Abstract
Leukemias are hematological neoplasms characterized by dysregulations in several cellular signaling pathways, prominently including the PI3K/AKT/mTOR pathway. Since this pathway is associated with several important cellular mechanisms, such as proliferation, metabolism, survival, and cell death, its hyperactivation significantly contributes to the development of leukemias. In addition, it is a crucial prognostic factor, often correlated with therapeutic resistance. Changes in the PI3K/AKT/mTOR pathway are identified in more than 50% of cases of acute leukemia, especially in myeloid lineages. Furthermore, these changes are highly frequent in cases of chronic lymphocytic leukemia, especially those with a B cell phenotype, due to the correlation between the hyperactivation of B cell receptors and the abnormal activation of PI3Kδ. Thus, the search for new therapies that inhibit the activity of the PI3K/AKT/mTOR pathway has become the objective of several clinical studies that aim to replace conventional oncological treatments that have high rates of toxicities and low specificity with target-specific therapies offering improved patient quality of life. In this review we describe the PI3K/AKT/mTOR signal transduction pathway and its implications in leukemogenesis. Furthermore, we provide an overview of clinical trials that employed PI3K/AKT/mTOR inhibitors either as monotherapy or in combination with other cytotoxic agents for treating patients with various types of leukemias. The varying degrees of treatment efficacy are also reported.
Keywords: Leukemia, PI3K/AKT/mTOR, target therapy, kinase inhibitors, molecular biomarkers, review
Leukemias are the main neoplasms that affect blood and bone marrow cells, inducing several malignant alterations, including abnormalities associated with cell proliferation and differentiation processes. Leukemias are broadly classified as acute or chronic. Additionally, leukemias can also be divided according to the specific cell lineages they impact, falling into four major groups: acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphoblastic leukemia (ALL), and chronic lymphoid leukemia (CLL) (1-3).
Acute leukemias are pathologies that have a rapid evolution and are strongly associated with a quantitative increase in blast cells in the peripheral blood and bone marrow, but they can also present with anemia and an increase in the number of leukocytes. AML is an acute leukemia primarily affecting myeloid progenitor cells and predominantly afflicting the adult population. On the other hand, ALL affects progenitor lineages of lymphoid origin and typically occurs in children and adolescents (4-6).
In the case of chronic leukemias, unlike the acute forms, the disease course has a slower progression and correlates with different stages of cell maturation. Among the types of these leukemias, CML affects patients at different ages, but is most common in adults aged between 40 and 60 years. It is a myeloproliferative dysfunction that affects myeloid cells at different stages of development and has three main clinical phases, namely the chronic phase, accelerated phase, and blast phase. Furthermore, there is also CLL, which mostly affects the elderly population whose blood count shows marked leukocytosis with the massive presence of mature lymphocytes (7-9).
Conventional oncological treatments that use drugs with high toxicity and low specificity have been proving less and less effective in the treatment of neoplastic malignancies. Thus, the therapeutic targeting of key molecules belonging to cellular signaling pathways involved in tumor development represents a promising alternative for the development of treatments aimed at improving the prognosis and quality of life of patients (10,11). Among these important pathways we highlight the PI3K/AKT/mTOR pathway, which is associated with several pivotal cell functions, such as the control of metabolism, proliferation, survival, and cell death, and has thus become the target of several studies aimed at developing inhibitory strategies (12,13).
A clinical study of 55 patients with CLL examined the effectiveness of Duvelisib, a PI3K δ and γ inhibitor, as a therapeutic alternative and reported an overall response rate (ORR) of 56.4% among individuals, of which 1.8% achieved complete response (CR) and 54.5% achieved partial responses (PR). Furthermore, it was also reported that 34.5% of patients achieved stable disease. It was also identified that this drug reduced the levels of some cytokines, chemokines, and growth factors, such as MMP-9, MMP-12, TNFα (Tumor necrosis factor α), interleukin (IL) 6, IL -10, IL-12p40, CCL1, CCL3, CCL4, CCL17, CCL22, CXCL10, and CXCL13 (14). Another important PI3K δ inhibitor is Idelalaisib, which was tested in association with Rituximab (anti-CD20 monoclonal antibody) in a study with 64 patients with relapsed and refractory CLL. As a result, an ORR of 96.9% was observed, of which 14.1% achieved CR and 82.8% achieved PR. Progress was also reported regarding the clinical conditions of patients including improvements in hemoglobin and platelet levels in individuals with anemia and thrombocytopenia, respectively, as well as normalization of the number of neutrophils for individuals with neutropenia (15).
Zeng et al. (16) tested the effects of Ginsenoside Rg3 on bone marrow stromal cells of individuals with acute leukemia and verified its effectiveness on cell growth and angiogenesis. They observed that Ginsenoside Rg3 interfered with the viability of these cells, in addition to inhibiting the expression of vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1α (HIF-1α), AKT and ERK 1 and 2.
For this reason, this review discusses the cellular roles of the PI3K/Akt/mTOR pathway in the development of leukemias and evaluates the currently available inhibitors as therapeutic options for these malignancies.
PI3K/AKT/MTOR Characterization and Cellular Roles
Phosphatidylinositol 3-kinase (PI3K) is a heterodimeric protein, which is activated mainly by tyrosine kinase receptors, but also by G protein-coupled receptors, and is classified into three main classes: Class I, II, and III. Class I are composed of subunits with regulatory functions, such as p85 and p101, and catalytic subunits, such as p110α, p110β, p110δ, and p110γ; they can be further divided into two types: classes IA, which includes PI3Kα, β, and δ, and classes IB, which includes PI3Kγ. Class II PI3K can be divided into three main types that are designated PI3KC2α, PI3KC2β, and PI3KC2γ. Class III has only one known isoform, PI3KC3. Activated PI3K catalyzes the conversion of phosphatidylinositol 4,5-bisphosphate (PIP2) molecules into phosphatidylinositol 3,4,5-triphosphate (PIP3), which will act as second messenger for several signaling molecules, such as 3-phosphoinositide-dependent kinase-1 (PDK-1) and AKT (17-19).
The AKT protein is a serine/threonine kinase, also known as protein kinase B (PKB), which can be regulated by hormonal and/or protein kinases (for example, PDK-1) and can be found in three isoforms: AKT1 (PKBα), AKT2 (PKBβ), and AKT3 (PKBγ). In general, AKT activation begins with PIP3 recruitment and binding tightly to PDK1, which have pleckstrin homology (PH) domains. Such an event acts as an anchor for AKT molecules and facilitates their phosphorylation. Furthermore, depending on the type of AKT, it will present different target residues for phosphorylation by PDK1, the main ones being Thr308, Thr309, and Thr305, which belong to the isoforms AKT1, AKT2, and AKT3, respectively (20-22).
In its active form, AKT acts as a mediator of the phosphorylation of several proteins that participate in cell signaling, such as the mammalian target of rapamycin (mTOR) complexes, which consist of proteins belonging to the serine/threonine kinase family and act in the promotion of genomic stability, cell cycle regulation and glucose metabolism. Through interaction with a wide range of proteins, mTOR forms two distinct cellular complexes: mTORC1 and mTORC2. Among the components of mTORC1 are the mTOR protein itself, regulatory-associated protein of mTOR (RAPTOR), DEP domain-containing mTOR-interacting protein (DEPTOR), mammalian lethal with Sec13 protein 8 (mLST8), and proline-rich Akt substrate 40kDa (PRAS40). In contrast, the mTORC2 has among its components mTOR, Rapamycin-insensitive companion of mTOR (RICTOR), protein observed with Rictor-1 (PROTOR), DEPTOR, mLST8, and mammalian stress activated protein kinase interacting protein 1 (mSin1) (23-25).
When mTORC1 is activated, it can act on several important metabolic processes for cell homeostasis, such as protein, lipid, glucose, and nucleic acid synthesis, in addition to participating in the stimulation of cell growth and inhibition of autophagic mechanisms. mTORC1 may also down-regulate mTORC2 activity through its downstream effector protein p70-S6 kinase 1 (S6K1), which also participates in the production of nucleic acids. mTORC2 has various essential functions, notably in lipid synthesis, regulating the actin cytoskeleton, and influencing glycolysis. Furthermore, it is noteworthy that mTORC2 activation can reactivate AKT molecules through phosphorylation on serine residues, such as Ser473, acting as a positive feedback for AKT, potentially leading to the reactivation of mTORC (23,26,27).
One way through which AKT activates mTOR1 complexes is through phosphorylation of the tuberous sclerosis complex 2 (TSC2) and prevention of its ability to inhibit GTPase Rheb. Furthermore, AKT can also activate the mTOR complex through activation of the Conserved helix-loop-helix ubiquitous kinase (CHUK), expressed by the CHUK gene and phosphorylation of PRAS40 (23,24,28).
AKT can also control the cell cycle by blocking and phosphorylating proteins that promote apoptosis, including proteins belonging to the B-cell lymphoma 2 (BCL-2) family, caspases 9, BCL-2-associated protein X (BAX) and BCL-2-associated cell death agonist (BAD). AKT can influence transcription factors that act in various metabolic processes and cell survival, such as nuclear factor kappa B (NF-kB), Forkhead box transcription factor (FOXO) and MDM2. Furthermore, it also has the ability to modulate the activity of glycogen synthase kinase 3 (GSK3), which is a protein that participates in the regulation of glycogen synthesis (Figure 1) (29-33).
When the physiological need for catalytic active PI3K/AKT/mTOR pathway, the organism needs a mechanism that works as negative feedback and for that it makes use of inhibitor proteins, which act mainly as catalysts for the conversion of PIP3 into PIP2. The main one is PTEN, a lipid phosphatase homologous to tensin that interferes both with metabolism and cell growth; however, it can also use other inhibitors such as the SHIP1 protein (34-37).
PI3K/AKT/mTOR and Leukemogenesis
The PI3K/AKT/mTOR pathway has a direct relationship with the hematopoiesis control mechanisms and for this reason, changes that promote overactivation of this pathway can induce aberrant hematopoiesis, leading to leukemogenesis onset (13,29,38).
When we refer to the involvement of PI3K in neoplastic development, the main class that is involved in this process is class I PI3K. The p110α and p110β subunits of class I PI3K are ubiquitously expressed in several cell types in the human body, however, the p110δ and p110γ subunits are expressed more specifically in hematopoietic cells, mainly in leukocytes. For this reason, PI3Kδ and γ are the most related to leukemogenesis and have become targets of many therapeutical approaches (39,40).
PI3Kδ is a prevalent PI3K isoform in leukocytes, mainly B lymphocytes, and many studies have examined the role of PI3Kδ in cases of malignancies with B-cell phenotype, especially in CLL cases. Extensive literature reports indicate that overactivation of B cell receptors (BCR) promotes the downstream activation of PI3Kδ, resulting in abnormal triggering of the PI3K/AKT/mTOR pathway and tumor development (41-44).
BCR is an immunoglobulin located in the cell membrane region of lymphocytes with a B cell phenotype that interacts through non-covalent bonds with the CD79a and CD79b heterodimers. The binding of specific BCR antigens to this receptor causes the phosphorylation of immunotyrosine-based activation motif (ITAMs) in the cytoplasmic domains of CD79a and CD79b by LYN protein kinase, which is a member of the Src family. This results in the recruitment, anchoring and activation of tyrosine kinase of the spleen (SYK) and promotion of the subsequent phosphorylation of PI3K BCAP and CD19 co-receptor, which are capable of activating PI3K, especially of the delta isoform. In addition, BCR is also responsible for activating other important pathways for cell metabolism and survival that are relevant to leukemogenesis development (41,42,45-47).
Alterations in the PI3K/AKT/mTOR pathway can also be identified in patients with AML, being observed in over 50% of these cases and may be associated with a worse prognosis. The p110δ catalytic subunit has been shown as relevant in this leukemic process, since its expression is high in AML blast cells and constant phosphorylation of AKT, which happens downstream, promotes enhanced cell proliferation, survival of neoplastic clones, aberrant DNA repair mechanisms and even dysfunctional glucose metabolism (28,48-51).
Despite over-activating mTORC, AKT also regulates other molecular targets to favor leukemogenesis, such as FOXO, which are transcription factors that act as tumor suppressors, participating in cell cycle, proliferation, and differentiation of the cells and, mainly, of in apoptosis, in which FOXO3 is the most relevant. Therefore, the modulation of FOXO3 by AKT favors the survival of malignant cells and inhibits cell death by apoptosis (52-55).
Disturbances in mTORC can also favor leukemia development affecting hematopoietic stem cells (HSC) and leukemic stem cells. Activation of S6K1 upon mTORC1 activation is a key mechanism that helps maintain HSC quiescence and influences mRNA translation. Thus, factors that lead to mTORC1 and S6K1 deficiency will not only reduce the quiescent state and the capacity for cell self-renewal, but they can also stimulate proliferation of these cells (56,57).
The cell culture experiment performed by Gosh et al. (58) demonstrated that S6K1 deficiency led to a reduction in the quiescence and self-renewal capacity of HSC that was correlated with a reduction in cyclin-dependent kinase 1A inhibitor (Cdkn1a) levels. In addition, there was also a greater propensity for myelotoxic stresses, a reduction in mTOR levels and maintenance and propagation of leukemias. Panuzzo et al. (59) revealed that, under hypoxic conditions, there is a reduction in mTORC1 activity in CML stem cells, while mTORC2 activity is maintained. They also reported an increase in phosphorylation of AKT molecules due to its positive feedback cycle, promoted by mTORC2, and an increase in the phosphorylation of PKCα, which is also a substrate of this complex. They also revealed that, despite the increase in AKT phosphorylation via mTORC2, the activation of some downstream targets of AKT are dependent on the phosphorylation of specific residues. For the activation of FOXO3a, there is a need for phosphorylation in the AKT molecule of both the Ser473 residue (mTORC2 target) and Thr308 (PDK-1 target). In contrast, GSK3β requires phosphorylation of only the Thr308 residue of AKT. With that, they reported a reduction in the levels of FOXO3a and GSK3β, which resulted in reduced self-renewal and cellular quiescence, cell cycle progression, decreased glycogen production and greater accumulation of cyclin D1.
In general, mTORC can contribute to leukemogenesis, mainly AML, in different ways depending on which of the complexes is acting. mTORC1 affects protein synthesis mechanisms through the modulation of effector molecules S6K1 and eIF4E binding protein (E4BP), regulates lipid synthesis where it can control the expression of genes that participate in the production of cholesterol and fatty acids, participates in metabolism of glucose by stimulating glycolysis and HIF1α synthesis, and blocks autophagy mainly by preventing the activation of Unc-51-like kinase 1 (ULK1), which is an important component for autophagosome formation. mTORC2, in addition to activating AKT, plays an important role in controlling cell survival and proliferation and participates in the organization of the actin cytoskeleton through the regulation of proteins that are members of the PKC family, especially PKCα (25,49,56,60,61).
Literature reports indicate that the RAPTOR component of mTORC1 is a key element in the involvement of this complex in the development of acute leukemias, as it helps to increase cellularity, reduce apoptosis, and stimulate cell proliferation, which may even lead to disease progression. Furthermore, mTORC1 overactivation can also be caused by increased activity of the TSC2/Rheb/mTORC1 axis. In cases of T-cell ALL, indirect activation of mTOR, mainly mTORC1, can also occur under the influence of Notch1 through its interaction with c-MYC. It is also important to note that Notch1 can directly activate the PI3K/AKT/mTOR pathway through its action on growth factor receptors and cytokine receptors (62-65).
Leukemogenesis related to the PI3K/AKT/mTOR pathway can also be triggered by alterations in other cytoplasmic elements and/or cell receptors, which interact with but are not related to the pathway itself, leading to a loss of balance in the bone marrow microenvironment, as is the case of FMS-like tyrosine kinase 3 (FLT3), insulin-like growth factor 1 (IGF-1), interleukin 6 (IL-6) and stroma derived factor 1 (SDF-1, also called CXCL12), which can promote an excessive stimulation of several different signal transduction pathways, including PI3K/AKT/mTOR (39,66,67).
PI3K/AKT/mTOR Pathway Inhibition in Leukemias
Due to the importance of the PI3K/AKT/mTOR pathway in the control and regulation of several important cellular functions, the molecules that comprise this signaling cascade have become relevant pharmaceutical targets since their blockade can be a potential therapeutic strategy in the treatment of leukemias (12,44).
Table I presents a set of clinical trials from the last ten years that address this theme and aim to find efficient pathway inhibitors for leukemias either as a monotherapy or in combination with other therapies. A total of 12 clinical trials are presented, and six of them examined the inhibition of AKT and mTOR components as treatment for acute leukemias whereas the other six examined the inhibition of PI3K as treatment for chronic leukemias, specifically CLL (68-79).
All studies described in Table I addressed the efficacy of PI3K/AKT/mTOR pathway inhibitors or their components, showing that changes in the pathway are very important for leukemia research, since they influence cell proliferation, metabolism, angiogenesis, and apoptosis, correlating with the clinical and hematological aspects of these diseases (68-79).
The results were more promising for CLL than acute leukemias, where even activation of secondary pathways that promote compensatory effects were reported (68-79).
Of the 12 clinical trials, two studies used Idelalisib as an inhibitor (monotherapy and in combination with Tirabrutinib) and two used Buparlisib showing beneficial results. Tensirolimus was also addressed in two studies, one of them in association with intensive re-induction chemotherapy, leading to high toxicity, and the other in combination with Cyclophosphamide and Etoposide, with generally beneficial results (69,70,73-75,79). Other inhibitors tested in the studies and which presented positive or partially positive results were: BEZ235, Umbralisib in combination with Ibrutinib and Duvelisib associated with FCR (Fludarabine, Cyclophosphamide and Rituximab) (72,77,78). Three studies did not show good results; two of them used Perifosine as an inhibitor (monotherapy and in combination with UCN-01) and the other used Trametinib plus GSK2141795 (68,71,76).
Idelalisib: Approved PI3K Inhibitor
Despite reports that the PI3K/AKT/mTOR pathway is activated in acute leukemias, many current studies focus on the development of inhibitors of this pathway in CLL cases, especially those with a B-cell phenotype, in large part owing to the correlation between BCR-mediated signal transduction and PI3Kδ activation (41,80-82). Dong et al. (83), found that mice with genetically inactivated p110δ showed a significant delay in the development of the disease indicating the relevance of PI3Kδ in the development and progression of CLL, since.
Idelalisib, also known as CAL-101 or GS-1101, is a selective inhibitor of the PI3Kδ isoform. It was approved by the Food and Drug Administration (FDA) in June 2014 for the treatment of patients with CLL, follicular non-Hodgkin lymphoma (LF) of relapsed B-cell and relapsed small lymphocytic lymphoma (SLL) (45,84,85).
Once administered, Idelalisib is metabolized primarily by aldehyde oxidase, and to a smaller scale by cytochrome P450 3A (CYP3A). Like many PI3K inhibitors, Idelalisib is a competitive drug for adenosine triphosphate (ATP) binding sites, in order to prevent PI3K-mediated phosphorylation of PIP2. Such binding sites are located in the protein’s catalytic domains, which are formed by the N-terminal and C-terminal domains. Between these domains there is a cleft where the hinge region is located, which contains valine residues (a conserved region among class I PI3K isoforms) and is the site where ATP interacts with the protein. Hydrogen bonds between the Val-828 and Val-826 residues in the PI3K hinge region and the N3 and N9 purine molecules of Idelalisib, respectively, are formed. Hydrogen bonds, mediated by water molecules, are also formed between purines N7, N1 and the side chain of Asp-911. In addition, this drug is also capable of making hydrophobic bonds with other protein residues, such as RS4, CS6, CS7, Val-827, Met-900, among others (Figure 2) (45,84,86-88).
Two of the studies presented in Table I used of Idelalisib as an inhibitor of the PI3K/AKT/mTOR pathway and showed satisfactory results regarding the effectiveness of pathway inhibition. Brown et al. (74) used it as monotherapy, while Danilov et al. (79) compared the combination of Idelalisib with Tirabrutinib and other combinations. Both studies showed significant clinical responses indicating that Idelalisib is a promising inhibitor of the PI3K/AKT/mTOR pathway in cases of CLL. Furthermore, its combination with Tirabrutinib seems to be more effective compared to the use of Idelalisib alone with an overall response rate (ORR) of approximately 93% and 72%, respectively. Staber et al. (89) examined Tafasitamab combined with Idelalisib or Venetoclax in individuals with relapsed/refractory CLL. Patients who received the combination of Tafasitamab with Idelalisib had an ORR of 90,9 and minimal residual disease (MRD) of 72,7% and two participants were negative for DRM in peripheral blood samples. However, Lampson et al. (90) reported that the combination of Idelalisib with Ofatumumab in patients with CLL generated high toxicity rates, making it impossible to complete the study.
Brown et al. (74) reported that treatment with Idelalisib reduced AKT phosphorylation, decreased plasma concentrations of chemokines, especially CCL3, CCL4, CCL17 and CCL22 and stromal-derived factors, such as CD40L (CD40 ligands), tumor necrosis factor α (TNFα), CCL2 and CXCL13. Hoellenriegel et al. (91), obtained similar results when examining the effects of Idelalisib in cultures of CLL cells and reported that there was a reduction in the phosphorylation of AKT and extracellular signal-regulated kinases (ERK), a reduction in the levels of CCL2, CCL3, CCL4, CCL7, CCL22, CD40L, CXCL13, TNF-α and interleukin-6 (IL-6) and a suspension of chemotaxis of CLL cells towards CXCL12 and CXCL13. Serrat et al. (92) also tested Idelalisib in cocultures of follicular lymphoma and follicular dendritic cells and observed that the inhibitor was capable of interfering with the CD40/CD40L pathway, modulating CCL22 levels and negatively regulating the pro-angiogenic factors VEGF-A and VEGF-C.
In the two studies with Idelalisib (monotherapy and in combination with Tirabrutinib) some adverse events (AEs) were described, with the most common being diarrhea, skin eruptions, pyrexia, and some cases of cytopenia. In addition, in more severe cases, the occurrence of pneumonia, febrile neutropenia and some bacterial infections have been reported (74,79). Other studies in the literature report similar AEs in their studies, highlighting the occurrence of hematological alterations (neutropenia, anemia, and thrombocytopenia) and biochemical alterations, such as increased transaminases, glycemic alterations (generally elevated), increased gamma glutamyl transferase and alkaline phosphatase (93-96).
Clinical Aspects of Other PI3K/AKT/mTOR Inhibitors Pathway
Although Idelalisib is an important PI3K inhibitor currently used as a therapeutic measure against hematological malignancies, other inhibitors of this protein have also been approved for combating hematological neoplasms, preventing signal transduction of a wide variety of downstream signaling pathways, including those linked to the BCR. Among these inhibitors we can highlight Duvelisib and Copanlisib (97-99).
Duvelisib, also known as IPI-145 and INK-1197, is a dual PI3K inhibitor which acts specifically on PI3Kδ and γ isoforms, having received FDA approval in 2018 and European Medicines Agency (EMA) approval in 2021 to be used as therapy in cases of relapsed and refractory CLL and small lymphocytic lymphomas (SLL) that have already undergone at least two previous treatments. In addition, there are reports that such medication can stimulate apoptosis, reduce the migration of neoplastic cells, reduce chemotaxis, decrease the levels of circulating cytokines and chemokines, and modulate several cellular signaling pathways (97,98,100-102). Flinn et al. (103) carried out a phase 1 study with patients with hematological malignancies, including CLL (relapsed/refractory and absent from previous treatment), indolent non-Hodgkin lymphoma (iNHL), and cutaneous and peripheral T-cell lymphoma (TCL). As a result of the therapy effectiveness, patients with relapsed/refractory CLL and those who were not previously submitted to treatments obtained an ORR of 56% and 83%, respectively, and, patients with iNHL cutaneous and peripheral TCL showed ORR 58%, 32%, and 50%, respectively.
Copanlisib is another PI3K inhibitor which was FDA approved in 2017 under the trade name of Aliqopa and acquired EMA approval in 2020. It is a class I pan PI3K inhibitor acting mainly on the PI3Kα and δ isoforms. Its application aims at the treatment of patients with relapsing follicular lymphomas (FL) who have already been submitted to at least two previous systemic therapeutic schemes. In addition, application of this medication for other B-cell malignancies, such as CLL has also been examined (99,104-106). Dreyling et al. (107) tested Copanlisib in patients with CLL and aggressive malignant lymphoma and obtained an ORR of 43.8% and 29.4%, respectively. In addition, they also found a reduction of at least 50% in lesion size in 66.7% of patients with indolent neoplasms and 42.5% in patients of the aggressive cohort.
mTOR inhibitors, which are classified into first and second generation inhibitors, have also received lot of attention. First-generation inhibitors are basically represented by Rapamycin and Rapamycin analogs (Rapalogs), whereas the second generation are considered as double inhibitors and are divided into mTORC1/mTORC2 inhibitors and PI3K/mTOR inhibitors (25,108,109).
First generation inhibitors show a good inhibitory activity of mTORC1. Inhibitors such as Rapamycin promote their effects by binding to intracellular receptors called FK506 binding protein 12 (FKBP12). This binding results in the formation of a complex formed by both elements that act on the FRB domains of mTOR. As a consequence, the interaction between Raptor and mTOR molecules is interrupted, preventing the action of downstream effector components mainly on cell proliferation and angiogenesis. Despite this, Rapamycin does not show such satisfactory effects, and there is even the possibility of AKT reactivation by positive feedback. The second generation inhibitors are considered dual inhibitors. The mTORC1 and 2 inhibitors are classes of inhibitors that act competitively on mTOR/ATP, promoting a complete blockade of 4E-BP1. In this way, they can prevent cell replication processes, but that can also promote cell death by apoptosis. In contrast, dual PI3K/mTOR inhibitors act as competitive compounds for ATP binding sites on these proteins due to the similarity between the catalytic sites of both, so that the pathway is blocked (25,108-111).
In general, PI3K/AKT/mTOR pathway inhibitors can affect the tumor microenvironment in several ways. One of them refers to their direct action on malignant cells present in the tumor stroma, by modulating cell signaling, preventing them from proliferating and stimulating cell death by apoptosis. In addition, they interfere and regulate cellular responses to various stimuli that are generated by inflammatory mediators that could favor the survival and maintenance of these malignant cells, such as molecules acting as costimulators, molecules involved with cell adhesion (for example, CXCR4 and CXCR5), cytokines and chemokines, such as interleukins (for example, IL-2, IL-4, IL-6, IL-10, and IL-17), interferon γ (IFNγ), TNFα, CD40L, CCL3, CCL4, CXCL10, and CXCL13. These inhibitors can also modulate the immune responses generated by TCD8+ and TCD4+ lymphocytes and reduce the levels of regulatory T cells (Treg cells), affecting antitumor immunity (45,112-114).
Maharaj et al. (115), found that inhibition of PI3Kδ and γ in cultured CLL cells using Idelalisib, Duvelisib and Umbralisib resulted in a reduction in the levels of AKT and cytokines. They also observed that Idelalisib and Duvelisib suppressed FOXP3 expression, significantly reduced the number of Treg cells and decreased the expression of immunosuppressive molecules PD-1 and CTLA-4 on the surface of Treg cells. Furthermore, they also associated the deleterious effects of Treg cells with certain immune-mediated toxicities, since the lower the percentage of Treg cells, the greater the possibility of toxicities occurring. Based on this, they noted that Umbralisib has a tendency to quantitatively and functionally preserve Treg cells, since it is a selective inhibitor of PI3Kδ and is also capable of inhibiting casein kinase-1-ε (CK1ε). Chellappa et al. (116), found that PI3Kδ inhibition in CLL cells treated with Idelalisib, also resulted in a reduction in AKT levels and the number of Treg cells, a decrease in the expression of FOXP3, CD-25, and PD-1 and a trend towards a decrease in CTLA-4 and CD-39 levels.
Two of the studies presented in Table I addressed the use of Buparlisib as an PI3K/AKT/mTOR pathway inhibitor that is a pan-PI3K inhibitor (69,75). In one of these studies, Assouline et al. (75) tested the inhibitor in CLL cases, specifically B-cell CLL and verified the effectiveness of Buparlisib in blocking the pathway and the relevance of the biomarkers RAPTOR and P70S6K expression with the responses to the treatment. It was observed that the quantitative reduction of leukemic cells positive for RAPTOR is related to a higher rate of tumor shrinkage. This occurs because in cases of signal transduction via BCR with the activation of PI3K-δ dependent on RAPTOR signaling, there is greater resistance to PI3K-δ inhibition. However, with the reduction of RAPTOR expression by tumor cells, there is an improvement in responsiveness to treatment. Amrein et al. (117) evaluated the effectiveness of Buparlisib in CLL B cells in in vitro studies and inhibitory growth effects, cell death of these leukemic lineages, a decrease in the basal RAPTOR and P70S6K expression and a reduction of AKT phosphorylation.
Ragon et al. (69) also evaluated the antileukemic potential of Buparlisib in acute leukemias and observed moderate efficacy, a decrease in the levels of p-S6K (target of mTORC) and p-FOXO3 (target of AKT), as well as PRAS40 down-regulation. Mehrpouri et al. (118) performed tests on cultures of ALL-derived cells treated with Buparlisib in combination with Panobinostat, a histone deacetylase (HDAC) inhibitor, and obtained positive results Specifically, they found induction of apoptosis, decreased cell proliferation and reduction in viability and metabolic activity of cells. Furthermore, they evidenced the capacity of the treatment to modulate FOXO3 and FOXO4 expression levels, increase cMYC suppression and increase p21 and p27 expression.
Although alterations in the PI3K/AKT/mTOR pathway are detected in many cases of acute leukemia, mainly AML, the development of efficient inhibitors of this pathway is a very complex, as there are reports predicting the overactivation of compensatory signaling pathways. For this reason, some studies addressed the PI3K/AKT/mTOR pathway inhibition while also targeting other secondary mechanisms that promote leukemogenesis (49,119,120). An example of the difficulty in identifying good inhibitors of this pathway for acute leukemias was demonstrated by Lang et al. (72), who tested the BEZ235 inhibitor in patients with acute leukemias. They found that only a small group of patients with ALL obtained good results, while patients with AML did not obtain significant benefits, including a progression of the disease in 91%.
Two other studies on acute leukemias, especially ALL, used Tensirolimus as an inhibitor. Rheingold et al. (70) used Tensirolimus in combination with intensive reinduction chemotherapy in three weekly doses. They observed excessive toxicity, non-tolerability in children with relapsing ALL and DLT (dose-limited toxicity) was present at all dose levels. Despite this, 47% of the evaluated participants had complete response (CR) or complete response with incomplete blood count recovery (CRi) and of these, five had minimal residual disease (MRD) less than 0.03% at the end of therapy. Tasian et al. (73) examined the effect of the inhibitor in combination with Cyclophosphamide and Etoposide and obtained a similar ORR of 47%, and 3 participants showed a MRD of less than 0.01%. In addition, they claimed less toxicity and greater therapeutic safety, since only two doses of treatment were administered per week.
Among the unsuccessful studies, two used Perifosine, both as monotherapy in CLL and in combination with UCN-01 in acute leukemias. Treatment of CLL with Perifosine generated lymphocytic cytotoxicity in vitro and absence of relevant clinical responses in vivo. In acute leukemias, DLT and lack of effectiveness in the maximum tolerated dose (MTD) were observed (68,76). Despite this, Perifosine alone and in combination with Sorafenib was examined in a study with lymphoproliferative diseases, including CLL; half of the patients with CLL treated with monotherapy and in 1/4 of the patients treated with the combination responded to medication (121).
Among the evaluated studies, there were some similarities regarding the occurrence of some of the AEs. The most common AEs were gastrointestinal problems (nausea, vomiting, diarrhea), biochemical alterations (mainly, hyperglycemia), hematological disturbances (neutropenia, anemia, thrombocytopenia), fatigue, mucositis, and an increased propensity to develop infections (68-79). Other studies reported, such as those of O’Brien et al. (14), Rigolin et al. (122) and Mato et al. (123) reported the occurrence of neutropenia, anemia, infections, fatigue, gastrointestinal problems, among other symptoms in patients with leukemia.
Conclusion
Our work emphasizes the major role of PI3K/AKT/mTOR pathway on leukemogenesis and leukemia development, highlighting its potential as a therapeutic target in hematological malignancies. Critical review of clinical studies conducted in the past ten years reveal a tendency for better responses to pathway inhibitors among CLL afflicted patients, supported by the FDA approval of treatment regimens including drugs, such as Idelalisib and Duvelisib. Further stratification for treatment efficacy may even be made among the CLL patient cohort, as B-cell CLL is better suited to be treated with PI3K inhibitors due to its upstream overactivation by the B-cell receptor (BCR). Furthermore, we observe increased difficulty in developing safe and effective treatment strategies using PI3K inhibitors to treat acute leukemias, primarily because of treatment-induced activation of compensatory pathways for neoplastic maintenance and consequent drug resistance. Overall, while the role of PI3K/AKT/mTOR pathway is well defined both in healthy and neoplastic scenarios, further studies are still need to properly translate its inhibition into potential therapies, aiming to prolong treatment efficacy and minimize cases of drug resistance, especially when considering its use for acute leukemias.
Conflicts of Interest
The Authors declare no conflict of interest in relation to this study. The funders had no role in the design of the study; in the collection, analyses, or data interpretation; in the writing of the manuscript, or in the decision to publish the results.
Authors’ Contributions
C.A.M.-N; Conceptualization, A.K.C.M, C.B.M., F.M.C.d.P.P., R.B.G, I.V.B and C.A.M.-N.; Provision of data and subsequent analysis and interpretation, A.K.C.M, C.B.M., F.M.C.d.P.P., R.B.G, I.V.B., C.A.M.-N., R.M.R., D.d.S.O., M.O.d.M.F., M.E.A.d.M., A.S.K; Writing – original draft preparation, A.K.C.M, C.B.M., F.M.C.d.P.P., R.B.G, I.V.B and C.A.M.-N.; Writing – review and editing, A.K.C.M, C.B.M., F.M.C.d.P.P and C.A.M.-N. Funding acquisition, C.A.M.-N. and A.S.K. All Authors have read and agreed to published version of the manuscript.
Funding
This study was supported by Brazilian funding agencies: Coordination for the Improvement of Higher Education Personnel (CAPES; to A.K.C.M, C.B.M and I.V.B), National Council of Technological and Scientific Development (CNPq grant number 404213/2021-9 to CAM-N; and Productivity in Research PQ scholarships to M.O.d.M.F, M.E.A.d.M, A.S.K. and CAM-N), Cearense Foundation of Scientific and Technological Support (FUNCAP grant number P20-0171-00078.01.00/20 to F.M.C.d.P.P, M.O.d.M.F and to CAM-N) and, we also thank PROPESP/UFPA for publication payment.
References
1
Nemkov T
,
D’Alessandro A
&
Reisz JA
. Metabolic underpinnings of leukemia pathology and treatment. Cancer Rep (Hoboken).
2(2)
e1139
2019.
DOI:
10.1002/cnr2.1139
2
Khoury JD
,
Solary E
,
Abla O
,
Akkari Y
,
Alaggio R
,
Apperley JF
,
Bejar R
,
Berti E
,
Busque L
,
Chan JKC
,
Chen W
,
Chen X
,
Chng WJ
,
Choi JK
,
Colmenero I
,
Coupland SE
,
Cross NCP
,
De Jong D
,
Elghetany MT
,
Takahashi E
,
Emile JF
,
Ferry J
,
Fogelstrand L
,
Fontenay M
,
Germing U
,
Gujral S
,
Haferlach T
,
Harrison C
,
Hodge JC
,
Hu S
,
Jansen JH
,
Kanagal-Shamanna R
,
Kantarjian HM
,
Kratz CP
,
Li XQ
,
Lim MS
,
Loeb K
,
Loghavi S
,
Marcogliese A
,
Meshinchi S
,
Michaels P
,
Naresh KN
,
Natkunam Y
,
Nejati R
,
Ott G
,
Padron E
,
Patel KP
,
Patkar N
,
Picarsic J
,
Platzbecker U
,
Roberts I
,
Schuh A
,
Sewell W
,
Siebert R
,
Tembhare P
,
Tyner J
,
Verstovsek S
,
Wang W
,
Wood B
,
Xiao W
,
Yeung C
&
Hochhaus A
. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and histiocytic/dendriticneoplasms. Leukemia.
36(7)
1703
- 1719
2022.
DOI:
10.1038/s41375-022-01613-1
3
Alaggio R
,
Amador C
,
Anagnostopoulos I
,
Attygalle AD
,
Araujo IBO
,
Berti E
,
Bhagat G
,
Borges AM
,
Boyer D
,
Calaminici M
,
Chadburn A
,
Chan JKC
,
Cheuk W
,
Chng WJ
,
Choi JK
,
Chuang SS
,
Coupland SE
,
Czader M
,
Dave SS
,
de Jong D
,
Du MQ
,
Elenitoba-Johnson KS
,
Ferry J
,
Geyer J
,
Gratzinger D
,
Guitart J
,
Gujral S
,
Harris M
,
Harrison CJ
,
Hartmann S
,
Hochhaus A
,
Jansen PM
,
Karube K
,
Kempf W
,
Khoury J
,
Kimura H
,
Klapper W
,
Kovach AE
,
Kumar S
,
Lazar AJ
,
Lazzi S
,
Leoncini L
,
Leung N
,
Leventaki V
,
Li XQ
,
Lim MS
,
Liu WP
,
Louissaint A Jr
,
Marcogliese A
,
Medeiros LJ
,
Michal M
,
Miranda RN
,
Mitteldorf C
,
Montes-Moreno S
,
Morice W
,
Nardi V
,
Naresh KN
,
Natkunam Y
,
Ng SB
,
Oschlies I
,
Ott G
,
Parrens M
,
Pulitzer M
,
Rajkumar SV
,
Rawstron AC
,
Rech K
,
Rosenwald A
,
Said J
,
Sarkozy C
,
Sayed S
,
Saygin C
,
Schuh A
,
Sewell W
,
Siebert R
,
Sohani AR
,
Tooze R
,
Traverse-Glehen A
,
Vega F
,
Vergier B
,
Wechalekar AD
,
Wood B
,
Xerri L
&
Xiao W
. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid neoplasms. Leukemia.
36(7)
1720
- 1748
2022.
DOI:
10.1038/s41375-022-01620-2
4
Rose-Inman H
&
Kuehl D
. Acute leukemia. Hematol Oncol Clin North Am.
31(6)
1011
- 1028
2017.
DOI:
10.1016/j.hoc.2017.08.006
5
Narayanan D
&
Weinberg OK
. How I investigate acute myeloid leukemia. Int J Lab Hematol.
42(1)
3
- 15
2020.
DOI:
10.1111/ijlh.13135
6
Fujita TC
,
Sousa-Pereira N
,
Amarante MK
&
Watanabe MAE
. Acute lymphoid leukemia etiopathogenesis. Mol Biol Rep.
48(1)
817
- 822
2021.
DOI:
10.1007/s11033-020-06073-3
7
Minciacchi VR
,
Kumar R
&
Krause DS
. Chronic myeloid leukemia: a model disease of the past, present and future. Cells.
10(1)
117
2021.
DOI:
10.3390/cells10010117
8
Hallek M
. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol.
94(11)
1266
- 1287
2019.
DOI:
10.1002/ajh.25595
9
Juliusson G
&
Hough R
. Leukemia. Res Tumor Prog.
43
87
- 100
2016.
DOI:
10.1159/000447076
10
Machado CB
,
Da Silva EL
,
De Moraes Filho MO
,
De Moraes MEA
&
Moreira-Nunes CA
. PARP inhibitors as therapeutic options for tyrosine kinase-dependent leukemia: a review. Anticancer Res.
40(6)
3055
- 3063
2020.
DOI:
10.21873/anticanres.14286
11
Poliaková M
,
Aebersold DM
,
Zimmer Y
&
Medová M
. The relevance of tyrosine kinase inhibitors for global metabolic pathways in cancer. Mol Cancer.
17(1)
27
2018.
DOI:
10.1186/s12943-018-0798-9
12
Bertacchini J
,
Heidari N
,
Mediani L
,
Capitani S
,
Shahjahani M
,
Ahmadzadeh A
&
Saki N
. Targeting PI3K/AKT/mTOR network for treatment of leukemia. Cell Mol Life Sci.
72(12)
2337
- 2347
2015.
DOI:
10.1007/s00018-015-1867-5
13
Yang J
,
Nie J
,
Ma X
,
Wei Y
,
Peng Y
&
Wei X
. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer.
18(1)
26
2019.
DOI:
10.1186/s12943-019-0954-x
14
O’Brien S
,
Patel M
,
Kahl BS
,
Horwitz SM
,
Foss FM
,
Porcu P
,
Jones J
,
Burger J
,
Jain N
,
Allen K
,
Faia K
,
Douglas M
,
Stern HM
,
Sweeney J
,
Kelly P
,
Kelly V
&
Flinn I
. Duvelisib, an oral dual PI3K-δ,γ inhibitor, shows clinical and pharmacodynamic activity in chronic lymphocytic leukemia and small lymphocytic lymphoma in a phase 1 study. Am J Hematol.
93(11)
1318
- 1326
2018.
DOI:
10.1002/ajh.25243
15
O’Brien SM
,
Lamanna N
,
Kipps TJ
,
Flinn I
,
Zelenetz AD
,
Burger JA
,
Keating M
,
Mitra S
,
Holes L
,
Yu AS
,
Johnson DM
,
Miller LL
,
Kim Y
,
Dansey RD
,
Dubowy RL
&
Coutre SE
. A phase 2 study of idelalisib plus rituximab in treatment-naïve older patients with chronic lymphocytic leukemia. Blood.
126(25)
2686
- 2694
2015.
DOI:
10.1182/blood-2015-03-630947
16
Zeng D
,
Wang J
,
Kong P
,
Chang C
,
Li J
&
Li J
. Original Article Ginsenoside Rg3 inhibits HIF-1α and VEGF expression in patient with acute leukemia via inhibiting the activation of PI3K/Akt and ERK1/2 pathways. Int J Clin Exp Pathol.
7
2172
- 2178
2014.
17
Noorolyai S
,
Shajari N
,
Baghbani E
,
Sadreddini S
&
Baradaran B
. The relation between PI3K/AKT signalling pathway and cancer. Gene.
698
120
- 128
2019.
DOI:
10.1016/j.gene.2019.02.076
18
Liu P
,
Cheng H
,
Roberts TM
&
Zhao JJ
. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov.
8(8)
627
- 644
2009.
DOI:
10.1038/nrd2926
19
Bilanges B
,
Posor Y
&
Vanhaesebroeck B
. PI3K isoforms in cell signalling andvesicle trafficking. Nat Rev Mol Cell Biol.
20(9)
515
- 534
2019.
DOI:
10.1038/s41580-019-0129-z
20
Jiang Q
,
Zhang X
,
Dai X
,
Han S
,
Wu X
,
Wang L
,
Wei W
,
Zhang N
,
Xie W
&
Guo J
. S6K1-mediated phosphorylation of PDK1 impairs AKT kinase activity and oncogenic functions. Nat Commun.
13(1)
1548
2022.
DOI:
10.1038/s41467-022-28910-8
23
Hua H
,
Kong Q
,
Zhang H
,
Wang J
,
Luo T
&
Jiang Y
. Targeting mTOR for cancer therapy. J Hematol Oncol.
12(1)
71
2019.
DOI:
10.1186/s13045-019-0754-1
24
Jhanwar-Uniyal M
,
Wainwright JV
,
Mohan AL
,
Tobias ME
,
Murali R
,
Gandhi CD
&
Schmidt MH
. Diverse signaling mechanisms of mTOR complexes: mTORC1 and mTORC2 in forming a formidable relationship. Adv Biol Regul.
72
51
- 62
2019.
DOI:
10.1016/j.jbior.2019.03.003
25
Saxton RA
&
Sabatini DM
. mTOR signaling in growth, metabolism, and disease. Cell.
168(6)
960
- 976
2017.
DOI:
10.1016/j.cell.2017.02.004
26
Fu W
&
Hall MN
. Regulation of mTORC2 signaling. Genes (Basel).
11(9)
1045
2020.
DOI:
10.3390/genes11091045
27
Szwed A
,
Kim E
&
Jacinto E
. Regulation and metabolic functions of mTORC1 and mTORC2. Physiol Rev.
101(3)
1371
- 1426
2021.
DOI:
10.1152/physrev.00026.2020
28
Wu F
,
Chen Z
,
Liu J
&
Hou Y
. The Akt-mTOR network at the interface of hematopoietic stem cell homeostasis. Exp Hematol.
103
15
- 23
2021.
DOI:
10.1016/j.exphem.2021.08.009
29
Jafari M
,
Ghadami E
,
Dadkhah T
&
Akhavan-Niaki H
. PI3k/AKT signaling pathway: Erythropoiesis and beyond. J Cell Physiol.
234
2373
- 2385
2019.
DOI:
10.1002/jcp.27262
30
Bang E
,
Kim DH
&
Chung HY
. Protease-activated receptor 2 induces ROS-mediated inflammation through Akt-mediated NF-ĸB and FoxO6 modulation during skin photoaging. Redox Biol.
44
102022
2021.
DOI:
10.1016/j.redox.2021.102022
31
Tzivion G
,
Dobson M
&
Ramakrishnan G
. FoxO transcription factors; Regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta Mol Cell Res.
1813(11)
1938
- 1945
2011.
DOI:
10.1016/j.bbamcr.2011.06.002
32
Chibaya L
,
Karim B
,
Zhang H
&
Jones SN
. Mdm2 phosphorylation by Akt regulates the p53 response to oxidative stress to promote cell proliferation and tumorigenesis. Proc Natl Acad Sci USA.
118(4)
e2003193118
2021.
DOI:
10.1073/pnas.2003193118
33
Hermida MA
,
Dinesh Kumar J
&
Leslie NR
. GSK3 and its interactions with the PI3K/AKT/mTOR signalling network. Adv Biol Regul.
65
5
- 15
2017.
DOI:
10.1016/j.jbior.2017.06.003
34
Papa A
&
Pandolfi PP
. The PTEN-PI3K axis in cancer. Biomolecules.
9(4)
153
2019.
DOI:
10.3390/biom9040153
35
Luongo F
,
Colonna F
,
Calapà F
,
Vitale S
,
Fiori ME
&
De Maria R
. PTEN tumor-suppressor: the dam of stemness in cancer. Cancers (Basel).
11(8)
1076
2019.
DOI:
10.3390/cancers11081076
36
Pedicone C
,
Meyer ST
,
Chisholm JD
&
Kerr WG
. Targeting SHIP1 and SHIP2 in cancer. Cancers (Basel).
13(4)
890
2021.
DOI:
10.3390/cancers13040890
37
Hibbs ML
,
Raftery AL
&
Tsantikos E
. Regulation of hematopoietic cell signaling by SHIP-1 inositol phosphatase: growth factors and beyond. Growth Factors.
36(5-6)
213
- 231
2018.
DOI:
10.1080/08977194.2019.1569649
38
Ahmed ESA
,
Ahmed NH
,
Medhat AM
,
Said UZ
,
Rashed LA
&
Abdel Ghaffar ARB
. Mesenchymal stem cells targeting PI3K/AKT pathway in leukemic model. Tumor Biology.
41(4)
101042831984680
2019.
DOI:
10.1177/1010428319846803
39
Piddock RE
,
Bowles KM
&
Rushworth SA
. The role of PI3K isoforms in regulating bone marrow microenvironment signaling focusing on acute myeloid leukemia and multiple myeloma. Cancers (Basel).
9(4)
29
2017.
DOI:
10.3390/cancers9040029
40
Gyori D
,
Chessa T
,
Hawkins PT
&
Stephens LR
. Class (I) phosphoinositide 3-kinases in the tumor microenvironment. Cancers (Basel).
9(3)
24
2017.
DOI:
10.3390/cancers9030024
41
Ondrisova L
&
Mraz M
. Genetic and non-genetic mechanisms of resistance to BCR signaling inhibitors in B cell malignancies. Front Oncol.
10
591577
2020.
DOI:
10.3389/fonc.2020.591577
42
Burger JA
&
Chiorazzi N
. B cell receptor signaling in chronic lymphocytic leukemia. Trends Immunol.
34(12)
592
- 601
2013.
DOI:
10.1016/j.it.2013.07.002
43
Wiestner A
. BCR pathway inhibition as therapy for chronic lymphocytic leukemia and lymphoplasmacytic lymphoma. Hematology.
2014(1)
125
- 134
2014.
DOI:
10.1182/asheducation-2014.1.125
44
Aoki M
&
Fujishita T
. Oncogenic roles of the PI3K/AKT/mTOR axis. Curr Top Microbiol Immunol.
407
153
- 189
2017.
DOI:
10.1007/82_2017_6
45
Vanhaesebroeck B
,
Perry MWD
,
Brown JR
,
André F
&
Okkenhaug K
. PI3K inhibitors are finally coming of age. Nat Rev Drug Discov.
20(10)
741
- 769
2021.
DOI:
10.1038/s41573-021-00209-1
46
Arnason JE
&
Brown JR
. Targeting B cell signaling in chronic lymphocytic leukemia. Curr Oncol Rep.
19(9)
61
2017.
DOI:
10.1007/s11912-017-0620-7
47
Cooney JD
,
Lin AP
,
Jiang D
,
Wang L
,
Suhasini AN
,
Myers J
,
Qiu Z
,
Wölfler A
,
Sill H
&
Aguiar RCT
. Synergistic targeting of the regulatory and catalytic subunits of PI3Kδ in mature B-cell malignancies. Clin Cancer Res.
24(5)
1103
- 1113
2018.
DOI:
10.1158/1078-0432.CCR-17-2218
48
Estruch M
,
Reckzeh K
,
Vittori C
,
Centio A
,
Ali M
,
Engelhard S
,
Zhao L
,
Won KJ
,
Liu P
,
Porse BT
&
Theilgaard-Mönch K
. Targeted inhibition of cooperative mutation- and therapy-induced AKT activation in AML effectively enhances response to chemotherapy. Leukemia.
35(7)
2030
- 2042
2021.
DOI:
10.1038/s41375-020-01094-0
49
Nepstad I
,
Hatfield KJ
,
Grønningsæter IS
&
Reikvam H
. The PI3K-Akt-mTOR signaling pathway in human acute myeloid leukemia (AML) Cells. Int J Mol Sci.
21(8)
2907
2020.
DOI:
10.3390/ijms21082907
50
Estruch M
,
Vittori C
,
Montesinos TM
,
Reckzeh K
&
Theilgaard-Mönch K
. Targeting of PI3K/AKT signaling and DNA damage response in acute myeloid leukemia: a novel therapeutic strategy to boost chemotherapy response and overcome resistance. Cancer Drug Resist.
4(4)
984
- 995
2021.
DOI:
10.20517/cdr.2021.76
51
Sujobert P
,
Bardet V
,
Cornillet-Lefebvre P
,
Hayflick JS
,
Prie N
,
Verdier F
,
Vanhaesebroeck B
,
Muller O
,
Pesce F
,
Ifrah N
,
Hunault-Berger M
,
Berthou C
,
Villemagne B
,
Jourdan E
,
Audhuy B
,
Solary E
,
Witz B
,
Harousseau JL
,
Himberlin C
,
Lamy T
,
Lioure B
,
Cahn JY
,
Dreyfus F
,
Mayeux P
,
Lacombe C
&
Bouscary D
. Essential role for the p110 isoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia. Blood.
106(3)
1063
- 1066
2005.
DOI:
10.1182/blood-2004-08-3225
52
Sykes SM
,
Lane SW
,
Bullinger L
,
Kalaitzidis D
,
Yusuf R
,
Saez B
,
Ferraro F
,
Mercier F
,
Singh H
,
Brumme KM
,
Acharya SS
,
Scholl C
,
Tothova Z
,
Attar EC
,
Fröhling S
,
DePinho RA
,
Armstrong SA
,
Gilliland DG
&
Scadden DT
. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell.
146(5)
697
- 708
2011.
DOI:
10.1016/j.cell.2011.07.032
53
Hu P
,
Li H
,
Yu X
,
Liu X
,
Wang X
,
Qing Y
,
Wang Z
,
Wang H
,
Zhu M
,
Xu J
,
Tan R
,
Guo Q
&
Hui H
. GL-V9 exerts anti-T cell malignancies effects via promoting lysosome-dependent AKT1 degradation and activating AKT1/FOXO3A/BIM axis. Free Radic Biol Med.
145
237
- 249
2019.
DOI:
10.1016/j.freeradbiomed.2019.09.028
54
Kapoor I
,
Li Y
,
Sharma A
,
Zhu H
,
Bodo J
,
Xu W
,
Hsi ED
,
Hill BT
&
Almasan A
. Resistance to BTK inhibition by ibrutinib can be overcome by preventing FOXO3a nuclear export and PI3K/AKT activation in B-cell lymphoid malignancies. Cell Death Dis.
10(12)
924
2019.
DOI:
10.1038/s41419-019-2158-0
55
Pagano MA
,
Tibaldi E
,
Molino P
,
Frezzato F
,
Trimarco V
,
Facco M
,
Zagotto G
,
Ribaudo G
,
Leanza L
,
Peruzzo R
,
Szabò I
,
Visentin A
,
Frasson M
,
Semenzato G
,
Trentin L
&
Brunati AM
. Mitochondrial apoptosis is induced by Alkoxy phenyl-1-propanone derivatives through PP2A-mediated dephosphorylation of Bad and Foxo3A in CLL. Leukemia.
33(5)
1148
- 1160
2019.
DOI:
10.1038/s41375-018-0288-5
56
Ghosh J
&
Kapur R
. Role of mTORC1-S6K1 signaling pathway in regulation of hematopoietic stem cell and acute myeloid leukemia. Exp Hematol.
50
13
- 21
2017.
DOI:
10.1016/j.exphem.2017.02.004
57
Herschbein L
&
Liesveld JL
. Dueling for dual inhibition: Means to enhance effectiveness of PI3K/Akt/mTOR inhibitors in AML. Blood Rev.
32(3)
235
- 248
2018.
DOI:
10.1016/j.blre.2017.11.006
58
Ghosh J
,
Kobayashi M
,
Ramdas B
,
Chatterjee A
,
Ma P
,
Mali RS
,
Carlesso N
,
Liu Y
,
Plas DR
,
Chan RJ
&
Kapur R
. S6K1 regulates hematopoietic stem cell self-renewal and leukemia maintenance. J Clin Invest.
126(7)
2621
- 2625
2016.
DOI:
10.1172/JCI84565
59
Panuzzo C
,
Pironi L
,
Maglione A
,
Rocco S
,
Stanga S
,
Riganti C
,
Kopecka J
,
Ali MS
,
Pergolizzi B
,
Bracco E
&
Cilloni D
. mTORC2 is activated under hypoxia and could support chronic myeloid leukemia stem cells. Int J Mol Sci.
24(2)
1234
2023.
DOI:
10.3390/ijms24021234
60
Tabe Y
,
Tafuri A
,
Sekihara K
,
Yang H
&
Konopleva M
. Inhibition of mTOR kinase as a therapeutic target for acute myeloid leukemia. Expert Opin Ther Targets.
21(7)
705
- 714
2017.
DOI:
10.1080/14728222.2017.1333600
61
Poulain L
,
Sujobert P
,
Zylbersztejn F
,
Barreau S
,
Stuani L
,
Lambert M
,
Palama TL
,
Chesnais V
,
Birsen R
,
Vergez F
,
Farge T
,
Chenevier-Gobeaux C
,
Fraisse M
,
Bouillaud F
,
Debeissat C
,
Herault O
,
Récher C
,
Lacombe C
,
Fontenay M
,
Mayeux P
,
Maciel TT
,
Portais JC
,
Sarry JE
,
Tamburini J
,
Bouscary D
&
Chapuis N
. High mTORC1 activity drives glycolysis addiction and sensitivity to G6PD inhibition in acute myeloid leukemia cells. Leukemia.
31(11)
2326
- 2335
2017.
DOI:
10.1038/leu.2017.81
62
Wang X
,
Chu Y
,
Wang W
&
Yuan W
. mTORC signaling in hematopoiesis. Int J Hematol.
103(5)
510
- 518
2016.
DOI:
10.1007/s12185-016-1944-z
63
Dinner S
&
Platanias LC
. Targeting the mTOR Pathway in Leukemia. J Cell Biochem.
117(8)
1745
- 1752
2016.
DOI:
10.1002/jcb.25559
64
McCarter AC
,
Wang Q
&
Chiang M
. Notch in leukemia. In: Advances in Experimental Medicine and Biology. New York, Springer LLC.
pp 355
- 394
2018.
DOI:
10.1007/978-3-319-89512-3_18
65
Hoshii T
,
Kasada A
,
Hatakeyama T
,
Ohtani M
,
Tadokoro Y
,
Naka K
,
Ikenoue T
,
Ikawa T
,
Kawamoto H
,
Fehling HJ
,
Araki K
,
Yamamura K
,
Matsuda S
&
Hirao A
. Loss of mTOR complex 1 induces developmental blockage in early T-lymphopoiesis and eradicates T-cell acute lymphoblastic leukemia cells. Proc Natl Acad Sci USA.
111(10)
3805
- 3810
2014.
DOI:
10.1073/pnas.1320265111
66
Chen W
,
Drakos E
,
Grammatikakis I
,
Schlette EJ
,
Li J
,
Leventaki V
,
Staikou-Drakopoulou E
,
Patsouris E
,
Panayiotidis P
,
Medeiros LJ
&
Rassidakis GZ
. mTOR signaling is activated by FLT3 kinase and promotes survival of FLT3-mutated acute myeloid leukemia cells. Mol Cancer.
9
292
2010.
DOI:
10.1186/1476-4598-9-292
67
Doepfner KT
,
Spertini O
&
Arcaro A
. Autocrine insulin-like growth factor-I signaling promotes growth and survival of human acute myeloid leukemia cells via the phosphoinositide 3-kinase/Akt pathway. Leukemia.
21(9)
1921
- 1930
2007.
DOI:
10.1038/sj.leu.2404813
68
Gojo I
,
Perl A
,
Luger S
,
Baer MR
,
Norsworthy KJ
,
Bauer KS
,
Tidwell M
,
Fleckinger S
,
Carroll M
&
Sausville EA
. Phase I study of UCN-01 and perifosine in patients with relapsed and refractory acute leukemias and high-risk myelodysplastic syndrome. Invest New Drugs.
31(5)
1217
- 1227
2013.
DOI:
10.1007/s10637-013-9937-8
69
Ragon BK
,
Kantarjian H
,
Jabbour E
,
Ravandi F
,
Cortes J
,
Borthakur G
,
DeBose L
,
Zeng Z
,
Schneider H
,
Pemmaraju N
,
Garcia-Manero G
,
Kornblau S
,
Wierda W
,
Burger J
,
DiNardo CD
,
Andreeff M
,
Konopleva M
&
Daver N
. Buparlisib, a PI3K inhibitor, demonstrates acceptable tolerability and preliminary activity in a phase I trial of patients with advanced leukemias. Am J Hematol.
92(1)
7
- 11
2017.
DOI:
10.1002/ajh.24568
70
Rheingold SR
,
Tasian SK
,
Whitlock JA
,
Teachey DT
,
Borowitz MJ
,
Liu X
,
Minard CG
,
Fox E
,
Weigel BJ
&
Blaney SM
. A phase 1 trial of temsirolimus and intensive re-induction chemotherapy for 2nd or greater relapse of acute lymphoblastic leukaemia: a Children’s Oncology Group study (ADVL1114). Br J Haematol.
177(3)
467
- 474
2017.
DOI:
10.1111/bjh.14569
71
Ragon BK
,
Odenike O
,
Baer MR
,
Stock W
,
Borthakur G
,
Patel K
,
Han L
,
Chen H
,
Ma H
,
Joseph L
,
Zhao Y
,
Baggerly K
,
Konopleva M
&
Jain N
. Oral MEK 1/2 inhibitor trametinib in combination with AKT inhibitor GSK2141795 in patients with acute myeloid leukemia with RAS mutations: a phase II study. Clin Lymphoma Myeloma Leuk.
19(7)
431
- 440.e13
2019.
DOI:
10.1016/j.clml.2019.03.015
72
Lang F
,
Wunderle L
,
Badura S
,
Schleyer E
,
Brüggemann M
,
Serve H
,
Schnittger S
,
Gökbuget N
,
Pfeifer H
,
Wagner S
,
Ashelford K
,
Bug G
&
Ottmann OG
. A phase I study of a dual PI3-kinase/mTOR inhibitor BEZ235 in adult patients with relapsed or refractory acute leukemia. BMC Pharmacol Toxicol.
21(1)
70
2020.
DOI:
10.1186/s40360-020-00446-x
73
Tasian SK
,
Silverman LB
,
Whitlock JA
,
Sposto R
,
Loftus JP
,
Schafer ES
,
Schultz KR
,
Hutchinson RJ
,
Gaynon PS
,
Orgel E
,
Bateman CM
,
Cooper TM
,
Laetsch TW
,
Sulis ML
,
Chi YY
,
Malvar J
,
Wayne AS
&
Rheingold SR
. Temsirolimus combined with cyclophosphamide and etoposide for pediatric patients with relapsed/refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium trial (TACL 2014-001). Haematologica.
107(10)
2295
- 2303
2022.
DOI:
10.3324/haematol.2021.279520
74
Brown JR
,
Byrd JC
,
Coutre SE
,
Benson DM
,
Flinn IW
,
Wagner-Johnston ND
,
Spurgeon SE
,
Kahl BS
,
Bello C
,
Webb HK
,
Johnson DM
,
Peterman S
,
Li D
,
Jahn TM
,
Lannutti BJ
,
Ulrich RG
,
Yu AS
,
Miller LL
&
Furman RR
. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood.
123(22)
3390
- 3397
2014.
DOI:
10.1182/blood-2013-11-535047
75
Assouline S
,
Amrein L
,
Aloyz R
,
Banerji V
,
Caplan S
,
Owen C
,
Hasegawa W
,
Robinson S
,
Shivakumar S
,
Prica A
,
Peters A
,
Hagerman L
,
Rodriguez L
,
Skamene T
,
Panasci L
,
Chen BE
&
Hay AE
. IND.216: a phase II study of buparlisib and associated biomarkers, raptor and p70S6K, in patients with relapsed and refractory chronic lymphocytic leukemia. Leuk Lymphoma.
61(7)
1653
- 1659
2020.
DOI:
10.1080/10428194.2020.1734594
76
Friedman DR
,
Lanasa MC
,
Davis PH
,
Allgood SD
,
Matta KM
,
Brander DM
,
Chen Y
,
Davis ED
,
Volkheimer AD
,
Moore JO
,
Gockerman JP
,
Sportelli P
&
Weinberg JB
. Perifosine treatment in chronic lymphocytic leukemia: results of a phase II clinical trial and in vitro studies. Leuk Lymphoma.
55(5)
1067
- 1075
2014.
DOI:
10.3109/10428194.2013.824080
77
Davids MS
,
Kim HT
,
Nicotra A
,
Savell A
,
Francoeur K
,
Hellman JM
,
Bazemore J
,
Miskin HP
,
Sportelli P
,
Stampleman L
,
Maegawa R
,
Rueter J
,
Boruchov AM
,
Arnason JE
,
Jacobson CA
,
Jacobsen ED
,
Fisher DC
,
Brown JR
&
Blood Cancer Research Partnership of the Leukemia and Lymphoma Society
. Umbralisib in combination with ibrutinib in patients with relapsed or refractory chronic lymphocytic leukaemia or mantle cell lymphoma: a multicentre phase 1-1b study. Lancet Haematol.
6(1)
e38
- e47
2019.
DOI:
10.1016/S2352-3026(18)30196-0
78
Davids MS
,
Fisher DC
,
Tyekucheva S
,
McDonough M
,
Hanna J
,
Lee B
,
Francoeur K
,
Montegaard J
,
Odejide O
,
Armand P
,
Arnason J
&
Brown JR
. A phase 1b/2 study of duvelisib in combination with FCR (DFCR) for frontline therapy for younger CLL patients. Leukemia.
35(4)
1064
- 1072
2021.
DOI:
10.1038/s41375-020-01010-6
79
Danilov AV
,
Herbaux C
,
Walter HS
,
Hillmen P
,
Rule SA
,
Kio EA
,
Karlin L
,
Dyer MJS
,
Mitra SS
,
Yi PC
,
Humeniuk R
,
Huang X
,
Zhou Z
,
Bhargava P
,
Jürgensmeier JM
&
Fegan CD
. Phase Ib study of tirabrutinib in combination with idelalisib or entospletinib in previously treated chronic lymphocytic leukemia. Clin Cancer Res.
26(12)
2810
- 2818
2020.
DOI:
10.1158/1078-0432.CCR-19-3504
80
Frustaci AM
,
Tedeschi A
,
Deodato M
,
Zamprogna G
,
Cairoli R
&
Montillo M
. Duvelisib: a new phosphoinositide-3-kinase inhibitor in chronic lymphocytic leukemia. Future Oncol.
15(19)
2227
- 2239
2019.
DOI:
10.2217/fon-2018-0881
81
Skånland SS
,
Cremaschi A
,
Bendiksen H
,
Hermansen JU
,
Thimiri Govinda Raj DB
,
Munthe LA
,
Tjønnfjord GE
&
Taskén K
. An in vitro assay for biomarker discovery and dose prediction applied to ibrutinib plus venetoclax treatment of CLL. Leukemia.
34(2)
478
- 487
2020.
DOI:
10.1038/s41375-019-0569-7
82
Murali I
,
Kasar S
,
Naeem A
,
Tyekucheva S
,
Khalsa JK
,
Thrash EM
,
Itchaki G
,
Livitz D
,
Leshchiner I
,
Dong S
,
Fernandes SM
,
Getz G
,
Johnson A
&
Brown JR
. Activation of the MAPK pathway mediates resistance to PI3K inhibitors in chronic lymphocytic leukemia. Blood.
138(1)
44
- 56
2021.
DOI:
10.1182/blood.2020006765
83
Dong S
,
Harrington BK
,
Hu EY
,
Greene JT
,
Lehman AM
,
Tran M
,
Wasmuth RL
,
Long M
,
Muthusamy N
,
Brown JR
,
Johnson AJ
&
Byrd JC
. PI3K p110δ inactivation antagonizes chronic lymphocytic leukemia and reverses T cell immune suppression. J Clin Invest.
129(1)
122
- 136
2019.
DOI:
10.1172/JCI99386
84
Zhang H
,
He F
,
Gao G
,
Lu S
,
Wei Q
,
Hu H
,
Wu Z
,
Fang M
&
Wang X
. Approved small-molecule ATP-competitive kinases drugs containing indole/azaindole/oxindole scaffolds: R&D and binding patterns profiling. Molecules.
28(3)
943
2023.
DOI:
10.3390/molecules28030943
85
Roskoski R Jr
. Properties of FDA-approved small molecule phosphatidylinositol 3-kinase inhibitors prescribed for the treatment of malignancies. Pharmacol Res.
168
105579
2021.
DOI:
10.1016/j.phrs.2021.105579
86
Smolewski P
&
Rydygier D
. Efficacy and safety of idelalisib for the treatment of indolent B-cell malignancies. Expert Opin Pharmacother.
21(15)
1915
- 1926
2020.
DOI:
10.1080/14656566.2020.1791083
87
Tripathy HK
,
Manju NS
,
Dittakavi S
,
Zakkula A
&
Mullangi R
. DBS assay with LC-MS/MS for the determination of idelalisib, a selective PI3K-δ inhibitor in mice blood and its application to a pharmacokinetic study. Drug Res.
71(01)
36
- 42
2021.
DOI:
10.1055/a-1252-2476
88
Somoza JR
,
Koditek D
,
Villaseñor AG
,
Novikov N
,
Wong MH
,
Liclican A
,
Xing W
,
Lagpacan L
,
Wang R
,
Schultz BE
,
Papalia GA
,
Samuel D
,
Lad L
&
McGrath ME
. Structural, biochemical, and biophysical characterization of idelalisib binding to phosphoinositide 3-kinase δ. J Biol Chem.
290(13)
8439
- 8446
2015.
DOI:
10.1074/jbc.M114.634683
89
Staber PB
,
Jurczak W
,
Greil R
,
Vucinic V
,
Middeke JM
,
Montillo M
,
Munir T
,
Neumeister P
,
Schetelig J
,
Stilgenbauer S
,
Striebel F
,
Dirnberger-Hertweck M
,
Weirather J
,
Brugger W
,
Kelemen P
,
Wendtner CM
&
Woyach JA
. Tafasitamab combined with idelalisib or venetoclax in patients with CLL previously treated with a BTK inhibitor. Leuk Lymphoma.
62(14)
3440
- 3451
2021.
DOI:
10.1080/10428194.2021.1964020
90
Lampson BL
,
Kim HT
,
Davids MS
,
Abramson JS
,
Freedman AS
,
Jacobson CA
,
Armand PA
,
Joyce RM
,
Arnason JE
,
Rassenti LZ
,
Kipps TJ
,
Fein J
,
Fernandes SM
,
Hanna JR
,
Fisher DC
&
Brown JR
. Efficacy results of a phase 2 trial of first-line idelalisib plus ofatumumab in chronic lymphocytic leukemia. Blood Adv.
3(7)
1167
- 1174
2019.
DOI:
10.1182/bloodadvances.2018030221
91
Hoellenriegel J
,
Meadows SA
,
Sivina M
,
Wierda WG
,
Kantarjian H
,
Keating MJ
,
Giese N
,
O’Brien S
,
Yu A
,
Miller LL
,
Lannutti BJ
&
Burger JA
. The phosphoinositide 3’-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia. Blood.
118(13)
3603
- 3612
2011.
DOI:
10.1182/blood-2011-05-352492
92
Serrat N
,
Guerrero-Hernández M
,
Matas-Céspedes A
,
Yahiaoui A
,
Valero JG
,
Nadeu F
,
Clot G
,
Di Re M
,
Corbera-Bellalta M
,
Magnano L
,
Rivas-Delgado A
,
Enjuanes A
,
Beà S
,
Cid MC
,
Campo E
,
Montero J
,
Hodson DJ
,
López-Guillermo A
,
Colomer D
,
Tannheimer S
&
Pérez-Galán P
. PI3Kδ inhibition reshapes follicular lymphoma-immune microenvironment cross talk and unleashes the activity of venetoclax. Blood Adv.
4(17)
4217
- 4231
2020.
DOI:
10.1182/bloodadvances.2020001584
93
Fukuhara N
,
Kinoshita T
,
Yamamoto K
,
Nagai H
,
Izutsu K
,
Yamamoto G
,
Bhargava P
,
Rajakumaraswamy N
,
Humeniuk R
,
Mathias A
,
Xing G
,
Fukui M
&
Tobinai K
. Phase 1b study to investigate the safety and tolerability of idelalisib in Japanese patients with relapsed/refractory follicular lymphoma and chronic lymphocytic leukemia. Jpn J Clin Oncol.
50(12)
1395
- 1402
2020.
DOI:
10.1093/jjco/hyaa153
94
Gopal AK
,
Fanale MA
,
Moskowitz CH
,
Shustov AR
,
Mitra S
,
Ye W
,
Younes A
&
Moskowitz AJ
. Phase II study of idelalisib, a selective inhibitor of PI3Kδ, for relapsed/refractory classical Hodgkin lymphoma. Ann Oncol.
28(5)
1057
- 1063
2017.
DOI:
10.1093/annonc/mdx028
95
Zelenetz AD
,
Barrientos JC
,
Brown JR
,
Coiffier B
,
Delgado J
,
Egyed M
,
Ghia P
,
Illés Á
,
Jurczak W
,
Marlton P
,
Montillo M
,
Morschhauser F
,
Pristupa AS
,
Robak T
,
Sharman JP
,
Simpson D
,
Smolej L
,
Tausch E
,
Adewoye AH
,
Dreiling LK
,
Kim Y
,
Stilgenbauer S
&
Hillmen P
. Idelalisib or placebo in combination with bendamustine and rituximab in patients with relapsed or refractory chronic lymphocytic leukaemia: interim results from a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol.
18(3)
297
- 311
2017.
DOI:
10.1016/S1470-2045(16)30671-4
96
Tomowiak C
,
Poulain S
,
Herbaux C
,
Perrot A
,
Mahé B
,
Morel P
,
Aurran T
,
Tournilhac O
,
Leprêtre S
,
Assaad S
,
Villemagne B
,
Casasnovas O
,
Nollet D
,
Roos-Weil D
,
Chevret S
&
Leblond V
. Obinutuzumab and idelalisib in symptomatic patients with relapsed/refractory Waldenström macroglobulinemia. Blood Adv.
5(9)
2438
- 2446
2021.
DOI:
10.1182/bloodadvances.2020003895
97
Hus I
,
Puła B
&
Robak T
. PI3K inhibitors for the treatment of chronic lymphocytic leukemia: current status and future perspectives. Cancers (Basel).
14(6)
1571
2022.
DOI:
10.3390/cancers14061571
98
Nikolaenko L
,
Liu T
&
Danilov AV
. Duvelisib (Copiktra) in relapsed or refractory chronic lymphocytic leukemia: safety and efficacy. Expert Rev Anticancer Ther.
21(5)
481
- 488
2021.
DOI:
10.1080/14737140.2021.1882857
99
Munoz J
,
Follows GA
&
Nastoupil LJ
. Copanlisib for the treatment of malignant lymphoma: Clinical experience and future perspectives. Target Oncol.
16(3)
295
- 308
2021.
DOI:
10.1007/s11523-021-00802-9
100
Patel K
,
Danilov AV
&
Pagel JM
. Duvelisib for CLL/SLL and follicular non-Hodgkin lymphoma. Blood.
134(19)
1573
- 1577
2019.
DOI:
10.1182/blood.2019001795
102
Balakrishnan K
,
Peluso M
,
Fu M
,
Rosin NY
,
Burger JA
,
Wierda WG
,
Keating MJ
,
Faia K
,
O’Brien S
,
Kutok JL
&
Gandhi V
. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia.
29(9)
1811
- 1822
2015.
DOI:
10.1038/leu.2015.105
103
Flinn IW
,
O’Brien S
,
Kahl B
,
Patel M
,
Oki Y
,
Foss FF
,
Porcu P
,
Jones J
,
Burger JA
,
Jain N
,
Kelly VM
,
Allen K
,
Douglas M
,
Sweeney J
,
Kelly P
&
Horwitz S
. Duvelisib, a novel oral dual inhibitor of PI3K-δ,γ, is clinically active in advanced hematologic malignancies. Blood.
131(8)
877
- 887
2018.
DOI:
10.1182/blood-2017-05-786566
104
Magagnoli M
,
Carlo-Stella C
&
Santoro A
. Copanlisib for the treatment of adults with relapsed follicular lymphoma. Expert Rev Clin Pharmacol.
13(8)
813
- 823
2020.
DOI:
10.1080/17512433.2020.1787829
105
Narkhede M
&
Cheson BD
. Copanlisib in the treatment of non-Hodgkin lymphoma. Future Oncology.
16(26)
1947
- 1955
2020.
DOI:
10.2217/fon-2020-0195
106
Eltantawy A
,
Vallejos X
,
Sebea E
&
Evans K
. Copanlisib: an intravenous phosphatidylinositol 3-kinase (PI3K) inhibitor for the treatment of relapsed follicular lymphoma. Ann Pharmacother.
53(9)
954
- 958
2019.
DOI:
10.1177/1060028019833992
107
Dreyling M
,
Morschhauser F
,
Bouabdallah K
,
Bron D
,
Cunningham D
,
Assouline SE
,
Verhoef G
,
Linton K
,
Thieblemont C
,
Vitolo U
,
Hiemeyer F
,
Giurescu M
,
Garcia-Vargas J
,
Gorbatchevsky I
,
Liu L
,
Koechert K
,
Peña C
,
Neves M
,
Childs BH
&
Zinzani PL
. Phase II study of copanlisib, a PI3K inhibitor, in relapsed or refractory, indolent or aggressive lymphoma. Ann Oncol.
28(9)
2169
- 2178
2017.
DOI:
10.1093/annonc/mdx289
108
Mirabilii S
,
Ricciardi MR
,
Piedimonte M
,
Gianfelici V
,
Bianchi MP
&
Tafuri A
. Biological aspects of mTOR in leukemia. Int J Mol Sci.
19(8)
2396
2018.
DOI:
10.3390/ijms19082396
109
Calimeri T
&
Ferreri AJM
. m‐TOR inhibitors and their potential role in haematological malignancies. Br J Haematol.
177(5)
684
- 702
2017.
DOI:
10.1111/bjh.14529
110
Grzybowska-Izydorczyk O
&
Smolewski P
. mTOR kinase inhibitors as a treatment strategy in hematological malignancies. Future Med Chem.
4(4)
487
- 504
2012.
DOI:
10.4155/fmc.12.14
111
Feng Y
,
Chen X
,
Cassady K
,
Zou Z
,
Yang S
,
Wang Z
&
Zhang X
. The role of mTOR inhibitors in hematologic disease: from bench to bedside. Front Oncol.
10
611690
2021.
DOI:
10.3389/fonc.2020.611690
112
Fan L
,
Wang C
,
Zhao L
,
Wang Z
,
Zhang X
,
Liu X
,
Cao L
,
Xu W
&
Li J
. SHC014748M, a novel selective inhi-bitor of PI3Kδ, demonstrates promising preclinical antitumor activity in B cell lymphomas and chronic lymphocytic leukemia. Neoplasia.
22(12)
714
- 724
2020.
DOI:
10.1016/j.neo.2020.10.004
113
Hanna BS
,
Roessner PM
,
Scheffold A
,
Jebaraj BMC
,
Demerdash Y
,
Öztürk S
,
Lichter P
,
Stilgenbauer S
&
Seiffert M
. PI3Kδ inhibition modulates regulatory and effector T-cell differentiation and function in chronic lymphocytic leukemia. Leukemia.
33(6)
1427
- 1438
2019.
DOI:
10.1038/s41375-018-0318-3
114
Eschweiler S
,
Ramírez-Suástegui C
,
Li Y
,
King E
,
Chudley L
,
Thomas J
,
Wood O
,
von Witzleben A
,
Jeffrey D
,
McCann K
,
Simon H
,
Mondal M
,
Wang A
,
Dicker M
,
Lopez-Guadamillas E
,
Chou TF
,
Dobbs NA
,
Essame L
,
Acton G
,
Kelly F
,
Halbert G
,
Sacco JJ
,
Schache AG
,
Shaw R
,
McCaul JA
,
Paterson C
,
Davies JH
,
Brennan PA
,
Singh RP
,
Loadman PM
,
Wilson W
,
Hackshaw A
,
Seumois G
,
Okkenhaug K
,
Thomas GJ
,
Jones TM
,
Ay F
,
Friberg G
,
Kronenberg M
,
Vanhaesebroeck B
,
Vijayanand P
&
Ottensmeier CH
. Intermittent PI3Kδ inhibition sustains anti-tumour immunity and curbs irAEs. Nature.
605(7911)
741
- 746
2022.
DOI:
10.1038/s41586-022-04685-2
115
Maharaj K
,
Powers JJ
,
Achille A
,
Mediavilla-Varela M
,
Gamal W
,
Burger KL
,
Fonseca R
,
Jiang K
,
Miskin HP
,
Maryanski D
,
Monastyrskyi A
,
Duckett DR
,
Roush WR
,
Cleveland JL
,
Sahakian E
&
Pinilla-Ibarz J
. The dual PI3Kδ/CK1ε inhibitor umbralisib exhibits unique immunomodulatory effects on CLL T cells. Blood Adv.
4(13)
3072
- 3084
2020.
DOI:
10.1182/bloodadvances.2020001800
116
Chellappa S
,
Kushekhar K
,
Munthe LA
,
Tjønnfjord GE
,
Aandahl EM
,
Okkenhaug K
&
Taskén K
. The PI3K p110δ isoform inhibitor idelalisib preferentially inhibits human regulatory T cell function. J Immunol.
202(5)
1397
- 1405
2019.
DOI:
10.4049/jimmunol.1701703
117
Amrein L
,
Shawi M
,
Grenier J
,
Aloyz R
&
Panasci L
. The phosphatidylinositol-3 kinase I inhibitor BKM120 induces cell death in B-chronic lymphocytic leukemia cells in vitro. Int J Cancer.
133(1)
247
- 252
2013.
DOI:
10.1002/ijc.27989
118
Mehrpouri M
,
Momeny M
&
Bashash D
. Synergistic effects of BKM120 and panobinostat on pre-B acute lymphoblastic cells: an emerging perspective for the simultaneous inhibition of PI3K and HDACs. J Recept Signal Transduct Res.
42(1)
100
- 108
2022.
DOI:
10.1080/10799893.2020.1853159
119
Li X
,
Su Y
,
Madlambayan G
,
Edwards H
,
Polin L
,
Kushner J
,
Dzinic SH
,
White K
,
Ma J
,
Knight T
,
Wang G
,
Wang Y
,
Yang J
,
Taub JW
,
Lin H
&
Ge Y
. Antileukemic activity and mechanism of action of the novel PI3K and histone deacetylase dual inhibitor CUDC-907 in acute myeloid leukemia. Haematologica.
104(11)
2225
- 2240
2019.
DOI:
10.3324/haematol.2018.201343
120
Riyahi N
,
Safaroghli-Azar A
,
Sheikh-Zeineddini N
,
Sayyadi M
&
Bashash D
. Synergistic effects of PI3K and c-Myc Co-targeting in acute leukemia: Shedding new light on resistance to selective PI3K-δ inhibitor CAL-101. Cancer Invest.
37(7)
311
- 324
2019.
DOI:
10.1080/07357907.2019.1651328
121
Guidetti A
,
Carlo-Stella C
,
Locatelli SL
,
Malorni W
,
Mortarini R
,
Viviani S
,
Russo D
,
Marchianò A
,
Sorasio R
,
Dodero A
,
Farina L
,
Giordano L
,
Di Nicola M
,
Anichini A
,
Corradini P
&
Gianni AM
. Phase II study of perifosine and sorafenib dual-targeted therapy in patients with relapsed or refractory lymphoproliferative diseases. Clin Cancer Res.
20(22)
5641
- 5651
2014.
DOI:
10.1158/1078-0432.CCR-14-0770
122
Rigolin GM
,
Cavazzini F
,
Piciocchi A
,
Arena V
,
Visentin A
,
Reda G
,
Zamprogna G
,
Cibien F
,
Vitagliano O
,
Coscia M
,
Farina L
,
Gaidano G
,
Murru R
,
Varettoni M
,
Paolini R
,
Sportoletti P
,
Pietrasanta D
,
Molinari AL
,
Quaglia FM
,
Laurenti L
,
Marasca R
,
Marchetti M
,
Mauro FR
,
Crea E
,
Vignetti M
,
Gentile M
,
Montillo M
,
Foà R
,
Cuneo A
&
GIMEMA Group
. Efficacy of idelalisib and rituximab in relapsed/refractory chronic lymphocytic leukemia treated outside of clinical trials. A report of the Gimema Working Group. Hematol Oncol.
39(3)
326
- 335
2021.
DOI:
10.1002/hon.2861
123
Mato AR
,
Ghosh N
,
Schuster SJ
,
Lamanna N
,
Pagel JM
,
Flinn IW
,
Barrientos JC
,
Rai KR
,
Reeves JA
,
Cheson BD
,
Barr PM
,
Kambhampati S
,
Lansigan F
,
Pu JJ
,
Skarbnik AP
,
Roeker L
,
Fonseca GA
,
Sitlinger A
,
Hamadeh IS
,
Dorsey C
,
LaRatta N
,
Weissbrot H
,
Luning Prak ET
,
Tsao P
,
Paskalis D
,
Sportelli P
,
Miskin HP
,
Weiss MS
,
Svoboda J
&
Brander DM
. Phase 2 study of the safety and efficacy of umbralisib in patients with CLL who are intolerant to BTK or PI3Kδ inhibitor therapy. Blood.
137(20)
2817
- 2826
2021.
DOI:
10.1182/blood.2020007376