CAL-101, a Potent Selective Inhibitor of the p110δ Isoform of Phosphatidylinositol 3-kinase, Attenuates Pathway Signaling, Induces Apoptosis, and Overcomes Signals From the Microenvironment In Cellular Models of Hodgkin Lymphoma

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3926-3926 ◽  
Author(s):  
Sarah A Meadows ◽  
Adam Kashishian ◽  
Dave Johnson ◽  
Volker Diehl ◽  
Brian Lannutti

Abstract Abstract 3926 Phosphatidylinositide 3-kinases (PI3Ks) are a family of lipid kinases that are involved in signaling events which control a diverse number of cellular processes. The class I kinases contain 4 isoforms designated p110α, β, δ, γ, and are activated by cell surface receptors. Aberrant regulation of the PI3K signaling pathway is frequently observed in human malignancies including those of hematological origin. CAL-101 is an oral p110δ-specific inhibitor which has shown preclinical and clinical activity in non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL). This compound is a potent p110δ inhibitor (EC50 of 65 nM in a whole-blood assay) with >200-fold selectivity over the other class I PI3K isoforms and no activity against Class II and III PI3K family members or other PI3K-related proteins, including mTOR and DNA-PK. Prior in vitro NHL studies revealed that CAL-101 induces caspase-dependent apoptosis, and inhibits CD40L-, BAFF-, CXCL12- and CXCL13-derived survival signals in cellular models (Lannutti BJ, et al., Blood 2010). To investigate the potential role of p110δ in Hodgkin lymphoma (HL) we screened a number of HL cell lines for p110δ isoform expression and constitutive PI3K pathway activation. We report high levels of p110δ protein and activated Akt in 5 of 5 HL cell lines evaluated (L428, L540, L591, L1236, KM-H2). Inhibition of p110δ with CAL-101 treatment of cell lines resulted in a reduction of Akt phosphorylation and a decrease in cellular viability. Because previous studies have established the importance of signals from the microenvironment for the survival and proliferation of malignant cells as well as for their resistance to standard therapies, we investigated the effect of p110δ inhibition by CAL-101 in HL cell line-stroma cocultures. In this setting, CAL-101 overcame tumor cell growth induced by coculture of HL cells with bone marrow stromal cells. In addition, CAL-101 induced dose-dependent apoptosis of HL cells at 48 hours. Furthermore, stromal cell coculture resulted in increased CCL5, CCL17, and CCL22 levels; productions of these chemokines by HL cells cultured in the presence of stromal cells were reduced by CAL-101 in a dose-dependent manner. These results indicate that specific inhibition of p110δ may disrupt signals between HL cells and their microenvironment, thereby providing the preclinical rationale for clinical evaluation of CAL-101 as a novel therapeutic approach in patients with Hodgkin lymphoma. Disclosures: Meadows: Calistoga Pharmaceuticals: Employment. Kashishian:Calistoga Pharmaceuticals: Employment. Johnson:Calistoga Pharmaceuticals: Employment. Lannutti:Calistoga Pharmaceutical Inc.: Employment.

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.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3169-3169
Author(s):  
Hugh Kikuchi ◽  
Amofa Eunice ◽  
Maeve McEnery ◽  
Farzin Farzaneh ◽  
Stephen A Schey ◽  
...  

Abstract Despite of newly developed and more efficacious therapies, multiple myeloma (MM) remains incurable as most patient will eventually relapse and become refractory. The bone marrow (BM) microenvironment provides niches that are advantageous for drug resistance. Effective therapies against MM should ideally target the various protective BM niches that promote MM cell survival and relapse. In addition to stromal mesenchymal/myofibroblastic cells, osteoclasts play a key supportive role in MM cell viability. Additionally, 80% of patients develop osteolytic lesions, which is a major cause of morbidity. Increased osteoclast activity is characteristic in these patients and targeting osteoclast function is desirable to improve therapies against MM. Osteoclasts need to form an F-actin containing ring along the cell margin that defines a resorbing compartment where protons and degradative enzymes are secreted for dissolution of bone mineral. Remodelling of F-actin and vesicle secretion are regulated by the class IA PI3K pathway during osteoclastic bone resorption. Additionally, it has recently been shown that inhibition of the class IA PI3K pathway in MM cells with GDC0941 induces apoptosis-mediated killing. We hypothesised that GDC0941 could be used as a therapeutic agent to overcome MM-induced osteoclast activation. GDC0941 inhibited maturation of osteoclasts derived from BM aspirates from MM patients in a dose dependent manner. This correlated with decreased bone resorption of osteoclasts cultured on dentine discs. Exposure of mature osteoclasts to GC0941 resulted in abnormal organisation of larger F-actin rings, suggesting a negative effect on the dynamics of the actin cytoskeleton required for bone resorption. We also found that GDC-0941 can prevent protection of the MM cell lines MM1.S and MM1.R by osteoclasts against killing. GDC-0941 alone blocked MM cell proliferation independently of the presence of BM stromal cells and synergised with other therapeutic agents including Lenalidomide, Pomalidomide, Bortezomid and Dexamethasone. We also found that in the presence of MM cells, Dexamethasone (a drug commonly used alone or in combination with new drugs against MM) induced the proliferation of BM stromal cells and adhesion of MM cells on this protective stroma in a dose dependent manner. Dexamethasone is highly effective at MM cell killing when cells are cultured alone. However, we found that at low doses (below 1 uM) and in the presence of BM stromal cells, Dexamethasone could induce MM cell proliferation. GDC0941 enhanced Dexamethasone killing even in the presence of BM stromal cells by blocking Dexamethasone-induced stromal cell proliferation and adhesion of MM cells on the stroma. Targeting individual the PI3K Class IA isoforms alpha, beta, delta or gamma proved to be a less efficient strategy to enhance Dexamethasone killing. Previous work has shown that efficacy of targeting individual PI3K Class I A isoforms would be low for activation of caspases in MM cells as it would be dependent on relative amounts of isoforms expressed by the MM patient. GDC-0941 also inhibited the proliferation of MM1.R and RPMI8266 MM cell lines, which are less sensitive to treatment to Dexamethasone. Co-culture of MM cells with BM stromal cells induced the secretion of IL-10, IL-6, IL-8, MCP-1 and MIP1-alpha. The dose-dependant increased proliferation of Dexamethasone-treated MM cells in the presence of the BM stroma correlated with the pattern of secretion of IL-10 (a cytokine that can induce B-cell proliferation) and this was blocked by the combination of Dexamethasone with GDC0941. GDC-0941 alone or in combination with Dexamethasone was more efficacious at inducing MM cell apoptosis in the presence of the BM stroma cells vs treatment of MM cells alone. These are very encouraging results as they suggest that GDC-0941 in combination with Dexamethasone would be potentially highly efficacious for targeting MM cells in the BM microenvironment. We are currently performing in vivo data using C57BL/KaLwRij mice injected with 5T33-eGFP MM cells that will be discussed at the meeting. We propose that MM patients with active bony disease may benefit from treatment with GDC0941 alone or in combination with currently used therapeutic drugs against MM. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 223 (2) ◽  
pp. 203-216 ◽  
Author(s):  
Yoshihiro Joshua Ono ◽  
Yoshito Terai ◽  
Akiko Tanabe ◽  
Atsushi Hayashi ◽  
Masami Hayashi ◽  
...  

Dienogest, a synthetic progestin, has been shown to be effective against endometriosis, although it is still unclear as to how it affects the ectopic endometrial cells. Decorin has been shown to be a powerful endogenous tumor repressor acting in a paracrine fashion to limit tumor growth. Our objectives were to examine the direct effects of progesterone and dienogest on the in vitro proliferation of the human ectopic endometrial epithelial and stromal cell lines, and evaluate as to how decorin contributes to this effect. We also examined DCN mRNA expression in 50 endometriosis patients. The growth of both cell lines was inhibited in a dose-dependent manner by both decorin and dienogest. Using a chromatin immunoprecipitation assay, it was noted that progesterone and dienogest directly induced the binding of the decorin promoter in the EMOsis cc/TERT cells (immortalized human ovarian epithelial cells) and CRL-4003 cells (immortalized human endometrial stromal cells). Progesterone and dienogest also led to significant induced cell cycle arrest via decorin by promoting production of p21 in both cell lines in a dose-dependent manner. Decorin also suppressed the expression of MET in both cell lines. We confirmed that DCN mRNA expression in patients treated with dienogest was higher than that in the control group. In conclusion, decorin induced by dienogest appears to play a crucial role in suppressing endometriosis by exerting anti-proliferative effects and inducing cell cycle arrest via the production of p21 human ectopic endometrial cells and eutopic endometrial stromal cells.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3919-3919
Author(s):  
Alfons Navarro ◽  
Tania Díaz ◽  
Antonio Martinez ◽  
Anna Gaya ◽  
Mariano Monzó

Abstract Abstract 3919 Background: The constitutive activation of the JAK/STAT pathway plays an important role in the pathogenesis and proliferation of Hodgkin Lymphoma (HL). Although somatic activating point mutations in the JAK2 gene have been reported in myeloproliferative disorders (MPD), they are rarely described in HL, where JAK2 amplification is associated with mutations of regulator genes such as SOCS-1, constitutive activation of STAT proteins or miRNA deregulation. Recently, many JAK2 inhibitors, including Lestaurtinib (CEP701), have been reported to have clinical efficacy in MPD. CEP701 is a multitargeted tyrosine kinase inhibitor that potently inhibits FLT3 at nanomolar concentrations. Recent studies in MPD have further shown that CEP701 inhibitory activity is not limited to FLT3 and can suppress JAK2/STAT5 signaling through JAK2 inhibition. As a first step towards elucidating the potential role of CEP701 in HL therapy, we have analyzed its efficacy in vitro. Methods: Four HL cell lines, L-428, L-1236, HDMYZ and L-540, were assayed for proliferation, apoptosis and levels of proteins in the JAK2/STAT pathway (pJAK2, JAK2, pSTAT5, STAT5, Bcl-xL) after CEP701 treatment. 100,000 cells were plated in a 96-well plate in 100 ml culture medium with CEP701 or DMSO (vehicle control) at concentrations of 30–300 nM. After 1 or 24 hours of incubation with CEP701, the levels of the proteins and of FLT3 were analyzed by Western blot. Proliferation was analyzed with CellTiter 96 AQueous One Solution Cell Proliferation Assay (MTS) and apoptosis by CaspaseGlo 3/7 after 48 hours of treatment. Results: The proliferation analysis showed an effective dose-dependent inhibition of cell growth in the 4 HL cell lines after treatment with increasing concentrations of CEP701. At 48h, in comparison to cells treated with DMSO alone (normalized to 100%), in cells treated with 100nM of CEP701, we observed a marked inhibition of 35% in L-428, 55% in L-1236, 15% in HDMYZ and 77% in L-540. Moreover, apoptosis increased by 38%, 31%, 21% and 25%, respectively. The protein analysis showed that after one hour, CEP701 inhibited phosphorylation of JAK2 (pJAK2) and its downstream target STAT5 (pSTAT5) in a dose-dependent manner, with no changes in the non-phosphorylated proteins. The downstream target Bcl-xL also decreased. Conclusions: Taken together, these data demonstrate that growth inhibition and apoptosis activation by CEP701 in HL cells correlates with the inhibition of the JAK2/STAT5-dependent signal transduction pathway. Here we present the first biological evidence that Lestaurtinib could be a promising new agent in the treatment of patients with HL. Supported by a FIS grant (PS09/00547). Disclosures: No relevant conflicts of interest to declare.


2015 ◽  
Vol 5 (1) ◽  
Author(s):  
Matteo Allegretti ◽  
Maria Rosaria Ricciardi ◽  
Roberto Licchetta ◽  
Simone Mirabilii ◽  
Stefania Orecchioni ◽  
...  

Abstract Aberrant activation of the PI3K/Akt/mTOR pathway is a common feature of acute myeloid leukemia (AML) patients contributing to chemoresistance, disease progression and unfavourable outcome. Therefore, inhibition of this pathway may represent a potential therapeutic approach in AML. The aim of this study was to evaluate the pre-clinical activity of NVP-BKM120 (BKM120), a selective pan-class I PI3K inhibitor, on AML cell lines and primary samples. Our results demonstrate that BKM120 abrogates the activity of the PI3K/Akt/mTOR signaling, promoting cell growth arrest and significant apoptosis in a dose- and time-dependent manner in AML cells but not in the normal counterpart. BKM120-induced cytotoxicity is associated with a profound modulation of metabolic behaviour in both cell lines and primary samples. In addition, BKM120 synergizes with the glycolitic inhibitor dichloroacetate enhancing apoptosis induction at lower doses. Finally, in vivo administration of BKM120 to a xenotransplant mouse model of AML significantly inhibited leukemia progression and improved the overall survival of treated mice. Taken together, our findings indicate that BKM120, alone or in combination with other drugs, has a significant anti-leukemic activity supporting its clinical development as a novel therapeutic agent in AML.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3730-3730 ◽  
Author(s):  
Sarah A Meadows ◽  
Adam Kashishian ◽  
David Johnson ◽  
Roger G Ulrich ◽  
Langdon L Miller ◽  
...  

Abstract Abstract 3730 Background: The PI3K/Akt/mTOR pathway plays a critical role in cellular proliferation and survival through transduction of signals from cell-surface receptors to proteins involved in cell cycling (eg, cyclin D1) and mRNA translation (eg, ribosomal protein S6 and translation initiation factor 4E-binding protein 1 [4E-BP1]). MCL is an aggressive B-cell non-Hodgkin lymphoma. Overexpression of cyclin D1 has prompted clinical evaluation of mTOR inhibitors (everolimus, temsirolimus) in MCL. While efficacy has been observed, the extent and duration of tumor control has been modest, encouraging assessment of additional methods of intervention. Among the several PI3K isoforms (p110α, β, δ, γ), we have previously shown a unique role for PI3Kδ in maintaining the survival of hematological cancers and have also shown that GS-1101, a highly selective oral PI3Kδ-isoform-specific inhibitor with no activity against mTOR, shows promising clinical activity in non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL). These considerations prompted us to assess the specific role of PI3Kδ in MCL and to determine if dual PI3Kδ/mTOR inhibition might enhance antitumor effects. Results: In all evaluated MCL cell lines (Jeko, Mino, Granta, and NCEB), PI3Kδ levels were high while expression of PI3Kα, β, and γ were variable. PI3Kδ was functionally active, inducing phosphorylation of Akt (pAkt) in all tested cell lines and also in 4 of 4 primary MCL samples. Single-agent GS-1101 decreased pAkt in cell lines and primary samples. The pAkt decrease was associated with growth suppression and induction of apoptosis in all MCL cell lines. Consistent with these effects, immunoblotting showed that GS-1101 decreased cyclin D1 levels in MCL cell lines. Because signals from the tumor microenvironment are essential for homing, survival, and proliferation of malignant B cells, we investigated the potential role of PI3Kδ in mediating these signals by stimulating MCL lines with CXCL12 (SDF-1), CXCL13 (BCA-1), BAFF, or BCR crosslinking in the presence or absence of GS-1101. These stimuli all resulted in the phosphorylation of Akt, which was inhibited by GS-1101 in a dose-dependent manner. Furthermore, selective inhibition of PI3Kd was able to attenuate the upregulation of Akt phosphorylation and the secreation of CCL17 and CCL22 associated with coculturing MCL cells and stromal cells. Treatment with GS-1101 also decreased S6 phosphorylation, but levels of phospho-4E-BP1 remained high, suggesting that mTOR signaling was not completely inhibited. However, the combination of GS-1101 and everolimus suppressed phosphorylation of both S6 and 4E-BP1, inhibiting MCL viability and enhancing apoptosis relative to treatment with either GS-1101 or everolimus alone. Conclusions: Our findings indicate that excessive PI3Kδ activity is characteristic in MCL and GS-1101-mediated PI3Kδ inhibition reduces MCL growth and survival. Combination therapy to address the molecular complexity associated with the convergence of the PI3Kδ-Akt and mTOR pathways may provide a novel treatment approach for MCL. Disclosures: Meadows: Gilead Sciences: Employment. Kashishian:Gilead Sciences: Employment. Johnson:Gilead Sciences: Employment. Ulrich:Gilead Sciences: Employment. Miller:Gilead Sciences: Employment. Lannutti:Gilead Sciences: Employment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1587-1587 ◽  
Author(s):  
Jennifer L. Cultrera ◽  
Lauren Rosenberg ◽  
David J. McConkey ◽  
Barbara Pro

Abstract Introduction: Epigenetic changes have been implicated in the pathogenesis of several human hematologic malignancies, including non-Hodgkin’s lymphomas (NHL). Vorinostat, a novel pan-histone deacetylase inhibitor (HDACi), has shown significant clinical activity in cutaneous T-cell lymphoma. This class of agents enhances transcription of target genes through histone hyperacetylation and through interactions with signal transduction mediators and transcription factors. Bortezomib is a reversible inhibitor of the chymotrypsin-like activity of the human 20s proteasome with established clinical activity in mantle cell lymphoma. The combination of vorinostat and bortezomib has exhibited synergism in several hematologic malignancy models. We sought to further explore the effects of these two agents in T-cell NHL cell lines and explore levels of apoptosis in response to combination treatment with vorinostat and bortezomib. Methods: A panel of T-cell NHL cell lines (SR, SUDHL1, SUPM2, SUPT1, L82, and HH), representing several different subtypes, were evaluated to assess the activity of vorinostat and bortezomib. Cells were plated at a concentration of 2 × 105 cells/mL and cultures were treated with varying concentrations of each agent (0.5 μM to 10 μM) and assessed after 24 hours for levels of apoptosis. Apoptosis and viability were analyzed through propidium iodide flow cytometry. Levels of apoptosis were considered significant at 30% cells in sub G0 phase. Further studies were undertaken to assess possible mechanisms of action contributing to this apoptosis. TRAIL, DR4, and DR5 concentrations were measured in these lines through ELISA and cell surface flow cytometry. Results: Heterogeneity was observed throughout the various T-cell NHL lines in response to vorinostat and bortezomib as single agents. After treatment with 1 μM of vorinostat and 5 nM of bortezomib, L82 cells (ALK negative anaplastic large cell lymphoma) exhibited approximately 40% and 60% apoptosis respectively (see figure 1). These concentrations of both vorinostat and bortezomib are consistent with clinically relevant maximum tolerated dose equivalents for each agent. IC50s were obtained for all cell lines tested within therapeutic ranges. Combination therapy resulted in additive levels of apoptosis in all cell lines, with a subset resulting with synergistic apoptotic levels as indicated by combination index values < 1 using the median effect method of Chou and Talalay (see figure 2). Apoptosis in L82 cells treated by a combination of suboptimal doses of each agent (vorinostat 0.5 μM and bortezomib 0.5 nM) was substantially higher at 35%. Similar results were noted for the SUPT1 and HH cell lines at levels of vorinostat 1 μM/bortezomib 1 nM and vorinostat 0.5 μM and bortezomib 1 nM respectively. In cells treated with vorinostat and bortezomib as single agents and in combination, upregulation in the components of the TRAIL dependent apoptotic system was noted. Transcriptional increases of DR5 and TRAIL mRNA was demonstrated in a dose dependent manner after single agent treatment of vorinostat and bortezomib in L82, SUDHL1, and HH cell lines. Further, flow cytometry and ELISA measurements demonstrated an increase in protein levels of DR5 and TRAIL in a dose dependent manner with both vorinostat and bortezomib. Conclusions: There is evidence that T-cell NHL cells are sensitive to both vorinostat and bortezomib as single agents and that there is synergy with combination therapy. TRAIL/DR5 expression may be altered by treatment with these agents and cause cell death through the extrinsic apoptotic pathway. These findings warrant further exploration of this combination in the clinical setting. FIGURE 1: Vorinostat and bortezomib activity in T-cell NHL T-cell NHL cell lines after treatment with vorinostat (A) and bortezomib (B) as single agents at varying concentrations. FIGURE 1:. Vorinostat and bortezomib activity in T-cell NHL . / T-cell NHL cell lines after treatment with vorinostat (A) and bortezomib (B) as single agents at varying concentrations. FIGURE 2: Synergism of vorinostat and bortezomib in T-cell NHL L82 cells after treatment with vorinostat and bortezomib in combination. FIGURE 2:. Synergism of vorinostat and bortezomib in T-cell NHL . / L82 cells after treatment with vorinostat and bortezomib in combination.


2004 ◽  
Vol 181 (3) ◽  
pp. 477-492 ◽  
Author(s):  
AA Fouladi Nashta ◽  
CV Andreu ◽  
N Nijjar ◽  
JK Heath ◽  
SJ Kimber

Decidualisation of uterine stromal cells is a prerequisite for implantation of the embryo in mice. Here we have used an in vitro culture system in which stromal cells decidualise as indicated by a number of markers, including an increase in alkaline phosphatase (ALP) activity. The latter was used as a quantitative marker of decidualisation in the presence of low (2%) fetal calf serum. Prostaglandin E(2) (PGE(2)), which is known to induce decidualisation, increased ALP activity, and this effect was blocked in a dose-dependent manner by indomethacin. Leukemia inhibitory factor (LIF) was then examined, but it had no effect on PGE(2) secretion. However, LIF suppressed ALP activity in a dose-dependent manner in the presence of 2% serum, while an inhibitor of LIF that competes for binding to its receptor reversed the effect of LIF and increased ALP activity above the control level. In serum-free cultures, stromal cells differentiated rapidly, and no differences were observed between LIF-treated and untreated cultures. Stromal cells produce LIF during in vitro culture, and this peaked at 48 h. Freshly collected stromal cells from both day-2 and -4 pregnant mice expressed mRNA for the LIF receptor, and the transcript level was higher in cells isolated on day 4. However, no differences were observed in the relative levels of transcripts in cells from day 2 and day 4 after culture, nor were there differences between the LIF-treated cultures and controls. Therefore, in this study, we have shown that LIF suppresses decidualisation of murine uterine stromal cells in the presence of serum, this is not due to the regulation of PGE(2) secretion by stromal cells.


2020 ◽  
Vol 13 (9) ◽  
pp. 208
Author(s):  
Min-Hee Kim ◽  
Tae Hyeong Lee ◽  
Jin Soo Lee ◽  
Dong-Jun Lim ◽  
Peter Chang-Whan Lee

Hypoxia-inducible factor (HIF)-1α plays an important role in cancer progression. In various cancers, including thyroid cancer, overexpression of HIF-1α is related to poor prognosis or treatment response. However, few studies have investigated the role of HIF-1α inhibition in thyroid cancer progression. We evaluated the utility of the HIF-1α inhibitor IDF-11774 in vitro utilizing two thyroid cancer cell lines, K1 and BCPAP. Both cell lines were tested to elucidate the effects of IDF-11774 on cell proliferation and migration using soft agar and invasion assays. Here, we found that a reduction of HIF-1α expression in BCPAP cells was observed after treatment with IDF-11774 in a dose-dependent manner. Moreover, cell proliferation, migration, and anchorage-independent growth were effectively inhibited by IDF-11774 in BCPAP cells but not in K1 cells. Additionally, invasion of BCPAP but not K1 cells was controlled with IDF-11774 in a dose-dependent manner. Our findings suggest that promoting the degradation of HIF-1α could be a strategy to manage progression and that HIF-1α inhibitors are potent drugs for thyroid cancer treatment.


Sign in / Sign up

Export Citation Format

Share Document