2.1. Interleukin-7 Signaling Pathway and Glucocorticoid Resistance in ALL
Interleukin-7 (IL-7) plays a crucial role in the T- and B-cell development. It is responsible for survival and regulating the functions of peripheral mature T-cells [
75,
76,
77,
78,
79]. IL-7 acts on cells via a heterodimer IL-7 receptor (IL-7R) comprised of subunit alpha (IL-7Rα, CD127) and the common γ chain (γc) subunit [
79]. Binding to IL-7R results in activation of associated tyrosine Janus kinases (JAK): JAK1 (by IL-7Rα) and JAK3 (by γc) [
79,
80]. JAK 1 and JAK3 induce phosphorylation of signal transducer and activator of transcription proteins (STAT) transcriptional factors, mainly STAT5A and STAT5B. Furthermore, STAT1 and STAT 3 are also activated. Next, STAT proteins translocate into the nucleus where they regulate cell-growth and survival via inter alia, increasing Bcl-2 expression. STAT5 proteins also activate the PI3K/AKT signaling pathway [
79,
80,
81]. Increased activation of the IL-7 pathway causes leukemogenesis in multiple in vitro studies. Upregulation of IL-7 signaling pathway was observed in the leukemic cells obtained from T-ALL patients, as well as in the T-ALL cell lines, and was associated with increased cell survival and proliferation [
81,
82,
83,
84,
85,
86,
87,
88]. Furthermore, activation of the IL-7 signaling pathway was showed to be associated with T-ALL resistance to glucocorticoid treatment [
89,
90,
91].
The findings described above are also important for the ALL treatment. Mutations in IL-7R/JAK signaling have been found in pre-B ALL, and thus are inhibitors of this pathway, as well as blockers of downstream signaling, which could be beneficial in pre-B ALL therapy [
87,
92,
93].
In around half of the Ph-like ALL rearrangement, the cytokine receptor-like factor 2
(CRLF2) gene is found, which leads to
CRLF2 overexpression. This, in turn, is frequently associated with activation of JAK/STAT cascade [
94,
95]. Ruxolitinib, in combination with vincristine, dexamethasone, and an
L-asparaginase (VXL) induction-type treatment regimen, was found to be efficient in mice engrafted with Ph-like ALL xenografts harboring
JAK activating mutations. This drug combination acted synergistically and prolonged disease remission [
96].
It is worth noting that a phase I clinical study of a monoclonal antibody against IL-7Rα (GSK2618960) has been completed. As indicated by this double-blind study conducted in 18 healthy individuals, GSK2618960 treatment is tolerated and efficiently blocks IL-7 signaling. However, it did not induce any effect on healthy T-cells. Nevertheless, GSK2618960 can still decrease pro-survival cascade activation in leukemic cells; thus, further studies are needed [
97]. Furthermore, another monoclonal antibody against IL-7Rα (named B12), which blocks both the wild-type and mutated IL-7Rα, was developed. B12 was found to block IL-7 and mutant IL-7Rα signaling, and to induce apoptosis in vitro. Further, B12 delayed T-ALL progression in vivo in the T-ALL engrafted mice. It was also found to potentialize dexamethasone-induced apoptosis in vitro [
98]. Given the good tolerance of IL-7Rα inhibitors during the phase I clinical study, as well as promising results of preclinical research, efficiency of these drugs in ALL treatment should be evaluated as soon as possible.
2.2. Activation of PI3K/AKT/mTOR Signaling Cascade Prevents GR from Translocation to the Nucleus
An alternate underlying mechanism of glucocorticoid resistance dependent on NRC3C1 activation could be the NR3C1 inhibitory phosphorylation, which reduces its nuclear localization and transactivation ability to activate critical downstream GTGs. Serine/threonine kinase (AKT) 1 binds to and phosphorylates the NR3C1 protein, inhibiting its nuclear translocation, according to Piovan et al. This finding suggests that activation of AKT1 may play a role in the development of glucocorticoid resistance in ALL. In vitro and in vivo, pharmacological inhibition of AKT with MK2206 significantly restores glucocorticoid-induced NR3C1 translocation to the nucleus, increases the sensitivity of T-ALL cells to the glucocorticoid therapy, and successfully reverses glucocorticoid resistance [
99].
AKT1 is a part of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of a rapamycin kinase (mTOR) signaling cascade. In leukemia, the PI3K/AKT/mTOR pathway is usually activated and plays a role in leukemogenesis, especially in T-ALL. Increased cell metabolism, proliferation, and decreased apoptosis are the result of continuous stimulation of this mechanism. Activating mutations in PI3K genes, as well as downstream effectors of the cascade, such as AKT and mTOR, causes overexpression of this pathway. Moreover, inactivating mutations in the
PTEN gene have been reported in T-ALL patients. PTEN, in turn, is a crucial inhibitor of the PI3K/AKT cascade [
100]. Activation of PI3K/AKT signaling has been also linked to the increase in the level of Bcl-2, which is an anti-apoptotic protein [
88]. Furthermore, the FBXW7 seems to be regulated by the PI3K/AKT, thus, blocking this pathway may result in increasing the GRα level [
101]. Therefore, this may be another mechanism of glucocorticoid resistance induced by the PI3K/AKT axis. However, this requires further laboratory studies, as so far, the influence of PI3K/AKT activation on FBXW7 in leukemic cells has not been clarified.
MTOR inhibitors might be beneficial in treating primary human ALL. In their studies, Teachey et al. demonstrated the activity of mTOR inhibitors in preclinical models of ALL [
111]. In addition, mTOR inhibitors improve methotrexate sensitivity by downregulating dihydrofolate reductase expression [
112]. In particular, the combined inhibition of PI3K and the mTOR complex may provide an effective treatment for acute leukemia. In fact, they had a much stronger cytostatic effect on ALL cells than everolimus, according to Wong et al. [
113]. Dactolisib (BEZ235) is an imidazoquinoline derivative that is a potent dual pan-class I PI3K and mTOR inhibitor. It inhibits downstream PI3K effectors in numerous preclinical models, including cell lines and xenografts, resulting in efficient reduction of tumor proliferation and growth [
114]. As showed by the phase I clinical study performed by Lang et al., dual inhibition of PI3K and mTOR by dactolisib induces responses in 30% of ALL cases. Dactolisib, at the dose of 400 mg/day, was poorly tolerated and severe toxicity events occurred, especially gastrointestinal (mainly stomatitis). Considering that most dose-limiting toxicities such as fatigue, diarrhea, nausea, and mucositis were noted with both PI3K and mTOR inhibitors, it is not unexpected that pan-PI3K and mTOR inhibition resulted in a high prevalence of adverse events at the dose of 400 mg/day. On the other hand, 300 mg/day was far better tolerated by the patients; therefore, this dose was established as recommended for phase II studies [
115]. It is worth noting that dactolisib was found to increase the incidence of grade 3–4 adverse events in evaluable patients in several other clinical studies [
116]. Regrettably, currently there is no ongoing clinical study of dactolisib, according to clinicaltrials.gov.
2.3. The MAPK-ERK Pathway
Glucocorticoid resistance of T-ALL leukemic cells, related to upregulation of the IL-7 pathway, was mainly associated with STAT5-induced increase in the anti-apoptotic Bcl-2. Furthermore, it was observed that glucocorticoid treatment results in upregulation of IL-7Rα, thus creating a vicious cycle [
89,
90]. However, recently, additional mechanisms of IL-7-pathway-related resistance have been described.
Li et al. discovered mutations of IL-7 pathway genes in 47 (32%) samples obtained from 146 pediatric T-ALL patients. These mutations affected
IL7R,
JAK1,
JAK3,
NF1,
NRAS,
KRAS, and
AKT genes. In 28 out of 97 prednisolone-treated samples, the IL-7 pathway mutations were associated with resistance to glucocorticoid treatment (
p = 0.033). Interestingly, patients with IL-7 pathway mutations were characterized by significantly (
p = 0.009) worse clinical outcomes, as compared to children without these mutations. Furthermore, those mutations caused activation of the PI3K/AKT pathway, resulting in an increase in Mcl-1 and Bcl-XL levels. Furthermore, higher levels of inactivated GSK3B, which is an important kinase that regulates BIM’s function, were found. Consequently, a higher ratio of phosphorylated/unphosphorylated BIM was observed. These changes were associated with mitogen-activated protein kinase (MAPK)—extracellular signal-regulated kinase (ERK) pathway activation [
89]. The MAPK-ERK pathway takes part in controlling cells’ growth, proliferation, survival, and division. Upon MAPK-ERK activation, ERK migrates to the nucleus where it directly phosphorylates target proteins or controls other kinase activity [
117]. When glucocorticoid-resistant cell lines were treated with the MAPK inhibitor (CI1040), there was a significant enhancement in GSK3B activation, as well as an increase in the non-phosphorylated BIM level [
89].
A recently published study by van der Zwet et al. demonstrated that SUPT-1 cells expressing cysteine mutants IL-7Rα
PILLT240−244RFCPH, IL-7R-α
PIL240−242QSPSC, and IL-7Rα
LT243−244LMCP, exhibited glucocorticoid resistance and activation of the downstream MAPK-ERK. In contrast, in the cells expressing the wild-type and the non-cysteine IL-7Rα mutant, MAPK-ERK upregulation and glucocorticoid resistance were not found. The MAPK-ERK pathway was also upregulated in the glucocorticoid-resistant SUPT-1 cells expressing
JAK1R724H, JAK1T901A,
NRASWT, or
NRASG12D, whereas in the glucocorticoid-sensitive SUPT-1 cells (expressing wild-type
JAK1) MAPK-ERK was not activated. It was demonstrated that MAPK-ERK upregulation causes increase in the phosphorylation of BIM
EL and BIM
L isoforms. Further, it was showed that ERK is responsible for the direct phosphorylation of BIM. BIM’s phosphorylation was found to cause impaired BIM’s binding to Bcl-2, Mcl-1, and Bcl-XL, which prevents their inactivation and, thus, promotes cells’ survival. Furthermore, in the
JAK1T901A-and-
NRASG12D-expressing SUPT-1 cells, MAPK 1/2 inhibitors selumetinib and trametinib prevented BIM’s phosphorylation in a dose-dependent manner. A similar effect was achieved in SUPT-1 cells expressing JAK1
T901A treated with JAK1/JAK2 inhibitor ruxolitinib. The response to the glucocorticoid treatment in the 46 T-ALL patient-derived xenografts with or without IL-7 was assessed. IL-7 addition induced glucocorticoid resistance in 12 (26%) of all xenografts. Interestingly, in those samples, MAPK-ERK signaling was activated by IL-7, indicating that physiological IL-7 signaling may activate the downstream MAPK-ERK pathway in T-ALL. Thus, in T leukemic cells, MAPK-ERK can be upregulated both by mutant and physiological IL-7 signaling [
118]. Interestingly, the MAPK-ERK signaling pathway is not activated by the IL-7 signaling in the healthy T-cells [
119]. Xenografts, in the absence of IL-7, were treated using MAPK inhibitors (selumetinib, trametinib, and binimetinib) and ruxolitinib. Ruxolitinib did not cause therapeutic effects, in contrast to MAPK inhibitors, which induced cytotoxicity in most of the samples. Six samples with IL-7-induced glucocorticoid resistance were then treated with ruxolitinib. The significant relationship (
p = 0.0039) between sensitivity to ruxolitinib treatment and IL-7-enhanced cells viability was found. Combined treatment with selumetinib and prednisolone was highly synergic, both in xenografts with IL-7-induced glucocorticoid resistance, and in the samples with not-IL-7-related glucocorticoid resistance. Furthermore, such a synergistic effect was also found in the glucocorticoid-sensitive xenografts. Ruxolitinib and prednisolone co-treatment was efficient only in one xenograft, with not- IL-7-related glucocorticoid resistance in the presence of IL-7, and in one xenograft with IL-7-induced glucocorticoid resistance without IL-7. In both samples, there was a significant STAT5 overexpression, which explains ruxolitinib efficiency. Therefore, ruxolitinib has limited clinical application, as it may efficiently decrease MAPK-ERK activation caused by IL-7R/JAK mutations or physiological IL-7 transmission, whereas it is not useful in the alterations occurring downstream of IL-7R/JAK. Such limitations are not encountered when using MAPK inhibitors [
118].
Mutations of genes encoding the rat sarcoma virus (Ras) protein family are found in around 30% of all cancers in humans, making them the most common genetical alterations occurring in cancer [
117,
119]. Ras’s proteins are involved in the Ras/rapidly accelerated fibrosarcoma (Raf)/MAPK/ERK signaling cascade; thus, Ras activation leads to ERK induction [
119]. Irving et al. examined 54 samples obtained from children with pre-B ALL at the time of diagnosis in search for activating mutations of Ras proteins (
KRAS and
NRAS), mutations of Ras regulators (
PTPN11), and alterations in genes encoding upstream signaling proteins
(FLT3). In 28 (51.9%) children, such mutations have been found. Ras/Raf/MAPK/ERK pathway activation was estimated in 80 pre-B ALL patients. Among 32 cases with Ras-related mutations, 27 (84.3%) had relevant Ras/Raf/MAPK/ERK signaling cascade activation. In the remaining patients without Ras mutations, activation of this pathway was found in 9 of 48 (18.7%) samples. The cells with Ras/Raf/MAPK/ERK activation were significantly more sensitive to selumetinib, regardless of the presence of Ras-related mutations [
120]. Jerchel et al. found Ras-related mutations (
NRAS,
KRAS,
FLT3,
PTPN11, and others) in 44.2% of 461 samples obtained from children with pre-B ALL at the time of diagnosis. Overall, the Ras-related mutations were most frequent in the high hyperdiploid (72.6%) and t (4;11)-rearranged (73.3%) pre-B ALL. Ras-related mutations were associated with significantly worse clinical outcomes in the HR patients. It was further established that Ras mutations are associated with glucocorticoid resistance, as samples with Ras-related mutations were 3-fold more resistant to prednisolone treatment than samples without these mutations (
p = 0.024). In this context, the clonal/subclonal
KRAS G13 were most harmful, as blasts with this mutation were characterized by more than 2000-fold-higher glucocorticoid resistance, as compared to wild-type leukemic cells. In contrast,
NRAS and
KRAS G12 were found not to increase glucocorticoid resistance in a significant manner. The treatment of Ras-mutated samples using MAPK inhibitor trametinib induced cytotoxicity, whereas wild-type blasts were unaffected (
p = 0.001) [
121]. Signaling cascades involved in the glucocorticoid resistance in ALL have been schematically presented on
Figure 1.
Figure 1. Signaling cascades involved in glucocorticoid resistance.
Matheson et al. demonstrated, that selemutinib and dexamethasone co-treatment is highly synergic, both in the in vitro and in vivo studies. In vitro, selemutinib and dexamethasone co-treatment exerted a highly synergic impact (mean combination index (CI) of 0.1) on primagrafts with Ras-related mutations (affecting
NRAS,
KRAS, and
CBL/FLT3). Combined therapy resulted in elevation of BIM levels, a decreased Mcl-1 level, and ERK phosphorylation. In vivo, it was demonstrated that selumetinib and dexamethasone co-treatment acts highly synergic in nonobese diabetic (NOD) SCID (severe combined immunodeficiency) gamma mice engrafted with a Ras-mutated, patient-derived xenograft.
KRAS G13D,
KRAS G12D, and
NRAS Q61R mice treated with drug combination had a spleen weight at the end of the therapy comparable with the healthy mice spleen (
p < 0.001) [
122]. Polak et al. obtained blast samples from 22 adults with newly diagnosed pre-B ALL. These cells were incubated with dexamethasone or dexamethasone in combination with selumetinib. Combined therapy managed to augment the dexamethasone-induced apoptosis in 17 samples [
123].
Ras-activating mutations have been also found in the relapsed T-ALL patients and were associated with highly unfavorable outcomes [
89,
124,
125,
126,
127]. Kerstjens et al. treated
Ras mutant and
Ras wild-type t (4;11)
+ infant ALL blasts with salirasib (Ras inhibitor), vemurafenib (serine/threonine-protein kinase B-raf inhibitor), sorafenib (pan-kinase inhibitor), temsirolimus (mTOR inhibitor), and MAPK inhibitors trametinib, selumetinib and binimetinib. MAPK inhibitors were significantly more efficient than the other tested drugs [
128]. This is particularly important, as
Ras-mutated mixed lineage leukemia (MLL)-rearranged leukemic cells are likely to be resistant to glucocorticoid treatment [
129]. MLL-rearranged ALL is also associated with far worse clinical outcomes as compared to patients without MLL translocations [
130].
Combining JAK inhibitors with blockers of the downstream signaling also seems to be a promising strategy for the ALL treatment. Ba/F3 cells harboring JAK3(L857Q) and JAK3(M511I) mutations were treated with tofacitinib (JAK1/JAK3 inhibitor) and selumetinib at various concentrations. All combinations were characterized by synergic, dose-dependent action (CI < 0.1–0.9). A tofacitinib and venetoclax combination was also synergistic, however to a lesser extent. Co-treatment with tofacitinib and buparlisib (PI3K inhibitor) also provided mediocre effects. Furthermore, in the blasts harboring a
JAK3 (M511I) mutation, obtained from the T-ALL patient, the combination of tofacitinib and trametinib was highly synergic (CI from <0.1 to 1, depending on the drugs concentrations). Further, tofacitinib and venetoclax co-treatment at low concentrations was also effective (CI < 0.1). Oral treatment consisting of venetoclax (20 mg/kg/day) and ruxolitinib (40 mg/kg/day) was effective in treating mice engrafted with blasts carrying the
JAK3 (M511I) mutation [
131]. Inhibiting the JAK/STAT pathway, combined with blocking MAPK/ERK signaling, seems to be a rational strategy. MAPK/ERK can be also activated by
Ras-activating mutations, therefore blocking JAK/STAT signaling may not be enough to overcome resistance to the treatment [
118,
119,
120,
121,
122,
123]. Furthermore, combining JAK/STAT inhibitors with Bcl-2 inhibitors is also rational, as STAT proteins induce Bcl-2. Unfortunately, MAPK inhibitors have not been used in co-treatment with venetoclax in this study, as this combination could also be beneficial. Further, using all of these drugs simultaneously could hypothetically provide the best results, as it would target various mechanisms of treatment resistance. Thus, further preclinical studies are required.
Agents capable of blocking signaling pathways involved in the ALL glucocorticoid resistance have been systematized in Table 1.
Table 1. Targeting signaling pathways in ALL treatment.
| Drugs |
Mechanism of Action |
Preclinical Studies |
Completed Clinical Studies |
Ongoing Clinical Studies |
| MK2206 |
Allosteric AKT 1 inhibition |
MK2206 and dexamethasone co-treatment of resistant ALL 2 cell lines in vitro and in vivo [99] |
Not applicable |
Not applicable |
Pictilisib Idelalisib Buparlisib ZSTK-474 AS-605240 CAL-101 Duvelisib |
pan-PI3K 3 inhibition |
Pictilisib in T-ALL 4 treatment [102,103,104,105] Idelalisib treatment of T-cell leukemia-lymphoma samples [107] |
Not applicable |
Phase I, chimeric antigen receptor T-cell followed by duvelisib for ALL, LL 5, and lymphosarcoma (NCT05044039). Phase I, duvelisib for relapsing/remitting T-LL 7 |
Buparlisib treatment of T-ALL cell lines [108,109] ZSTK-474, AS-605240, CAL-101, and IPI-145 for the treatment of pre-B ALL 8 cell lines and ex vivo blasts from pre-B ALL patients [110] |
|
| Dactolisib |
pan-PI3K inhibition, mTOR 9 inhibition |
Dactolisib treatment of T-ALL and pre-B ALL cell lines [132] |
Phase I, relapsed/remitting ALL [115] |
Not applicable |
| Ruxolitinib |
JAK1/2 inhibition |
Ruxolitinib and dexamethasone co-treatment of T-ALL cells [90] Ruxolitinib and VXL 10 chemotherapy in mice engrafted with Ph-like 11 ALL [96] Ruxolitinib treatment of cell lines with IL-7 12-signaling, mediated steroid resistance. Ruxolitinib alone or in co-treatment with prednisolone in the T-ALL xenografts ex vivo [118] Venetoclax and ruxolitinib co-treatment in vivo in treating mice engrafted with blasts carrying JAK3 mutation [131] |
Phase II—part 1, ruxolitinib and consolidation chemotherapy for pediatric Ph-like ALL [133] |
Phase I, newly diagnosed pediatric Ph-like ALL (NCT03571321) Phase II—part 2, pediatric Ph-like ALL (NCT02723994) Phase II, relapsing/remitting Ph-like ALL (NCT02420717) Phase II/III, pediatric T-ALL/T-LL or pre-B ALL/B-LL 13 (NCT03117751) |
| CI1040 |
MAPK 14 inhibition |
CI1040 treatment of glucocorticoid-resistant cell lines [89] CI1040 treatment of cell lines with IL-7-signaling, mediated steroid resistance [118] |
Not applicable |
Not applicable |
Selumetinib Trametinib Binimetinib |
MAPK inhibition |
Selumetinib and trametinib treatment of cell lines with IL-7-signaling, mediated steroid resistance. Selumetinib, trametinib and binimetinib alone in the T-ALL xenografts ex vivo. Selumetinib in co-treatment with prednisolone alone in the T-ALL xenografts ex vivo [118] Selumetinib ex vivo treatment of pre-B ALL samples. Selumetinib in vivo treatment of xenografts with Ras 15 pathway mutant/wild-type ALL cells [120] Selumetinib in vivo and in vitro treatment of xenografts with Ras pathway mutant ALL cells [122] Selumetinib in vivo and in vitro treatment of xenografts with Ras pathway mutant/wild-type ALL cells [122] Selumetinib in co-treatment with dexamethasone for pre-B ALL samples ex vivo and in vitro pre-B ALL and T-ALL cell lines [123] Trametinib treatment ex vivo for pre-B ALL samples [121] Selumetinib, trametinib and binimetinib treatment of Ras mutated/wild-type MLL 16 rearranged ALL cell lines [128] Tofacitinib and MAPK inhibitors co-treatment for JAK 6-mutated ALL cell lines and ex vivo blasts from T-ALL patients [131] |
Not applicable |
Phase I/II, selumetinib for the relapsing/remitting ALL (NCT03705507) |
Table 2. BH3 mimetics in ALL treatment.
| Drug |
Preclinical Studies |
Completed Clinical Studies |
Ongoing Clinical Studies |
| Venetoclax |
Venetoclax treatment of T-ALL 1 and ETP ALL 2 cell lines/blasts from patients [143] Venetoclax treatment of T-ALL cell lines/blasts from patients [144] |
Series of cases, relapsing/remitting ALL 3 [145] Retrospective study, relapsing/remitting T-ALL [146] Phase I, relapsing/remitting ALL [147] |
Phase Ib-II, navitoclax and venetoclax co-treatment for pre-transplant and post-transplant treatment of adult T-ALL patients (NCT05054465) Phase I, adult pre-B ALL 4 (NCT05157971) Phase I/phase II, relapsing/remitting ALL (NCT03808610, NCT03504644, NCT03576547, NCT03319901, NCT04872790, NCT05016947, NCT03808610, NCT04752163, and NCT05149378) |
| Venetoclax and tofacitinib ex vivo co-treatment of JAK 5-mutated ALL cell lines and blasts from T-ALL patients [137] |
| Navitoclax |
Navitoclax treatment of T-ALL and ETP ALL cell lines/blasts from patients [143] |
Phase I, relapsing/remitting ALL [147] |
Phase Ib-II, navitoclax and venetoclax co-treatment for pre-transplant and post-transplant treatment of adult T-ALL patients (NCT05054465) |