ANP-vessel Dilator Enhances the Effect of Trametinib on Colorectal Cancer Cells via the MAPK Pathway

All published articles of this journal are available on ScienceDirect.

RESEARCH ARTICLE

ANP-vessel Dilator Enhances the Effect of Trametinib on Colorectal Cancer Cells via the MAPK Pathway

The Open Biotechnology Journal 25 Jun 2024 RESEARCH ARTICLE DOI: 10.2174/0118740707313891240614065219

Abstract

Background

Colorectal cancer (CRC) is a common malignant disease that involves the interaction of both genetic and environmental factors. Trametinib (TNB) has been approved alone or in combination with dabrafenib to treat melanoma. Vessel dilator (VDL) is a cardiac hormone that possesses anticancer properties. This study evaluates TNB with VDL combined effects in CRC cells and explores the role of Mutagen-activated protein kinases (MAPK, ERK1/2) in this effect.

Methods

The HCT-15 CRC cells were treated with TNB and VDL. The MTT and ELISA assays were used to assess p-ERK, VEGF, and VEGFR levels.

Results

The results showed that combined treatment of TNB with VDL produced a significant and synergistic inhibition of HCT-15 cell growth with a combination index of less than 1. A combination treatment demonstrated no change in the expression of VEGF, inhibition of VEGFR2, and reduction of p-ERK1/2 in HCT-15 cells.

Conclusion

The findings encourage further evaluation of this combination as the combined effect is not mediated through p-ERK, VEGF, and VEGFR.

Keywords: Trametinib, Vessel dilator, Combination, MAPK pathway, Colorectal cancer, CRC.

1. INTRODUCTION

Colorectal cancer (CRC) is a major public health concern worldwide. It is the third most diagnosed cancer in males and females and the second most common cause of mortality worldwide [1]. Among Jordanians, it was the second most frequent cancer in both genders and the second killer among cancers [2]. Treatment options depend on the stage of therapy, as early disease is managed surgically with adjuvant pyrimidines-based chemotherapy [3]. However, in metastatic disease, other targeted therapies may be added to conventional chemotherapy, with little or no role in surgical intervention. Targeted therapies include drugs that antagonize epidermal growth factor (EGF) or vascular endothelial growth factor (VEGF) [4]. Recently, additional genetic tests have been required to assess the involvement of specific targets, such as KRAS, NRAS, and BRAF muta tions, human epidermal receptor-2 (HER-2) expression, assessment of genetic mutations by assessing microsate llite stability, mismatch Repair proficiency, tumor muta tion burden, and Neurotrophic Tyrosine Receptor Kinase (NTRK) fusions [3].

Genetic mutations are a major cause of the malignant transformation of CRC. One of the major pathways affected is the Mitogen-Activated Protein Kinase (MAPK) pathway [5], in which RAS, RAF, and mitogen-activated extracellular kinase (MEK) are the major components. A large body of evidence shows that the activation of this pathway plays an essential role in the progression of CRC [6], which induces tumor cell proliferation, survival, orientation, and angiogenesis. This pathway is triggered by ligands, such as EGF binding to its tyrosine kinase receptor (RTK) and EGFR, activating downstream RAS, RAF, and MEK1/2. Extracellular signal-regulated kinase (ERK)1/2. Phosphory lated ERK1/2 translocates to the nucleus, activating various targets that consecutively regulate genes essential for many cellular processes, leading to proliferation, survival, invasion, invasion, and metastasis [7]. Mutations in KRAS, NRAS, and V-RAF murine sarcoma viral oncogene homolog B (BRAF) are frequently encountered in this pathway. Approximately 40% of KRAS mutations [8] and 11% of BRAF mutations have been reported in CRC [9]. The most recent NCCN guidelines include many targeted therapies for KRAS (sotorasib or Adagrasib in combination with cetuximab or panitumumab) and BRAFV600E (Encorafenib + panitumumab) mutations [3]. Mutations in MEK1/2 are less frequently encountered than RAS and RAF mutations [9]. Therefore, the latter component of this pathway is an attractive target for novel treatment approaches, as MEK is a MAPK pathway component that is downstream to both KRAS and BRAF.

Trametinib (TNB) is an orally active allosteric inhibitor of MEK1/2 [10]. This eventually inhibits cancer cell proliferation, motility, and survival [10]. The Food and Drug Administration (FDA) approved TNB for the treatment of metastatic melanoma [11], metastatic non-small cell lung cancer (NSCLC), and anaplastic thyroid cancer [12]. TNB has been investigated as a monotherapy and was found to be ineffective in patients with CRC with BRAF mutations [13] primarily because of many concomitant mechanisms of resistance, including activation of RTK and stimulation of parallel signaling pathways bypassing MEK inhibition to reactivate ERK [14]. Therefore, combining TNB with an agent that targets several signals inside the cell may improve therapeutic efficacy and enhance CRC treatment. Clinically, dabrafenib combined with TNB has been evaluated in patients with BRAF-mutant metastatic CRC (mCRC) [15] and in patients with BRAF-mutant CRC [16]. Unfor- tunately, its effectiveness is limited, unlike that in BRAF-mutant melanoma [15, 16]. In phase I/II study, TNB was investigated in combination with dabrafenib and an anti-EGFR antibody, panitumumab, in patients with BRAF V600E mCRC, and the clinical activity of the combination was confirmed [17]. Therefore, further investigation of TNB in CRC is required to achieve optimal inhibition of MAPK.

The human body synthesizes four peptide hormones within the 126-aa Atrial natriuretic peptide (ANP) prohormone that is encoded by the atrial natriuretic peptide prohormone proANP gene, long-acting natriuretic peptide (LANP), vessel dilator (VDL), kaliuretic peptide (KP) and atrial natriuretic peptide (ANP). VDL, ANP, KP, and LANP have shown anticancer effects in various types of cancer in vitro [18-23] and in vivo [24-26]. The MAPK pathway is blocked by these hormones, as they target multiple kinases in this cascade [27]. VDL, ANP, KP, and LANP exert 95%, up to 98% inhibition of the phosphorylation of MEK1/2 [25, 28-30] and up to 96% inhibition of the phosphorylation of ERK1/2 [31, 32]. Notably, those hormones did not inhibit the ERK1/2 in the normal cells [33]. The ability of those hormones to inhibit these kinases as well as inhibit DNA synthesis is mediated by intracellular messenger cyclic GMP (cGMP) inhibition [25, 28, 29, 31, 32, 34].

Vascular endothelial growth factor A (VEGF-A) promotes cancer cell growth by acting as a growth factor and stimulating the MAPK pathway [35]. These four cardiac hormones were the first dual inhibitors of VEGF and VEGFR2. VDL has the most significant anticancer properties of ANPs in many types of cancers in vitro [19, 21-23, 36] and in vivo [24-26]. In CRC, VDL treatment significantly decreased the number and proliferation of cells within 24 hrs [34]. No previous studies have investigated the combination of VDL with other agents.

In this study, we hypothesized that combining TNB and VDL would produce a significant synergistic effect on the MAPK/ERK pathway inhibition. In addition, the effect on the levels of p-ERK, VEGF, and VEGFR was assessed.

2. MATERIALS AND METHODS

In this study, the possible synergistic effect of the combination of TNB and VDL on CRC was investigated using HCT-15 cells. This study was performed using different concentrations of TNB or VDL, alone and in combination, to study their synergistic effects.

2.1. Cell Line and Materials

HCT-15 colorectal cancer cell line (ATCC® CCL-225TM) was purchased from the American Type Culture Collection (ATCC). TNB and VDL used in this study were obtained from Santa Cruz Biotechnology (Catalog # sc-364639) and Phoenix Pharmaceuticals (Catalog # 005-20), respectively. 3- [4, 5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) was obtained from Duchefa Biochemie (Cat#298-93-1), and all ELISA kits (p-ERK1/2, VEGFR2, and VEGF) were purchased from Abcam (ab176640, ab213476, ab100662, respectively).

2.2. Cell Culture

HCT-15 cells were cultured under sterile conditions in a biological safety cabinet in RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. The cells were incubated at 37°C with 5% CO2. For subculturing, the cells were washed with Ca2+-and Mg2+-free phosphate-buffered saline (Dulbecco's PBS W/O Calcium & Magnesium, Cat# ECB4004L) and dissociated using 0.25% EDTA-trypsin (ECB3057D) for 5-15 minutes at 37°C. Afterward, trypsin was neutralized with 10% FBS RPMI-1640 medium, and the detached cells were centrifuged, resuspended, and counted using a hemocytometer. The cells were then seeded at an appropriate number, depending on the experiment.

2.3. Drug Preparation

TNB 5 mg was stored at -20°C until use, and 200 µg of VDL was stored at 4°C until it was used. The DMSO cell culture reagent was stored at room temperature. TNB was dissolved in 0.81 ml DMSO to prepare a 10 mM stock solution, and VDL was dissolved in 1 ml sterile distilled water to prepare a 200 µg/ml (51.569 µM) stock solution. Both drugs were aliquoted and stored at -20°C.

2.4. MTT Cell Proliferation Assay

Briefly, HCT-15 cells were seeded at a density of 5×103 cells/well in a 96-well plate in 10% FBS RPMI-1640 media and allowed to attach overnight. The next day, the medium was replaced with 100 µL of fresh serum-free medium (SFM) containing various concentrations of TNB (0.39, 0.78, 1.56, 3.12, 6.24, 12.5,25 and 50 µM) and 100 µL containing various concentrations of VDL (15.6, 31.25, 62.5, 125, 250, 500, 1000, and 2000 nM) in triplicate. The control wells contained 100 µL of 0.5% DMSO and 100 µL of SFM for TNB and VDL, respectively. The cells were cultured for 48 h. Then, 10 µL of MTT solution (5 mg/ml in PBS) was added to each well, and the plates were incubated at 37°C for 2 h. The medium was then removed, and 100 µL of DMSO (solubilization solution) was added to each well to dissolve the formazan crystals. The absorbance was measured at 570 nm using a microplate reader (Biochrom EZ Read 400).

2.5. Enzyme-linked Immunosorbent Assay sample preparation

HCT-15 cells were cultured at 5×104 cells/well in 12 well-plate for 24 h. The following day, variable drug concentrations were added: 2 µM VDL, 50 µM TNB, and a combination of both and were treated vs. control. After 48 h, the medium was collected for VEGF quantification. Furthermore, to quantitate p-ERK and VEGFR2, cells were lysed by adding 250 µL/well of lysis buffer provided in the ELISA kits, followed by the collection of cell lysates for the next step.

2.6. Enzyme-linked Immunosorbent Assay VEGF, VEGFR2, and p-ERK1/2 Measurement

The quantification of VEGF, VEGFR2, and p-ERK1/2 was performed using VEGF, VEGFR2, and p-ERK1/2-ELISA kits, respectively. All measurements were performed in accordance with the manufacturer’s instructions.

2.7. Statistical Analysis

All data are presented as the mean ± SEM. Additionally, to determine significant differences between the treatment and control values, we used parametric one-way ANOVA. The significance level was set at p < 0.05. The IC50 value (the concentration that induced 50% cell growth inhibition) was obtained by applying a nonlinear regression curve fit analysis using GraphPad Prism version 9.0.0 for Windows, GraphPad Software, Boston, Massachusetts USA, www.graphpad.com.

The level of interaction between TNB and VDL was assessed using a combination index (CI), isobologram analysis, and dose reduction index.

The combination index (CI) quantitatively represents the pharmacological interactions between two compounds. A CI of 1 indicated an additive interaction, a CI of less than 1 indicated a synergistic effect, and a CI of more than 1 suggested antagonism. CI was calculated as follows [37, 38]:

C and X represent the IC50 values of TNB and VDL when used alone for cell growth studies. Cc and Xc are the IC50 values of TNB and VDL, respectively, which also inhibited cell growth by 50% when used in combination.

Drug reduction index (DRI) values represent a fold decrease in the dose of individual drugs when combined compared to the dose of a single drug required to induce the same effect level. DRI values > 1 are considered favorable, allowing less toxicity while retaining the therapeutic efficacy of the individual compounds. The DRI value in this study was calculated as follows:

Where X is the IC50 value of the compound when used alone for cell growth studies, and Xc is the IC50 value for the same compound when used in combination [37, 38].

Isobologram analysis is a graphical method for evaluating the effect of equally effective concentration pairs at a single-effect level in a simple manner. It was created on a coordinate system comprising the individual drug concentrations and a straight line representing the additive effects for data points on the line was shown. The data points above the line indicate antagonistic interactions, whereas those below the line represent synergistic interactions between the two compounds when combined. A straight line in each isobologram was constructed by plotting the IC50 concentrations of TNB and VDL on the x- and y-axes, respectively. The solid straight line linking these points represents the drug concentrations for each drug that induced relatively similar growth inhibition when combined if these drugs interacted in an additive manner. The data points for each isobologram indicate the combined IC50 values of TNB and VDL. If the data point is on the line, this indicates an additive treatment effect, which lies below or above the line and represents synergistic or antagonistic effects, respectively [39].

3. RESULTS

3.1. Effect of Trametinib, Vessel Dilator, or the Combination on the Proliferation of HCT-15 Cells

Fig. (1A) shows the effect of TNB treatment on HCT-15 cell viability after 48 hrs of treatment. The results showed a dose-dependent decrease in cell viability and a significant reduction in proliferation starting at 6.25 µM. The IC50 of TNB for HCT-15 cells was 13.56 µM. Fig. (1B) shows the effect of VDL treatment on HCT-15 cell viability after 48 hrs of incubation. The results showed a dose-dependent reduction in cell viability and a significant reduction in proliferation, starting at 1000nM. The IC50 of VDL for HCT-15 cells was 1017 nM.

Fig. (1C) shows the effect of the combined treatment with TNB and VDL on the viability of HCT-15 cells after 48 hrs of incubation. However, there was no significant reduction in cell proliferation after treatment with 4 µM TNB alone. Interestingly, the combination of VDL (62.5, 125 nM) and 4 µM TNB resulted in a significant reduction in cell proliferation compared to the control.

Fig. (1).

(A). Effect of TNB versus control on Proliferation of HCT-15 cells. The cell viability reduces dramatically with an increase in TNB dose. The significant effect appears at 50 μM. (* P˂ 0.05; ** P˂ 0.005;*** P˂ 0.0005;**** P˂ 0.0001) ordinary one-way ANOVA test followed by Holm-Sidak's test. (B). The effect of Vessel dilator versus control on the proliferation of HCT-15 cells. The cell viability reduces dramatically with an increase in vessel dilator dose. The significant effect appears at 2000 nM. (* P˂ 0.05; ** P˂ 0.005;*** P˂ 0.0005;**** P˂ 0.0001) ordinary one-way ANOVA test followed by Holm-Sidak's test. (C). Comparison of the effect of using 4 μM TNB separately and in combination with the different concentrations of VDL on the viability of HCT-15 cells. (*P˂0.05;** P˂ 0.005;*** P˂ 0.0005;**** P˂ 0.0001) ordinary one-way ANOVA test followed by Holm-Sidak's test. (D). Isobologram of combined antiproliferative effects of TNB and VDL in HCT-15. (D). Combination index and dose-reduction index values for combined treatment of TNB and VDL resulted in a 50% reduction in the growth of HCT-15 cells.

The isobologram analysis of the effect of the combined treatment on HCT-15 cells is shown in Fig. (1D). Isobologram indicated a synergistic interaction between TNB and VDL, as indicated by data points falling below the line of the additive effect for the combined treatments. Combination index (CI) analysis for the effect on viability of the combined treatment of TNB and VDL on HCT-15 cells is shown in Fig. (1D). The CI data agree with the isobologram analysis, in which synergistic interactions are indicated with a CI value less than 1 for combining TNB with VDL in HCT-15 cells. The dose-reduction index (DRI) also showed a multifold reduction in the growth-inhibitory dose of TNB combined with VDL (Fig. 1D).

For the next part of the experiments, we used “sub-effective” concentrations of both drugs, 2 µM for TNB and 50 nM of VDL, to study the synergistic effect of the combination.

3.2. Effect of Trametinib, Vessel Dilator, and their Combination on the Expression of VEGF, VEGFR2, and p-ERK1/2

In addition, to investigate the effects of TNB and VDL alone and in combination on the expression of VEGF, VEGFR2, and p-ERK1/2, HCT-15 cells were treated the cells with 2 µM TNB, 50 nM of VDL, or a combination of these drugs for 48 h. The results showed a significant decrease in VEGF levels with both TNB and combination treatment, with no additional effect on VDL (Fig. 2A).

Fig. (2B) shows a significant reduction in the expression of VEGFR2 in both TNB alone and in combination treatment, with no effect on VDL.

Regarding the phosphorylation of ERK1/2, the results showed a significant reduction in p-ERK1/2 expression with both TNB alone and the combination treatment, with no effect of VDL alone, as shown in Fig. (2C).

Fig. (2A-C).

Comparison of the impact of using 2 μM TNB and 50 nM VDL separately and in combination on the expression of VEGF, VEGFR2, and p-ERK1/2. (* P˂ 0.05;** P˂ 0.005;*** P˂ 0.0005;**** P˂ 0.0001) ordinary one-way ANOVA test followed by Holm-Sidak's test.

4. DISCUSSION

The MAPK pathway plays an essential role in the progression of CRC [6]; thus, effective targeting of this pathway is a successful strategy for CRC management. Our findings showed that TNB, which inhibits MEK1/2, reduced HCT-15 cell viability in a dose-dependent manner, with a significant effect observed starting at 3.125 µM (P< 0.005). This might be expected as HCT-15 cells are RAS-mutant [40], and thus, inhibition of downstream MEK signaling will be effective in the reduction of viability of cells. In addition, VDL, which inhibits each step of the MAPK pathway, reduced the viability of HCT-15 cells in a dose-dependent manner, with a significant effect observed starting at 2 µM (P<0.05).

One group reported that TNB combined with other targeted and cytotoxic chemotherapies, especially in tumors that contain genetic alterations leading to activation of the MAPK pathway, could be more effective than monotherapy [13]. The advantages of combined treatment approaches include achieving a synergistic effect, thus allowing the use of smaller doses of each compound while maintaining a high level of effectiveness. The combination regimens of multiple drug therapies also minimize the toxicity of larger doses of single agents, overcome drug resistance, and maximize quality of life [41]. In this study, we evaluated the level of interaction between TNB and VDL treatments using CI, DRI, and isobologram. The results showed for the first time a synergistic effect of the combination on the viability of HCT-15 cells harboring a KRAS G13D activating mutation, as indicated by Ahmed, Eide [42].

Contrary to our expectations, the results of our study showed that the use of TNB, VDL, or a combination of both in HCT-15 cells did not have significant effects on VEGF, VEGFR2, or p-ERK1/2 concentrations. One study found that four cardiac hormones (VDL, LANP, KP, and ANP) inhibit the vascular endothelial growth factor (VEGF) and VEGFR2 receptor in human prostate cancer, small-cell lung cancer, and human pancreatic adenocarcinoma cells [43]. In addition, VDL can decrease VEGF by inhibiting VEGF itself or inhibiting Ras-MEK1/2-ERK1/2 kinase activity [29, 32, 44], which plays a role in the enhancement of VEGF expression [45].

Unexpectedly, in our study, VDL did not decrease the concentration of VEGF or VEGFR2 in HCT-15 cells. This study supports the findings of Hsu et al. that TNB exerts antiangiogenic activity by inhibiting VEGF expression [46]. We showed that TNB significantly inhibited the expression of both VEGF and VEGFR2 in HCT-15 cells, with no additional benefits of combination with VDL. Hsu et al. also reported that other important targets may contribute to the anti-angiogenic activity of TNB [46]. The ability of TNB to inhibit VEGFR in CRC was investigated for the first time.

In a study conducted by Sun Y and his colleagues, VDL (1 µM) was investigated on prostate cancer cells to inhibit phosphorylation of ERK1/2. Results showed that up to 96% of phosphorylation of ERK1/2 had been inhibited, and which lasted up to two hours [32]. In this study, we examined for the first time the ability of VDL to inhibit ERK1/2 phosphorylation in HCT-15 cells. The results demonstrated that VDL did not inhibit the phosphorylation of ERK 1/2 compared to the control. Similar to our previous findings, TNB blocked ERK phosphorylation, with no additional benefits for the combination.

Although combination studies using the MTT viability assay showed a synergistic effect of the combination, similar effects were not observed for VEGF and VEGFR expression or p-ERK levels. These results suggest the presence of other mechanisms that lead to increased expression of VEGF, activation of VEGFR2, and increase in p-ERK1/2, such as the PI3K/Akt, Wnt, and notch pathway [47-53]. Therefore, we postulate that blocking the ERK signaling pathway alone is not sufficient or prevents the expression of VEGF, and VEGFR2, or decreasing p-ERK levels. To this end, future gene expression studies are needed to prove this postulation using qPCR, western blotting, or IHC.

CONCLUSION

In conclusion, our study revealed that adding TNB to the vessel dilator produced a significant synergistic effect on the viability of HCT-15 cells, which was not associated with a significant decrease in ERK1/2 phosphorylation or VEGF or VEGFR2 expression. Therefore, this study encourages further investigation of the effects of this combination treatment on other signaling pathways in CRC.

AUTHOR’S CONTRIBUTION

It is hereby acknowledged that all authors have accepted responsibility for the manuscript's content and consented to its submission. They have meticulously reviewed all results and unanimously approved the final version of the manuscript.

LIST OF ABBREVIATIONS

ANP = Atrial Natriuretic Peptide
ATCC = American Type Culture Collection
BRAF = V-RAF Murine Sarcoma Viral Oncogene Homolog B
cGMP = cyclic GMP
CI = Combination Index
CRC = Colorectal Cancer
DRI = Drug Reduction Index
EGF = Epidermal Growth Factor
FBS = Fetal Bovine Serum
HER-2 = Human Epidermal Receptor-2
KP = Kaliuretic Peptide
LANP = Long-acting Natriuretic Peptide
MAPK = Mitogen-Activated Protein Kinase
mCRC = metastatic CRC
NTRK = Neurotrophic Tyrosine Receptor Kinase
RTK = Tyrosine Kinase Receptor
TNB = Trametinib
VDL = Vessel Dilator
VEGF = Vascular Endothelial Growth Factor

ETHICS APPROVAL AND CONSENT TO PARTICIPATE

Not applicable.

HUMAN AND ANIMAL RIGHTS

Not applicable.

CONSENT FOR PUBLICATION

Not applicable.

AVAILABILITY OF DATA AND MATERIALS

The data supporting the findings of the article is available from the corresponding author [K.A] upon reasonable request.

FUNDING

This study was funded by Deanship of Research of Jordan University of Science and Technology. (Awards/Grant number: 20170303).

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

Declared none.

REFERENCES

1
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
3
National Comprehensive Cancer Network. Colon Cancer (Version 3.2024). 2024. Available from: https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf
4
Holle LM, Clement JM, Davis LE. Colorectal Cancer. In: DiPiro JT, Talbert RL, Yee GC, Matzke GR, Wells BG, Posey LM, Eds. Pharmacotherapy: A Pathophysiologic Approach 2017.
5
Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature 2002; 417(6892): 949-54.
6
Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol 2005; 6(5): 322-7.
7
Reddy KB, Nabha SM, Atanaskova N. Role of MAP kinase in tumor progression and invasion. Cancer Metastasis Rev 2003; 22(4): 395-403.
8
Hong S, Choi Y, Kwon H-S, Park YB, Ki M-H, Shin HJ. Abstract 5162: Development of a dual inhibitor of pan-RAF and angiogenesis for the treatment of metastatic colorectal cancer with mutant K-RAS. Cancer Res 2017; 77(13): 5162.
9
Mandal R, Becker S, Strebhardt K. Stamping out RAF and MEK1/2 to inhibit the ERK1/2 pathway: an emerging threat to anticancer therapy. Oncogene 2016; 35(20): 2547-61.
10
Gilmartin AG, Bleam MR, Groy A, Moss KG, Minthorn EA, Kulkarni SG. GSK1120212 (JTP-74057) is an inhibitor of MEK activity and activation with favorable pharmacokinetic properties for sustained in vivo pathway inhibition. Clin Cancer Res 2011; 17(5): 989-1000.
11
Wu P, Nielsen TE, Clausen MH. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol Sci 2015; 36(7): 422-39.
12
Subbiah V, Kreitman RJ, Wainberg ZA, et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study. Ann Oncol 2022; 33(4): 406-15.
13
Infante JR, Fecher LA, Falchook GS, et al. Safety, pharmacokinetic, pharmacodynamic, and efficacy data for the oral MEK inhibitor trametinib: a phase 1 dose-escalation trial. Lancet Oncol 2012; 13(8): 773-81.
14
Geng R, Tan X, Zuo Z, et al. Synthetic lethal short hairpin RNA screening reveals that ring finger protein 183 confers resistance to trametinib in colorectal cancer cells. Chin J Cancer 2017; 36(1): 63.
15
Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK Inhibition With Dabrafenib and Trametinib in BRAF V600–Mutant Colorectal Cancer. J Clin Oncol 2015; 33(34): 4023-31.
16
Corcoran RB, Atreya CE, Falchook GS, Infante JR, Hamid O, Messersmith WA. Phase 1-2 trial of the BRAF inhibitor dabrafenib (D) plus MEK inhibitor trametinib (T) in BRAF V600 mutant colorectal cancer (CRC): Updated efficacy and biomarker analysis. JCO 2014; 32: 3517.
17
Bendell JC, Atreya CE, André T, Tabernero J, Gordon MS, Bernards R. Efficacy and tolerability in an open-label phase I/II study of MEK inhibitor trametinib (T), BRAF inhibitor dabrafenib (D), and anti-EGFR antibody panitumumab (P) in combination in patients (pts) with BRAF V600E mutated colorectal cancer (CRC). JCO 2014; 32: 3515.
18
Eichelbaum EJ, Vesely BA, Alli AA, Sun Y, Gower WR Jr, Vesely DL. Four cardiac hormones eliminate up to 82% of human medullary thyroid carcinoma cells within 24 hours. Endocrine 2006; 30(3): 325-32.
19
Vesely B, Eichelbaum E, Alli A, Sun Y, Gower W Jr, Vesely D. Four cardiac hormones cause cell death in 81% of human ovarian adenocarcinoma cells. Cancer Ther 2007; 5: 97-104.
20
Vesely BA, Eichelbaum EJ, Alli AA, Sun Y, Gower WR Jr, Vesely DL. Four cardiac hormones eliminate 4-fold more human glioblastoma cells than the green mamba snake peptide. Cancer Lett 2007; 254(1): 94-101.
21
Vesely BA, Eichelbaum EJ, Alli AA, Sun Y, Gower WR Jr, Vesely DL. Urodilatin and four cardiac hormones decrease human renal carcinoma cell numbers. Eur J Clin Invest 2006; 36(11): 810-9.
22
Vesely BA, Eichelbaum EJ, Sun Y, Vesely DL, Alli AA, Gower WR Jr. Four cardiac hormones cause cell death of melanoma cells and inhibit their DNA synthesis. Am J Med Sci 2007; 334(5): 342-9.
23
Vesely BA, Song S, Sanchez-Ramos J, et al. Five cardiac hormones decrease the number of human small‐cell lung cancer cells. Eur J Clin Invest 2005; 35(6): 388-98.
24
Eichelbaum EJ, Sun Y, Alli AA, Gower WR Jr, Vesely DL. Cardiac and kidney hormones cure up to 86% of human small‐cell lung cancers in mice. Eur J Clin Invest 2008; 38(8): 562-70.
25
Vesely DL, Eichelbaum EJ, Sun Y, et al. Elimination of up to 80% of human pancreatic adenocarcinomas in athymic mice by cardiac hormones. In Vivo 2007; 21(3): 445-51.
26
Vesely DL, Vesely BA, Eichelbaum EJ, Sun Y, Alli AA, Gower WR Jr. Four cardiac hormones eliminate up to two-thirds of human breast cancers in athymic mice. In Vivo 2007; 21(6): 973-8.
27
Vesely DL. Cardiac Hormones Target the Ras-MEK 1/2-ERK 1/2 Kinase Cancer Signaling Pathways. Cancers (Basel) 2011; 3(1): 1182-94.
28
Sun Y, Eichelbaum EJ, Lenz A, Skelton WP IV, Wang H, Vesely DL. Atrial natriuretic peptide and long-acting natriuretic peptide inhibit ras in human prostate cancer cells. Anticancer Res 2009; 29(6): 1889-93.
29
Sun Y, Eichelbaum EJ, Skelton WP IV, Lenz A, Wang H, Vesely DL. Vessel dilator and kaliuretic peptide inhibit Ras in human prostate cancer cells. Anticancer Res 2009; 29(4): 971-5.
30
Sun Y, Eichelbaum EJ, Wang H, Vesely DL. Atrial natriuretic peptide and long acting natriuretic peptide inhibit MEK 1/2 activation in human prostate cancer cells. Anticancer Res 2007; 27(6B): 3813-8.
31
Sun Y, Eichelbaum EJ, Wang H, Vesely DL. Atrial natriuretic peptide and long acting natriuretic peptide inhibit ERK 1/2 in prostate cancer cells. Anticancer Res 2006; 26(6B): 4143-8.
32
Sun Y, Eichelbaum EJ, Wang H, Vesely DL. Vessel dilator and kaliuretic peptide inhibit ERK 1/2 activation in human prostate cancer cells. Anticancer Res 2006; 26(5A): 3217-22.
33
Sun Y, Eichelbaum E, Wang H, Vesely D. Cardiac hormones activate ERK 1/2 kinases in human fibroblasts. Horm Metab Res 2009; 41(3): 197-201.
34
Gower WR, Vesely BA, Alli AA, Vesely DL. Four peptides decrease human colon adenocarcinoma cell number and DNA synthesis via cyclic GMP. Int J Gastrointest Cancer 2005; 36(2): 77-88.
35
Doanes AM, Hegland DD, Sethi R, Kovesdi I, Bruder JT, Finkel T. VEGF stimulates MAPK through a pathway that is unique for receptor tyrosine kinases. Biochem Biophys Res Commun 1999; 255(2): 545-8.
36
Vesely BA, Fitz SR, Gower WR Jr, Vesely DL. Vessel dilator: Most potent of the atrial natriuretic peptides in decreasing the number and DNA synthesis of human squamous lung cancer cells. Cancer Lett 2006; 233(2): 226-31.
37
Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 2006; 58(3): 621-81.
38
Chou T-C. Drug combination studies and their synergy quantification using the Chou-Talalay Method. Cancer Res 2010; 70(2): 440-6.
39
Tallarida RJ. An overview of drug combination analysis with isobolograms. J Pharmacol Exp Ther 2006; 319(1): 1-7.
40
Buck E, Eyzaguirre A, Brown E, et al. Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non–small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther 2006; 5(11): 2676-84.
41
Lee JH, Nan A. Combination drug delivery approaches in metastatic breast cancer. J Drug Deliv 2012; 2012: 1-17.
42
Ahmed D, Eide PW, Eilertsen IA, et al. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013; 2(9): e71.
43
Nguyen JP, Frost CD, Lane ML, Skelton WP IV, Skelton M, Vesely DL. Novel dual inhibitors of vascular endothelial growth factor and VEGFR2 receptor. Eur J Clin Invest 2012; 42(10): 1061-7.
44
Sun Y, Eichelbaum EJ, Wang H, Vesely DL. Vessel dilator and kaliuretic peptide inhibit MEK 1/2 activation in human prostate cancer cells. Anticancer Res 2007; 27(3B): 1387-92.
45
Grugel S, Finkenzeller G, Weindel K, Barleon B, Marmé D. Both v-Ha-Ras and v-Raf stimulate expression of the vascular endothelial growth factor in NIH 3T3 cells. J Biol Chem 1995; 270(43): 25915-9.
46
Hsu CW, Huang R, Khuc T, et al. Identification of approved and investigational drugs that inhibit hypoxia-inducible factor-1 signaling. Oncotarget 2016; 7(7): 8172-83.
47
Jiang BH, Zheng JZ, Aoki M, Vogt PK. Phosphatidylinositol 3-kinase signaling mediates angiogenesis and expression of vascular endothelial growth factor in endothelial cells. Proc Natl Acad Sci USA 2000; 97(4): 1749-53.
48
Phng LK, Gerhardt H. Angiogenesis: a team effort coordinated by notch. Dev Cell 2009; 16(2): 196-208.
49
Roca C, Adams RH. Regulation of vascular morphogenesis by Notch signaling. Genes Dev 2007; 21(20): 2511-24.
50
Woods SA, McGlade CJ, Guha A. Phosphatidylinositol 3′-kinase and MAPK/ERK kinase 1/2 differentially regulate expression of vascular endothelial growth factor in human malignant astrocytoma cells. Neuro-oncol 2002; 4(4): 242-52.
51
Yoon S, Seger R. The extracellular signal-regulated kinase: Multiple substrates regulate diverse cellular functions. Growth Factors 2006; 24(1): 21-44.
52
Zhang X, Gaspard JP, Chung DC. Regulation of vascular endothelial growth factor by the Wnt and K-ras pathways in colonic neoplasia. Cancer Res 2001; 61(16): 6050-4.
53
Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res 2000; 60(6): 1541-5.