Page 47 - Read Online
P. 47

Page 726                 Bijnsdorp et al. Cancer Drug Resist 2021;4:719-27  https://dx.doi.org/10.20517/cdr.2021.21

               combination with TPI would prevent such an effect. Therefore, it is of interest to explore combinations of
               TAS-102 (which contains TPI) with rapamycin, which should include mechanistic studies evaluating, e.g.,
               the phosphorylation status of Akt, mTOR, and p70/S6k.


               In conclusion, resistance to rapamycin may be related to the activation of autophagy by TdR. To optimize
               the use of mTOR inhibitors, it seems important to study the levels of TdR combined with intratumoral TP
               expression.


               DECLARATIONS
               Authors’ contributions
               Concept: Bijnsdorp IV, Peters GJ
               Data collection: Bijnsdorp IV
               Writing: Bijnsdorp IV, Peters GJ

               Availability of data and materials
               Not applicable.


               Financial support and sponsorship
               This study was supported by an educational grant from Taiho Pharmaceuticals, Tokushima, Japan.


               Ethical approval and consent to participate
               Not applicable.


               Conflicts of interest
               Both authors declared that there are no conflicts of interest.


               Consent for publication
               Not applicable.


               Copyright
               © The Author(s) 2021.

               REFERENCES
               1.       Bruin M, Temmink OH, Hoekman K, Pinedo HM, Peters GJ. Role of platelet derived endothelial cell growth factor/thymidine
                   phosphorylase in health and disease. Cancer Therapy 2006;4:99-124.
               2.       Bronckaers A, Gago F, Balzarini J, Liekens S. The dual role of thymidine phosphorylase in cancer development and chemotherapy.
                   Med Res Rev 2009;29:903-53.  DOI  PubMed  PMC
               3.       Ackland SP, Peters GJ. Thymidine phosphorylase: its role in sensitivity and resistance to anticancer drugs. Drug Resist Updat
                   1999;2:205-14.  DOI  PubMed
               4.       Brown NS, Bicknell R. Thymidine phosphorylase, 2-deoxy-D-ribose and angiogenesis. Biochem J 1998;334:1-8.  DOI  PubMed  PMC
               5.       Liekens S, Bronckaers A, Pérez-Pérez MJ, Balzarini J. Targeting platelet-derived endothelial cell growth factor/thymidine
                   phosphorylase for cancer therapy. Biochem Pharmacol 2007;74:1555-67.  DOI  PubMed
               6.       Matsushita S, Nitanda T, Furukawa T, et al. The effect of a thymidine phosphorylase inhibitor on angiogenesis and apoptosis in
                   tumors. Cancer Res 1999;59:1911-6.  PubMed
               7.       Mayer RJ, Van Cutsem E, Falcone A, et al. Randomized trial of TAS-102 for refractory metastatic colorectal cancer. N Engl J Med
                   2015;372:1909-19.  DOI  PubMed
               8.       Peters GJ. Therapeutic potential of TAS-102 in the treatment of gastrointestinal malignancies. Ther Adv Med Oncol 2015:7;340-56.
                   DOI  PubMed  PMC
               9.       Peters GJ, Bijnsdorp IV. TAS-102: more than an antimetabolite. Lancet Oncol 2012;13:e518-9.  DOI  PubMed
               10.      Bijnsdorp IV, Azijli K, Jansen EE, et al. Accumulation of thymidine-derived sugars in thymidine phosphorylase overexpressing cells.
                   Biochem Pharmacol 2010;80:786-92.  DOI  PubMed
               11.      Hotchkiss KA, Ashton AW, Schwartz EL. Thymidine phosphorylase and 2-deoxyribose stimulate human endothelial cell migration by
                   specific activation of the integrins alpha 5 beta 1 and alpha V beta 3. J Biol Chem 2003;278:19272-9.  DOI  PubMed
   42   43   44   45   46   47   48   49   50   51   52