Page 305 - Read Online
P. 305

Robinson et al. J Cancer Metastasis Treat 2019;5:39  I  http://dx.doi.org/10.20517/2394-4722.2019.15                         Page 5 of 9

               TELOMERE-DIRECTED THERAPIES FOR METASTATIC BREAST CANCER: CURRENT AND
               FUTURE PERSPECTIVES
               The functions of telomerase in tumorigenesis have been rigorously interrogated over the last several decades,
               as  has  the  potential  to  target telomerase therapeutically [65,81] .  The  telomerase  inhibitors BIBR1532 and
               GRN163L (also known as Imetelstat) display high efficacy in depleting the CSC pool and disrupting breast
               cancer metastasis [82-85] . Indeed, Imetelstat was assessed in a Phase I clinical trial for recurrent or metastatic
               breast cancer, although the trial was suspended due to dose-limiting toxicity . In addition to such toxicity
                                                                                [81]
               concerns, the success of telomerase inhibitors in clinical trials has thus far been moderated by the inherent
               complexity of telomere homeostasis. First, telomere shortening-induced senescence can be bypassed in
                                                                                [86]
               the absence of functional p53 or other components of the DDR machinery . Second, the critically short
               telomeres and chromosomal instability associated with telomere crisis are disproportionately associated with
               metastasis [87,88] . Thus, the evolution of DTCs that underlie metastatic disease may be enhanced unwittingly
               by therapies that promote telomere shortening. Despite these challenges, telomerase remains an appealing
               therapeutic objective in need of innovative targeting approaches in which these evolutionary considerations
               are taken into account.

               Emerging telomerase-targeting strategies include cytotoxic small molecules that act as substrates for
               telomerase as well as anti-telomerase immunotherapies [89-92] . Current immunotherapeutic platforms are
               primarily centered on telomerase peptide or dendritic cell vaccines, which can be engineered to elicit either
               CD4+ or CD8+ T cell antitumor responses . These strategies are being assessed in diverse preclinical
                                                     [93]
               settings, including breast cancer. Indeed, the telomerase peptide vaccine Vx-001 is progressing through
                                                   [90]
               clinical trials for advanced solid tumors . More recent investigations have examined the feasibility of
               adoptive transfer of anti-telomerase chimeric antigen receptor (CAR) T cells for treating triple-negative
               breast cancer . Future studies into the generalizability of anti-telomerase CAR T cell therapy to other
                           [94]
               breast cancer subtypes, as well as the efficacy of these diverse immunotherapeutic approaches in clinical
               settings will be of tremendous value.

               Although the functions of specific ALT-associated proteins have been elucidated, their utility as therapeutic
               targets for ALT-driven cancers has only recently been investigated. For example, the DNA damage-
               responsive kinase ataxia-telangectasia and Rad3-related (ATR) is activated secondary to depletion of
               ATRX, which leads to persistent retention of replication protein A (RPA) at telomeres and generation of
                                                                                                  [95]
               a recombinogenic substrate. Inhibition of ATR, in turn, triggers apoptosis of ALT-positive cells . BLM,
               a RecQ DNA helicase, unwinds telomeric G-quadruplex structures and coordinates 5'→3' end resection
               during telomere recombination [96,97] . Accordingly, a recently-developed small molecule inhibitor of BLM
               may possess great potential as an anticancer agent against ALT-driven tumors . Finally, topoisomerase
                                                                                    [98]
               IIIα (Topo IIIα) associates with BLM and regulates the topology of telomeric recombination intermediates.
               Interestingly, genetic inactivation of Topo IIIα selectively reduces the survival of ALT-positive compared
                                      [99]
               to telomerase-positive cells . Moreover, telomerase activity is enhanced in the surviving fraction of Topo
               IIIα-deficient cells , suggesting that telomerase activation provides a pathway for chemoresistance. Thus,
                               [100]
               targeting TMMs may best be achieved using a multidrug regimen consisting of multiple anti-TMM agents
               or an anti-TMM agent in combination with chemotherapy or other targeted agents . The effectiveness of
                                                                                      [101]
               these therapeutic modalities in eliminating breast CSCs and in treating metastatic breast cancers remain
               intriguing and important open questions.



               CONCLUSION
               By overseeing multiple pathways that promote breast cancer stemness, EMT, and metastasis, telomeres
               function as critical nodes in the nexus between cellular immortalization, tumor evolution, and disease
               progression. The selection of TMM likely exhibits a high degree of plasticity in different tumor cell types
   300   301   302   303   304   305   306   307   308   309   310