Abstract

Simple SummaryChronic myeloid leukemia (CML) is a blood cancer with a very good long-term prognosis for the majority of patients. Still, some patients relapse or progress under the standard therapy. Therefore, we performed an in-depth computational gene expression analysis to determine similarities and differences between the CML phases at the level of single genes, signaling pathways and gene regulatory networks and further compared treatment-resistant patients to the individual phases to identify associated expression differences. Our study provides several lines of evidence that CML development represents a three rather than a two step process. The identified characteristic gene expression alterations in advanced phases indicate that affected patients could potentially profit from other existing drugs. Moreover, characteristic signaling pathway changes identified for patients for which the standard therapy was inefficient may allow to predict such patients before treatment start and could also provide a basis to develop treatment strategies for them.Chronic myeloid leukemia (CML) is a slowly progressing blood cancer that primarily affects elderly people. Without successful treatment, CML progressively develops from the chronic phase through the accelerated phase to the blast crisis, and ultimately to death. Nowadays, the availability of targeted tyrosine kinase inhibitor (TKI) therapies has led to long-term disease control for the vast majority of patients. Nevertheless, there are still patients that do not respond well enough to TKI therapies and available targeted therapies are also less efficient for patients in accelerated phase or blast crises. Thus, a more detailed characterization of molecular alterations that distinguish the different CML phases is still very important. We performed an in-depth bioinformatics analysis of publicly available gene expression profiles of the three CML phases. Pairwise comparisons revealed many differentially expressed genes that formed a characteristic gene expression signature, which clearly distinguished the three CML phases. Signaling pathway expression patterns were very similar between the three phases but differed strongly in the number of affected genes, which increased with the phase. Still, significant alterations of MAPK, VEGF, PI3K-Akt, adherens junction and cytokine receptor interaction signaling distinguished specific phases. Our study also suggests that one can consider the phase-wise CML development as a three rather than a two-step process. This is in accordance with the phase-specific expression behavior of 24 potential major regulators that we predicted by a network-based approach. Several of these genes are known to be involved in the accumulation of additional mutations, alterations of immune responses, deregulation of signaling pathways or may have an impact on treatment response and survival. Importantly, some of these genes have already been reported in relation to CML (e.g., AURKB, AZU1, HLA-B, HLA-DMB, PF4) and others have been found to play important roles in different leukemias (e.g., CDCA3, RPL18A, PRG3, TLX3). In addition, increased expression of BCL2 in the accelerated and blast phase indicates that venetoclax could be a potential treatment option. Moreover, a characteristic signaling pathway signature with increased expression of cytokine and ECM receptor interaction pathway genes distinguished imatinib-resistant patients from each individual CML phase. Overall, our comparative analysis contributes to an in-depth molecular characterization of similarities and differences of the CML phases and provides hints for the identification of patients that may not profit from an imatinib therapy, which could support the development of additional treatment strategies.

Highlights

  • Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm that accounts for about 15% of all leukemias diagnosed in adulthood [1]

  • We found that chronic phase samples were clearly separated from blast crisis samples

  • The phase-wise development of CML known from histology is reflected in the global gene expression profiles of CML samples at the molecular level

Read more

Summary

Introduction

Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm that accounts for about 15% of all leukemias diagnosed in adulthood [1]. About one to two cases occur per year among 100,000 adults [1]. As described in [3], CML development is initiated in about 95% of cases by a characteristic reciprocal translocation between the chromosomes 9 and 22 (t(9;22)(q34.1;q11.2)), which produces a shortened chromosome 22 that is known as Philadelphia chromosome. The breakpoints of this translocation are located within the breakpoint cluster region (BCR). The resulting BCR-ABL1 fusion gene is an oncogene that encodes a constitutively active tyrosine kinase, which triggers uncontrolled cell proliferation

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.