Next Article in Journal
Metabolic Flexibility Is a Determinant of Breast Cancer Heterogeneity and Progression
Next Article in Special Issue
Pharmacologic Strategies for Post-Transplant Maintenance in Acute Myeloid Leukemia: It Is Time to Consider!
Previous Article in Journal
Cathepsin S Evokes PAR2-Dependent Pain in Oral Squamous Cell Carcinoma Patients and Preclinical Mouse Models
Previous Article in Special Issue
Therapeutic Targeting of Acute Myeloid Leukemia by Gemtuzumab Ozogamicin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions

1
Hematology Service and Romagna Transplant Network for HSCT, 48121 Ravenna, Italy
2
Department of Internal Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(18), 4698; https://doi.org/10.3390/cancers13184698
Submission received: 9 September 2021 / Accepted: 14 September 2021 / Published: 19 September 2021
Acute myeloid leukemia (AML) is a clonal disorder resulting from acquired somatic mutations in hematopoietic progenitor cells that lead to the dysregulation of differentiation and the proliferation of hematopoietic cells [1,2]. There is accumulating evidence that a great number of genomic alterations, such as chromosomal rearrangements; gene amplifications, deletions, and mutations are essential for AML classification [1,2,3,4,5,6]. Furthermore, the identification of genetic lesions plays an increasing role in the prognosis and treatment of AML patients [1,2,3,4]. Next-generation sequencing (NGS) as well as whole-genome sequencing (WGS) has been recently integrated into clinical practice, allowing for a better risk stratification of AML patients. In fact, the routine use of NGS methodology has made possible the identification of one or more somatic mutations in more than 90% of patients with AML [1,2,3,4,5,6,7,8,9]. The most frequently mutated genes include NPM1, FLT3, DNMT3A, IDH1, IDH2, TET2, RUNX1, TP53, WT1, NRAS, and CEBPα. However, the prognostic-predictive significance of genetic mutations is more important in normal karyotype AML [4,10,11].
These aberrations might help to identify AML pathways of clonal dominance and shifts that could assist hematologists in targeting precision medicine therapies [7,8,9].
Over the last few years, there has been an increasing understanding of molecular aberrations that trigger the development of AML, and an increase in the use of novel molecular biology technologies, thus facilitating the development of investigational drugs targeting driver genetic mutations [7,8,9]. Based on this consideration, it could be postulated that identifying “druggable” mutations paves the way for the use of novel targeted therapies [12]. This Special Issue of Cancers focuses on the novel diagnostic and therapeutic tools for the management of AML with the main aim of improving our knowledge in the field of AML [10,11,12,13,14,15,16,17].
Twenty years ago, the discovery of imatinib for the treatment of chronic myeloid leukemia and its remarkable activity raised hopes for similar benefits in targeted therapies for AML. Over the last few years, precision medicine has been proposed for several hematological malignancies including acute leukemia, and more than 100 different targets have been identified in AML, making it an optimal candidate for experimental clinical studies [18,19,20,21,22].
Targeting fms, such as tyrosine kinase-3 (FLT-3), have been the first example of clinically actionable mutations, making them an attractive target for hematologists, as well as for pharmaceutical and biotechnology companies developing novel drugs [23]. Over the last few years, a great number of FLT-3-targeting drugs have been developed and tested in clinical trials [24,25]. The main consideration for the use of FLT-3 targeting compounds is related to the notion that the internal tandem duplications of the FLT3 gene (FLT3- ITD-mut) characterize a significant number (25–30%) of AML cases and represent an independent predictor of poor prognosis associated with an increased risk of relapse. In the Ratify trial, the addition of midostaurin to daunorubucine- and cytarabine- based induction therapy (the so-called “3 + 7”regimen) resulted in a significant improvement in patient outcome for the very first time [23,24,25]. More recent data have shown that FLT3 inhibitors produced better results in those patients carrying a specific insertion point of the ITD of FLT3 (located in the beta1-sheet region) compared to patients with other ITD insertion sites. Moreover, the use of midostaurin is associated with longer survival in patients with a high allelic FLT3-ITD ratio who lack NPM1 mutations (NPM1wt/FLT-3-ITDhigh). Interestingly, even after allogeneic stem cell transplantation (allo-SCT), FLT3-mutated AML is associated with a higher risk of early relapse (up to 60%) [26].Chemotherapy or FLT-3 inhibitors alone, or combined with donor lymphocyte infusions, show unsatisfactory long-term therapeutic effects. Therefore, several studies have investigated the use of post-transplant maintenance with FLT-3 inhibitors as a strategy to reduce the risk of relapse after allo-SCT. Some of these studies showed very promising results, making the use of FLT-3 inhibitors relatively popular in this clinical setting [26,27,28,29,30,31].
More recently, a great number of experimental drugs targeting single metabolic pathways have been developed and tested in phase 1, 2 and 3 clinical trials [12,17,18,19,20,21,22]. The question remains: what is the main reason for using these agents in AML?
Metabolism is a complex biochemical process that mammalian cells use to generate energy and maintain their growth and survival. Metabolism encompasses the generation of energy (ATP), the synthesis and breakdown of amino acids, glucose, fatty acids, and oxidative phosphorylation. In AML cells, metabolism can promote tumor growth and cellular proliferation. As a consequence, these alterations have emerged as a potential therapeutic target [21,22].
Some of these compounds proved to be effective in targeting altered metabolism, such as in the Krebs/citric acid cycle, hedgehog pathway (glasdegib), and the mitochondrial oxidative and carrier-associated secretory membrane proteins (so-called SCAMP), making the AML therapeutic armamentarium more flexible and intriguing. The identification of actionable bioenergetic cell pathways is rapidly expanding, allowing us to hypothesize new potential therapeutic modalities in AML [18].
More recently, the use of Bcl-2 inhibitors (venetoclax), and IDH inhibitors data have raised hopes for the benefits of targeted therapies in AML. For example, in older/unfit AML patients, hypomethylating agents (HMAs) and venetoclaxare now the new standard of care in this setting [12,13,14].
AML is also known to be an epigenetically heterogeneous disease. Based on this consideration, hypomethylating agents such as 5-azacitidine, decitabine, and guadecitabine; HDAC inhibitors (pracinostat, bromodomain and extraterminal (BET) family protein, among others), as well as novel agents targeting the aberrant activation of gene signaling (m-TOR-CDK9; syk inhibitors; Toll-like receptor signaling) have been tested in clinical trials.
Oncoprotein degradation is the basis of the curative effect of the combination of arsenic trioxide and all-trans retinoic acid in acute promyelocytic leukemia and has shown promising results in NPM1-mutated AML.
As far as immunotherapy is concerned, immunotherapy-based protocols have demonstrated modest activity in AML which needs to be confirmed in larger series. In contrast, encouraging results have been observed in acute lymphoblastic leukemia and in non-Hodgkin and Hodgkin lymphoma [12,13,14].
Interestingly, novel drugs and small compounds have been developed that can activate p53 in AML cells expressing the wild-type form of p53; the clinical implication related to the use of these molecules is under investigation [12].
A comprehensive list of druggable genetic lesions and targeted therapy in AML is shown in Table 1.
The role of minimal residual disease detection for the sharper management of acute leukemia is also considered in this issue, provided that the authors include a clinical setting in which one or multiple druggable genetic lesions are presented [3,8,31].
With this background, the journal Cancers is launching a collection of articles dealing with NGS data and the use of targeted therapies, as well as cellular therapies [32,33,34].

Funding

This research was funded by the Utah Department of Transportation, grant number UDOT 08-9117.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Papaemmanuil, E.; Gerstung, M.; Bullinger, L.; Gaidzik, V.I.; Paschka, P.; Roberts, N.D.; Potter, N.E.; Heuser, M.; Thol, F.; Bolli, N.; et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N. Engl. J. Med. 2016, 374, 2209–2221. [Google Scholar] [CrossRef]
  2. Ley, T.J.; Miller, C.; Ding, L.; Raphael, B.J.; Mungall, A.J.; Robertson, A.; Hoadley, K.; Triche, T.J., Jr.; Laird, P.W.; Baty, J.D.; et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar] [CrossRef] [Green Version]
  3. Patel, J.P.; Gonen, M.; Figueroa, M.E.; Fernandez, H.; Sun, Z.; Racevskis, J.; Van Vlierberghe, P.; Dolgalev, I.; Thomas, S.; Aminova, O.; et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 2012, 366, 1079–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mack, E.K.M.; Marquardt, A.; Langer, D.; Ross, P.; Ultsch, A.; Kiehl, M.G.; Mack, H.I.D.; Haferlach, T.; Neubauer, A.; Brendel, C. Comprehensive genetic diagnosis of acute myeloid leukemia by next-generation sequencing. Haematologica 2019, 104, 277–287. [Google Scholar] [CrossRef] [PubMed]
  5. Issa, G.C.; DiNardo, C.D. Acute myeloid leukemia with IDH1 and IDH2 mutations: 2021 treatment algorithm. Blood Cancer J. 2021, 11, 107. [Google Scholar] [CrossRef]
  6. Borthakur, G.; Kantarjian, H. Core binding factor acute myelogenous leukemia-2021 treatment algorithm. Blood Cancer J. 2021, 11, 114. [Google Scholar] [CrossRef] [PubMed]
  7. Haferlach, T.; Hutter, S.; Meggendorfer, M. Genome Sequencing in Myeloid Cancers. N. Engl. J. Med. 2021, 384, e106. [Google Scholar] [CrossRef] [PubMed]
  8. Tsai, C.-H.; Tang, J.-L.; Tien, F.-M.; Kuo, Y.-Y.; Wu, D.-C.; Lin, C.-C.; Tseng, M.-H.; Peng, Y.-L.; Hou, M.-F.; Chuang, Y.-K.; et al. Clinical implications of sequential MRD monitoring by NGS at 2 time points after chemotherapy in patients with AML. Blood Adv. 2021, 5, 2456–2466. [Google Scholar] [CrossRef]
  9. Sahajpal, N.S.; Mondal, A.K.; Ananth, S.; Njau, A.; Ahluwalia, P.; Jones, K.; Ahluwalia, M.; Okechukwu, N.; Savage, N.M.; Kota, V.; et al. Clinical performance and utility of a comprehensive next-generation sequencing DNA panel for the simultaneous analysis of variants, TMB and MSI for myeloid neoplasms. PLoS ONE 2020, 15, e0240976. [Google Scholar] [CrossRef]
  10. Heuser, M.; Ofran, Y.; Boissel, N.; Brunet Mauri, S.; Craddock, C.; Janssen, J.; Wierzbowska, A.; Buske, C.; ESMO Guidelines Committee. Acute myeloid leukaemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020, 31, 697–712. [Google Scholar] [CrossRef]
  11. Blum, W.G.; Mims, A.S. Treating acute myeloid leukemia in the modern era: A primer. Cancer 2020, 126, 4668–4677. [Google Scholar] [CrossRef]
  12. Cucchi, D.G.J.; Polak, T.B.; Ossenkoppele, G.J.; Uyl-De Groot, C.A.; Cloos, J.; Zweegman, S.; Janssen, J.J.W.M. Two decades of targeted therapies in acute myeloid leukemia. Leukemia 2021, 35, 651–660. [Google Scholar] [CrossRef] [PubMed]
  13. Canaani, J. Management of AML Beyond “3 + 7” in 2019. Clin. Hematol. Int. 2019, 1, 10–18. [Google Scholar] [CrossRef]
  14. Kantarjian, H.M.; Kadia, T.M.; DiNardo, C.D.; Welch, M.A.; Ravandi, F. Acute myeloid leukemia: Treatment and research outlook for 2021 and the MD Anderson approach. Cancer 2021, 127, 1186–1207. [Google Scholar] [CrossRef]
  15. Short, N.J.; Tallman, M.S.; Pollyea, D.A.; Ravandi, F.; Kantarjian, H. Optimizing Risk Stratification in Acute Myeloid Leukemia: Dynamic Models for a Dynamic Therapeutic Landscape. J. Clin. Oncol. 2021. [Google Scholar] [CrossRef]
  16. Mer, A.S.; Heath, E.M.; MadaniTonekaboni, S.A.; Dogan-Artun, N.; Nair, S.K.; Murison, A.; Garcia-Prat, L.; Shlush, L.; Hurren, R.; Voisin, V.; et al. Biological and therapeutic implications of a unique subtype of NPM1 mutated AML. Nat. Commun. 2021, 12, 1054. [Google Scholar] [CrossRef]
  17. Falini, B.; Brunetti, L.; Sportoletti, P.; Martelli, M.P. NPM1-mutated acute myeloid leukemia: From bench to bedside. Blood 2020, 136, 1707–1721. [Google Scholar] [CrossRef] [PubMed]
  18. Kent, A.; Vasu, S.; Schatz, D.; Monson, N.; Devine, S.; Smith, C.; Gutman, J.A.; Pollyea, D.A. Glasdegib as maintenance therapy for patients with AML and MDS patients at high risk for postallogeneic stem cell transplant relapse. Blood Adv. 2020, 4, 3102–3108. [Google Scholar] [CrossRef]
  19. Shi, A.; Murai, M.G.; He, S.; Lund, G.; Hartley, T.; Purohit, T.; Reddy, G.; Chruszcz, M.; Grembecka, J.; Cierpicki, T. Structural insights into inhibition of the bivalent menin-MLL interaction by small molecules in leukemia. Blood 2012, 120, 4461–4469. [Google Scholar] [CrossRef]
  20. Dzama, M.M.; Steiner, M.; Rausch, J.; Sasca, D.; Schönfeld, J.; Kunz, K.; Taubert, M.C.; McGeehan, G.M.; Chen, C.-W.; Mupo, A.; et al. Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition. Blood 2020, 136, 2442–2456. [Google Scholar] [CrossRef] [PubMed]
  21. Nachmias, B.; Schimmer, A.D. Targeting nuclear import and export in hematological malignancies. Leukemia 2020, 34, 2875–2886. [Google Scholar] [CrossRef]
  22. Latif, A.-N.; Newcombe, A.; Li, S.; Gilroy, K.; Robertson, N.A.; Lei, X.; Stewart, H.J.; Cole, J.; Terradas, M.; Rishi, L.; et al. BRD4-mediated repression of p53 is a target for combination therapy in AML. Nat. Commun. 2021, 12, 241. [Google Scholar] [CrossRef]
  23. Stone, R.M.; Manley, P.W.; Larson, R.A.; Capdeville, R. Midostaurin: Its odyssey from discovery to approval for treating acute myeloid leukemia and advanced systemic mastocytosis. Blood Adv. 2018, 2, 444–453. [Google Scholar] [CrossRef] [Green Version]
  24. Bazarbachi, A.; Labopin, M.; Battipaglia, G.; Djabali, A.; Forcade, E.; Arcese, W.; SociÈ, G.; Blaise, D.; Halter, J.; Gerull, S.; et al. Allogeneic Stem Cell Transplantation for FLT3-Mutated Acute Myeloid Leukemia: In Vivo T-Cell Depletion and Posttransplant Sorafenib Maintenance Improve Survival. A Retrospective Acute Leukemia Working Party-European Society for Blood and Marrow Transplant Study. Clin. Hematol. Int. 2019, 1, 58–74. [Google Scholar] [CrossRef] [Green Version]
  25. Schiller, G.J.; Tuttle, P. Desai Allogeneic Hematopoietic Stem Cell Transplantation in FLT3-ITD-Positive Acute Myelogenous Leukemia: The Role for FLT3 Tyrosine Kinase Inhibitors Post-Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 982–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bazarbachi, A.; Bug, G.; Baron, F.; Brissot, E.; Ciceri, F.; Dalle, I.A.; Döhner, H.; Esteve, J.; Floisand, Y.; Giebel, S.; et al. Clinical practice recommendation on hematopoietic stem cell transplantation for acute myeloid leukemia patients with FLT3-internal tandem duplication: A position statement from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Haematologica 2020, 105, 1507–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Duarte, R.F.; Labopin, M.; Bader, P.; Basak, G.W.; Bonini, C.; Chabannon, C.; Corbacioglu, S.; Dreger, P.; Dufour, C.; Gennery, A.R.; et al. Indications for haematopoietic stem cell transplantation for haematological diseases, solid tumours and immune disorders: Current practice in Europe, 2019. Bone Marrow Transplant. 2019, 54, 1525–1552. [Google Scholar] [CrossRef]
  28. Nelson, M.A.; McLaughlin, K.L.; Hagen, J.T.; Coalson, H.S.; Schmidt, C.; Kassai, M.; Kew, K.A.; McClung, J.M.; Neufer, P.D.; Brophy, P.; et al. Intrinsic OXPHOS limitations underlie cellular bioenergetics in leukemia. eLife 2021, 10, e63104. [Google Scholar] [CrossRef]
  29. Borthakur, G.; Odenike, O.; Aldoss, I.; Rizzieri, D.A.; Prebet, T.; Chen, C.; Popovic, R.; Modi, D.A.; Joshi, R.H.; Wolff, J.E.; et al. A phase 1 study of the pan-bromodomain and extraterminal inhibitor mivebresib (ABBV-075) alone or in combination with venetoclax in patients with relapsed/refractory acute myeloid leukemia. Cancer 2021, 127, 2943–2953. [Google Scholar] [CrossRef]
  30. Guolo, F.; Fianchi, L.; Minetto, P.; Clavio, M.; Gottardi, M.; Galimberti, S.; Rizzuto, G.; Rondoni, M.; Bertani, G.; Dargenio, M.; et al. CPX-351 treatment in secondary acute myeloblastic leukemia is effective and improves the feasibility of allogeneic stem cell transplantation: Results of the Italian compassionate use program. Blood Cancer J. 2020, 10, 96. [Google Scholar] [CrossRef]
  31. Short, N.J.; Zhou, S.; Fu, C.; Berry, D.A.; Walter, R.B.; Freeman, S.D.; Hourigan, C.S.; Huang, X.; Nogueras Gonzalez, G.; Hwang, H.; et al. Association of Measurable Residual Disease with Survival Outcomes in Patients with Acute Myeloid Leukemia: A Systematic Review and Meta-analysis. JAMA Oncol. 2020, 6, 1890–1899. [Google Scholar] [CrossRef] [PubMed]
  32. Lanza, F.; Maffini, E. Issue Highlights—July 2020. Cytom. B Clin. Cytom. 2020, 98, 295–298. [Google Scholar] [CrossRef] [PubMed]
  33. Uy, G.L.; Aldoss, I.; Foster, M.C.; Sayre, P.H.; Wieduwilt, M.J.; Advani, A.S.; Godwin, J.E.; Arellano, M.L.; Sweet, K.L.; Emadi, A.; et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021, 137, 751–762. [Google Scholar] [CrossRef] [PubMed]
  34. Comoli, P.; Chabannon, C.; Koehl, U.; Lanza, F.; Urbano-Ispizua, A.; Hudecek, M.; Ruggeri, A.; Secondino, S.; Bonini, C.; Pedrazzoli, P. European Society for Blood and Marrow Transplantation, Cellular Therapy & Immunobiology Working Party—Solid Tumor Sub-committee. Development of adaptive immune effector therapies in solid tumors. Ann. Oncol. 2019, 30, 1740–1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Druggable genetic lesions and targeted therapy in AML.
Table 1. Druggable genetic lesions and targeted therapy in AML.
NumberType of Genetic Lesion or Metabolic Pathways Which Can Be Used for Targeted Therapy
1FLT3 mutation: FLT3 inhibitors (midostaurin, gilteritinib, crenolanib, sorafenib, quizartinib, etc.)
2IDH1 and 2 mutation: IDH1/2 mutant inhibitors (ivosidenib, enasidenib, olutasidenib) – BCL2 inhibitors (venetoclax)
3NPM1 mutation: BCL2 inhibitor (venetoclax); All-trans retinoic acid; Arsenic trioxide, Actinomycin D, Menin inhibitors.
4KMT2A rearrangement: Menin inhibitors (SNDX-5613 and KO-539)
5PIM 1-2-3 kinase inhibitors
6Metabolic Pathway Targets: drugs targeting altered metabolism (such as Krebs/citric acid cycle); hedgehog pathway inhibitor (such as glasdegib); mitochondrial-targeted chemotherapeutics; SCAMP-carrier-associated secretory membrane proteins, and interfering drugs
7BCL2 pathway: BCL2 inhibitors (venetoclax and new drug formulations); MCL1 inhibitors
8Cell Surface Antigens: CD33 Antibody drug conjugate (gentuzumab ozogamicin, Vadastuximab talirine); anti-CD47 monoclonal antibody (Magrolimab); anti-CD70 monoclonal antibody (ARGX-110), and bi-specific antibodies such as anti-CD123 (flotetuzumab, etc.)
9Epigenetic Pathway: Hypomethylating agents (5-azacitidine, decitabine, guadecitabine, CC-486), HDAC inhibitors (pracinostat, bromodomain and extraterminal (BET) family proteins; etc) and the targeting of aberrant activation of gene signaling (m-TOR-CDK9; syk inhibitor: entospletinib; Toll-like receptor signalling: CA-4948, small molecule inhibitor of IRAK4)
10Immunotherapy: Wilms Tumor 1-WT1 target therapy (Galinpepimut-S); Checkpoint inhibitors targeting PD1/PDL1 (Nivolumab and pembrolizumab); CTLA4 (ipilimumab), and TIM3 inhibitor (sabatolimab)
11Disruption of adhesion molecules and heparinoids: dociparstat, CX-01: CXCL12/CXCR4 axis), and uproleselan.
12TP53 mutation: MDM2 inhibtors (such as Idasanutlin, miladematan, siremadlin, RG7112, etc.)
13Cellular therapies: CAR-T, CAR-NK, and related CAR molecules
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lanza, F.; Bazarbachi, A. Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions. Cancers 2021, 13, 4698. https://doi.org/10.3390/cancers13184698

AMA Style

Lanza F, Bazarbachi A. Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions. Cancers. 2021; 13(18):4698. https://doi.org/10.3390/cancers13184698

Chicago/Turabian Style

Lanza, Francesco, and Ali Bazarbachi. 2021. "Targeted Therapies and Druggable Genetic Anomalies in Acute Myeloid Leukemia: From Diagnostic Tools to Therapeutic Interventions" Cancers 13, no. 18: 4698. https://doi.org/10.3390/cancers13184698

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