scholarly journals The metastasis suppressor protein NM23-H1 modulates the PI3K-AKT axis through interaction with the p110α catalytic subunit

Oncogenesis ◽  
2021 ◽  
Vol 10 (4) ◽  
Author(s):  
Francesco Paolo Pennino ◽  
Masanao Murakami ◽  
Massimo Zollo ◽  
Erle S. Robertson

AbstractThe PI3K pathway is one of the most deregulated pathways in cancer, which is predominantly due to gain of function mutations or altered expression of the PI3KCA gene. This is codified by what is seen for the class I PI3K catalytic subunit p110α, a common feature of many cancers. The metastasis suppressor protein NM23-H1 (NME1), whose ability to suppress the metastasis activities of different tumors has been widely described and was previously reported to alter phosphatidylinositol signaling. Here, we show interaction of NM23-H1 with the p110α subunit and the functional consequence of this interaction. This interaction is predominantly localized at the plasma membrane with some signals seen in the cytoplasmic compartment. Analysis of NM23-H1 levels showed a negative correlation between NM23-H1 expression and Akt phosphorylation, the key marker of PI3K pathway activation. Investigating the functional consequence of this interaction using cell motility and clonogenicity assays showed that expression of NM23-H1 reversed the enhanced migration, invasion, adhesion, and filopodia structure formation in cells expressing the p110α catalytic subunit. A similar trend was seen in anchorage-independent assays. Notably, differential analyses using NM23-H1 mutants which lacked the enzymatic and metastasis suppressor activity, showed no detectable interaction between p110α and the NM23-H1 mutant proteins P96S, H118F, and S120G, as well as no dysregulation of the PI3K-AKT axis.

Blood ◽  
2012 ◽  
Vol 119 (8) ◽  
pp. 1897-1900 ◽  
Author(s):  
Sarah A. Meadows ◽  
Francisco Vega ◽  
Adam Kashishian ◽  
Dave Johnson ◽  
Volker Diehl ◽  
...  

Abstract GS-1101 (CAL-101) is an oral PI3Kδ-specific inhibitor that has shown preclinical and clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. To investigate the potential role of PI3Kδ in Hodgkin lymphoma (HL), we screened 5 HL cell lines and primary samples from patients with HL for PI3Kδ isoform expression and constitutive PI3K pathway activation. Inhibition of PI3Kδ by GS-1101 resulted in the inhibition of Akt phosphorylation. Cocultures with stroma cells induced Akt activation in HL cells, and this effect was blocked by GS-1101. Conversely, production of the stroma-stimulating chemokine, CCL5, by HL cells was reduced by GS-1101. GS-1101 also induced dose-dependent apoptosis of HL cells at 48 hours. Reductions in cell viability and apoptosis were enhanced when combining GS-1101 with the mTOR inhibitor everolimus. Our findings suggest that excessive PI3Kδ activity is characteristic in HL and support clinical evaluation of GS-1101, alone and in combination, as targeted therapy for HL.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Ziying Lin ◽  
Lixia Huang ◽  
Shao Li Li ◽  
Jincui Gu ◽  
Xiaoxian Cui ◽  
...  

Abstract Background Recent evidences had shown that loss in phosphatase and tensin homolog deleted on chromosome 10 (PTEN) was associated with immunotherapy resistance, which may be attributed to the non-T-cell-inflamed tumor microenvironment. The impact of PTEN loss on tumor microenvironment, especially regarding T cell infiltration across tumor types is not well understood. Methods Utilizing The Cancer Genome Atlas (TCGA) and publicly available dataset of immunotherapy, we explored the correlation of PTEN expressing level or genomic loss with tumor immune microenvironment and response to immunotherapy. We further investigated the involvement of PI3K-AKT-mTOR pathway activation, which is known to be the subsequent effect of PTEN loss, in the immune microenvironment modulation. Results We reveal that PTEN mRNA expression is significantly positively correlated with CD4/CD8A gene expression and T cells infiltration especially T helpers cells, central memory T cell and effector memory T cells in multiples tumor types. Genomic loss of PTEN is associated with reduced CD8+ T cells, type 1 T helper cells, and increased type 2 T helper cells, immunosuppressed genes (e.g. VEGFA) expression. Furthermore, T cell exclusive phenotype is also observed in tumor with PI3K pathway activation or genomic gain in PIK3CA or PIK3CB. PTEN loss and PI3K pathway activation correlate with immunosuppressive microenvironment, especially in terms of T cell exclusion. PTEN loss predict poor therapeutic response and worse survival outcome in patients receiving immunotherapy. Conclusion These data brings insight into the role of PTEN loss in T cell exclusion and immunotherapy resistance, and inspires further research on immune modulating strategy to augment immunotherapy.


Cells ◽  
2019 ◽  
Vol 8 (5) ◽  
pp. 442 ◽  
Author(s):  
Ana Raquel-Cunha ◽  
Diana Cardoso-Carneiro ◽  
Rui M. Reis ◽  
Olga Martinho

Lung cancer is the most deadly neoplasm with the highest incidence in both genders, with non-small cell lung cancer (NSCLC) being the most frequent subtype. Somatic mutations within the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene are key drivers of NSCLC progression, with EGFR inhibitors being particularly beneficial for patients carrying the so-called “EGFR-sensitizing mutations”. However, patients eventually acquire resistance to these EGFR inhibitors, and a better knowledge of other driven and targetable proteins will allow the design of increasingly accurate drugs against patients’ specific molecular aberrations. Raf kinase inhibitory protein (RKIP) is an important modulator of relevant intracellular signaling pathways, including those controlled by EGFR, such as MAPK. It has been reported that it has metastasis suppressor activity and a prognostic role in several solid tumors, including lung cancer. In the present review, the potential use of RKIP in the clinic as a prognostic biomarker and predictor of therapy response in lung cancer is addressed.


2017 ◽  
Vol 216 (12) ◽  
pp. 4255-4270 ◽  
Author(s):  
Brajendra K. Tripathi ◽  
Tiera Grant ◽  
Xiaolan Qian ◽  
Ming Zhou ◽  
Philipp Mertins ◽  
...  

We report several receptor tyrosine kinase (RTK) ligands increase RhoA–guanosine triphosphate (GTP) in untransformed and transformed cell lines and determine this phenomenon depends on the RTKs activating the AKT serine/threonine kinase. The increased RhoA-GTP results from AKT phosphorylating three serines (S298, S329, and S567) in the DLC1 tumor suppressor, a Rho GTPase-activating protein (RhoGAP) associated with focal adhesions. Phosphorylation of the serines, located N-terminal to the DLC1 RhoGAP domain, induces strong binding of that N-terminal region to the RhoGAP domain, converting DLC1 from an open, active dimer to a closed, inactive monomer. That binding, which interferes with the interaction of RhoA-GTP with the RhoGAP domain, reduces the hydrolysis of RhoA-GTP, the binding of other DLC1 ligands, and the colocalization of DLC1 with focal adhesions and attenuates tumor suppressor activity. DLC1 is a critical AKT target in DLC1-positive cancer because AKT inhibition has potent antitumor activity in the DLC1-positive transgenic cancer model and in a DLC1-positive cancer cell line but not in an isogenic DLC1-negative cell line.


2009 ◽  
Vol 126 (5) ◽  
pp. 1121-1131 ◽  
Author(s):  
Elena López-Knowles ◽  
Sandra A. O'Toole ◽  
Catriona M. McNeil ◽  
Ewan K.A. Millar ◽  
Min R. Qiu ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 286-286 ◽  
Author(s):  
Brian J Lannutti ◽  
Sarah A Meadows ◽  
Adam Kashishian ◽  
Bart Steiner ◽  
Galina Pogosov ◽  
...  

Abstract Abstract 286 It is well established that deregulation of the PI3K signaling pathway plays an important role in the etiology of human malignancies including those of hematologic origin. In 30–50% of solid tumors, oncogenic activation of the PI3K pathway is the result of gain-of-function mutations in the PI3K p110α isoform or due to the loss-of-function of the PTEN phosphatase that is responsible for PI3K downregulation. In B cell malignancies these mutations are rarely observed in spite of the fact that PI3K pathway activation is commonly observed and often essential for tumor cell growth and survival. In this case, PI3K pathway activation has been shown to result from constitutive B cell receptor (BCR) activation and/or from exposure to survival factors present in the microenvironment. The activation of the PI3K pathway by cell surface receptors is directly mediated by the Class I isoforms (α, β, δ, and γ), however, their relative contribution in B cell tumors is unknown. Interestingly, genetic and pharmacological approaches that specifically inactivate the p110δ isoform have demonstrated its important role in normal B cell signaling in response to multiple factors including antigen, CD40L, BAFF, SDF-1 and CXCR13 making it an attractive target for therapeutic intervention in B cell malignancies. CAL-101 is an oral p110δ specific inhibitor which is currently being evaluated in a phase I clinical trial for the treatment of patients with select hematologic malignancies. This compound is a novel potent p110δ inhibitor with an IC50 of 2.5 nM against purified p110δ and EC50 of 65 nM in p110δ-mediated basophil activation in whole blood. CAL-101 demonstrates 300-, 200-, and 40-fold selectivity over the other class I family members (α, β, and, γ respectively) and no activity against class II and III PI3K family members or other PI3K-related proteins including mTOR and DNA-PK. Furthermore, a kinome-wide screen failed to detect activity against any additional kinases. To investigate the potential role of p110δ in B cell hematologic tumors we screened a wide range of leukemia and lymphoma cell lines for constitutive PI3K pathway activation. The expression of p110δ was observed in >90% of these cell lines and in many cases was accompanied by constitutive Akt phosphorylation. In this context, CAL-101 was able to reduce p-Akt levels and block additional downstream effectors such as p-S6, and GSK-3β in cells that represent a range of tumor types including follicular lymphoma, acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma, and mantle cell lymphoma (MCL). Furthermore, treatment with CAL-101 resulted in growth suppression and induction of apoptosis which was accompanied by PARP and caspase-3 cleavage. Growing evidence suggests that signals from the microenviroment are essential for the expansion, homing, and survival of malignant B cells, in addition to promoting resistance to conventional drug therapy. To investigate the potential role p110δ plays during B cell signaling via interactions with the microenvironment, we examined invoked stimulation of leukemia and lymphoma cell lines with CXCL12, CXCL13, BAFF, or BCR crosslinking in the presence or absence of CAL-101. Stimulation with either chemokines or growth factors resulted in the phosphorylation of Akt which was inhibited by CAL-101 in a dose dependent manner. Moreover, p110δ inhibition with CAL-101 inhibits the chemotaxis of ALL and MCL cell lines to CXCL12. These studies have now been extended to the analysis of primary patient B-ALL and MCL cells to further establish preclinical proof of concept for therapeutic application of CAL-101. In summary, CAL-101 is a potent and selective p110δ kinase inhibitor with broad anti-tumor activity against cancer cells of hematologic origin. Our results demonstrate that selective inhibition of p110δ with CAL-101 inhibits malignant B cell growth, survival, and migration. Furthermore, p110δ inhibition may enhance the effect of cytotoxic drugs or overcome cell mediated drug resistance by inhibiting the protective signals of the microenviroment, providing a rational for combination therapy. These data suggest that p110δ may play an important role in regulating signals between malignant B cells and their microenvironment thereby providing the preclinical rationale for clinical evaluation of CAL-101 alone or in combination with cytotoxics or biologics in patients with hematologic malignancies. Disclosures: Lannutti: Calistoga Pharmaceuticals: Employment. Meadows:Calistoga Pharmaceuticals: Employment. Kashishian:Calistoga Pharmaceuticals: Employment. Steiner:Calistoga Pharmaceuticals: Employment. Johnson:Calistoga Pharmaceuticals: Research Funding. Giese:Calistoga Pharmaceuticals: Employment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1493-1493 ◽  
Author(s):  
Anas Younes ◽  
Gilles Salles ◽  
Giovanni Martinelli ◽  
R. Gregory Bociek ◽  
Dolores Caballero Barrigon ◽  
...  

Abstract Background: Patients (pts) with relapsed or refractory (R/R) non-Hodgkin's lymphoma (NHL), including DLBCL, MCL, and FL, have poor outcomes and few therapy options. Activation of the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway plays an important role in NHL pathogenesis. The relative expression of aberrant PI3K isoforms can change over the course of disease: for example in MCL, PI3Kd (frequently overexpressed in B-cell malignancies) is more common initially, while PI3Kα plays a more significant role in disease progression and has been identified as an escape mechanism to selective PI3Kd inhibition (Iyengar et al. 2013). Therefore, an agent that targets multiple PI3K isoforms might elicit more durable responses, particularly in later stages of disease. The oral pan-class I PI3K inhibitor buparlisib (BKM120) showed activity in DLBCL cells in vitro (decreased phosphorylation of downstream PI3K signaling effectors, reduced cell proliferation and survival (Zang et al. 2014)) and has shown clinical activity in solid tumors. This study evaluated the safety and efficacy of buparlisib in pts with R/R NHL. Methods: In this multicenter, parallel-arm, open-label, global Phase II study (NCT01693614), pts with DLBCL, MCL, or FL received buparlisib (100 mg QD) continuously until progression, intolerance, or withdrawal of consent. Key inclusion criteria were R/R disease following ≥1 prior therapy, ≥1 measurable lesion per local assessment using standard criteria (Cheson et al. 2007), and Eastern Cooperative Oncology Group (ECOG) performance status ≤2. Pts with DLBCL were eligible if they had received or were ineligible for autologous stem cell transplant (ASCT). The primary endpoint was overall response rate (ORR). Secondary endpoints were progression-free survival (PFS), duration of response, overall survival, safety, and tolerability. PI3K pathway activation status, an exploratory endpoint, was assessed by mutation analysis (PIK3CA, PTEN) and immunohistochemistry (PTEN loss) in archival tumor tissue samples. Results for the 6-month primary efficacy and safety analysis of buparlisib in pts with DLBCL and FL were previously presented (Younes et al. ASH 2014). Here, we report results for pts enrolled in the MCL cohort. Results: At this 6-month primary analysis (data cut-off, February 25, 2015), 22 pts with MCL were enrolled. The median age was 68.5 years (range 47-79 years). The median number of prior therapies was 2 (range 1-6), which included intensive combination chemoimmunotherapy regimens, radiotherapy, and/or ASCT. The median duration of exposure was 20.6 weeks (range 1.7-54.1 weeks). Overall, 17 (77.3%) pts discontinued study treatment for the following reasons: 9 (40.9%) adverse events (AEs); 5 (22.7%) disease progressions, 2 (9.1%) protocol deviations, and 1 (4.5%) physician decision. Four pts died during the study: 1 due general deterioration and 3 due to underlying disease (with 1 on-treatment death due to disease progression). The most common AEs regardless of causality were hyperglycemia (n=10 [45.5%]), and fatigue, anxiety, depression, and weight decrease (n=7 [31.8%] each). The most frequent grade 3/4 AEs were asthenia, confusional state, leukopenia, and febrile neutropenia (n=2 [9.1%] each). Abnormal laboratory values grade 3/4 included elevated levels of fasting glucose (18.2%), aspartate transaminase and alanine transaminase (4.5% each), and neutropenia (18.2%). ORR was 22.7% (95% CI, 7.8-45.4), with 1 (4.5%) complete response and 4 (18.2%) partial responses (Figure 1). Thirteen (59.1%) pts had stable disease. The disease control rate was 81.8% (95% CI, 59.7-94.8). With 7 PFS events, the median PFS was 11.3 months (95% CI, 3.8-not estimable).The estimated PFS rate at 6 months was 68.6% (95% CI, 39.8-85.7). Of the 11 pts with evaluable samples, none showed PI3K pathway activation at baseline. Conclusions: Buparlisib treatment was moderately well tolerated, had an acceptable safety profile, and elicited sustained reduction in tumor burden in pts with R/R MCL with a median PFS of 11.3 months. Despite the limited ORR (22.7%), the observed disease control with targeted inhibition of all 4 PI3K isoforms in this cohort of pts with MCL is encouraging, and warrants further exploration of buparlisib in pts with NHL. Updated results for the DLBCL and FL cohorts will be presented at this meeting. Disclosures Younes: Celgene: Honoraria; Sanofi-Aventis: Honoraria; Bayer: Honoraria; Bristol Meyer Squibb: Honoraria; Curis: Research Funding; Novartis: Research Funding; Incyte: Honoraria; Janssen: Honoraria; Takeda Millenium: Honoraria; Seattle Genetics: Honoraria, Research Funding; Johnson and Johnson: Research Funding. Salles:Roche: Consultancy, Honoraria, Research Funding; Janssen: Honoraria; Gilead: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Gerecitano:AbbVie: Consultancy, Other: Advisory Board; Genentech: Consultancy, Other: Advisory Board. Herbst:Novartis: Employment. Williams:Novartis: Employment. Mukherjee:Novartis: Employment. Tavorath:Novartis: Employment, Equity Ownership.


Author(s):  
Helga B. Salvesen ◽  
Henrica Maria Werner ◽  
Camilla Krakstad

Alterations in PI3K signaling are common in gynecologic malignancies. Alterations detected vary with gynecologic cancer type, histologic subtypes within these, and clinical phenotypes. The distinction into type I and type II endometrial and ovarian carcinomas is reflected in distribution of changes detected in several of the PI3K members. PIK3CA mutations and amplifications are common in endometrial, ovarian, and cervical cancers. PTEN mutations and deletions are frequent in endometrial cancers. Several immunohistochemical studies of protein expression have explored these and other potential surrogate markers for PI3K pathway activation. Biomarkers to measure level of PI3K activity in clinical samples are not established. Whether amplifications, mutations, and deletions of the PI3K pathway members, and in particular change in their expression levels, result in clinically relevant pathway activation needs to be further explored. Also, to what extent these alterations drive the tumor behavior and are critical targets for therapeutics to improve patient survival needs to be further tested to establish predictive biomarkers for response to PI3K inhibition.


Sign in / Sign up

Export Citation Format

Share Document