Next Article in Journal
Combinations of Anti-Angiogenic Agents and Immune Checkpoint Inhibitors in Renal Cell Carcinoma: Best Option?
Next Article in Special Issue
CD22 Exon 12 Deletion as an Independent Predictor of Poor Treatment Outcomes in B-ALL
Previous Article in Journal
Practical Application of Circulating Tumor-Related DNA of Human Papillomavirus in Liquid Biopsy to Evaluate the Molecular Response in Patients with Oropharyngeal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Multi-Omic Approaches to Classify, Predict, and Treat Acute Leukemias

by
Maria Hernandez-Valladares
1,2,3
1
Department of Physical Chemistry, University of Granada, 18071 Granada, Spain
2
Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
3
Institute of Biotechnology, School of Sciences, University of Granada, 18071 Granada, Spain
Cancers 2023, 15(4), 1049; https://doi.org/10.3390/cancers15041049
Submission received: 30 January 2023 / Accepted: 1 February 2023 / Published: 7 February 2023
(This article belongs to the Special Issue Novel Insights in Acute Lymphoblastic and Myeloblastic Leukemia)

1. Current ALL and AML Classification and Therapeutical Strategies

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer, in which nearly 5% of the cases are diagnosed before the first year of age [1]. Around 75% of the pediatric ALL cases are characterized by a high frequency of lysine methyltransferase 2A (KTM2A, previously known as MLL) gene rearrangement KMT2A-r, which is associated with neprilysin (CD10)-negative immature B-cell precursor phenotype and a very poor prognosis [2]. Recently, an unbiased molecular classification of KTM2A-r based on transcription factors (iroquois-class homeodomain protein IRX and homeobox protein HOXA), fusion partners, and corresponding stages of B-lymphopoietic and hemato-endothelial development has been suggested for genomic-driven diagnostics and potential therapeutic strategies in infant ALL [3]. Interestingly, the combined mutations of KMT2D and phosphatidylinositol 3,4,5-triphosphate 3-phosphatase and dual-specificity protein phosphatase (PTEN) defined a group of T-cell lymphoblastic lymphoma patients with a high incidence of relapse [4]. Nearly 70% of childhood B-cell ALL (B-ALL) is currently characterized by well-known cytogenetic abnormalities, which are associated with good or poor outcomes [5]. Recently, genomic approaches have identified new genetic features among the remaining 30%, including double homeobox protein 4 (DUX4)-rearranged (DUX4-r), tyrosine-protein kinase ABL (ABL)-class fusions, and myocyte-specific enhancer factor 2 D (MEF2D)-rearranged (MED2D-r) [6,7,8]. A short while ago, whole-genome sequencing (WGS) on 210 childhood B-ALL cases detected 294 subtype-defining genetic abnormalities in 96% of patients. These included fusions affecting genes in the mitogen-activated protein (MAP) kinase pathway and improved detection of DUX4-r [9]. Last year, RNA-sequencing (RNA-seq) technology contributed remarkably to ALL classifications. RNA-seq data from leukemic cells of 1,988 patients with B-ALL provided a revised taxonomy of B-ALL, incorporating 23 subtypes defined by chromosomal rearrangements, sequence mutations, or heterogeneous genomic alterations [10]. To classify RNA-seq data according to 18 of these 23 subtypes, ALLSorts software has recently been developed to analyze B-ALL gene expression data and attribute study samples to the described subtypes [11].
ALL treatment typically involves several phases: induction (e.g., vincristine, prednisone, and anthracycline), prophylaxis (e.g., methotrexate, cytarabine, and radiation therapy), consolidation (allogeneic stem cell transplant (SCT)) and maintenance (e.g., methotrexate and 6-mercaptopurine). Targeted therapy protocols including the use of tyrosine kinase inhibitors to Philadelphia chromosome-positive ALL patients and of immunotherapeutic agents such as antibody and chimeric antigen receptor (CAR) T-cell therapies are now being examined to be part of the upfront setting [12,13].
Acute myeloid leukemia (AML) is an aggressive and heterogeneous hematological cancer. Although most patients with newly diagnosed AML achieve complete remission (CR) after intensive induction and consolidation therapy, more than half of them relapse within the next three years [14,15]. Because of the advances in cytogenetics, molecular biology, next-generation sequencing (NGS), and an increasing number of prognostic markers and therapeutic targets (i.e., Fms-like tyrosine kinase 3 (FLT3), B-cell lymphoma 2 (BCL2), and isocitrate dehydrogenase 1 and 2 (IDH1/2)), AML cases are now classified according to the World Health Organization (WHO) system that integrates clinical, molecular/genetic, and pathologic parameters, providing the separation of AML with defining genetic abnormalities from AML defined by differentiation [16]. This WHO classification has been recently supported by transcriptomics and differentiation hierarchies in the largest cohort of AML patients in China [17]. The relevance of genomic characterization has also been reflected in the new International Consensus Classification (ICC, [18]) and the European LeukemiaNet (ELN) risk stratification [19]. Recently, the molecular genetic characterization of Philadelphia chromosome-positive AML has been published, which showed that patients with this type of AML shared similar genetic profiles and clinical outcomes with those with chronic myeloid leukemia in myeloid blast crisis (CML-MBC) [20]. Within a national context, the cooperative initiative PETHEMA has established the first nationwide diagnostic network to provide standardized NGS studies for AML patients [21]. Moreover, it reported a distinct molecular profile between age groups at diagnosis and sex. In such a national effort, the clinical validation of genomic classifications in the PETHEMA cohort consisting of seven reference laboratories is carried out to demonstrate or adjust the correlation of the molecular subgroups with clinical prognosis.
Besides intensive induction and consolidation therapy, AML treatment has improved in recent years, and several new therapeutic options have been approved. Most of them include mutation-specific approaches (e.g., gilteritinib for AML patients with activating FLT3 mutations), combined epigenetic therapy with the BCL-2 inhibitor venetoclax, immunotherapy or restricted approaches for AML with myeloid-related changes (AML-MRC), or therapy-related AML (CPX-351) cases [22].

2. Single-Omics and Integration of Multi-Omics

The genetic and clinical heterogeneity of ALL and AML cases represent a challenge even for the new therapies. Moreover, relapse and treatment resistance seriously hinder ALL and AML treatment. To understand the biology underlying those differences, omic sciences have increasingly been utilized to produce useful knowledge.
Multi-omic analyses of 49 childhood ALL cell lines using proteomics, transcriptomics, and pharmacoproteomic characterization (arranged as a database for the interactive online Functional Omics Resource of ALL (FORALL), at https://proteomics.se/forall accessed on 8 January 2023) identified the diacylglycerol-analog bryostatin-1 as a therapeutic candidate in the myocyte enhancer factor 2D (MEF2D)-heterogeneous nuclear ribonucleoprotein U-like protein 1 (HNRNPUL1) fusion high-risk subtype, for which this drug activates pro-apoptotic mitogen-activated protein kinase (ERK) signaling associated with the molecular mediators of pre-B-cell negative selection [23].
To uncover the molecular changes allowing AML cells to escape treatment, two proteomic studies with liquid chromatography–mass spectrometry (LC–MS) and serial time-point samples during the disease progression of patients have shown that the proteomic profile at relapse is enriched for mitochondrial ribosomal proteins and subunits of the respiratory chain complex, indicative of reprogrammed energy metabolism from diagnosis to relapse [14,24]. Using a proteogenomic strategy, the most recent study on the latter detected 370 novel peptides, which represent a promising repertoire in the search for biomarkers and tumor-specific druggable targets. A multi-omic approach involving genomics, transcriptomics, LC–MS proteomics, and phosphoproteomics identified examples of post-transcriptionally regulated proteins and phosphorylation events both in all studied AML samples and also in patients with recurrent AML driver mutations (e.g., samples with IDH1/2 mutations displayed a high expression of the 2-oxoglutarate–dependent histone demethylases KDM4A/B/C, despite no changes in messenger RNA levels for these genes, and samples with FLT3-tyrosine kinase domain (TKD) mutationsassociated with the activation of the SRC-family tyrosine kinases FGR and HCK) [25]. Using systems medicine and multi-omics, an approach involving transcriptomics, proteomics, and metabolomics identified new molecular references such as mir-484, miR-519d-3p, activin receptor type-1 (ACVR1), receptor-type tyrosine-protein phosphatase gamma (PTPRG), PR domain zinc finger protein 14 (PRDM14), trans-acting T-cell-specific transcription factor GATA-3 (GATA3), and amino acid derivatives [26].
Altogether, these studies show us the importance and strength of single- and multi-omic approaches to decode the complex and heterogenous molecular basis of acute leukemias and to improve the current treatment and survival of ALL and AML patients.

3. Implementation and Future Use

In the past few decades, single-omic strategies have been used for research leading to ALL- and AML-related biomarker discovery. Genomic approaches have described several causative variants that are now part of international diagnosis guidelines. However, the integration of genome sequencing into a healthcare setting still lags behind. Nevertheless, examples of genomic incorporation, such as the reported initiative across multiple clinical entities of rare diseases [27] and the PETHEMA initiative described earlier, should promote the implementation of genomic approaches into clinical diagnosis. To facilitate the identification of genomic alterations of clinical significance in ALL and encourage the implementation of RNA-seq analysis in the clinic, the RaScALL platform (codes and scripts) is publicly available from the GitHub repository (https://github.com/j-rehn/RaScALL accessed on 10 January 2023) [28].
Numerous single-omic studies with patient cohorts have revealed a high number of leukemia biomarkers [29,30,31]. Regarding MS-based omic studies for biomarker discovery, very few of them have undergone validation to support their use in the clinic (Figure 1). The real bottleneck when considering the clinical use of new biomarkers is their validation using external cohorts. Because of the remarkable efforts and funds employed in both single- and multi-omic projects, it is hoped that stronger validation initiatives and closer collaborations at national and international levels can be established soon so that the introduction and use of new biomarkers in the clinic unveil more accurate diagnostic and treatment tools to defeat acute leukemias.
Multi-omic approaches that integrate the data from distinct levels of the cellular organization, i.e., from genes to metabolites, have recently been reported, which offer new perspectives for innovation in ALL and AML diagnostics and therapeutics. Multi-omic strategies bring together researchers with different expertise, including data managers and bioinformaticians, to provide protein and metabolite targets of specific genomic backgrounds. The incorporation of more clinicians in research teams appears to be more necessary than ever to transfer clinical innovation into practice.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Downing, J.R.; Wilson, R.K.; Zhang, J.; Mardis, E.R.; Pui, C.H.; Ding, L.; Ley, T.J.; Evans, W.E. The Pediatric Cancer Genome Project. Nat. Genet. 2012, 44, 619–622. [Google Scholar] [CrossRef] [PubMed]
  2. Tomizawa, D.; Miyamura, T.; Imamura, T.; Watanabe, T.; Moriya Saito, A.; Ogawa, A.; Takahashi, Y.; Hirayama, M.; Taki, T.; Deguchi, T.; et al. A risk-stratified therapy for infants with acute lymphoblastic leukemia: A report from the JPLSG MLL-10 trial. Blood 2020, 136, 1813–1823. [Google Scholar] [CrossRef]
  3. Isobe, T.; Takagi, M.; Sato-Otsubo, A.; Nishimura, A.; Nagae, G.; Yamagishi, C.; Tamura, M.; Tanaka, Y.; Asada, S.; Takeda, R.; et al. Multi-omics analysis defines highly refractory RAS burdened immature subgroup of infant acute lymphoblastic leukemia. Nat. Commun. 2022, 13, 4501. [Google Scholar] [CrossRef]
  4. Khanam, T.; Sandmann, S.; Seggewiss, J.; Ruether, C.; Zimmermann, M.; Norvil, A.B.; Bartenhagen, C.; Randau, G.; Mueller, S.; Herbrueggen, H.; et al. Integrative genomic analysis of pediatric T-cell lymphoblastic lymphoma reveals candidates of clinical significance. Blood 2021, 137, 2347–2359. [Google Scholar] [CrossRef]
  5. Schwab, C.J.; Murdy, D.; Butler, E.; Enshaei, A.; Winterman, E.; Cranston, R.E.; Ryan, S.; Barretta, E.; Hawking, Z.; Murray, J.; et al. Genetic characterisation of childhood B-other-acute lymphoblastic leukaemia in UK patients by fluorescence in situ hybridisation and Multiplex Ligation-dependent Probe Amplification. Br. J. Haematol. 2022, 196, 753–763. [Google Scholar] [CrossRef]
  6. Gu, Z.; Churchman, M.; Roberts, K.; Li, Y.; Liu, Y.; Harvey, R.C.; McCastlain, K.; Reshmi, S.C.; Payne-Turner, D.; Iacobucci, I.; et al. Genomic analyses identify recurrent MEF2D fusions in acute lymphoblastic leukaemia. Nat. Commun. 2016, 7, 13331. [Google Scholar] [CrossRef]
  7. Schwab, C.; Cranston, R.E.; Ryan, S.L.; Butler, E.; Winterman, E.; Hawking, Z.; Bashton, M.; Enshaei, A.; Russell, L.J.; Kingsbury, Z.; et al. Integrative genomic analysis of childhood acute lymphoblastic leukaemia lacking a genetic biomarker in the UKALL2003 clinical trial. Leukemia 2022. [Google Scholar] [CrossRef]
  8. Zhang, J.; McCastlain, K.; Yoshihara, H.; Xu, B.; Chang, Y.; Churchman, M.L.; Wu, G.; Li, Y.; Wei, L.; Iacobucci, I.; et al. Deregulation of DUX4 and ERG in acute lymphoblastic leukemia. Nat. Genet. 2016, 48, 1481–1489. [Google Scholar] [CrossRef] [PubMed]
  9. Ryan, S.L.; Peden, J.F.; Kingsbury, Z.; Schwab, C.J.; James, T.; Polonen, P.; Mijuskovic, M.; Becq, J.; Yim, R.; Cranston, R.E.; et al. Whole genome sequencing provides comprehensive genetic testing in childhood B-cell acute lymphoblastic leukaemia. Leukemia 2023. [Google Scholar] [CrossRef] [PubMed]
  10. Gu, Z.; Churchman, M.L.; Roberts, K.G.; Moore, I.; Zhou, X.; Nakitandwe, J.; Hagiwara, K.; Pelletier, S.; Gingras, S.; Berns, H.; et al. PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia. Nat. Genet. 2019, 51, 296–307. [Google Scholar] [CrossRef] [PubMed]
  11. Schmidt, B.; Brown, L.M.; Ryland, G.L.; Lonsdale, A.; Kosasih, H.J.; Ludlow, L.E.; Majewski, I.J.; Blombery, P.; Ekert, P.G.; Davidson, N.M.; et al. ALLSorts: An RNA-Seq subtype classifier for B-cell acute lymphoblastic leukemia. Blood Adv. 2022, 6, 4093–4097. [Google Scholar] [CrossRef] [PubMed]
  12. Biondi, A.; Gandemer, V.; De Lorenzo, P.; Cario, G.; Campbell, M.; Castor, A.; Pieters, R.; Baruchel, A.; Vora, A.; Leoni, V.; et al. Imatinib treatment of paediatric Philadelphia chromosome-positive acute lymphoblastic leukaemia (EsPhALL2010): A prospective, intergroup, open-label, single-arm clinical trial. Lancet Haematol. 2018, 5, e641–e652. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, D.W.; Kochenderfer, J.N.; Stetler-Stevenson, M.; Cui, Y.K.; Delbrook, C.; Feldman, S.A.; Fry, T.J.; Orentas, R.; Sabatino, M.; Shah, N.N.; et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: A phase 1 dose-escalation trial. Lancet 2015, 385, 517–528. [Google Scholar] [CrossRef]
  14. Aasebo, E.; Berven, F.S.; Hovland, R.; Doskeland, S.O.; Bruserud, O.; Selheim, F.; Hernandez-Valladares, M. The Progression of Acute Myeloid Leukemia from First Diagnosis to Chemoresistant Relapse: A Comparison of Proteomic and Phosphoproteomic Profiles. Cancers 2020, 12, 1466. [Google Scholar] [CrossRef] [PubMed]
  15. Aasebo, E.; Berven, F.S.; Bartaula-Brevik, S.; Stokowy, T.; Hovland, R.; Vaudel, M.; Doskeland, S.O.; McCormack, E.; Batth, T.S.; Olsen, J.V.; et al. Proteome and Phosphoproteome Changes Associated with Prognosis in Acute Myeloid Leukemia. Cancers 2020, 12, 709. [Google Scholar] [CrossRef]
  16. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F.; Bejar, R.; Berti, E.; Busque, L.; Chan, J.K.C.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
  17. Cheng, W.Y.; Li, J.F.; Zhu, Y.M.; Lin, X.J.; Wen, L.J.; Zhang, F.; Zhang, Y.L.; Zhao, M.; Fang, H.; Wang, S.Y.; et al. Transcriptome-based molecular subtypes and differentiation hierarchies improve the classification framework of acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2022, 119, e2211429119. [Google Scholar] [CrossRef] [PubMed]
  18. Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.M.; Wang, S.A.; Bagg, A.; Barbui, T.; Branford, S.; et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef]
  19. Dohner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef]
  20. Zhou, Q.; Zhao, D.; Eladl, E.; Capo-Chichi, J.M.; Kim, D.D.H.; Chang, H. Molecular genetic characterization of Philadelphia chromosome-positive acute myeloid leukemia. Leuk. Res. 2023, 124, 107002. [Google Scholar] [CrossRef]
  21. Sargas, C.; Ayala, R.; Larrayoz, M.J.; Chillon, M.C.; Carrillo-Cruz, E.; Bilbao-Sieyro, C.; Prados de la Torre, E.; Martinez-Cuadron, D.; Rodriguez-Veiga, R.; Boluda, B.; et al. Molecular Landscape and Validation of New Genomic Classification in 2668 Adult AML Patients: Real Life Data from the PETHEMA Registry. Cancers 2023, 15, 438. [Google Scholar] [CrossRef]
  22. Fleischmann, M.; Schnetzke, U.; Hochhaus, A.; Scholl, S. Management of Acute Myeloid Leukemia: Current Treatment Options and Future Perspectives. Cancers 2021, 13, 5722. [Google Scholar] [CrossRef] [PubMed]
  23. Leo, I.R.; Aswad, L.; Stahl, M.; Kunold, E.; Post, F.; Erkers, T.; Struyf, N.; Mermelekas, G.; Joshi, R.N.; Gracia-Villacampa, E.; et al. Integrative multi-omics and drug response profiling of childhood acute lymphoblastic leukemia cell lines. Nat. Commun. 2022, 13, 1691. [Google Scholar] [CrossRef]
  24. Stratmann, S.; Vesterlund, M.; Umer, H.M.; Eshtad, S.; Skaftason, A.; Herlin, M.K.; Sundstrom, C.; Eriksson, A.; Hoglund, M.; Palle, J.; et al. Proteogenomic analysis of acute myeloid leukemia associates relapsed disease with reprogrammed energy metabolism both in adults and children. Leukemia 2022. [Google Scholar] [CrossRef]
  25. Kramer, M.H.; Zhang, Q.; Sprung, R.; Day, R.B.; Erdmann-Gilmore, P.; Li, Y.; Xu, Z.; Helton, N.M.; George, D.R.; Mi, Y.; et al. Proteomic and phosphoproteomic landscapes of acute myeloid leukemia. Blood 2022, 140, 1533–1548. [Google Scholar] [CrossRef]
  26. Kelesoglu, N.; Kori, M.; Turanli, B.; Arga, K.Y.; Yilmaz, B.K.; Duru, O.A. Acute Myeloid Leukemia: New Multiomics Molecular Signatures and Implications for Systems Medicine Diagnostics and Therapeutics Innovation. OMICS 2022, 26, 392–403. [Google Scholar] [CrossRef] [PubMed]
  27. Stranneheim, H.; Lagerstedt-Robinson, K.; Magnusson, M.; Kvarnung, M.; Nilsson, D.; Lesko, N.; Engvall, M.; Anderlid, B.M.; Arnell, H.; Johansson, C.B.; et al. Integration of whole genome sequencing into a healthcare setting: High diagnostic rates across multiple clinical entities in 3219 rare disease patients. Genome Med. 2021, 13, 40. [Google Scholar] [CrossRef] [PubMed]
  28. Rehn, J.; Mayoh, C.; Heatley, S.L.; McClure, B.J.; Eadie, L.N.; Schutz, C.; Yeung, D.T.; Cowley, M.J.; Breen, J.; White, D.L. RaScALL: Rapid (Ra) screening (Sc) of RNA-seq data for prognostically significant genomic alterations in acute lymphoblastic leukaemia (ALL). PLoS Genet. 2022, 18, e1010300. [Google Scholar] [CrossRef]
  29. Aasebo, E.; Forthun, R.B.; Berven, F.; Selheim, F.; Hernandez-Valladares, M. Global Cell Proteome Profiling, Phospho-signaling and Quantitative Proteomics for Identification of New Biomarkers in Acute Myeloid Leukemia Patients. Curr. Pharm. Biotechnol. 2016, 17, 52–70. [Google Scholar] [CrossRef]
  30. Bardelli, V.; Arniani, S.; Pierini, V.; Di Giacomo, D.; Pierini, T.; Gorello, P.; Mecucci, C.; La Starza, R. T-Cell Acute Lymphoblastic Leukemia: Biomarkers and Their Clinical Usefulness. Genes 2021, 12, 1118. [Google Scholar] [CrossRef]
  31. Prada-Arismendy, J.; Arroyave, J.C.; Rothlisberger, S. Molecular biomarkers in acute myeloid leukemia. Blood Rev. 2017, 31, 63–76. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Single- and multi-omic workflow (e.g., genomics, transcriptomics, proteomics, and metabolomics) from new biomarker discovery to new biomarker applications in the clinic.
Figure 1. Single- and multi-omic workflow (e.g., genomics, transcriptomics, proteomics, and metabolomics) from new biomarker discovery to new biomarker applications in the clinic.
Cancers 15 01049 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hernandez-Valladares, M. Multi-Omic Approaches to Classify, Predict, and Treat Acute Leukemias. Cancers 2023, 15, 1049. https://doi.org/10.3390/cancers15041049

AMA Style

Hernandez-Valladares M. Multi-Omic Approaches to Classify, Predict, and Treat Acute Leukemias. Cancers. 2023; 15(4):1049. https://doi.org/10.3390/cancers15041049

Chicago/Turabian Style

Hernandez-Valladares, Maria. 2023. "Multi-Omic Approaches to Classify, Predict, and Treat Acute Leukemias" Cancers 15, no. 4: 1049. https://doi.org/10.3390/cancers15041049

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop