Page 74 - Read Online
P. 74

Page 68                   Leung et al. J Transl Genet Genom 2023;7:79-86  https://dx.doi.org/10.20517/jtgg.2023.09

                                                                                [1]
               heterogeneity that tends to evolve during the course of cancer treatment . This information has also
               formed the theoretical basis of personalized cancer treatment based on the unique mutation compositions
               of cancers at diagnosis and relapse.


               Leukaemia has long been the foundational paradigm for new concepts in cancer biology and innovation in
               therapeutic targeting. Phenomenal observations of chromosomal translocations in chronic myeloid
                               [2]
                                                                       [3]
               leukaemia (CML)  and acute promyelocytic leukaemia (APL) , and the subsequent identification of
               pathogenic fusion genes in these diseases, have led to the development of tyrosine kinase inhibitors and
               differentiation agents. Both of these therapies have transformed the outcomes of patients, who can now be
               cured based on chemotherapy-free regimens.


               Acute myeloid leukaemia (AML) is a devastating disease worldwide, for which treatment outcomes are
               unsatisfactory overall. The considerable inter- and intra-tumoral heterogeneities in AML have hampered
               the development of effective treatments applicable to the multitude of AML subtypes. However, advances in
               cancer genetics and genomics in recent years have empowered clinicians and scientists with the ability to
               analyze leukaemia heterogeneities and clonal evolution at different treatment stages. This information has
                                                                                                   [4]
               begun to influence the clinical management of AML, improving outcomes for some of these patients .

               We review the history of genomic research in AML in the past and how this knowledge has led to
               improvements in the treatment outcomes of patients. We also discuss the future development of a
               personalized approach to AML management.


               ACUTE MYELOID LEUKAEMIA IN THE CLINICS – CURRENT STATE OF AFFAIRS
               AML is a group of heterogeneous diseases with diverse morphologies, immunophenotypes, cytogenetics,
               and genetics, sharing in common an abnormal increase in blasts in blood and bone marrow (BM). It occurs
               in 3-5 patients per 100,000 individuals every year, and its incidence has increased in the past few decades. It
               is a highly lethal disease and is the fifth deadliest cancer of all kinds, particularly in elderly patients who are
               unfit to receive standard treatment. About 50% of AML cases carry normal cytogenetics, and they show on
               average 2-4 recurrent mutations in different combinations, some of which are considered drivers and others
                                         [5]
               passengers in leukemogenesis . Some of these mutations are of prognostic significance. In particular,
               NPM1 mutation and bZIP in-frame mutation of CEBPa are associated with a favorable response to
               conventional chemotherapy, whereas FLT3-ITD is associated with a less favorable response. About
               10%-15% of AML cases carry translocation t(8;21) (RUNX1::RUNX1T1) or inversion of chromosome 16
               (CBFβ::MYH11), both of which involve components of the core-binding factor (CBF), which is a
               heterodimeric transcription factor comprising the non-DNA-binding CBFβ chain and the DNA-binding
               CBFα chain RUNX1. Half of these patients carry KIT mutations, which confer an inferior prognosis in this
               AML subtype, which hitherto had a favorable response to conventional chemotherapy . Another 10% of
                                                                                          [6]
               AML cases carry complex (≥ 3 karyotypic abnormalities) or monosomy karyotype (≥ 2 monosomies or 1
               monosomy and one structural abnormality), and this subtype portends an extremely poor prognosis,
               particularly those carrying TP53 mutations, which happen in half of the patients with this subtype . The
                                                                                                    [7]
               rest of AML cases are made up of diverse diseases with different karyotypic and genetic abnormalities
               [Figure 1].


               Despite the heterogeneity, AML has been managed by a one-size-fits-all approach in the past 4 decades. In
               young and fit patients, intensive chemotherapy and allogeneic hematopoietic stem cell transplantation
               (HSCT) are the mainstays of treatment. However, this approach has reached an impasse, and only 40% of
               patients can survive long-term. A number of agents, when added to the chemotherapy regimen, were shown
   69   70   71   72   73   74   75   76   77   78   79