Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematologic
neoplasm with poor survival outcomes. However, the routine clinical
features are not sufficient to accurately predict the prognosis of AML.
The expression of hypoxia-related genes was associated with survival
outcomes of a variety of hematologic and lymphoid neoplasms. We
established an 18-gene signature-based hypoxia-related prognosis model
(HPM) and a complex model that consisted of the HPM and clinical risk
factors using machine learning methods. Both two models were able to
effectively predict the survival of AML patients, which might
contribute to improving risk classification. Differentially expressed
genes analysis, Gene Ontology (GO) categories, and Kyoto Encyclopedia
of Genes and Genomes (KEGG) pathway enrichment analysis were performed
to reveal the underlying functions and pathways implicated in AML
development. To explore hypoxia-related changes in the bone marrow
immune microenvironment, we used CIBERSORT to calculate and compare the
proportion of 22 immune cells between the two groups with high and low
hypoxia-risk scores. Enrichment analysis and immune cell composition
analysis indicated that the biological processes and molecular
functions of drug metabolism, angiogenesis, and immune cell
infiltration of bone marrow play a role in the occurrence and
development of AML, which might help us to evaluate several
hypoxia-related metabolic and immune targets for AML therapy.
Keywords: acute myeloid leukemia, prognostic model, hypoxia,
metabolism, immunity, bone marrow microenvironment
Introduction
Acute myeloid leukemia (AML) is a clonal malignant aggressive
hematological tumor, resulting in the accumulation of acquired
chromosomal, genetic, and epigenetic abnormalities in highly
heterogeneous myeloid precursors. It is the most common acute leukemia
and accounts for approximately 80% of cases in adults. In the United
States, the age-adjusted incidence of AML is 4.3 per 100,000 population
annually ([40]Shallis et al., 2019), which has a high mortality rate
and variable prognosis. In recent years, the incidence rate of AML is
getting increasingly serious and poses an enormous threat to human
health. In the research of AML, there remain several challenges,
advances in treatment for AML have remained quite limited, and the
current prognostic evaluation system cannot completely distinguish the
prognosis of AML patients. One of the hotspots and critical points for
medical research is to identify specific prognostic factors that may
help predict the outcomes.
Hypoxia is a common condition in the solid-tumor microenvironment
([41]Kim et al., 2007), playing an important role in various biological
processes, such as metabolic alteration, angiogenesis, and metastasis
([42]Cosse and Michiels, 2008; [43]Godet et al., 2019). However, the
role of the pathophysiological implications of hypoxia in AML remains
controversial, and the mechanism is still not clear. In bone marrow
(BM), the low oxygen partial pressure (pO[2]) is physiological
([44]Harrison et al., 2002). And the hypoxic microenvironmental niches
within leukemic BM compared with those of the normal BM were expanded,
accompanied by leukemia stem cell (LSC) proliferation ([45]Keith and
Simon, 2007; [46]Benito et al., 2011; [47]Benito et al., 2013; [48]Zhou
et al., 2016). The hypoxic BM microenvironment has also been shown to
contribute to acute leukemic progression, resistance to chemotherapy,
and minimal residual disease (MRD) ([49]Frolova et al., 2012;
[50]Matsunaga et al., 2012; [51]Tabe and Konopleva, 2015). In response
to hypoxia, cells change their hypoxia-related gene expression, which
was proved to be correlated with prognosis for various solid tumors.
However, the European LeukemiaNet (ELN) 2017 risk classification (ELN
2017) ([52]Dohner et al., 2017), an important AML risk stratification
standard that has been widely used to estimate prognosis of AML, is
based on cytogenetic and molecular features. Mutations in the FMS-like
tyrosine kinase 3 gene (FLT3-ITD) are quite common in AML and have been
associated with poorer overall survival (OS) ([53]Kayser et al., 2009).
Nucleophosmin (NPM1) gene mutations have been associated with improved
outcomes in patients with AML ([54]Becker et al., 2010). Mutations of
the CCAAT/enhancer binding protein alpha (CEBPA) gene have been
associated with a favorable outcome in patients with AML, but mainly in
those patients with cytogenetically normal AML ([55]Renneville et al.,
2009; [56]Rockova et al., 2011). Rare studies to date have developed a
hypoxia-related prognosis model (HPM) of AML based on gene expression
profiles. AML-suitable hypoxia gene signatures still need to be
developed.
To evaluate the potential utility of hypoxia-related gene expression
profiles in AML prognosis, The Cancer Genome Atlas (TCGA) and MsigDB
databases were analyzed, and the clinical features of patients were
considered to construct an 18-gene-based hypoxia risk classifier. The
model could be useful for the prognostic evaluations and development of
novel therapeutic modalities aimed at interfering with hypoxia-sensing
pathways and modifying the hematopoietic microenvironment.
Materials and Methods
Acquiring and Pre-Processing of Sample Data and Primary Screening of Acute
Myeloid Leukemia Hypoxia-Related Genes
In this study, three public accessible transcriptome datasets of
BEATAML1.0 (Cohort 1,
[57]https://portal.gdc.cancer.gov/projects/BEATAML1.0-COHORT;
[58]Supplementary Data Sheet S1), TARGET-AML (Cohort 2,
[59]https://portal.gdc.cancer.gov/projects/TARGET-AML;
[60]Supplementary Data Sheet S2), and TCGA-LAML (Cohort 3,
[61]https://portal.gdc.cancer.gov/projects/TCGA-LAML; [62]Supplementary
Data Sheet S3) were used throughout the training and validation stages.
The latest clinical follow-up information was also obtained from Vizome
([63]http://www.vizome.org/; [64]Supplementary Data Sheet S4)
([65]Tyner et al., 2018) or TCGA database
([66]https://cancergenome.nih.gov/; [67]Supplementary Data Sheet S5,
S6). A total of 315 hypoxia-related genes defined in the Molecular
Signatures Database ([68]http://www.gsea-msigdb.org/gsea/msigdb/;
[69]Supplementary Table S1) were used as the initial candidates for Cox
and least absolute shrinkage and selection operator (LASSO) survival
analysis. We applied strict quality control (QC) for these datasets on
sample and gene levels, respectively. On the sample level, we removed
patients diagnosed with myelodysplastic syndromes (MDSs),
myeloproliferative neoplasms (MPNs), or other non-AML diseases; we also
filtered out individuals with no survival information. On the gene
level QC, we kept genes having expression information in all three
transcriptome datasets; genes with low expression quantity in all
samples [reads per kilobase of transcript per million mapped reads
(RPKM) < 1] were removed from downstream analysis. The principal
component analysis (PCA) was also performed to identify the outlier; we
removed individuals who deviated from the study samples. The ComBat
method was used to correct the potential batch effects of RNA
sequencing ([70]Supplementary Figure S1). At this point, a total of 419
samples in Cohort 1, 156 samples in Cohort 2, and 151 samples in Cohort
3 (315 hypoxia-related genes) were kept for survival analysis
([71]Supplementary Data Sheet S7–S9).
Identification of Hypoxia-Related Signatures and Establishment and
Verification of a Hypoxia Risk Score Model
Univariate Cox proportional hazards regression was first used to
preliminarily screen the AML prognostic genes (p < 0.05). Next, Cohort
1 was randomly divided into a training set of 293 cases and a test set
of 126 cases (7:3 ratio). To narrow down the prognostic genes for
prediction, a Cox proportional hazards regression model combined with
the LASSO ([72]Goeman, 2010) using the “glmnet” package was applied to
select the most important hypoxia-related signatures, and the optimal
values of the penalty parameter λ were determined by 10-fold
cross-validations at which the minimal mean squared error (MSE) is
achieved in the training set ([73]Simon et al., 2011). Afterward, the
multivariate Cox regression analysis was performed to estimate
independent prognostic factors associated with patient survival.
Finally, the stepwise method was employed to select the best subset of
predictors in a risk score model. To this, a hypoxia-related prognostic
risk (HRS) score model was built, with the regression coefficients (β)
weighted by the multivariate Cox proportional hazards regression model
in the training set. The HRS model formula was as follows:
[MATH:
Hypoxia
risk s<
/mi>core=∑i=1n
munderover>(βi
∗ xi)<
/mo> :MATH]
β[i] are coefficients (β) weighted by the multivariate Cox proportional
hazards regression model, and x[i] is the RNA expression level.
We calculate the HRS of the study samples and use the median HRS of the
training group as the cutoff point to label the low-risk or high-risk
individual of the three cohorts. The trained model was then tested
using the test and validation sets (other independent cohorts: Cohort 2
and Cohort 3). The “survminer” package was used for the Kaplan–Meier
(KM) survival analysis of patients in the high-risk and low-risk
groups, while the “timeROC” package was used to construct
time-dependent receiver operating characteristic (ROC) curves and
calculate the area under the ROC curve (AUC) at 1-, 3-, and 5-years OS.
Construction and Evaluation of the Nomogram Model
We explored the relationship between HPM and other clinical parameters
for AML patient outcomes. Univariable Cox analysis and multivariate Cox
analysis were performed with all patients’ clinical covariates in the
BEATAML1.0 cohort by the “rms” package. Samples with incomplete data
about potential prognostic factors were excluded from the multivariable
Cox analyses. Pearson’s correlation between HPM and different clinical
characteristics was calculated by “stats” package and plotted by
“corplot” package. A nomogram was formulated using the “rms” r package
based on the results of the multivariate analyses; and calibration
plots and time-dependent ROC plots were performed to assess the
prognostic accuracy of the nomogram. The predicted outcomes and
observed outcomes of the nomogram were plotted in the calibration curve
to evaluate the degree of fitting of the nomogram, and the 45° line
represented the best prediction.
Biological Phenotypes Associated With the Hypoxia Risk Score Model
Hypoxia-Related Metabolic Alterations
We procured 3,695 human metabolic genes concerning 145 metabolic
subsystems from the Recon 3D ([74]http://vmh.life) ([75]Brunk et al.,
2018). Among them, 3,224 metabolic genes were matched to our Cohort 1
data. The empirical Bayes algorithm of the R package “limma”
([76]Ritchie et al., 2015) was used to identify differentially
expressed genes (DEGs) between the top 25% samples of the high- and
low-risk groups (totally 210 samples). All gene expression values were
log2 transformed to identify the metabolic genes with significant
differential expression during hypoxia stress [logarithmically
transformed fold change (log2(FC)) ≥ 1 or (log2(FC)) ≤ −1 and p-value <
0.05]. Besides, we performed Kyoto Encyclopedia of Genes and Genomes
(KEGG) and Gene Ontology (GO) enrichment analyses of DEGs with
clusterProfiler package ([77]Yu et al., 2012). KEGG pathway enrichment
analysis utilized the KEGG database ([78]http://www.genome.jp/kegg)
while GO enrichment was utilized ([79]http://www.geneontology.org).
Hypoxia-Related Immune Alterations
Regarding the association between the hypoxia risk score and immune
cells in the BM microenvironment, CIBERSORT algorithm ([80]Newman et
al., 2015) was used to estimate the relative immune cell fractions in
the BM samples of Cohort 1, based on the standard LM22 leukocyte
signature matrix that distinguishes 22 immune cell subtypes and 1,000
permutations (CIBERSORT R script v1.03 is available on
[81]http://cibersort.stanford.edu/). We performed the following
analyses in CIBERSORT: B cells, CD4^+ T cells, CD8^+ T cells, Tregs, NK
T cells, γδ-T cells, lymphocytes, and macrophages. Further, xCell
([82]http://xcell.ucsf.edu/) was used to validate the result.
Result
A flowchart overviewing the procedures of this study is presented in
[83]Figure 1.
FIGURE 1.
[84]FIGURE 1
[85]Open in a new tab
Flowchart of data collection, modeling, and further analysis.
Patient Clinical Characteristics
We analyzed clinical characteristics of patients with AML from Cohort
1, including age, gender, ELN 2017, the mutations of NPM1 and FLT3, and
CEBPA Biallelic status ([86]Table 1). Differences in general clinical
information of two sets are not statistically significant. Among the
419 AML patients with complete clinical information in BEATAML1.0, the
mean diagnosis age was 56.22 ± 18.23 years, while the proportion of
males was 55.61% (233/419).
TABLE 1.
Patients’ basic characteristics in BEATAML1.0 cohort.
Total n = 419 Training set n = 293 Test set n = 126 p-value
Age (mean (SD)) (%) 56.22 (18.25) 55.98 (18.29) 56.77 (18.23) 0.685
<65 256 (61.1) 185 (63.1) 71 (56.35) 0.231
≥65 163 (38.9) 108 (36.9) 55 (43.7)
Gender (%) 0.582
Female 186 (44.4) 127 (43.3) 59 (46.8)
Male 233 (55.6) 166 (56.7) 67 (53.2)
ELN 2017 (%) 0.846
Favorable 108 (25.8) 77 (26.3) 31 (24.6)
Intermediate 140 (33.4) 93 (31.7) 47 (37.3)
Adverse 149 (35.6) 108 (36.9) 41 (32.5)
Favorable or intermediate 14 (3.3) 9 (3.1) 5 (4.0)
Intermediate or adverse 7 (1.7) 5 (1.7) 2 (1.6)
Not available 1 (0.2) 1 (0.3) 0 (0.0)
NPM1 (%) 0.439
Negative 309 (73.7) 213 (72.7) 96 (76.2)
Positive 107 (25.5) 77 (26.3) 30 (23.8)
Not available 3 (0.7) 3 (1.0) 0 (0.0)
FLT3-ITD (%) 0.233
Negative 321 (76.6) 218 (74.4) 103 (81.7)
Positive 97 (23.2) 74 (25.3) 23 (18.3)
Not available 1 (0.2) 1 (0.3) 0 (0.0)
CEBPA Biallelic (%) 1
No 412 (98.3) 124 (98.4) 288 (98.3)
Yes 7 (1.7) 2 (1.6) 5 (1.7)
[87]Open in a new tab
Establishment of Hypoxia Risk Score Model
A univariate Cox regression was performed to identify prognostic
hypoxia-related genes associated with OS in BEATAML1.0 dataset. A total
of 33 genes (ALDH1A1, ALDOC, BACE2, BATF3, CA9, CALD1, COL5A1, DR1,
EGLN3, ELOB, HBP1, HK1, ID2, KRT14, LRP8, NOS1, NOS2, PDK3, PLOD2,
PSMA2, PSMA7, PSMB6, PSMC1, PSMC4, PTGS1, RPS27A, SIAH2, SLC16A1,
SORL1, TGM2, THBS1, TPD52, and UBA52) were identified to have a
significant prognostic value in patients with AML (p < 0.05). Then,
LASSO-penalized Cox analysis with 10-fold cross-validation ([88]Figures
2A,B) was performed for further screening, and 23 genes were left.
Finally, a total of 18 hub genes (ALDOC, BATF3, COL5A1, DR1, ELOB,
HBP1, HK1, KRT14, NOS2, PSMA2, PSMA7, PSMB6, PSMC1, PTGS1, SIAH2,
SORL1, THBS1, and UBA52) were identified from the stepwise multivariate
Cox regression ([89]Figure 2C), and the formula to calculate the
hypoxia risk score was as follows:
[MATH: Hypoxia risk score=0
mtext>.2737×ALDOC+<
/mo>0.1343×BATF<
/mtext>3×0.0762×COL5A1+<
/mo>0.2795×DR1+0.6469<
mo>×ELOB-0.<
mn>5492×HBP1-0.4513×HK1
-0.1064×KRT14+0
.0740×NOS2
+0.3421×PSM
A2+0.8385
mn>×PSMA7-1.<
/mtext>1489×PSMB6+0.4238×PSMC1+0.1434×
PTGS1+0.3213×SIAH2-0.1447×SORL<
mn>1+0.0994×
mo>THBS1-
0.4732×UBA5
2 :MATH]
FIGURE 2.
[90]FIGURE 2
[91]Open in a new tab
(A) Tenfold cross-validation for tuning parameter selection in the
LASSO model. (B) Least absolute shrinkage and selection operator
(LASSO) coefficient profiles of the 33 prognostic genes. (C) Forest
plot of 18 hypoxia-related genes significantly associated with overall
survival according to multivariate Cox regression analysis.
Prognostic Value of the Hypoxia Risk Score
The KM survival curves revealed that patients in the high-risk group
exhibited a significantly lower OS rate than the low-risk group in all
cohorts (training set, test set, Cohort 2, and Cohort 3; p < 0.0001, p
= 0.04, p < 0.001, and p < 0.01, respectively; [92]Figure 3). In the
training dataset of BEATAML1.0 Cohort, the median OS of low-risk
patients was 1.970 years (95% CI: 1.734–3.775), whereas the median OS
of high-risk patients was 0.718 years (95% CI: 0.575–0.912), and the HR
is 3.261 (95% CI: 2.398–4.435). In comparison, in the test dataset of
BEATAML1.0 Cohort (validation Cohort 1), the median OS of patients with
low-risk scores was 1.592 years (95% CI: 1.230–NA), and the median OS
of patients with high-risk scores was 0.978 years (95% CI:
0.860–1.556), while HR is 1.626 (95% CI: 1.007–2.626). In TARGET-AML
cohort (validation Cohort 2), the median OS of patients with low-risk
scores was NR (not reached), and the median OS of patients with
high-risk scores was 2.227 years (95% CI: 1.689–5.195), and the HR is
2.283 (95% CI: 1.455–3.582). In TCGA-LAML cohort (validation Cohort 3),
the median OS of patients with low-risk scores was 2.170 years (95% CI:
1.581–3.838), the median OS of patients with high-risk scores was
0.833 years (95% CI: 0.586–1.003), and the HR is 1.773 (95% CI:
1.190–2.642). Furthermore, the prognostic accuracy of the hypoxia risk
score was assessed with time-dependent ROC analysis for OS at 1, 3, and
5 years in all datasets, and the results were as follows. In the
training set, the AUC was 0.813 at 1 year, 0.788 at 3 years, and 0.899
at 5 years ([93]Figure 3E); 0.675 at 1 year, 0.767 at 3 years, and
0.753 at 5 years ([94]Figure 3F) in the test set; 0.616 at 1 year,
0.684 at 3 years, and 0.690 at 5 years ([95]Figure 3G) in Cohort 2; and
0.712 at 1 year, 0.657 at 3 years, and 0.640 at 5 years ([96]Figure 3H)
in Cohort 3, which revealed that the hypoxia risk score was a valuable
predictor. We next studied whether the HRS could improve prognostic
assessment in AML patients on the 2017 ELN genetic risk stratification
([97]Dohner et al., 2017) basis. In each ELN2017 stratification, the
HPM high-risk group had a poorer prognosis than the HPM low-risk group
([98]Figure 3I). Furthermore, we found that 37 out of 108 ELN2017
favorable patients (51.7%) were high-risk for the HPM and had
significantly worse survival. In this patient subgroup (ELN
favorable–HPM high), representing 9.3% (37/397, only contains patients
who had clear ELN2017 stratification) of the BEATAML1.0 cohort, their
survival was similar to that of ELN intermediate/adverse-risk patients
with HPM low-risk. To further evaluate the performance of the HPM, we
compared our HPM with other gene expression based AML prognostic
models, which were published within the last 5 years, including
PMID29138577 ([99]Huang et al., 2017), PMID32268820 ([100]Zhang and
Xiao, 2020), PMID29956722 ([101]Zhao et al., 2018), and PMID34282207
([102]Jiang et al., 2021). Risk score was calculated based on formulas
from the corresponding literatures. We used the p-value of the KM
survival analysis to reflect the discrimination and the AUC value to
evaluate the accuracy. The survival analysis showed that the HPM was
significantly correlated with the survival of the patients in all three
datasets. In contrast, other models could only perform well in at most
two of the three datasets ([103]Supplementary Figure S2). The AUC
values of the HPM were more stable, which means a wider range of
suitable population ([104]Supplementary Figure S3; [105]Supplementary
Table S2).
FIGURE 3.
FIGURE 3
[106]Open in a new tab
Prognostic value of the hypoxia risk score Kaplan–Meier survival curves
of overall survival (OS) between the low- and high-risk group patients
in all three cohorts. (A) Training dataset of Cohort 1 and (B) test
dataset of Cohort 1, (C) Cohort 2, and (D) Cohort 3. ROC curves of the
hypoxia risk score model based on the 18 characteristic genes. (E)
Training dataset of Cohort 1 and (F) test dataset of Cohort 1, (G)
Cohort 2, and (H) Cohort 3. ROC, receiver operating characteristic;
AUC, area under the curve. (I) Comparison of survival between six
different ELN2017 and hypoxia-related prognosis model (HPM) risk
subgroups of patients.
Hypoxia-Related Characteristics of High- and Low-Risk Patients, Based on the
Prognostic Risk Score Model
Association Between the Hypoxia Risk Score and the Clinical Characteristics
We ranked the risk scores of patients in the training and test sets,
and the distributions associated with gene expression, survival time,
and status are shown in [107]Figures 4A,B. Hypoxia risk score increased
with higher age, worse 2017 ELN genetic risk stratification, NPM1 wild
type, and FLT3-ITD wild type and have no CEBPA double mutation
([108]Figure 4C).
FIGURE 4.
[109]FIGURE 4
[110]Open in a new tab
Risk score distributions, genes expression, survival time, and status
profiles in the (A) training and (B) test set. (C) Hypoxia risk score
group by stratification factors.
Hypoxia-Related Prognosis Model-Related Metabolic Alterations
To study the relationship between HRS and metabolic flux, we performed
differential gene expression analysis using “limma” package, 115 DEGs
were shortlisted from the raw dataset, and 93 genes were upregulated in
the high-risk group while 22 genes were downregulated. Results were
visualized into volcano plot ([111]Figure 5A) and heatmap ([112]Figure
5B). To evaluate the molecular mechanisms of DEGs, KEGG metabolic
pathways enrichment analyses, and GO functional annotation were
conducted ([113]Table 2; [114]Figure 5). Unsurprisingly, pathways
associated with oxidoreduction activity were enriched. Unexpectedly, we
found 12 immune-related pathways such as “leukocyte mediated immunity”
and “cell activation involved in immune response,” which were
significantly overrepresented. Besides, two pathways of angiogenesis
were significantly enriched in the upregulated genes for the high-risk
group. And the enrichment of “heme binding” and “tetrapyrrole binding”
might be associated with oxygen-carrying capacity. Moreover, drug
catabolic process pathways were markedly enriched, such as “xenobiotic
metabolic process,” “drug catabolic process,” “Drug metabolism—other
enzymes,” “Drug metabolism—cytochrome P450,” and “Metabolism of
xenobiotics by cytochrome P450.” Moreover, the enrichment of the
pathway “Chemical carcinogenesis” suggests different disease
susceptibility between the high- and low-risk groups.
FIGURE 5.
FIGURE 5
[115]Open in a new tab
Differential gene expression analysis of metabolic genes. And Kyoto
Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO)
analysis of differentially expressed genes (DEGs). (A) Volcano plot
showing DEGs between the top 25% samples of the high- and low-risk
groups. Each dot represents one gene. Red dot represents upregulated
gene (log2 (fold change) > 1 and p-value < 0.05). Blue dot represents
downregulated gene (log2 (fold change) < −1 and p-value < 0.05). And
black dot represents non-differentially expressed gene. (B) Heatmap of
DEGs associated with hypoxia-related prognosis model (HPM) risk group.
(C) Bubble graph of the enrichment KEGG pathways for the downregulated
genes (there are no significantly enriched KEGG pathways for
upregulated genes). (D) Enrichment map of the enrichment KEGG pathways
for the downregulated genes showing the association between different
pathways. (E) Heatmap of the enrichment KEGG pathways for the
downregulated genes, showing the association between pathways and
genes. (F) Lollipop plot of GO term enrichment for Biological Process,
Cellular Component, and Molecular Function. In the figure, the size of
the dot indicates the number of DEGs that were enriched in the pathway,
the color of the dot corresponds to the different p-values, and the
solid line is used for GO terms enriched for upregulated genes, while
dashed line is used for GO terms enriched for downregulated genes.
TABLE 2.
KEGG pathway enrichment analyses and GO functional annotation result.
Terms ID Description
KEGG hsa00140 Steroid hormone biosynthesis
hsa00983 Drug metabolism—other enzymes
hsa00982 Drug metabolism—cytochrome P450
hsa00980 Metabolism of xenobiotics by cytochrome P450
hsa05204 Chemical carcinogenesis
hsa04976 Bile secretion
GO: BP GO:0002283 Neutrophil activation involved in immune response
GO:0042119 Neutrophil activation
GO:0002446 Neutrophil mediated immunity
GO:0043312 Neutrophil degranulation
GO:0043299 Leukocyte degranulation
GO:0002366 Leukocyte activation involved in immune response
GO:0002443 Leukocyte mediated immunity
GO:0036230 Granulocyte activation
GO:0002274 Myeloid leukocyte activation
GO:0002275 Myeloid cell activation involved in immune response
GO:0002444 Myeloid leukocyte mediated immunity
GO:0002263 Cell activation involved in immune response
GO:0045055 Regulated exocytosis
GO:0006887 Exocytosis
GO:0045766 Positive regulation of angiogenesis
GO:1904018 Positive regulation of vasculature development
GO:0016192 Vesicle-mediated transport
GO:0022603 Regulation of anatomical structure morphogenesis
GO:0051187 Cofactor catabolic process
GO:0071466 Cellular response to xenobiotic stimulus
GO:0009410 Response to xenobiotic stimulus
GO:0006805 Xenobiotic metabolic process
GO:0042737 Drug catabolic process
GO:0042743 Hydrogen peroxide metabolic process
GO: CC GO:1904724 Tertiary granule lumen
GO:0070820 Tertiary granule
GO:0034774 Secretory granule lumen
GO:0060205 Cytoplasmic vesicle lumen
GO:0031983 Vesicle lumen
GO:0042581 Specific granule
GO:0035580 Specific granule lumen
GO:0030141 Secretory granule
GO:0099503 Secretory vesicle
GO:0031225 Anchored component of membrane
GO: MF GO:0020037 Heme binding
GO:0046906 Tetrapyrrole binding
GO:0048037 Cofactor binding
GO:0004497 Monooxygenase activity
GO:0005506 Iron ion binding
GO:0016709 Oxidoreductase activity, acting on paired donors, with
incorporation or reduction of molecular oxygen, NAD(P)H as one donor,
and incorporation of one atom of oxygen
GO:0016705 Oxidoreductase activity, acting on paired donors, with
incorporation or reduction of molecular oxygen
[116]Open in a new tab
Note: BP, biological process; CC, cellular component; GO, gene
ontology; KEGG, kyoto encyclopedia of genes and genomes; MF, molecular
function.
Hypoxia-Related Prognosis Model-Related Immune Alterations
Based on the foregoing GO enrichment analysis results, we conjecture
that hypoxia is related to immunity. Using CIBERSORT, we found the
proportion of resting mast cells and plasma cells in the high-risk
group significantly to be lower than that of the low-risk group,
whereas the proportion of neutrophils, monocytes, M0 macrophages, γδ-T
cells, and regulatory T cells (Tregs) increased in high-risk patients
([117]Figure 6A; [118]Supplementary Data Sheet S10). Cell types were
also predicted and visualized using xCell, giving similar results.
Further, xCell also shows that the Immune Score and Microenvironment
Score of the high-risk group were significantly higher than of the
low-risk group ([119]Figure 6B). An overview of the predictive result
of mechanism exploration is presented in [120]Figure 7.
FIGURE 6.
[121]FIGURE 6
[122]Open in a new tab
Landscape of immune infiltration in the bone marrow (BM) of acute
myeloid leukemia (AML) patients, as estimated from gene-expression data
(Cohort 1, BM samples n = 216) using CIBERSORT. (A) Boxplots
visualizing significantly different immune cell infiltrations between
high- and low-risk patients. The p-values calculated from Wilcoxon test
are shown: *p-values < 0.05; **p-values < 0.01; ***p-values < 0.001.
(B) Immune Score and Microenvironment Score between high- and low-risk
patients scoring by xCell based on estimated immune cell proportion.
Higher hypoxia risk is associated with higher Immune Score and
Microenvironment Score.
FIGURE 7.
[123]FIGURE 7
[124]Open in a new tab
The hypoxia-related mechanisms that are involved in a poor prognosis of
acute myeloid leukemia (AML) patients. Hypoxia may affect the prognosis
of patients with AML by affecting angiogenesis, drug metabolism, and
bone marrow immune microenvironment.
A Complex Model for Prognostic Evaluation of Acute Myeloid Leukemia
To explore the independent prognostic factors for AML, univariate and
multivariate Cox analyses were sequentially performed in the BEATAML1.0
dataset ([125]Figure 8A), including the hypoxia risk score and other
available clinical characteristics, such as age, gender, ELN 2017, the
mutations of NPM1 and FLT3, and CEBPA Biallelic status. The HRS, ELN
2017, and age remained statistically significant (p < 0.05) in both the
univariate and multivariate Cox analyses, indicating that HRS, ELN
2017, and age were independent prognostic factors. NPM1 status was an
independent factor of prognosis after adjustment for other clinical
factors. The correlation between clinical factors was analyzed and
visualized in [126]Figure 8B. HRS was positively correlated with
survival status, age, and ELN2017 while negatively correlated with
survival time and NPM1 status. Furthermore, [127]Figures 8C–E indicate
that the HRS had a higher AUC than other clinical factors in 1-, 3-,
and 5-years survival prediction. Overall, these results demonstrated
that the HPM can predict the AML prognosis independently and
effectively. To reveal the prognostic value, maximize practicability,
and facilitate clinicians’ usage of our model, we constructed a
nomogram that was composed of both the hypoxia risk score and available
clinical risk factors based on BEATAML1.0 cohort ([128]Figure 8F). A
combination of HRS and clinical risk factors was found to improve its
prognostic value with a markedly better AUC than the 2017 ELN genetic
risk stratification ([129]Figure 8G). It is shown that the complex
model can predict more accurately in the long term with the increasing
tendency of AUC. To validate the predicted and actual probabilities at
1, 3, and 5 years, calibration plots were constructed ([130]Figures
8H–J), and the nomogram performs well. These findings demonstrated that
the nomogram is an optimal model for predicting the survival
probability of AML patients than individual prognostic factors.
FIGURE 8.
[131]FIGURE 8
[132]Open in a new tab
The establishment of a nomogram to predict the overall survival of
acute myeloid leukemia (AML) patients. (A) Forest plot of univariate
and multivariate Cox regression analysis of clinical characteristics in
AML patients. The red dot with bar indicates statistical significance
(p < 0.05). (B) Pearson’s correlation between hypoxia-related prognosis
model (HPM) and different clinical characteristics (*p-values < 0.05;
**p-values < 0.01; ***p-values < 0.001). Receiver operating
characteristic (ROC) plot of HRM and important clinical characteristics
at 1 (C), 3 (D), and 5 years (E). (F) Survival analysis for HPM risk
and other prognostic factors in AML. (G) Nomogram of the complex model
for overall survival at 1, 3, and 5 years in AML patients. (H)
Time-dependent ROC curves for complex model and ELN 2017. Calibration
plot at 1 (I), 3 (J), and 5 years (K) for validation of prognostic
nomogram.
Discussion
AML is a highly aggressive and heterogeneous hematologic malignancy
([133]Döhner et al., 2015). Hypoxia is a significant outcome factor of
leukemia patients, which could be reflected by the changed expression
of related genes ([134]Benito et al., 2016). Here, in our analysis, we
identified an 18-hypoxia related gene-based prognosis model that could
independently predict the survival probability of AML patients. To
facilitate clinical application, we combined our HPM, 2017 ELN genetic
risk stratification, and clinical risk factors to construct a complex
model and nomogram, which outperformed 2017 ELN genetic risk
stratification in the prediction of survival rate. Thus, the
comprehensive nomogram could be utilized by clinicians in the near
future. Further, we surmise that hypoxia may affect the prognosis of
AML patients by affecting the angiogenesis, drug metabolism, and BM
immune microenvironment.
The risk model created in this study consisted of 18 hypoxia-related
genes, many of which were reported in cancer. ALDOC encodes a member of
the class I fructose-biphosphate aldolase gene family and is involved
in HIF-1 signaling pathway. ALDOC was identified as activators of Wnt
signaling, a signaling pathway involved in cancer genesis and
progression when it was over-activated ([135]Caspi et al., 2014).
Meanwhile, ALDOC was overexpressed in gallbladder carcinoma ([136]Fan
et al., 2020), melanoma ([137]Izraely et al., 2020), and lung cancer
([138]Yuan et al., 2021), associated with their growth or pathogenesis.
BATF3 is an AP-1 family transcription factor that controls the
differentiation of CD8(+) thymic conventional dendritic cells in the
immune system. According to the KEGG database, it was involved in PD-L1
expression and PD-1 checkpoint pathway in cancer. Immunotherapy with
immunomodulatory monoclonal antibodies targeting PD-1 or CD137 requires
Batf3-dependent dendritic cells ([139]Sanchez-Paulete et al., 2016).
COL5A1 is a member of the fibrous subfamily of collagen. Overexpression
of COL5A1 may promote metastasis of lung adenocarcinoma ([140]Liu et
al., 2018) and the progression of muscle-invasive bladder cancer
([141]Ewald et al., 2013) and may increase the risk of hematogenous and
lymphatic metastasis in serous ovarian cancer ([142]Yue et al., 2019).
COL5A1 is also overexpressed in gastric cancer, which may regulate the
proliferation of gastric cancer cells by affecting the tumor
microenvironment and is associated with poor prognosis ([143]Wei et
al., 2020). HBP1 plays a role in the regulation of the cell cycle and
is a tumor suppressor ([144]Bollaert et al., 2019). Downregulating HBP1
promotes the migration and invasion of oral squamous cell carcinoma
([145]Li K. et al., 2020) and breast cancer ([146]Li et al., 2011). HK1
encodes hexokinase 1, which is the first rate-limiting enzyme in
glycolysis, is related to the progression of ovarian cancer ([147]Li Y.
et al., 2020) and colorectal cancer ([148]Li S. et al., 2020). PSMA7
interacts with proteins such as HIF-1α, EMAP II, c-Abl, and Arg
tyrosine kinases, which participated in tumorigenesis. Studies reported
that PSMA7 expression was elevated in testicular, liver, breast,
prostate, cervical, gastric, and colorectal cancers ([149]Xia et al.,
2019), while UBA52 is overexpressed in the colon ([150]Barnard et al.,
1995), and renal cancers ([151]Kanayama et al., 1991). THBS1 was also
implicated in the development of several cancers, including breast,
gastric, melanoma, and cervical cancers and glioblastoma ([152]Qi et
al., 2021).
To probe into the mechanism of hypoxia in leukemia, differential gene
expression analysis and enrichment analysis were implemented. The
enriched biological process like “leukocyte mediated immunity” and
“cell activation involved in immune response” suggested that hypoxia
might affect cell-mediated immunity against cancer cells. Angiogenesis,
which was particularly important for tumor survival in the hypoxic
condition ([153]Hanahan and Weinberg, 2011), was significantly enriched
in the upregulated genes for the high-risk group. Although there are
few studies about angiogenesis in hematological malignancy, BM
angiogenesis in AML patients has been observed and may play a role in
the pathogenesis ([154]Hussong et al., 2000; [155]Padro et al., 2000;
[156]Testa et al., 2020). BM microvessel density of AML patients is
higher than that of healthy individuals at the time of diagnosis and
decreases after remission ([157]Padro et al., 2000; [158]Song et al.,
2015). The vascular niches could support the survival of leukemic cells
and protect AML by regulating AML cell cycle through paracrine
secretion and adhesive contact with endothelial cells, helping to
resist chemotherapy ([159]Cogle et al., 2016). Moreover, higher
microvessel density at the time of diagnosis was associated with poor
prognosis ([160]Kuzu et al., 2004; [161]Rabitsch et al., 2004). In
addition, drug metabolism is significantly affected by changes in
pharmacokinetics, expression, and function of drug metabolic enzymes
and transporters under hypoxia ([162]Donovan et al., 2010). Hypoxia
affects the transcription and function of cytochrome P450 (CYP450)
through HIF-1α ([163]Min et al., 2019). CYP enzymes and other
drug-metabolizing enzymes are expressed in BM stroma ([164]Alonso et
al., 2015). CYP450 is involved in the stroma-mediated resistance of AML
cells to chemotherapy ([165]Alonso et al., 2015). Drug catabolic
process pathways, such as “xenobiotic metabolic process,” “drug
catabolic process,” “Drug metabolism—other enzymes,” “Drug
metabolism—cytochrome P450,” and “Metabolism of xenobiotics by
cytochrome P450,” were markedly enriched, which might correlate with
chemotherapeutic drugs concentration and sensitivity change in a
hypoxic microenvironment of AML. Hypoxia might promote angiogenesis,
disturb cell-mediated immunity balance, and affect pharmacokinetics to
result in a bad prognosis.
To explore hypoxia-related changes in the BM immune microenvironment,
immune cell composition analysis was performed. We found that the
proportion of resting mast cells in the high-risk group was
significantly lower than in the low-risk group, while activated mast
cells in the high-risk group were higher than in the low-risk group,
even though the difference did not reach statistical significance (mean
proportion 0.12 vs. 0.02, p = 0.22). The present study shows that mast
cells can promote cancer growth by stimulation of neoangiogenesis and
modulation of the immune response ([166]Dyduch et al., 2012). This
might indicate that hypoxia affects angiogenesis and immune response by
affecting the proportion of mast cells. The proportion of monocytes and
M0 macrophages in the high-risk group were significantly higher than
the low-risk group, which contribute to the poor prognosis of high-risk
group patients. Tumor-infiltrating monocytes and macrophages have
well-recognized facilitative roles in the initiation, migration, and
invasion of solid tumors ([167]Condeelis and Pollard, 2006; [168]Qian
and Pollard, 2010; [169]Richards et al., 2013). Although not widely
researched, there are indications of a similar pro-tumor function in
hematological malignancy. Lee et al. demonstrated that monocytes have a
promoting effect on migration and invasion of human B-cell precursor
acute lymphoblastic leukemia (BCP-ALL) in vitro ([170]Lee et al.,
2012). Al-Matary et al. showed an increase of monocytes/macrophages in
the BM of AML patients and AML mouse models, which might support the
proliferation of AML cells in vitro, and it was also observed that the
grade of macrophage infiltration was correlated with the survival of
AML mice ([171]Al-Matary et al., 2016). The proportion of monocytes and
M0 macrophages in the high-risk group was significantly higher than in
the low-risk group, which contributes to the poor prognosis of
high-risk group patients. Tregs, which were regarded as suppressor T
cells preventing autoimmunity, were found to be aberrantly accumulated
in some types of tumor, playing a crucial role in dampening antitumor
immunity and establishing an immunosuppressive microenvironment
([172]Wang et al., 2017). Higher Treg percentages could indicate poor
prognosis in a variety of cancer types ([173]Overacre-Delgoffe and
Vignali, 2018), and AML is no exception. Williams et al. found that an
increased amount of Tregs in peripheral blood of AML patients is also
associated with an increased risk of relapse ([174]Williams et al.,
2019). These studies are consistent with our findings that Treg
proportion is higher in high-risk patients than low-risk patients. The
detailed mechanism needs further investigation in the future. And the
scRNA-seq transcriptome data can provide precision and details of the
interaction between the tumor cells and the microenvironment. Zhang et
al. developed a novel scRNA-seq data-based approach to reconstruct a
multilayer signaling network that contains pathways from intercellular
ligand–receptor interactions, intracellular transcriptional factors,
and their target genes ([175]Zhang et al., 2020). Meanwhile, the
single-cell RNA-sequencing data based on multilayer network method
(scMLnet) ([176]Cheng et al., 2021) also help to resolve
tumor–microenvironment interactions and dissect the
microenvironment-mediated intercellular and intracellular signaling
pathways of tumor cells, which might help to investigate the influence
of microenvironment on the tumor growth, drug resistance, and patient
prognosis.
Concerning treatment, Tregs have been targeted in the clinic, although
the efficacy is limited ([177]Overacre-Delgoffe and Vignali, 2018).
Targeting macrophages with bisphosphonate could reduce angiogenesis and
tumor growth in melanoma-bearing mice ([178]Gazzaniga et al., 2007). In
a mouse model of AML, Tregs accumulate at the site of disease and
suppress the function of adoptively transferred cytotoxic T cells
(CTL), and depletion of Tregs restored CTL function and reduced
leukemia progression ([179]Zhou et al., 2009). Anti-CD47 monoclonal
antibody can inhibit the immune escape of AML leukemic stem cells to
macrophages and play an antileukemic role by phagocytosis of leukemic
stem cells through macrophages ([180]Majeti et al., 2009). Clinical
trials of CD47 mAb magrolimab (Hu5F9-G4) as a single agent or in
combination for the treatment of relapsed refractory AML have shown
promising efficacy (magrolimab, Phase I, [181]ClinicalTrials.gov ID:
[182]NCT02678338; magrolimab + atezolizumab, Phase I,
[183]ClinicalTrials.gov ID: [184]NCT03922477; magrolimab + azacitidine,
Phase Ib, [185]ClinicalTrials.gov ID: [186]NCT03248479). Antiangiogenic
therapy may be an effective method in AML patients. Reduced BM
angiogenesis may help to restore drug sensitivity of drug-resistant AML
([187]Lin et al., 2019). Antiangiogenic therapy may be an effective
method in AML patients. Lin et al. demonstrated that wogonoside, one of
the metabolites of traditional Chinese medicine Huangqin, could inhibit
the BM angiogenesis and tumor progression of AML in vivo and in vitro
([188]Lin et al., 2019). Based on the results of clinical trials, some
antiangiogenic drugs that inhibit vascular endothelial growth factor
(VEGF), such as bevacizumab ([189]Karp et al., 2004), cediranib
([190]Fiedler et al., 2010), AG-013736 ([191]Giles et al., 2006), and
SU5416 ([192]Fiedler et al., 2003), could be an effective treatment for
AML, either alone or in addition to chemotherapy that works
independently on different targets. Hypoxia is becoming an emerging
target in AML. Hypoxia-activated prodrug, a new class of anti-cancer
agents, selectively deliver cytostatic or cytotoxic agents to hypoxic
subregions, uncloak at low oxygen pressure, and release the active
drug. Small-scale clinical trials about hypoxia-activated prodrugs
PR-104 and TH-302 treating patients with relapsed and/or refractory AML
were conducted, showing a definite antileukemia activity
([193]Konopleva et al., 2015; [194]Badar et al., 2016) (TH-302, Phase
1, [195]clinicaltrials.gov ID: [196]NCT01149915; PR-104, Phase 1,
[197]ClinicalTrials.gov ID: [198]NCT01037556). BCL-2, a proapoptotic
protein, could be overexpressed in hypoxia conditions; its inhibitors
can reduce oxidative phosphorylation and eradicate quiescent
chemo-resistant AML stem cells ([199]Ashton et al., 2018). Echinomycin,
a hypoxia-inducible factor HIF-1α inhibitor, can selectively kill the
leukemia-initiating cell without affecting host HSCs in relapsed AML
mice ([200]Wang et al., 2014). The preliminary pathophysiological
observation of this study may provide a perspective for further
investigation and a potential therapeutic target in the future.
Although large cohorts were utilized to establish our model, there are
still certain limitations in our study. The study was based on
retrospective cohorts, lacking prospective cohorts, and experimental
evidence. Data from different centers and various platforms are
necessary to validate the performance of our model. Further studies
including animal experiments and in vitro cellular experiments will be
needed to confirm our findings and delineate the pathophysiological
mechanism.
Conclusion
In summary, our HPM, complex model, and nomogram had excellent
predictive power for clinical applications, helping clinicians in
making clinical decisions. Our hypoxia-related immune and metabolic
alterations might help to find a potential therapeutic target.
Acknowledgments