Abstract

BackgroundClinical studies have shown that all-trans retinoic acid (RA), which is often used in treatment of cancer patients, improves hemostatic parameters and bleeding complications such as disseminated intravascular coagulation (DIC). However, the mechanisms underlying this improvement have yet to be elucidated. In vitro studies have reported that RA upregulates thrombomodulin (TM) expression on the endothelial cell surface. The objective of this study was to investigate how and to what extent the TM concentration changes after RA treatment in cancer patients, and how this variation influences the blood coagulation cascade.ResultsIn this study, we introduced an ordinary differential equation (ODE) model of gene expression for the RA-induced upregulation of TM concentration. Coupling the gene expression model with a two-compartment pharmacokinetic model of RA, we obtained the time-dependent changes in TM and thrombomodulin-mRNA (TMR) concentrations following oral administration of RA. Our results indicated that the TM concentration reached its peak level almost 14 h after taking a single oral dose (110 frac{mg}{m^2} ) of RA. Continuous treatment with RA resulted in oscillatory expression of TM on the endothelial cell surface. We then coupled the gene expression model with a mechanistic model of the coagulation cascade, and showed that the elevated levels of TM over the course of RA therapy with a single daily oral dose (110 frac{mg}{m^2} ) of RA, reduced the peak thrombin levels and endogenous thrombin potential (ETP) up to 50 and 49%, respectively. We showed that progressive reductions in plasma levels of RA, observed in continuous RA therapy with a once-daily oral dose (110 frac{mg}{m^2} ) of RA, did not affect TM-mediated reduction of thrombin generation significantly. This finding prompts the hypothesis that continuous RA treatment has more consistent therapeutic effects on coagulation disorders than on cancer.ConclusionsOur results indicate that the oscillatory upregulation of TM expression on the endothelial cells over the course of RA therapy could potentially contribute to the treatment of coagulation abnormalities in cancer patients. Further studies on the impacts of RA therapy on the procoagulant activity of cancer cells are needed to better elucidate the mechanisms by which RA therapy improves hemostatic abnormalities in cancer.

Highlights

  • Clinical studies have shown that all-trans retinoic acid (RA), which is often used in treatment of cancer patients, improves hemostatic parameters and bleeding complications such as disseminated intravascular coagulation (DIC)

  • For the first time, we developed a mechanistic model to investigate the role of thrombomodulin (TM) in RA therapy of cancer-induced coagulation disorders

  • Our results indicate that RA-induced TM upregulation reduces thrombin generation significantly

Read more

Summary

Introduction

Clinical studies have shown that all-trans retinoic acid (RA), which is often used in treatment of cancer patients, improves hemostatic parameters and bleeding complications such as disseminated intravascular coagulation (DIC). RA therapy can improve blood clotting disorders such as thrombosis and disseminated intravascular coagulation (DIC) in cancer patients [6,7,8,9,10,11,12]. DIC, a life-threatening coagulation disorder associated with uncontrolled clot formation and/or excessive bleeding, was reported in patients with different types of cancer [13,14,15,16]. Expression of the cancer procoagulant (CP), a specific enzyme that directly activates factor X, by tumor cells is another important mechanism for the initiation of the coagulation cascade in cancer [21, 22]. Platelet aggregation and induction of inflammatory cytokine release are the other phenomena which can be responsible for blood clotting system abnormalities in cancer patients [26, 27]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call