Efficacy of adavosertib therapy against anaplastic thyroid cancer

2021 ◽  
Author(s):  
Yu-Ling Lu ◽  
Yu-Tung Huang ◽  
Ming-Hsien Wu ◽  
Ting-Chao Chou ◽  
Richard J Wong ◽  
...  

Wee1 is a kinase that regulates the G2/M progression by inhibition of CDK1, which is critical for ensuring DNA damage repair before initiation of mitotic entry. Targeting Wee1 may be a potential strategy in the treatment of anaplastic thyroid cancer, a rare but lethal disease. The therapeutic effects of adavosertib, a Wee1 inhibitor for anaplastic thyroid cancer was evaluated in this study. Adavosertib inhibited cell growth in three anaplastic thyroid cancer cell lines in a dose-dependent manner. Cell cycle analysis revealed cells were accumulated in the G2/M phase. Adavosertib induced caspase-3 activity and led to apoptosis. Adavosertib monotherapy showed significant retardation of the growth of two anaplastic thyroid cancer tumor models. The combination of adavosertib with dabrafenib and trametinib revealed strong synergism in vitro and demonstrated robust suppression of tumor growth in vivo in anaplastic thyroid cancer xenograft models with BRAFV600E mutation. The combination of adavosertib with either sorafenib or lenvatinib also demonstrated synergism in vitro and had strong inhibition of tumor growth in vivo in an anaplastic thyroid cancer xenograft model. No appreciable toxicity appeared in mice treated with either single agent or combination treatment. Our findings suggest adavosertib holds the promise for the treatment of patients with anaplastic thyroid cancer.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2274-2274
Author(s):  
Jessie Hao-Ru Hsu ◽  
Sasha Borodovsky ◽  
Yanjun Wang ◽  
Maryann San Martin ◽  
Minhui Shen ◽  
...  

Abstract The polycomb repressive complex 2(PRC2), consisting of the core subunits EED, EZH2, and SUZ12, is frequently overexpressed and deregulated in hematological and solid malignancies. Clinical validation for targeting the PRC2 complex has been demonstrated with several small molecule inhibitors of EED or EZH2 that are either approved or being evaluated in clinical trials in different cancer settings. Like other treatment modalities, single-agent activity is limited. Given that the PRC2 complex is involved in multiple diverse oncogenic/immune pathways, we thus explored the combination potential of inhibiting PRC2 in a wide array of biological contexts. To this end, we have developed a potent, selective, and orally bioavailable EED ligand. The compound binds to EED (K D =3.2 nM) and inhibits PRC2 enzyme activity (IC 50= 7nM). The compound inhibits proliferation of an EZH2 dependent Karpas422 GCB-DLBCL cell line (GI 50= 27nM) in vitro. In vivo, the inhibitor is well tolerated and drives tumor regression at 10 mpk and 50 mpk doses in a DLBCL xenograft model. Given the compound has favorable selectivity, potency and pharmacokinetic profile, we next evaluated the combinability of this EEDi with select compounds targeting diverse oncogenic pathways. We find that EED and PI3K/AKT inhibitors show marked combination activity in DLBCL, in part through downregulation of AKT signaling. Combination activity is also observed with the EED inhibitor in combination with the BTK inhibitor, acalabrutinib, in DLBCL, although this appears to be context dependent. In addition to DLBCL models, we have also assessed the therapeutic potential of EED inhibitor combinations in solid tumors. In SCLC, the EED inhibitor increased expression of SLFN11, a biomarker that has been linked to clinical response to a PARP inhibitor / temozolomide combination (Pietanza et al., 2018). Therefore, we evaluated the EED inhibitor in combination with the PARP inhibitor, olaparib, in an aggressive SCLC xenograft model and observed reduced tumor growth in the combination arm but not in monotherapy arms. In ovarian cancer, ARID1A mutations are proposed to sensitize to EZH2 and ATR inhibitors (Bitler et al., 2015, Williamson et al., 2016) which prompted evaluation of the EED inhibitor with the ATR inhibitor, AZD6738. Indeed, the combination resulted in tumor stasis whereas the monotherapies produced modest tumor growth inhibition. Last, we have evaluated the EED inhibitor alone, and in combination with a CTLA4 antibody, in a tumor immunity setting using an immunologically cold syngeneic melanoma model. Interestingly, we find that EEDi has significant anti-tumor activity alone, as compared with either control or CTLA4 antibody treatment alone. Together, these data show that while our EEDi has strong single-agent activity in vitro and in vivo, combining the EEDi together with inhibitors of select oncogenic pathways may bring deeper therapeutic response in a context dependent manner. Disclosures Hsu: AstraZeneca: Current Employment. Borodovsky: AstraZeneca: Other: Past employment. Wang: AstraZeneca: Current Employment. San Martin: AstraZeneca: Current Employment. Shen: AstraZeneca: Current Employment. Woods: AstraZeneca: Current Employment. Rosen: AstraZeneca: Current Employment, Other: may hold equity, stock, or stock options. Bagal: AstraZeneca: Current Employment. Rawlins: AstraZeneca: Current Employment. Robinson: AstraZeneca: Current Employment. Code: AstraZeneca: Current Employment. Prickett: AstraZeneca: Current Employment. Bloecher: AstraZeneca: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company.


Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3487
Author(s):  
Yu-Ling Lu ◽  
Ming-Hsien Wu ◽  
Yi-Yin Lee ◽  
Ting-Chao Chou ◽  
Richard J. Wong ◽  
...  

Differentiated thyroid cancer (DTC) patients are usually known for their excellent prognoses. However, some patients with DTC develop refractory disease and require novel therapies with different therapeutic mechanisms. Targeting Wee1 with adavosertib has emerged as a novel strategy for cancer therapy. We determined the effects of adavosertib in four DTC cell lines. Adavosertib induces cell growth inhibition in a dose-dependent fashion. Cell cycle analyses revealed that cells were accumulated in the G2/M phase and apoptosis was induced by adavosertib in the four DTC tumor cell lines. The sensitivity of adavosertib correlated with baseline Wee1 expression. In vivo studies showed that adavosertib significantly inhibited the xenograft growth of papillary and follicular thyroid cancer tumor models. Adavosertib therapy, combined with dabrafenib and trametinib, had strong synergism in vitro, and revealed robust tumor growth suppression in vivo in a xenograft model of papillary thyroid cancer harboring mutant BRAFV600E, without appreciable toxicity. Furthermore, combination of adavosertib with lenvatinib was more effective than either agent alone in a xenograft model of follicular thyroid cancer. These results show that adavosertib has the potential in treating DTC.


2017 ◽  
Vol 58 (1) ◽  
pp. 15-23 ◽  
Author(s):  
Chen-Tian Shen ◽  
Wei-Jun Wei ◽  
Zhong-Ling Qiu ◽  
Hong-Jun Song ◽  
Xin-Yun Zhang ◽  
...  

More aggressive thyroid cancer cells show a higher activity of glycometabolism. Targeting cancer cell metabolism has emerged as a novel approach to prevent or treat malignant tumors. Glucose metabolism regulation effect of metformin in papillary thyroid cancer was investigated in the current study. Human papillary thyroid carcinoma (PTC) cell lines BCPAP and KTC1 were used. Cell viability was detected by CCK8 assay. Glucose uptake and relative gene expression were measured in metformin (0–10 mM for 48 h)-treated cells by 18F-FDG uptake assay and western blotting analysis, respectively. MicroPET/CT imaging was performed to detect 18F-FDG uptake in vivo. After treatment with metformin at 0, 2.5, 5 and 10 mM for 48 h, the ratio of p-AMPK to total AMPK showed significant rising in a dose-dependent manner in both BCPAP and KTC1, whereas p-AKT and p-mTOR expression level were downregulated. 18F-FDG uptake reduced after metformin treatment in a dose-dependent manner, corresponding to the reduced expression level of HK2 and GLUT1 in vitro. Xenograft model of PTC using BCPAP cells was achieved successfully. MicroPET/CT imaging showed that in vivo 18F-FDG uptake decreased after treatment with metformin. Immunohistochemistry staining further confirmed the reduction of HK2 and GLUT1 expression in the tumor tissue of metformin-treated PTC xenograft model. In conclusion, metformin could reduce glucose metabolism of PTC in vitro and in vivo. Metformin, by targeting glycometabolism of cancer cells, could be a promising adjuvant therapy alternative in the treatment modality of advanced thyroid carcinoma.


Endocrinology ◽  
2008 ◽  
Vol 149 (11) ◽  
pp. 5357-5365 ◽  
Author(s):  
Zhaowei Meng ◽  
Norisato Mitsutake ◽  
Masahiro Nakashima ◽  
Dmytro Starenki ◽  
Michiko Matsuse ◽  
...  

Nuclear factor κB (NF-κB), as an antiapoptotic factor, crucially affects the outcomes of cancer treatments, being one of the major culprits of resistance to chemotherapy. In this study, we investigated whether dehydroxymethylepoxyquinomicin (DHMEQ), a novel NF-κB inhibitor, can enhance antitumor activities of taxanes in anaplastic thyroid cancer (ATC) cells. Taxanes induced NF-κB activation in ATC cells, which could compromise the therapeutic effect of the drugs. However, DHMEQ, by inhibiting the nuclear translocation of NF-κB, completely suppressed the DNA binding capacities of NF-κB and lowered the levels of nuclear NF-κB protein. Compared with single treatment (either taxane or DHMEQ), the combined treatment strongly potentiated apoptosis, confirmed by cell survival assay; Western blotting for poly (ADP-ribose) polymerase, caspase 3, X-linked inhibitor of apoptosis, and survivin; and flow cytometry for annexin V. Furthermore, we also demonstrate for the first time that the combined treatment showed significantly greater inhibitory effect on tumor growth in a nude mice xenograft model. These findings suggest that taxanes are able to induce NF-κB activation in ATC cells, which could attenuate antitumor activities of the drugs, but inhibition of NF-κB by DHMEQ creates a chemosensitive environment and greatly enhances apoptosis in taxanes-treated ATC cells in vitro and in vivo. Thus, DHMEQ may emerge as an attractive therapeutic strategy to enhance the response to taxanes in ATCs.


2018 ◽  
Vol 66 (5) ◽  
pp. 1.4-9 ◽  
Author(s):  
Chih-Yuan Wang ◽  
Hao-Ai Shui ◽  
Tien-Chun Chang

This study tested the hypothesis that the effects of lovastatin on anaplastic thyroid cancer cell growth are mediated by upregulation of transketolase (TKT) expression. The effects of lovastatin on TKT protein levels in ARO cells were determined using western blot and proteomic analyses. After treatment with lovastatin and oxythiamine, the in vitro and in vivo growth of ARO cells was determined using 3-(4,5-Dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide (MTT) assays and tumor xenografts in nude mice. TKT protein expression in the ARO tumors was assessed using immunohistochemistry analysis. Proteomic analysis revealed that 25 µM lovastatin upregulated TKT expression. Co-treatment of ARO cells with 1 µM lovastatin + 1 µM oxythiamine increased TKT protein expression compared with control levels; however, no differences were observed with 10 µM lovastatin + 1 µM oxythiamine. Furthermore, treatment with either oxythiamine or lovastatin alone reduced ARO tumor expression of TKT, as well as decreased ARO cell proliferation in vitro and tumor growth in vivo. However, mice treated with both lovastatin and oxythiamine at the same time had tumor volumes similar to that of the untreated control group. We conclude that either lovastatin or oxythiamine reduced ARO cell growth; however, the combination of these drugs resulted in antagonism of ARO tumor growth.


2019 ◽  
Vol 26 (8) ◽  
pp. 727-738 ◽  
Author(s):  
Shu-Fu Lin ◽  
Jen-Der Lin ◽  
Chun-Nan Yeh ◽  
Yu-Tung Huang ◽  
Ting-Chao Chou ◽  
...  

Polo-like kinases (PLKs) are pivotal regulators of cell proliferation and cell survival; therefore, PLKs may be potential targets in the treatment of malignancy. The therapeutic effects of volasertib, a PLKs inhibitor for papillary and follicular thyroid cancer (known as well-differentiated thyroid cancer (WDTC)), were evaluated in this study. Volasertib inhibited cell proliferation in two papillary and two follicular thyroid cancer cell lines in a dose-dependent manner. Volasertib treatment reduced cells in the S phase and increased cells in the G2/M phase. Volasertib activated caspase-3 activity and induced apoptosis. Drug combinations of volasertib and sorafenib showed mostly synergism in four well-differentiated thyroid carcinoma cell lines in vitro. Volasertib treatment in vivo retarded the growth of a papillary thyroid tumor model. Furthermore, the combination of volasertib with sorafenib was more effective than a single treatment of either in a follicular thyroid cancer xenograft model. Promising safety profiles appeared in animals treated with either volasertib alone or volasertib and sorafenib combination therapy. These findings support volasertib as a potential drug for the treatment of patients with WDTC.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3743-3743
Author(s):  
James Tsai ◽  
Elizabeth A Burton ◽  
Gaston Habets ◽  
Brian West ◽  
Paul Lin ◽  
...  

Abstract Introduction: While clinical studies using targeted therapies as single agents in AML have shown promising results in recent years, long-term durable responses in this aggressive cancer may require combination therapies to overcome disease progression and single agent resistance mechanisms. PLX3397 is an orally active, selective small molecule inhibitor of the constitutively activated FLT3-ITD mutant kinase. In cellular assays PLX3397 effectively inhibited FLT3-ITD autophosphorylation and FLT3-ITD driven proliferation with IC50s in the 10-100nM range. A clinical study to evaluate the pharmacokinetics (PK), safety and efficacy of PLX3397 in patients with FLT3-ITD AML is currently ongoing. In order to determine if combination therapy could improve efficacy, we evaluated the combination of PLX3397 with the hypomethylating agent decitabine (DEC; 5-aza-2’-deoxycytidine) in preclinical models of FLT-ITD AML. Decitabine, a drug originally indicated for myelodysplastic syndrome, is approved in Europe for the treatment of adult patients (≥65 years of age) with newly diagnosed or secondary AML. Methods: For the in vitro growth assays, cells were pre-treated with decitabine for 0-3 days prior to the addition of PLX3397. Following a 3-day incubation, cell viability was measured based on quantification of the ATP present. The resulting data were analyzed for synergy and combination indices were calculated using CalcuSyn software. Apoptosis was analyzed by measuring caspase 3/7 activity following a 24h incubation with both compounds. For the in vivo study, MV-4-11 cells were grown as subcutaneously implanted xenografts in SCID mice. When tumors reached a size of ~500 mm3 the mice were randomized into equal-sized treatment groups by body weight and tumor size (the day on which this was done was counted as day 0). Decitabine was dosed at 20mg/kg on days 1, 7, 13 and 20 after randomization. PLX3397 was dosed at 20mg/kg on day 2, and continued for 20 days. The combination followed the same dosing schemes as the two single agents. Results: In vitro viability experiments in two AML cell lines (MV-4-11 and MOLM14) using a dose matrix format demonstrated a combination benefit of PLX3397 and decitabine over a range of concentrations. Pre-incubation with decitabine for 3 days prior to the addition of PLX3397 enhanced the synergy observed. PLX3397 alone was more effective than decitabine at inducing apoptosis. Adding both compounds together slightly enhanced the induction of apoptosis, though there did not appear to be an added benefit to pre-treating the cells with decitabine, as was seen in the viability assays. To confirm the synergy observed in vitro we tested the in vivo efficacy of the two agents in the MV-4-11 xenograft model. By day 19, both decitabine and PLX3397 delayed tumor growth, resulting in tumor growth inhibition (TGI) of 89% and 42%, respectively. The combination of decitabine and PLX3397 showed striking antitumor activity, causing tumor regression and reducing tumor volume by 88%. This tumor suppression was maintained for 15 days after the treatment was stopped. Consistent with clinical experience, decitabine treatment was associated with bone marrow toxicity. This toxicity was not worsened by PLX3397. After 2 weeks of recovery bone marrow cellularity rebounded to pre-dosing levels in the combination, with the exception of red blood cell count. Conclusion: Preclinical studies of PLX3397 and decitabine in FLT3-ITD AML cell lines and a xenograft model demonstrated beneficial effects when used in combination. Single agent treatment inhibited MV-4-11 xenograft tumor growth, while the combination resulted in tumor regression. PLX3397 did not further enhance the bone marrow toxicity induced by decitabine. PLX3397 exposures in these preclinical studies are similar to those achieved in AML patients in the on-going single agent clinical trial. Figure 1. Preclinical combination of PLX3397 and decitabine in an MV-4-11 xenograft model. Figure 1. Preclinical combination of PLX3397 and decitabine in an MV-4-11 xenograft model. Disclosures Zhang: Plexxikon: Employment.


Molecules ◽  
2020 ◽  
Vol 25 (8) ◽  
pp. 1960 ◽  
Author(s):  
Erkang Zhang ◽  
Yani Zhang ◽  
Zhuoyan Fan ◽  
Lei Cheng ◽  
Shiwen Han ◽  
...  

Apigenin is a natural flavone with anti-inflammatory and antioxidant properties and antitumor abilities against several types of cancers. Previous studies have found that the antitumor effects of apigenin may be due to its similar chemical structure to 17β-estradiol (E2), a main kind of estrogen in women. However, the precise mechanism underlying the antitumor effects of apigenin in cervical cancer remains unknown. On the other hand, there is increasing evidence that describes a histamine role in cancer cell proliferation. In this study, we examined whether apigenin can attenuate the effects of histamine on tumors by regulating the expression level of estrogen receptors (ERs) to inhibit cervical cancer growth. Our in vitro data indicates that apigenin inhibited cell proliferation in a dose-dependent manner in human cervical cancer cells (HeLa), while histamine shows the opposite effects. After that, the xenograft model was established to explore the antitumor effects of apigenin in vivo, the results show that apigenin inhibited cervical tumor growth by reversing the abnormal ER signal in tumor tissue which was caused by histamine. We also demonstrate that apigenin inhibited cell proliferation via suppressing the PI3K/Akt/mTOR signaling pathway. Collectively, our results suggest that apigenin may inhibit tumor growth through the ER-mediated PI3K/Akt/mTOR pathway and that it can also attenuate the effects of histamine on tumors.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1502-1502
Author(s):  
Yang Yang ◽  
Telisha Swain ◽  
Annamaria Naggi ◽  
Giangiacomo Torri ◽  
Benito Casu ◽  
...  

Abstract Heparanase is an enzyme that cleaves heparan sulfate chains of proteoglycans and promotes the growth and metastasis of many types of human tumors. Our previous work demonstrates that enzymatically active heparanase is present in the bone marrow of myeloma patients and is associated with a poor prognosis, substantially enhances tumor growth and spontaneous metastasis to bone in an animal model of myeloma, and increases the synthesis and shedding of syndecan-1 by myeloma cells, this in turn contributes to myeloma progression by elevating levels of syndecan-1 in the tumor microenvironment. Thus, we hypothesized that inhibitors of heparanase activity would have a dramatic impact on the growth of myeloma tumors. To test this we used a chemically modified form of heparin that is 100% N-acetylated and 25% glycol-split (designated 100NA,RO-H). This form heparin is a potent inhibitor of heparanase enzyme activity but lacks anticoagulant activity thus enabling use of relatively high doses of the drug in vivo. Delivery of the 100NA,RO-H to animals bearing established myeloma tumors dramatically blocked tumor growth and progression in a dose-dependent manner(P<0.04). To understand the mechanism of action of 100NA,RO-H, a series of experiments were performed on tumor tissue harvested from the animals. Results demonstrate that the modified heparin significantly inhibits proliferation of cells within the tumor (33 ± 8/mm2 Ki-67 positive cells in treated vs. 688 ± 164/mm2 positive cells in controls, P = 0.002). 100NA,RO-H also dramatically inhibits angiogenesis as compared to controls as assessed by CD34 staining of tumor tissue. This anti-angiogenic effect may be due at least in part to its regulation of hepatocyte growth factor and/or vascular endothelial growth factor, two angiogenic factors that are detected by immunohistochemistry at high levels in controls but virtually absent from tumors of animals treated with 100NA,RO-H. In vitro studies demonstrate that 100NA,RO-H blocks syndecan-1 shedding from cells, consistent with the role of heparanase in promoting syndecan-1 shedding. In addition, in contrast to our finding that 100NA,RO-H blocks tumor growth in vivo, it only slightly inhibited proliferation, cell cycle progression and growth factor signaling in myeloma cells growing in vitro. Thus, the compound does not appear to have substantial direct effects on tumor cells. Although it is not yet clear if all of the effects of 100NA,RO-H are due to its anti-heparanase activity, we conclude that this modified heparin disrupts the myeloma tumor microenvironment thereby blocking in vivo growth and progression of the cancer. Therefore, its use as a single agent or in combination with agents having direct anti-tumor cell activity may constitute a potent new anti-myeloma therapy.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Keisuke Enomoto ◽  
Fuyuki Sato ◽  
Shunji Tamagawa ◽  
Mehmet Gunduz ◽  
Naoyoshi Onoda ◽  
...  

Abstract A novel therapeutic approach is urgently needed for patients with anaplastic thyroid cancer (ATC) due to its fatal and rapid progress. We recently reported that ATC highly expressed MYC protein and blocking of MYC through its selective inhibitor, JQ1, decreased ATC growth and improved survival in preclinical models. One of the important roles of MYC is regulation of L-neutral amino acid transporter 1 (LAT1) protein and inhibition of LAT1 would provide similar anti-tumor effect. We first identified that while the human ATC expresses LAT1 protein, it is little or not detected in non-cancerous thyroidal tissue, further supporting LAT1 as a good target. Then we evaluated the efficacy of JPH203, a LAT1 inhibitor, against ATC by using the in vitro cell-based studies and in vivo xenograft model bearing human ATC cells. JPH203 markedly inhibited proliferation of three ATC cell lines through suppression of mTOR signals and blocked cell cycle progression from the G0/G1 phase to the S phase. The tumor growth inhibition and decrease in size by JPH203 via inhibition of mTOR signaling and G0/G1 cell cycle associated proteins were further confirmed in xenograft models. These preclinical findings suggest that LAT1 inhibitors are strong candidates to control ATC, for which current treatment options are highly limited.


Sign in / Sign up

Export Citation Format

Share Document