, HSPC behavior can be influenced by signaling dynamics, which are tightly regulated during hematopoietic differentiation and enable cell type-specific responses to the same signaling inputs.
2672. Asciminib: a new therapeutic option in chronic-phase CML with treatment failure.
Asciminib, a first-in-class allosteric inhibitor of BCR::ABL1 kinase activity, is now approved for the treatment of patients with chronic-phase chronic myeloid leukemia who failed 2 lines of therapy or in patients with the T315I mutation. Promising attributes include high specificity and potency against BCR::ABL1, activity against most kinase domain mutations, and potential for combination therapy with ATP-competitive tyrosine kinase inhibitors. Clinicians now have expanded third-line options, which in most cases will involve a choice between asciminib and ponatinib.
2673. Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL.
作者: Kristina Müller.;Fotini Vogiatzi.;Dorothee Winterberg.;Thies Rösner.;Lennart Lenk.;Lorenz Bastian.;Carina L Gehlert.;Marie-Pauline Autenrieb.;Monika Brüggemann.;Gunnar Cario.;Martin Schrappe.;Andreas E Kulozik.;Cornelia Eckert.;Anke K Bergmann.;Beat Bornhauser.;Jean-Pierre Bourquin.;Thomas Valerius.;Matthias Peipp.;Christian Kellner.;Denis M Schewe.
来源: Blood. 2022年140卷1期45-57页
Acute lymphoblastic leukemia (ALL) is the most common malignant disease affecting children. Although therapeutic strategies have improved, T-cell acute lymphoblastic leukemia (T-ALL) relapse is associated with chemoresistance and a poor prognosis. One strategy to overcome this obstacle is the application of monoclonal antibodies. Here, we show that leukemic cells from patients with T-ALL express surface CD38 and CD47, both attractive targets for antibody therapy. We therefore investigated the commercially available CD38 antibody daratumumab (Dara) in combination with a proprietary modified CD47 antibody (Hu5F9-IgG2σ) in vitro and in vivo. Compared with single treatments, this combination significantly increased in vitro antibody-dependent cellular phagocytosis in T-ALL cell lines as well as in random de novo and relapsed/refractory T-ALL patient-derived xenograft (PDX) samples. Similarly, enhanced antibody-dependent cellular phagocytosis was observed when combining Dara with pharmacologic inhibition of CD47 interactions using a glutaminyl cyclase inhibitor. Phase 2-like preclinical in vivo trials using T-ALL PDX samples in experimental minimal residual disease-like (MRD-like) and overt leukemia models revealed a high antileukemic efficacy of CD47 blockade alone. However, T-ALL xenograft mice subjected to chemotherapy first (postchemotherapy MRD) and subsequently cotreated with Dara and Hu5F9-IgG2σ displayed significantly reduced bone marrow infiltration compared with single treatments. In relapsed and highly refractory T-ALL PDX combined treatment with Dara and Hu5F9-IgG2σ was required to substantially prolong survival compared with single treatments. These findings suggest that combining CD47 blockade with Dara is a promising therapy for T-ALL, especially for relapsed/refractory disease harboring a dismal prognosis in patients.
2674. Clinical features of thrombosis and bleeding in COVID-19.
Infection with the SARS-CoV-2 virus, resulting in COVID-19 disease, has presented a unique scenario associated with high rates of thrombosis. The risk of venous thrombosis is some three- to sixfold higher than for patients admitted to a hospital for other indications, and for patients who have thrombosis, mortality appears to increase. Thrombosis may be a presenting feature of COVID-19. Pulmonary thrombi are the most frequent events, some related to deep vein thrombosis, but also to in situ microvascular and macrovascular thrombosis. Other venous thromboses include catheter- and circuit-associated in patients requiring hemofiltration and extracorporeal membrane oxygenation. Arterial thrombosis is less commonly documented, with 3% of patients in intensive care units having major arterial strokes and up to 9% having myocardial infarction, both of which are most likely multifactorial. Risk factors for thrombosis above those already documented in hospital settings include duration of COVID-19 symptoms before admission to the hospital. Laboratory parameters associated with higher risk of thrombosis include higher D-dimer, low fibrinogen, and low lymphocyte count, with higher factor VIII and von Willebrand factor levels indicative of more severe COVID-19 infection. All patients should receive thromboprophylaxis when admitted with COVID-19 infection, but the dose and length of treatment are still debated. Thrombosis continues to be treated according to standard VTE guidelines, but adjustments may be needed depending on other factors relevant to the patient's admission.
|