Targeting NLRP3 Inflammasome-Induced Therapy Resistance in ALL

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 46-46
Author(s):  
Zhongbo Hu ◽  
Michael Sporn ◽  
John Letterio

Background: Pediatric acute lymphoblastic leukemia (ALL) is the most common childhood cancer and the most frequent cause of death from cancer before 20 years of age. Survival rates for patients with relapsed ALL remain less than 50% due to the emergence of chemoresistance, particularly steroid resistance. Several lines of evidence suggest the importance of therapy-related activation of the NLRP3 inflammasome in ALL treatment resistance. The NLRP3 inflammasome has been shown to convey steroid resistance, through epigenetic mechanisms leading to NLRP3 promoter hypomethylation and upregulation of NLRP3 expression, and through inflammasome-dependent modulation of the expression of the glucocorticoid receptor. Objectives: The primary objective of this study is to define the kinetics of activation of the NLRP3 inflammasome in ALL patients following systemic chemotherapy. A secondary objective is to determine whether inhibition of the NLRP3 inflammasome enhances the ALL response to conventional chemotherapy in established, patient-derived xenograft (PDX) preclinical models of pediatric ALL. Specifically, we will investigate whether a newly developed synthetic triterpenoid, CDDO-2P-Im (which we have shown to inhibit activation of NLRP3), can suppress the in vivo growth of steroid-resistant human ALL PDX lines and augment their response to the steroid dexamethasone (DEX), and other conventional chemotherapy agents. Methods: We obtained 1-3 ml of peripheral blood from newly diagnosed ALL patients (0-40 years of age) before and after induction chemotherapy and from normal controls without significant inflammatory or infectious diseases under the UH Hospital Cleveland Medical Center IRB-approved protocol STUDY20190453. Either the Ella Simple Plex protein assay or ELISA were used to determine the expression levels of inflammasome associated proteins such as IL-1β, IL-18, caspase-1 and ASC. Western blot was used to analyze the inflammasome signaling proteins such as casepase-1, IL-1β, IL-18, ASC, NLRP3, AIM2, and LonP1 in the cell lysates. Cell viability assay with Cell-Titer-Glo was performed with different leukemia cell lines, such CEM-1, CEM-7, NALP-3 to determine effects of CDDO-2P-Im and DEX on the leukemia growth. Flow cytometry with Annex-V combined with 7-AAD staining was used for apoptosis assay to detect the effect of CDDO-2P-Im and chemotherapy reagents such as DEX. Leukemia PDX models were established to evaluate the in vivo effects of DEX combined with CDDO-2P-Im. Results: Currently 22 ALL patients and 13 controls were included, with plasma samples from 7 ALL patients. The post chemotherapy expression levels of IL-18, which is one of the important inflammasome proteins in the plasma, are significantly elevated compared with pre-chemotherapy. The expression of NLRP3 mRNA and that of downstream proteins caspase-1 and IL-18 in ALL culture supernatants are upregulated after leukemia cells are exposed to doxorubicin and dexamethasone at 12 and 18 hours. The newly synthesized triterpenoid analog CDDO-2P-Im, which inhibits the activation of NLRP3 inflammasome, inhibited the in vitro growth of ALL cells in culture, induced apoptosis of both steroid-sensitive and steroid-resistant leukemia cells, enhanced the anti-leukemia effect of dexamethasone in steroid-resistant leukemia cells and significantly prolonged the survival of mice bearing human PDX lines. Conclusions: Our preliminary data indicate that chemotherapy activates the NLRP3 inflammasome and downstream signals in patients undergoing therapy for ALL. Importantly, the novel triterpenoid analog, CDDO-2P-Im blocks NLRP3 activation, suppresses ALL viability and exhibits significant single agent activity in steroid-resistant ALL. Disclosures No relevant conflicts of interest to declare.

Haematologica ◽  
2021 ◽  
Author(s):  
Karen L. Bride ◽  
Hai Hu ◽  
Anastasia Tikhonova ◽  
Tori J. Fuller ◽  
Tiffaney L. Vincent ◽  
...  

Despite improvements in outcomes for children with B and T-cell acute lymphoblastic leukemia (B-ALL and T-ALL), patients with resistant or relapsed disease fare poorly. Previous studies have demonstrated the essential role of cyclin D3 in T-ALL disease initiation and progression and that targeting of the CDK4/6-cyclin D complex can suppress T-ALL proliferation, leading to efficient cell death in animal models. Studies in leukemia and other malignancies, suggest that schedule is important when combining CDK4/6 inhibitors (CDKis) with cytotoxic agents. Based on these observations, we broadened evaluation of two CDKis, palbociclib (PD-0332991, Pfizer) and ribociclib (LEE011, Novartis) in B and T-ALL as single agent and in combination with conventional cytotoxic chemotherapy, using different schedules in preclinical models. As monotherapy, CDKis caused cell cycle arrest with a significant decrease in S phase entry and were active in vivo across a broad number of patient-derived xenograft samples. Prolonged monotherapy induces resistance, for which we identified a potential novel mechanism using transcriptome profiling. Importantly, simultaneous but not sequential treatment of CDKis with conventional chemotherapy (dexamethasone, L-asparaginase and vincristine) led to improved efficacy compared to monotherapy in vivo. We provide novel evidence that combining CDKis and conventional chemotherapy can be safe and effective. These results led to the rational design of a clinical trial.


2021 ◽  
Vol 12 ◽  
Author(s):  
Pengxiao Chen ◽  
Qi Bai ◽  
Yanting Wu ◽  
Qiongzhen Zeng ◽  
Xiaowei Song ◽  
...  

Artemisia argyi H. Lév. and Vaniot is a traditional medical herb that has been used for a long time in China and other Asian counties. Essential oil is the main active fraction of Artemisia argyi H. Lév. and Vaniot, and its anti-inflammatory potential has been observed in vitro and in vivo. Here, we found that the essential oil of Artemisia argyi H. Lév. and Vaniot (EOAA) inhibited monosodium urate (MSU)- and nigericin-induced NLRP3 inflammasome activation. EOAA suppressed caspase-1 and IL-1β processing and pyroptosis. NF-κB p65 phosphorylation and translocation were also inhibited. In addition, EOAA suppressed nigericin-induced NLRP3 inflammasome activation without blocking ASC oligomerization, suggesting that it may inhibit NLRP3 inflammasome activation by preventing caspase-1 processing. Our study thus indicates that EOAA inhibits NLRP3 inflammasome activation and has therapeutic potential against NLRP3-driven diseases.


2020 ◽  
Author(s):  
Chuan-jiang Liu ◽  
Qiang Fu ◽  
Wenjing Zhou ◽  
Xu Zhang ◽  
Rui Chen ◽  
...  

Abstract Background: Methylprednisolone (MP) is a synthetic corticosteroid with potent anti-inflammatory and antioxidant properties used as therapy for a variety of diseases. The underlying mechanism of MP to reduce acute pancreatitis still needs to be elucidated.Methods: Twenty-four male C57BL/6 mice (6-8 weeks) were used to establish SAP mouse model by administering an intraperitoneal injection of Cae and LPS. Amylase expression levels of serum and PLF were measured with an amylase assay kit. The concentrations of IL-1β and TNF-α in the serum and PLF were detected by ELISA. The level of pancreatic and lung tissue damage and inflammation was assessed by H&E staining and immunofluorescence staining. Western blot and qPCR were used to detect the expression levels of NLRP3, IL-1β and TNF-αin vivo and in vitro.Results: In this study, we found MP, used in the early phase of SAP, decreased the levels of IL-1β and TNF-α in serum and peritoneal lavage fluids (PLF), reduced the level of serum amylase and the expression of MPO in lung tissue, attenuated the pathological injury of the pancreas and lungs in a dose-dependent manner. The expression of NLRP3 and IL-1β in pancreas and lungs was down-regulated significantly depending on the MP concentration. In vitro, MP reduced the levels of IL-1β and TNF-α by down-regulating the expression of NLRP3, IL-1β and p-NF-κB in isolated peritoneal macrophages. Conclusion: MP can attenuate the injury of pancreas and lungs, and the inflammatory response in SAP mice by down-regulating the activation of NF-κB and the NLRP3 inflammasome.


2011 ◽  
Vol 35 (6) ◽  
pp. 800-807 ◽  
Author(s):  
Johan Jansson ◽  
Yu-Chiao Hsu ◽  
Igor I. Kuzin ◽  
Andrew Campbell ◽  
Craig A. Mullen

2019 ◽  
Vol 2019 ◽  
pp. 1-16 ◽  
Author(s):  
Bin Leng ◽  
Yingjie Zhang ◽  
Xinran Liu ◽  
Zhen Zhang ◽  
Yang Liu ◽  
...  

Long-term exposure to high glucose induces vascular endothelial inflammation that can result in cardiovascular disease. Astragaloside IV (As-IV) is widely used for anti-inflammatory treatment of cardiovascular diseases. However, its mechanism of action is still not fully understood. In this study, we investigated the effect of As-IV on high glucose-induced endothelial inflammation and explored its possible mechanisms. In vivo, As-IV (40 and 80 mg/kg/d) was orally administered to rats for 8 weeks after a single intraperitoneal injection of streptozotocin (STZ, 65 mg/kg). In vitro, human umbilical vein endothelial cells (HUVECs) were treated with high glucose (33 mM glucose) in the presence or absence of As-IV, NPS2143 (CaSR inhibitor), BAY 11-7082 (NF-κB p65 inhibitor), and INF39 (NLRP3 inhibitor), and overexpression of CaSR was induced by infection of CaSR-overexpressing lentiviral vectors to further discuss the anti-inflammatory property of As-IV. The results showed that high glucose increased the expression of interleukin-18 (IL-18), interleukin-1β (IL-1β), NLRP3, caspase-1, and ASC, as well as the protein level of TLR4, nucleus p65, and CaSR. As-IV can reverse these changes in vivo and in vitro. Meanwhile, NPS2143, BAY 11-7082, and INF39 could significantly abolish the high glucose-enhanced NLRP3, ASC, caspase-1, IL-18, and IL-1β expression in vitro. In addition, both NPS2143 and BAY 11-7082 attenuated high glucose-induced upregulation of NLRP3, ASC, caspase-1, IL-18, and IL-1β expression. In conclusion, this study suggested that As-IV could inhibit high glucose-induced NLRP3 inflammasome activation and subsequent secretion of proinflammatory cytokines via inhibiting TLR4/NF-κB signaling pathway and CaSR, which provides new insights into the anti-inflammatory activity of As-IV.


2015 ◽  
Vol 8 (1) ◽  
Author(s):  
Ilaria Iacobucci ◽  
Andrea Ghelli Luserna Di Rorà ◽  
Maria Vittoria Verga Falzacappa ◽  
Claudio Agostinelli ◽  
Enrico Derenzini ◽  
...  

2016 ◽  
Vol 64 (3) ◽  
pp. 740-744 ◽  
Author(s):  
Olivia L Francis ◽  
Terry-Ann M Milford ◽  
Cornelia Beldiman ◽  
Kimberly J Payne

Many leukemias are characterized by well-known mutations that drive oncogenesis. Mice engineered with these mutations provide a foundation for understanding leukemogenesis and identifying therapies. However, data from whole genome studies provide evidence that malignancies are characterized by multiple genetic alterations that vary between patients, as well as inherited genetic variation that can also contribute to oncogenesis. Improved outcomes will require precision medicine approaches–targeted therapies tailored to malignancies in each patient. Preclinical models that reflect the range of mutations and the genetic background present in patient populations are required to develop and test the combinations of therapies that will be used to provide precision medicine therapeutic strategies. Patient-derived xenografts (PDX) produced by transplanting leukemia cells from patients into immune deficient mice provide preclinical models where disease mechanisms and therapeutic efficacy can be studied in vivo in context of the genetic variability present in patient tumors. PDX models are possible because many elements in the bone marrow microenvironment show cross-species activity between mice and humans. However, several cytokines likely to impact leukemia cells are species-specific with limited activity on transplanted human leukemia cells. In this review we discuss the importance of PDX models for developing precision medicine approaches to leukemia treatment. We illustrate how PDX models can be optimized to overcome a lack of cross-species cytokine activity by reviewing a recent strategy developed for use with a high-risk form of B-cell acute lymphoblastic leukemia (B-ALL) that is characterized by overexpression of CRLF2, a receptor component for the cytokine, TSLP.


Blood ◽  
2015 ◽  
Vol 125 (2) ◽  
pp. 273-283 ◽  
Author(s):  
Duohui Jing ◽  
Vivek A. Bhadri ◽  
Dominik Beck ◽  
Julie A. I. Thoms ◽  
Nurul A. Yakob ◽  
...  

Key Points The glucocorticoid receptor coordinately regulates the antiapoptotic BCL2 and proapoptotic BIM genes in pediatric ALL cells in vivo. GR binding at a novel intronic region is associated with BIM transcription and dexamethasone sensitivity in pediatric ALL cells in vivo.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4682-4682
Author(s):  
Jason Lickliter ◽  
Chris Arthur ◽  
James D’Rozario ◽  
Chi-hung Hui ◽  
Jeffrey Szer ◽  
...  

Abstract CML in blast crisis (BC) and Philadelphia chromosome-positive (Ph+) ALL are incurable with conventional chemotherapy and in most cases respond only transiently to the Bcr-Abl kinase inhibitor imatinib mesylate. We hypothesized that pretreatment with imatinib would overcome inherent drug resistance to induction chemotherapy mediated by Bcr-Abl, and evaluated the combination in 17 patients with Ph+ acute leukemias (5 CML in BC, 6 de-novo Ph+ ALL, 6 relapsed Ph+ ALL). All patients received a 1-week prephase of imatinib alone at 600 mg daily. In CML in BC and relapsed Ph+ ALL, imatinib was continued concomitantly with induction chemotherapy (standard-dose idarubicin/ara-C with vincristine/prednisone added for lymphoid leukemias) for an additional week. Imatinib was then ceased, but recommenced as a single agent after blood-count recovery. Patients with de-novo Ph+ ALL received the imatinib prephase and then commenced the French LALA94 protocol without concomitant imatinib. Neutrophils recovered to ≥ 1.0 x 109/L at a median of day 28 (range 21–56) of induction chemotherapy. During induction, 1 patient died from sepsis and multi-organ failure, and 1 patient died on day 7 from CNS leukemia. Subsequently, 1 patient died on day 37 from massive hemoptysis and 1 patient developed pulmonary fibrosis and died on day 144. Among 14 evaluable patients, combined imatinib and chemotherapy induction resulted in 9 (64%) complete hematological responses and there were 5 complete and 3 major cytogenetic responses. Notably, 2 out of 3 patients with CML in myeloid BC achieved major cytogenetic responses. The in-vivo effects of imatinib on Bcr-Abl activity were monitored in leukemic blasts during the imatinib-only prephase. Western blots revealed a decrease in CrkL phosphorylation to < 25% of the pretreatment level in peripheral-blood samples from 5 out of 6 evaluable cases and in marrow samples from 1 of 2 evaluable cases. Bcl-xL expression levels revealed no consistent in-vivo responses to imatinib. However, all 7 cases evaluable for Bcl-2 protein showed strong expression levels, which were unaffected by imatinib in 5 cases and decreased in 2 cases. Imatinib potently inhibited Bcr-Abl activity in vivo in Ph+ acute leukemic blasts and its combination with induction chemotherapy was tolerable and shows promising activity.


Sign in / Sign up

Export Citation Format

Share Document