Abstract
Oral azacitidine (oral-Aza) treatment results in longer median overall survival (OS) (24.7 vs. 14.8 months in placebo) in patients with acute myeloid leukemia (AML) in remission after intensive chemotherapy. The dosing schedule of oral-Aza (14 days/28-day cycle) allows for low exposure of Aza for an extended duration thereby facilitating a sustained therapeutic effect. However, the underlying mechanisms supporting the clinical impact of oral-Aza in maintenance therapy remain to be fully understood. In this preclinical work, we explore the mechanistic basis of oral-Aza/extended exposure to Aza through in vitro and in vivo modeling. In cell lines, extended exposure to Aza results in sustained DNMT1 loss, leading to durable hypomethylation, and gene expression changes. In mouse models, extended exposure to Aza, preferentially targets immature leukemic cells. In leukemic stem cell (LSC) models, the extended dose of Aza induces differentiation and depletes CD34+CD38- LSC. Mechanistically, LSC differentiation is driven in part by increased myeloperoxidase (MPO) expression. Inhibition of MPO activity either by using an MPO-specific inhibitor or blocking oxidative stress, a known mechanism of MPO, partly reverses the differentiation of LSC. Overall, our preclinical work reveals novel mechanistic insights into oral-Aza and its ability to target LSC.
Introduction
Acute myeloid leukemia (AML) is a disease of the elderly with a median age at diagnosis >68 years. Unfavorable cytogenetics and associated comorbidities are among the factors that make elderly patients unfit/ineligible for intensive chemotherapy, known as the 7+3 regimen.1-3 As a result, targeted and less intensive treatments are used as first line therapy for many of these patients. Venetoclax/ azacitidine (Ven/Aza) is one such treatment regimen that has improved outcomes in chemotherapy-ineligible newly diagnosed AML patients.4,5 The backbone of this regimen, Aza, is a hypomethylating agent that incorporates into RNA and DNA. DNA-incorporated Aza leads to loss of DNMT1 and therefore, genome-wide hypomethylation.6 Through hypomethylation-dependent/independent effects, Aza impacts multiple cellular processes such as apoptosis, DNA damage response, tumor suppressor re-expression, immune modulation, and more.7 Given the genomic and epigenomic dysregulation and heterogeneity in AML, the broad hypomethylating effect and multitude of mechanisms impacted by Aza could be one of the keys to its clinical efficacy. Venetoclax selectively inhibits the anti-apoptotic protein, BCL-2.8 On the other hand, many AML patients eligible for intensive chemotherapy achieve complete remission with the 7+3 regimen.3 However, most of these patients on chemotherapy as well as Ven/Aza therapy eventually relapse.9-11
A multicenter study identified longer relapse-free survival following first remission as one of the key parameters for better prognosis.12 Thus, better maintenance therapy is critical for AML patients. The QUAZAR AML-001 study demonstrated that maintenance therapy with oral-Aza significantly prolonged overall and relapse-free survival.13,14 Interestingly, single agent injectable-Aza did not provide a significant overall survival benefit in maintenance therapy in a similar patient population.15,16 The key translational question posed by this observation is, what specific mechanisms are associated with the oral formulation that may contribute to the observed clinical benefit in AML maintenance. In order to further explore this, we modeled injectable- and oral-Aza in our preclinical studies and explored the mechanism of action of oral-Aza. Equally important is to identify similarities between the two dosing regimens for potential future investigations of oral-Aza with agents that could combine well with injectable-Aza.
Methods
Human and mouse cell lines
AML cell lines were cultured in RPMI1640 (SKM1), Iscove’s Modified Dulbecco’s Medium (IMDM; OCI-AML2, OCI-AML20, MV411) or α-MEM (OP9, OCI-AML3, OCI-AML5) supplemented with 10% or 20% fetal bovine serum, sodium pyruvate, glutamine, and penicillin-streptomycin. OP9 cells were further supplemented with β-mercaptoethanol (β-ME) and OCI-AML20 with |3-ME and granulocyte-macrophage colony-stimulating factor (GM-CSF).
Flow cytometry
For OCI-AML20, CD33, and CD45, stains were used to distinguish OP9 cells (mouse) and OCI-AML20 cells (human). OCI-AML20 were then assessed for differentiation and changes in the leukemic stem cell (LSC) population with the following markers: live/dead stain, CD34 and CD38. Flow cytometry was performed in a BD LSRFortessaTM X-20.
Myeloperoxidase inhibition
OCI-AML20 cells were treated with Aza (conventional or extended dose) alone or in combination with 10 μM myeloperoxidase (MPO)-IN-28 (dosed on days 1 and 4). Flow analysis was done on day 7.
Cytokine array
OCI-AML20 cells were seeded at 0.5x106 cells/mL and treated with Aza, conventional or extended dosing. Supernatant from the cells was collected on days 3, 5, and 7. Cytokine array was performed using Proteome Profiler Human XL Cytokine Array Kit (biotechne, ARY022B), according to the manufacturer’s recommendation except that membranes were incubated with IRDye 800CW Streptavidin (Licor, 926-32230) to allow fluorescent detection of the cytokines. Membranes were imaged with Odyssey CLx (Licor).
Primary acute myeloid leukemia cell culture and CD34+ cells enrichment
Frozen primary patient samples (mutations in BCOR, BCORL1, CDKN2A, IKZF1, NOTCH1 RUNX1, SF3B1, STAG2, TET2, TP53) were quickly thawed and followed by CD34+ cells isolation (Stemcell Technologies, 17856) according to the manufacturer’s protocol. Flow cytometry was used to verify enrichment of the CD34+ population before treatment initiation. Cells were resuspended to a concentration of 0.5x106 cells/mL in X-VIVO 10 medium (Lonza, 04-380Q) supplemented with 20% BIT 9500 serum substitute (STEMCELL Technologies, 09500) and cytokines: interleukin-6 (IL-6) (10 ng/mL; PeproTech, 200-06), IL-3 (10 ng/mL; PeproTech, 200-03), stem cell factor (50 ng/mL; Peprotech, 300-07), FLT3-ligand (50 ng/mL; PeproTech, 300-19), granulocyte colony-stimulating factor (10 ng/mL; PeproTech, 300-23), and TPO (25 ng/mL; PeproTech, 300-18). The cells were dosed with Aza (extended or conventional) and flow analysis was performed on day 7 day with live/dead stain, CD34, and CD38 antibodies.
In vivo studies
For in vivo intrafemoral engraftment studies, we modeled injectable-Aza and oral-Aza doses with 3 mg/kg once daily for 5 days (QDx5) and 1 mg/kg QDx15, respectively (intraperitonially in both cases, modified from Vu et al.)13 Twelve-week female immunodeficient NOD.CB17-Prkdcscid/J (NOD-SCID)17 mice used for the transplantation of primary AML cells were obtained from the Ontario Cancer Institute (Toronto, Ontario, Canada). All animal studies were performed in accordance with the Ontario Cancer Institute Animal Use Protocol (AUP): # 1251.33 (NOD-SCID).
For the syngenic mouse model: C1498 (ATCC® TIB-49™) murine acute leukemia cells (American Tissue Culture Collection) expressing firefly luciferase and GFP (designated C1498-Luc3-GFP) were developed at Charles River Laboratories. Mice were injected with 1x106 C1498-Luc3-GFP tumor cells via the tail vein. Animals were randomized into treatment groups based on body weight on day 1 post-injection. All drug treatments were initiated on day 5 and were administered QD. Vehicle (phosphate-buffered saline [PBS]) and Aza were administered intraperitoneally. For the chimeric AML model: FLT3-internal tandem duplication (ITD), TET2, and LysM-cre mice were bred to generate a heterologous genotype of FLT3-ITD-/+TET2-/+LysM-cre-/+. Bone marrow chimeras were generated by irradiating mice in an X-ray irradiator at a lethal dose of 9 Gy. Bone marrow from FLT3-ITD-/+TET2-/+LysM-cre-/+ (CD45.2) was adoptively transferred via intravenous injection into 8-week-old CD45.1 congenic disparate recipient mice. Mice were monitored for chimerism after week 8 post-adoptive transfer. Aza was administered intraperitonially.
Results
Extended-dose Aza leads to durable hypomethylation and sustained gene expression independent of the integrated stress response pathway
While both injectable- and oral-Aza have the same active ingredient, their clinical dose and schedules are different, and they do not have similar bioavailability, pharmacokinetic (PK) or pharmacodynamic (PD) profiles.18,19 In a phase I study, compared with non-responders, patients who achieved clinical response with oral-Aza had greater hypomethylation as measured by global demethylation scores.19 This indicated that the dosing schedule might impact the hypomethylative effect of Aza. In order to test this hypothesis, we modeled injectable- and oralAza based on a recent study by Vu and colleagues.20 They modeled the clinical exposures of injectable- and oral-Aza using a “high exposure, limited duration” (HE-LD) regimen mimicking injectable-Aza and a “low exposure, extended duration” (LE-ED) mimicking oral-Aza hereafter referred to as conventional and extended dose respectively.
In order to understand the mechanistic basis of greater hypomethylation by oral-Aza, we treated three different AML cell lines with the in vitro modeling of the conventional- versus extended-dosing regimens. In these Aza-treated samples, genome-wide methylation was measured by whole-genome bisulfite sequencing and gene expression changes by RNA sequencing (RNA-seq) (Figure 1A) on days 3 and 5. While day 3, showed greater genome-wide hypomethylation in the conventional dose in two of the three cell lines (Figure 1B-D, upper panels), by the end of treatment (day 5), the extended-dose Aza regimen demonstrated durable hypomethylation in all three cell lines relative to the conventional Aza (P<0.001) (Figure 1B-D, lower panels, based on b score greater than 0.7; Online Supplementary Figure S1A; Online Supplementary Table S1). This suggested progressive hypomethylation in the extended dose but not in the conventional dose. Similarly, gene expression changes were greater/progressive in the extended dose at the end of treatment (day 5) (Figure 1E). Intrigued by the sustained effects of the low-dose Aza, we explored the status of DNMT1, which is degraded through entrapment by Aza thereby facilitating hypomethylation.6 Biochemical analysis revealed loss of DNMT1 (90% reduction) at early time points (Figure 1F). On day 7, DNMT1 levels recovered to nearly 90% with the conventional dose (Figure 1F) but not in the extended dose. Thus, the extended dose, albeit at lower exposures of Aza, facilitates prolonged loss of DNMT1 and thereby durable hypomethylation. Of note, while up to day 10, DNMT1 levels continue to be low in extended Aza treatment, there is about 20-50% cell death at these time points raising the possibility that at time points beyond day 7, cell death could contribute to loss of DNMT1 (data not shown).
In order to further understand the sustained hypomethylation of the extended-dose Aza, we analyzed the PD basis in an in vivo study measuring [14C] Aza incorporation in peripheral blood mononucelar cells (PBMC) after intraperitoneal administration to male C57BL/6 mice. These mice were treated with the two dosing regimens to mimic the conventional dose (3 mg/kg, QDx5) and extended dose (1 mg/kg, QDx15) regimens of Aza (Online Supplementary Figure S1B) with both regimens receiving the same cumulative dose of Aza. Results showed that the relative incorporation of Aza into DNA was greater in the extended dose based on relative area under the curve (AUC) (Figure 1G). Thus, the continued exposure of Aza resulted in a sustained therapeutic effect through greater nucleic acid incorporation thereby explaining the prolonged loss of DNMT1.
RNA incorporation of Aza is a potential mechanism through which, integrated stress response (ISR), one of the known mechanisms of action of Aza,21 is activated. In a recent study, this has been postulated as a mechanistic basis for synergy with venetoclax.22 In order to investigate if the extended dose also activates ISR, we treated MV-4-11, SKM-1, and AML-193 cell lines with 0.2 µM or 1 µM Aza in short-term (24 hours) experiments (Figure 1H). As expected, 1 µM Aza, mimicking conventional Aza, resulted in increased levels of eIF2α phosphorylation as well as ATF4, the core and effector of ISR, respectively.23 With the 0.2 µM dose, we observed slightly lower levels of phospho-eIF2α and ATF4. In addition, treatment with IS-RIB,24 a known inhibitor of ISR, reversed ATF4 expression without impacting DNMT1 loss. In order to investigate if ISR could be activated at later stages of the extended dose, we performed a time-course experiment over a period of 7 days (Figure 1I). While the extended dose was able to induce ATF4 expression, it did not result in activation of C/EBP Homologous Protein (CHOP), a downstream effector of ATF4 that leads to cell death25 which was observed in the 1 µM dose at 24 hours. These results indicate that while both the conventional and extended dosing trigger a stress response, the extended dose is not robust enough to mediate cell death through the ISR pathway.
Taken together, the above-stated observations indicate that while conventional-Aza works through activation of stress response and hypomethylation mediated changes, the extended dosing of oral-Aza results in sustained gene expression driven temporal effects that could be a key differentiating factor between injectable- and oral-Aza.
Extended dosing of Aza preferentially targets bone marrow blasts than conventional-Aza
In order to further investigate the therapeutic impact of the above-observed differences, we investigated the effect of the two dosing regimens in an in vivo syngeneic and patient-derived xenograft (PDX) model of AML. C1498-Luc3-GFP cells were injected into the tail vein of syngeneic mice which were then treated with the two dosing regimens of Aza. Ventral bioluminescence imaging (BLI) of luciferase-expressing tumor cells demonstrated that both regimens of Aza significantly decreased tumor burden at 15, 18 and 21-days post-injection (Online Supplementary Figure S2A). At day 25 post-injection, flow cytometry analysis demonstrated that both dosing regimens significantly decreased the percentage of GFP+ C1498 tumor cells in peripheral blood (Online Supplementary Figure S2B). Both Aza regimens similarly increased survival of tumor-bearing mice, compared to vehicle controls (Figure 2A) suggesting that both regimens are equally effective in reducing disease burden.
In order to further validate these in vivo observations, we used an IDH1R132H PDX AML model, CTG-2227.26 Cells were injected into the tail vein of sublethally irradiated, immunocompromised mice (NOG-EXL) and the animals were treated with the two dosing regimens. Tumor burden was assessed by measuring spleen weight at the end of the study. Similar to the syngenic model, both dosing regimens caused significant and comparable decreases in spleen weight compared to vehicle-treated animals (Figure 2B). Taken together, the syngeneic and PDX models indicate that despite the difference in dosing schedule, both the Aza dosing regimens are equally effective against bulk AML cells and contribute to lowering disease burden.
Given that oral-Aza is effective in maintenance therapy where leukemic stem/progenitor cells are one of the drivers of relapse, we tested the two dosing regimens in a chimeric AML model. Bone marrow from FLT3-ITD; TET2; LysM-cre compound heterozygous mice on a CD 45.2 background was adoptively transferred to CD45.1 wild-type (WT) recipient mice. Again, the extended-Aza dosing was as effective as the conventional dosing at resolving splenomegaly (Figure 2C), a strong indicator of AML progression; However, the extended-Aza dosing significantly decreased blasts in the bone marrow, whereas conventional dosing did not (Figure 2D). These data suggest that the extended dosing regimen more effectively controls bone marrow disease in this model.
Extended-dose Aza induces differentiation/elimination of leukemic stem cells
LSC are primitive hematopoietic cells related to the origin and relapse in AML.27,28 In order to investigate the impact of the oral-Aza/extended dosing on the LSC compartment, we utilized an in vitro LSC model, OCI-AML20.29 Extended-Aza dosing resulted in depletion of LSC (CD34+/CD38- or CD38 low) and concomitant enrichment of CD34+/CD38+ cells at day 7 (Figure 3A, representative flow plot; Online Supplementary Figure S3A, replicate data). While the conventional-Aza dosing also resulted in depletion of CD34+CD38-cells, the extended-Aza dosing had greater depletion of LSC in response to the extended-dose Aza. Notably, no overt cell death/apoptosis observed in these cells (Online Supplementary Figure S3B). In order to further validate the differentiation phenotype, we treated a CD34+ cell-enriched primary AML sample (TP53- and TET2-mutated) with both dosing regimens and identified differentiation of CD34+ cells to CD34- cells (Figure 3B; Online Supplementary Figure S3C). In order to further substantiate these observations, we performed single cell RNA-seq in OCI-AML20 cells at different time points (3, 5, and 7 days). Data were analyzed using a previously described classifier to identify leukemic myeloid cell lineages.30 Compared to untreated cells, the extended dosing resulted in an increase of granulocyte-monocyte progenitor (GMP) and myeloid cells indicating induction of differentiation in immature cells. While the conventional dosing also resulted in similar change in cell type composition, the extended-dosing regimen was associated with significantly greater differentiation compared with the conventional regimen (Figure 3C).
In order to test the effects of oral-Aza like dosing on leukemic progenitors in vivo, we leveraged a mouse xenograft model (NOD/SCID). Following injection of AML patient-derived PBMC into the right femur, the mice were treated with Aza regimens mimicking the injectable/conventional dose (3 mg/kg x5 days) or the oral/extended dose (1 mg/ kg x15/21 days) (Figure 4A). At day 42, animals were sacrificed, and primary engraftment was measured in the left femur/distal site. Both conventional and extended dosing reduced engraftment by about 10-fold relative to the control treatment (Figure 4B).
In order to further validate the differentiation phenotype observed with the extended dose, we analyzed the human PBMC that remained at the site of injection (right femur) in the primary engraftment experiment. Similar to the engraftment, the degree of differentiation induction on progenitors was similar in both regimens. Most of the cells in the sample tested were arrested in the GMP-like phase and both regimens of Aza induced the differentiation of >50% of these cells into a common myeloid progenitor (CMP) or megakaryocyte-erythrocyte progenitor (MEP) like phenotype (Figure 4C-E). There was also reduction of CD33+CD123+ stem/progenitor cells at the site of injection (Figure 4F). Notably, there was no overt loss of weight in the animals (Figure 4G).
Taken together, our data based on an in vitro LSC model suggest that the efficacy of oral-Aza in AML maintenance is in part linked to the ability of the extended dosing schedule to target LSC. In addition, the in vivo model demonstrates that oral-Aza induces differentiation of LSC and leukemic progenitor cells.
Extended-dose Aza induces differentiation through increased myeloperoxidase activity
In order to understand the molecular determinants of the greater degree of differentiation occurring in the LSC compartment in the extended dose, we sorted CD34+CD38- cells from OCI-AML20 cells (Figure 5A) and treated them with the two Aza dosing regimens and performed bulk RNA-seq at day 5. As expected, pathway analysis (GO process) revealed that the most significantly upregulated pathway was cellular differentiation in the CD34+CD38- cells upon 5 days of extended dosing but not in the conventional dosing (Figure 5B).
In order to investigate drivers of the upregulated cell differentiation pathway, we analyzed secreted factors from cell culture supernatants of OCI-AML20 cells treated with both the Aza-dosing regimens in a feeder layer-free culture using the Proteome Profiler™ antibody array. Among the differentially secreted factors, MPO, a hallmark enzyme of the myeloid lineage was the secreted factor that was also the most upregulated by gene expression analysis in LSC that were treated with the extended-dose Aza (Online Supplementary Figure S4A). In addition, western blot analysis confirmed that MPO is upregulated in OCI-AML20 cells as well as in CD34+ cells from a primary sample (TP53- and TET2-mutated) in response to the extended dose (Figure 5C). Given MPO’s association to myeloid differentiation, we hypothesized that upregulation of MPO could be implicated in extended Aza-induced LSC differentiation. In order to test this hypothesis, we used an inhibitor of MPO, MPO-IN-2831 and treated OCI-AML20 cells with MPO-IN-28 and the both the Aza doses. Inhibition of MPO countered the differentiation induction by extended-dose Aza (Figure 5D; Online Supplementary Figure S4B) by more than 20% reduction in the non-LSC population (CD34+CD38+ or CD34-). Being a peroxidase, MPO has been shown to induce reactive oxygen species (ROS) generation in myeloid cells,32 a potential mechanism that has been shown to induce differentiation of LSC.33,34 In order to investigate if MPO-induced ROS could facilitate differentiation in LSC, OCI-AML20 cells were concurrently treated with N-acetylcysteine (NAC) and the two Aza dosing regimens. ROS scavenging with NAC, blocked differentiation of LSC with the extended dose but not with the conventional dose further validating our hypothesis (Figure 5E; Online Supplementary Figure S4C). Taken together, our results suggest that extended dose Aza induces differentiation of LSC through upregulation of MPO and ROS.
Discussion
In AML maintenance therapy, the primary goal is to eliminate residual leukemic cells in the bone marrow thereby eliminating minimal residual disease (MRD) and reducing the risk of relapse. However, rare therapy-resistant cells that reside within the MRD, also known as LSC, can drive clonal leukemic repopulation resulting in relapse.28,35 Thus, understanding the biology of these LSC and identifying therapeutic agents that effectively target LSC during induction or maintenance therapy would be important in reducing rates of relapse in AML. While oral-Aza is effective in AML maintenance therapy, the mechanistic basis was not clear. Our preclinical work, for the first time demonstrates oral-Aza’s ability to target LSC.
Even though both injectable- and oral-Aza have the same active ingredient, the dosing schedule results in varying PK and PD profiles as well as therapeutic effects. Based on the preclinical data presented here, oral-Aza acts primarily through sustained hypomethylation-mediated gene expression changes and injectable-Aza acts through stress response as well as hypomethylation (not as sustained as oral-Aza). Consistent with our observations, a study with low-risk MDS patients reported sustained hypomethylation imparted by oral-Aza over the entire treatment cycle (up to 21 days). In addition, patients administered with oral-Aza and with resultant reduction in DNA methylation had better hematologic response.36 Similarly, sustained hypomethylation with oral-Aza were observed in another independent study.19
Given the biochemical differences observed (Figure 1) between the two dosing regimens, our in vivo studies yielded interesting similarities and differences (Figure 2) highlighting the functional impact of the two dosing regimens based on differentiation state of the cell. Both oral- and injectable-Aza were equally efficacious in targeting bulk AML cells in the syngeneic and the PDX model, both of which used bulk AML cells. However, when we looked at less differentiated bone marrow blast cells in the FLT3-ITD; TET2 model, the extended dose was effective in targeting this less differentiated population. When we applied this finding to an in vitro LSC model (OCI-AML20), we observed similar results and demonstrated that oral-Aza is more effective in eliminating immature myeloid cells and inducing differentiation of leukemic stem cells through upregulation of MPO. MPO, being a ROS generator, in turn likely acts through ROS to induce differentiation of LSC. Inhibition of MPO activity as well as ROS scavenging both rescue/block oral-Aza-mediated differentiation of LSC. Thus, a key understanding from our work is that the impact of oral-Aza is cell type specific.
Aza in combination with other agents has been shown to target LSC and induce differentiation.37,38 However, the specific Aza-associated mechanism was not clear. A key understanding from our work is the differential effect of oral-Aza on LSC versus non-LSC. In the in vitro model (OCI-AML20) enriched in LSC, the extended dose was more effective than the conventional dose. In order to recapitulate the in vitro findings, we performed engraftment studies of primary AML samples using bulk mononuclear cells which predominantly includes progenitors and blasts. The two dosing regimens performed similarly in reducing disease burden. Notably, conventional Aza is administered for 7 days in the clinic while our animal models received Aza only for 5 days based on our modeling. While we attempted to perform secondary engraftment, a measure of the capacity of re-initiation of leukemia by LSC when serially transplanted into mice, we did not achieve any engraftment in the conventional or extended-dose regimens even at the site of injection in the secondary mice likely because of the robust effect of both dosing regimens in the primary engraftment (data not shown). Thus, we could not leverage our transplant models to interrogate effects on LSC in vivo. Multiple studies have demonstrated that LSC rely on oxidative phosphorylation, a process that could generate more ROS and result in loss of stemness.33,39,40 However, LSC have developed unique mechanisms to maintain stemness by countering increased ROS such as increased antioxidant enzymes, increased levels of mitophagy and others.41-44 Thus, when oxidative stress/damage/signaling increases, stem cells tend to differentiate.45,46,33 MPO is a hallmark enzyme of the myeloid lineage and patients with high MPO have been shown to have superior overall survival47 and has been shown to be expressed at high levels in cells committed to granulomonocytic differentiation and activation of MPO facilitates oxidative damage.48 In addition, MPO has been shown to be increased in response to Aza in a few cases through promoter demethylation by DNMT-inhibitors.49,50 Our work uniquely demonstrates MPO upregulation in the LSC compartment upon Aza treatment which induce differentiation through oxidative stress as evidenced by a block in differentiation through ROS scavenging. A limitation of our study is that the cell lines and the LSC model that we have used are not truly representative of de novo AML and do not carry the common molecular mutations observed in elderly AML patients. To this end we attempted to replicate our key findings in commercially available multiple primary AML samples. However, we were successful in culturing CD34+ cells from only one primary sample (TET2- and TP53-mutated and not the commonly observed DNMT3A/NPM1/FLT3-ITD mutation) for 7 days and replicated our key findings.
Overall, we have highlighted mechanistic insights of oral-Aza that make it an effective maintenance therapy. Aza is known to have additional mechanisms of action beyond the ones described here including immune modulation, viral mimicry, and others. While we have primarily focused on tumor intrinsic mechanisms as part of this work, our data warrant exploring other mechanisms where Aza is implicated and identify further unique properties of oral-Aza.
Footnotes
- Received May 2, 2023
- Accepted October 30, 2023
Correspondence
Disclosures
DVJ, MA, AP, AR, PS, AA, KG, CK, DH, DLM, PRH and AG are current employees of Bristol Myers Squibb and received equity compensation. ADS has received research funding from Takeda Pharmaceuticals, BMS and Medivir AB, and consulting fees/honorarium from Takeda, Novartis, Jazz, and Otsuka Pharmaceuticals; and is named on a patent application for the use of DNT cells to treat AML. AGT and AA are former employees of BMS and received equity compensation. EFL received research funding from Celgene/BMS.
Contributions
DVJ, MA, AP, AR, PS, CK, DH, RH, XW, MG, AA, GT and HC designed and performed the research, analyzed data, and wrote the paper. AA, EFL, KG, DLM, ADS, PRH, AG and AGT designed the research, analyzed data and wrote the paper.
References
- Appelbaum FR, Gundacker H, Head DR. Age and acute myeloid leukemia. Blood. 2006; 107(9):3481-3485. Google Scholar
- Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020; 70(1):7-30. Google Scholar
- Pollyea DA, Bixby D, Perl A. NCCN guidelines insights: acute myeloid leukemia, Version 2.2021. J Natl Compr Canc Netw. 2021; 19(1):16-27. Google Scholar
- DiNardo CD, Wang J, Pratz KW. Azacitidine and venetoclax in AML. Reply. N Engl J Med. 2020; 383(21):2088-2089. Google Scholar
- Mustafa Ali MK, Corley EM, Alharthy H. Outcomes of newly diagnosed acute myeloid leukemia patients treated with hypomethylating agents with or without venetoclax: a Propensity Score-Adjusted Cohort Study. Front Oncol. 2022; 12:858202. Google Scholar
- Patel K, Dickson J, Din S. Targeting of 5-aza-2’-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme. Nucleic Acids Res. 2010; 38(13):4313-4324. Google Scholar
- Issa JP, Kantarjian H. Azacitidine. Nat Rev Drug Discov. 2005. Google Scholar
- Souers AJ, Leverson JD, Boghaert ER. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013; 19(2):202-208. Google Scholar
- Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015; 373(12):1136-1152. Google Scholar
- Lowenberg B, Ossenkoppele GJ, van Putten W. High-dose daunorubicin in older patients with acute myeloid leukemia. N Engl J Med. 2009; 361(13):1235-1248. Google Scholar
- Reville PK, Kadia TM. Maintenance therapy in AML. Front Oncol. 2020; 10:619085. Google Scholar
- Breems DA, Van Putten WL, Huijgens PC. Prognostic index for adult patients with acute myeloid leukemia in first relapse. J Clin Oncol. 2005; 23(9):1969-1978. Google Scholar
- Wei AH, Dohner H, Pocock C. Oral azacitidine maintenance therapy for acute myeloid leukemia in first remission. N Engl J Med. 2020; 383(26):2526-2537. Google Scholar
- Roboz GJ, Montesinos P, Selleslag D. Design of the randomized, phase III, QUAZAR AML Maintenance trial of CC-486 (oral azacitidine) maintenance therapy in acute myeloid leukemia. Future Oncol. 2016; 12(3):293-302. Google Scholar
- Burnett A, Russell N, Freeman S. A comparison of limited consolidation chemotherapy therapy or not, and demethylation maintenance or not in older patients with Aml and high risk Mds: long term results of the Uk Ncri Aml16 Trial. EHA Library. 2015; 103225:S513. Google Scholar
- Huls G, Chitu DA, Havelange V. Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood. 2019; 133(13):1457-1464. Google Scholar
- Nicolini FE, Cashman JD, Hogge DE. NOD/SCID mice engineered to express human IL-3, GM-CSF and Steel factor constitutively mobilize engrafted human progenitors and compromise human stem cell regeneration. Leukemia. 2004; 18(2):341-347. Google Scholar
- Garcia-Manero G, Gore SD, Cogle C. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. J Clin Oncol. 2011; 29(18):2521-2527. Google Scholar
- Laille E, Shi T, Garcia-Manero G. Pharmacokinetics and pharmacodynamics with extended dosing of CC-486 in patients with hematologic malignancies. PLoS One. 2015; 10(8):e0135520. Google Scholar
- Vu T, Straube J, Porter AH. Hematopoietic stem and progenitor cell-restricted Cdx2 expression induces transformation to myelodysplasia and acute leukemia. Nat Commun. 2020; 11(1):3021. Google Scholar
- Lee TT, Karon MR. Inhibition of protein synthesis in 5-azacytidine-treated HeLa cells. Biochem Pharmacol. 1976; 25(15):1737-1742. Google Scholar
- Jin S, Cojocari D, Purkal JJ. 5-Azacitidine induces NOXA to prime AML cells for venetoclax-mediated apoptosis. Clin Cancer Res. 2020; 26(13):3371-3383. Google Scholar
- Costa-Mattioli M, Walter P. The integrated stress response: from mechanism to disease. Science. 2020; 368(6489):eaat5314. Google Scholar
- Sidrauski C, Tsai JC, Kampmann M. Pharmacological dimerization and activation of the exchange factor eIF2B antagonizes the integrated stress response. Elife. 2015; 4:e07314. Google Scholar
- Zinszner H, Kuroda M, Wang X. CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 1998; 12(7):982-995. Google Scholar
- Gruber E, So J, Lewis AC. Inhibition of mutant IDH1 promotes cycling of acute myeloid leukemia stem cells. Cell Rep. 2022; 40(7):111182. Google Scholar
- Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997; 3(7):730-737. Google Scholar
- Shlush LI, Mitchell A, Heisler L. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 2017; 547(7661):104-108. Google Scholar
- Luciani GM, Xie L, Dilworth D. Characterization of inv(3) cell line OCI-AML-20 with stroma-dependent CD34 expression. Exp Hematol. 2019; 69:27-36. Google Scholar
- van Galen P, Hovestadt V, Wadsworth Ii MH. Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019; 176(6):1265-1281. Google Scholar
- Soubhye J, Chikh Alard I, Aldib I. Discovery of novel potent reversible and irreversible myeloperoxidase inhibitors using virtual screening procedure. J Med Chem. 2017; 60(15):6563-6586. Google Scholar
- Nakazato T, Sagawa M, Yamato K. Myeloperoxidase is a key regulator of oxidative stress mediated apoptosis in myeloid leukemic cells. Clin Cancer Res. 2007; 13(18 Pt 1):5436-5445. Google Scholar
- Lagadinou ED, Sach A, Callahan K. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 2013; 12(3):329-341. Google Scholar
- Owusu-Ansah E, Banerjee U. Reactive oxygen species prime Drosophila haematopoietic progenitors for differentiation. Nature. 2009; 461(7263):537-541. Google Scholar
- Ng SW, Mitchell A, Kennedy JA. A 17-gene stemness score for rapid determination of risk in acute leukaemia. Nature. 2016; 540(7633):433-437. Google Scholar
- Garcia-Manero G, Gore SD, Kambhampati S. Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes. Leukemia. 2016; 30(4):889-896. Google Scholar
- MacBeth KJ, Chopra VS, Tang L. Combination of azacitidine and enasidenib enhances leukemic cell differentiation and cooperatively hypomethylates DNA. Exp Hematol. 2021; 98:47-52. Google Scholar
- Pollyea DA, Stevens BM, Jones CL. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018; 24(12):1859-1866. Google Scholar
- Sriskanthadevan S, Jeyaraju DV, Chung TE. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood. 2015; 125(13):2120-2130. Google Scholar
- Culp-Hill R, D’Alessandro A, Pietras EM. Extinguishing the embers: targeting AML metabolism. Trends Mol Med. 2021; 27(4):332-344. Google Scholar
- Pei S, Minhajuddin M, Adane B. AMPK/FIS1-mediated mitophagy is required for self-renewal of human AML stem cells. Cell Stem Cell. 2018; 23(1):86-100. Google Scholar
- Tothova Z, Gilliland DG. FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell. 2007; 1(2):140-152. Google Scholar
- Aloum L, Brimson CA, Zhyvoloup A. Coenzyme A and protein CoAlation levels are regulated in response to oxidative stress and during morphogenesis in Dictyostelium discoideum. Biochem Biophys Res Commun. 2019; 511(2):294-299. Google Scholar
- Folkerts H, Hilgendorf S, Wierenga ATJ. Inhibition of autophagy as a treatment strategy for p53 wild-type acute myeloid leukemia. Cell Death Dis. 2017; 8(7):e2927. Google Scholar
- Diehn M, Cho RW, Lobo NA. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009; 458(7239):780-783. Google Scholar
- Kim YS, Kang MJ, Cho YM. Low production of reactive oxygen species and high DNA repair: mechanism of radioresistance of prostate cancer stem cells. Anticancer Res. 2013; 33(10):4469-4474. Google Scholar
- Matsuo T, Kuriyama K, Miyazaki Y. The percentage of myeloperoxidase-positive blast cells is a strong independent prognostic factor in acute myeloid leukemia, even in the patients with normal karyotype. Leukemia. 2003; 17(8):1538-1543. Google Scholar
- Yu G, Liang Y, Zheng S. Inhibition of myeloperoxidase by N-Acetyl lysyltyrosylcysteine amide reduces oxidative stress-mediated inflammation, neuronal damage, and neural stem cell injury in a murine model of stroke. J Pharmacol Exp Ther. 2018; 364(2):311-322. Google Scholar
- Aumer T, Gremmelmaier CB, Runtsch LS. Comprehensive comparison between azacytidine and decitabine treatment in an acute myeloid leukemia cell line. Clin Epigenetics. 2022; 14(1):113. Google Scholar
- Leung KK, Nguyen A, Shi T. Multiomics of azacitidine-treated AML cells reveals variable and convergent targets that remodel the cell-surface proteome. Proc Natl Acad Sci U S A. 2019; 116(2):695-700. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.