Page 33 - Read Online
P. 33

Page 12 of 16        Merhi et al. J Cancer Metastasis Treat 2021;7:42  https://dx.doi.org/10.20517/2394-4722.2021.80

                                                             [94]
               humans have demonstrated oral bioavailability of HF . However, two aspects limit the pharmacological
               use of purified HF, which concern: (1) its poor solubility and instability [30,95] ; and (2) its interference with
               other therapeutic drugs via activation of liver CYP450 enzymes involved in the metabolism of
               pharmaceutical drugs (notably, CYP3A4) [94,96,97] . Oral administration by healthy volunteers of SJW extracts
               (300-2700 mg containing 14.8-70 mg of HF) results in the detection of 0.15-1.45 μg/mL (0.28-2.68 μM) HF
               concentrations in blood 3 h after absorption [94,98-100] . An SJW treatment period of 12.5-21 days using high HF
               extracts (> 27 mg/day) demonstrates a significant CYP3A induction . Altogether, these data strongly
                                                                            [97]
               suggest that the maximal concentration of HF in blood should not exceed 0.3 μg/mL (0.56 μM) to reduce the
               frequency of drug interactions. With regard to our data obtained in vitro, it remains to examine whether or
               not apoptosis and inhibition of secretion of AML cells in vivo can be triggered by a such low dose of HF. In
               the last years, various chemical derivatives of HF have been developed with improved stability and
               solubility, in the hope of removing HF drug metabolism stimulatory activities without affecting its
               antitumor properties. Thus, HF-DCHA, tetrahydro-HF-DCHA, octahydro-HF-DCHA, and O-(carboxy
               methyl)-HF have been shown to retain the antitumor and antiangiogenic properties of HF without inducing
               toxicity in laboratory animals [28,68,101,102] . Whether these HF derivatives retain the antileukemic properties of
               parental HF in vitro in AML primary cells and in experimental animals should be established. In parallel,
               the effects of these analogs on CYP3A activity have yet to be evaluated on healthy volunteers. Ongoing
               therapeutic strategies involve encapsulation of HF into polymeric nanoparticles for overcoming hurdles that
               hamper the bioactivity and bioavailability of the drug [103-106] . In conclusion, although further studies are
               undoubtedly required, all these data highlight that HF and its derivatives deserve further study to evaluate
               them in association with chemotherapy in the treatment of AML.

               DECLARATIONS
               Acknowledgments
               The authors thank Fanny Fava (Hôpital Saint-Antoine, Paris, France) for her expert technical assistance.
               This work is dedicated to Jean-Pierre Kolb, a pionner in the research on antitumoral activity of HF in CLL.


               Authors’ contributions
               Conceptualization: Merhi F, Bauvois B
               Formal analysis: Merhi F, Tang R, Bauvois B
               Investigation: Merhi F, Bauvois B
               Methodology: Tang R, Merhi F, Bauvois B
               Resources: Legrand O
               Writing, review and editing: Merhi F, Tang R, Legrand O, Nguyen-Khac F, Susin SA, Bauvois B
               All authors have read and agreed to the published version of the manuscript.


               Availability of data and materials
               Not applicable.


               Financial support and sponsorship
               This work was supported by the French National Institute of Health and Medical Research (INSERM). This
               research received no external funding.

               Conflicts of interest
               All authors declared that there are no conflicts of interest.
   28   29   30   31   32   33   34   35   36   37   38