Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 2.
Published in final edited form as: Semin Cancer Biol. 2023 Jan 2;88:172–186. doi: 10.1016/j.semcancer.2022.12.011

Tumor hypoxia: From basic knowledge to therapeutic implications

Chengheng Liao a,1, Xijuan Liu b,1, Cheng Zhang a,1, Qing Zhang a,c,*
PMCID: PMC9929926  NIHMSID: NIHMS1864352  PMID: 36603793

Abstract

Diminished oxygen availability, termed hypoxia, within solid tumors is one of the most common characteristics of cancer. Hypoxia shapes the landscape of the tumor microenvironment (TME) into a pro-tumorigenic and pro-metastatic niche through arrays of pathological alterations such as abnormal vasculature, altered metabolism, immune-suppressive phenotype, etc. In addition, emerging evidence suggests that limited efficacy or the development of resistance towards antitumor therapy may be largely due to the hypoxic TME. This review will focus on summarizing the knowledge about the molecular machinery that mediates the hypoxic cellular responses and adaptations, as well as highlighting the effects and consequences of hypoxia, especially for angiogenesis regulation, cellular metabolism alteration, and immunosuppressive response within the TME. We also outline the current advances in novel therapeutic implications through targeting hypoxia in TME. A deep understanding of the basics and the role of hypoxia in the tumor will help develop better therapeutic avenues in cancer treatment.

Keywords: Hypoxia, Hypoxia-inducible factors, Tumor microenvironment, angiogenesis, metabolism, immunotherapy

1. Introduction

The essence of tumor hypoxia is the imbalance between the oxygen demand of cancer cells and insufficient oxygen supply due to the dysfunctional tumor vasculature. Cancer cells exploit hypoxia to defend against the attack of immune cells or therapeutic interventions, therefore contributing to immune evasion and therapy resistance. Almost all solid tumors show a phenotype of hypoxia in their intratumoral area, which is often referred to as the tumor microenvironment (TME) [1]. It is challenging to thoroughly eliminate the negative effects of tumor hypoxia, partially because hypoxia not only induces the proliferative, invasive, or resistant properties of tumor cells but also elicits the dysfunction of immune cells or agitates the stromal environment to further facilitate tumor development.

The exploration of hypoxia and delineation of the related oxygen sensing signaling has a long-term history [2], and was well recognized by the awarding of the 2019 Nobel Prize in Physiology or Medicine to Drs. William G. Kaelin Jr, Sir Peter J. Ratcliffe, and Gregg L. Semenza [3]. Along with the discovery of the oxygen sensing signaling, numerous molecular pathways or biological processes that are associated with hypoxia were later uncovered and elucidated, eliciting an era of biomedical interventions on tumor hypoxia [4]. In this review, we will focus on summarizing the basic knowledge of the oxygen-sensing pathway in cancer, the major molecular mechanisms, and signaling pathways and machinery, including the proline hydroxylases (PHDs or EglNs) and novel oxygen sensor proteins from the 2-oxoglutarate (2-OG) dependent dioxygenases, hypoxia-inducible factors (HIFs), as well as the von Hippel-Lindau (VHL) E3 ubiquitin ligase system involved in different stages of oxygen sensing and cellular adaptation. In addition, the effects of hypoxia on the tumor microenvironment (TME), especially from the perspective of how hypoxia regulates angiogenesis, metabolism, and immune response in the TME, will be summarized. We will also discuss the current preclinical or clinical applications of cancer therapy by targeting hypoxia and hypoxic biological processes or signaling pathways.

2. Oxygen sensing in the tumor microenvironment

The crucial role of oxygen (O2) in radiation response was first acknowledged in 1953 [5]. More recently, investigations have demonstrated that hypoxia is a characteristic feature of most solid tumors. Solid tumors are dynamic heterogeneous structures and display significantly lower oxygen tensions compared to their surrounding normal tissue. Cancer cells typically outgrow their vascular supply and develop hypoxic and necrotic tumor areas located at a distance of more than 100 μm away from blood vessels [6]. Hypoxic conditions bordering necrotic areas select for cancer cells with reduced apoptotic potential [6] (Fig. 1). In addition, this region tends to be more resistant towards radiotherapy as radiotherapy normally needs oxygen to produce fresh radicals to kill cancer cells [7].

Fig. 1.

Fig. 1.

The hypoxia signaling in tumor.

Tumors contain regions of normoxic cells that are close to blood vessels, developing hypoxic and necrotic tumor areas with increased distance from a functional blood supply. (Poorly oxygenated tumors are characterized with increasing chemotherapy/radiation resistance and metastasis risk that cause therapy failure. In normoxia cells, HIF-αs are hydroxylated by proline hydroxylases (EglN1/2/3) in the presence of O2. Hydroxylated HIF-αs are recognized by pVHL, which mediates the proteasome degradation of HIF-as. In response to hypoxia, proline hydroxylation is inhibited. VHL is no longer able to bind and target HIF-αs for proteasomal degradation, which leads to HIF-αs accumulation and translocation to the nucleus. There, HIF-1α dimerizes with ARNT, binds to hypoxia-response elements (HREs) of target genes and recruits transcriptional co-activators such as p300/CBP for full transcriptional activity.

Hypoxia in solid tumors can be caused by three primary pathogenetic mechanisms: chronic or diffusion-limited hypoxia, acute or perfusion-limited hypoxia and anemia hypoxia [5]. Chronic hypoxia is caused by an increase in diffusion distances, chronically hypoxic cells are far away from capillaries which can result in a more clinically aggressive phenotype. Acute hypoxia is caused by inadequate blood flow in tissues when aberrant blood vessels are shut down. This is often transient and causes activation of stress-response, tissue damage and an increase in free-radical concentrations. Anemic hypoxia is a “relative” anemia caused by the decreased O2-carrying capacity of the blood, which implies that fewer hemoglobin molecules are available for binding oxygen. Anemic hypoxia could be tumor-associated or therapy-induced anemia, this type of hypoxia is particularly intensified in tumors or tumor areas exhibiting low perfusion rates.

Tumor hypoxia is a hallmark of increased malignant progression and diminished therapeutic response. A large amount of clinical and experimental evidence shows that poorly oxygenated tumors have a negative impact on the effectiveness of curative treatment [8]. Facilitation of cancer cell survival by tumor hypoxia will: 1) reduces the effectiveness of radiotherapy; 2) cause resistance to chemotherapy and chemoradiation and 3) increases metastasis risk that may facilitate patient mortality (Fig. 1). Hypoxia regions tend to harbor cancer stem cells (CSC), which correlates strongly with both treatment failure and tumor recurrence. In breast tumors, hypoxic response in CSCs is modulated by signaling pathways including HIF-1α signaling and p-AKT/WNT/β-catenin signaling essential for phenotype and self-renewal of breast CSC [9, 10]. To meet metabolic demands, hypoxic tumors continually produce angiogenic factors, such as VEGF [11, 12]. However, newly formed tumor vessels are chaotically organized, immature and fail to support adequate blood supply, which further exacerbates local hypoxia [13]. Prolonged treatment of anti-angiogenic therapies has been known to further reduce oxygen tension, resulting in subsequent treatment failures. The upregulation of VEGF also facilitates both intravasation and extravasation of circulating tumor cells and even CSC [14]. In addition to the reasons above, there are some other complicated pathogenetic mechanisms for hypoxia to cause bad clinical outcomes and poor treatment response in cancers, as we will outline later, HIF-α subunits (Hypoxia-inducible factor α, mainly including HIF-1α and HIF-2α) are the key ones.

3. The regulatory machinery of the oxygen-sensing pathway

3.1. Hydroxylation and regulation of HIF-αs

HIF-αs are regulated by oxygen-dependent posttranslational modifications, which are mediated by three 2-oxoglutarate-dependent (2-OG) dioxygenases Egl nine homologs in vivo (EglNs, EglN1, EglN2, and EglN3, also called PHD2, PHD1 and PHD3 respectively). Under normoxic condition, HIF-1α is hydroxylated mainly on proline residues 402 and 564 (in HIF-2α – Pro405 and Pro531) by EglNs, which is vital for von Hippel-Lindau (encoding pVHL) E3 ubiquitin ligase complex recognition and binding. The pVHL complex adds ubiquitin to HIF-αs, which leads to the degradation of HIF-αs by proteasomes. Under hypoxic condition, due to the decreasing oxygen concentration, EglNs activity is reduced, which leads to limitation of hydroxylation of HIF-αs, subsequent ubiquitination and degradation. As a result, accumulated HIF-αs form the dimer with a constitutively expressed protein ARNT (aryl hydrocarbon receptor nuclear translocator, or HIF-1β) and associates with coactivator(s) p300/CBP to form an active transcription complex [15]. HIF complex is functional and binds to target genes containing hypoxia-responsive elements (HREs) across the genome, which promotes transcriptional regulation of targets in human cell lines and has been revealed by a cell line pan-genomic analyses of HIF [16, 17]. Proteins encoded by HIF target genes are involved in many aspects of cancer progression. For instance, VEGF, as we described above, is involved in angiogenesis and other examples including TGFα and EGFR in cell proliferation; GLUT1 in glucose uptake and metabolism; SDF1 and its receptor CXCR4 in chemotaxis [18] (Fig. 1). Both HIF-1α and HIF-2α dimerizes with ARNT but may activate transcription of a similar but distinct set of target genes depending on the cellular context. For example, HIF-2α potentates, while HIF-1α antagonizes c-Myc and β-catenin activity [19]. Mounting evidence suggests that HIF2α promotes pVHL-defective renal carcinogenesis. However, HIF1α suppresses tumor formation by VHL−/− renal carcinoma cells, therefore HIF1α has the potential of being a kidney cancer suppressor gene [19]. Recently, Cowman S et al. found that HIF-1α but not HIF-2α is increased in ccRCC tumor cells. Compared with HIF-2α, HIF-1α is expressed highest in tumor-associated macrophages and is significantly associated with clinical benefit from antiangiogenic therapy [20]. TAD (the amino-terminal transactivation) domains of the HIF-α subunits may determine the specificity of targets of HIF-1α or HIF-2α [21]. For example, JMJD2c binds with TAD of HIF-1α but not HIF-2α and stimulates HIF-1α mediated transcription in Hela cells [22]. Both HIF-1α and HIF-2α transactivation function is negatively regulated by FIH (factor-inhibiting HIF)-dependent hydroxylation on asparagine site, which blocks the HIF-α recruitment of p300/CBP [23].

Even though most of the studies regarding HIF-1α and epigenetic regulators is about the interaction with p300/CBP, growing evidence has revealed that HIF-1α can also recruit other epigenetic factors. The H3K4 methyltransferases SET9 can bind to HIF-1α and promote HIF-1α protein stability in hypoxia [24]; the histone methyltransferase SET1B is recruited by HIF-1α to facilitate activation of the HIF response [25]; JMJD2C functions as a coactivator for HIF-1α, and HIF-1α can recruit JMJD2C into the hypoxia response elements of HIF-1 target genes [22].

3.2. The VHL-E3 ubiquitination system

VHL (encoding pVHL) is a critical tumor suppressor in clear cell renal cell carcinoma (ccRCC), which accounts for approximately 85% of all kidney cancer cases [26]. pVHL consists of two main protein binding domains, an α- and a β-domain; the α-domain of pVHL binds to elongin C, which in turn recruits elongin B and Cullin-2, whereas the latter recognizes and binds to substrates for ubiquitination and degradation by the 26S proteasome [15]. The canonical role of pVHL is its involvement in oxygen sensing and adaptive response to hypoxia. pVHL complexes recognize hydroxylated HIFαs, targeting them for proteasome degradation by ubiquitination [27, 28].

Growing evidence suggests that while HIF represents one of most important effectors for pVHL response, there exist other HIF-independent signaling pathways. For example, the inhibition of HIF-2α-mediated pathways by a specific HIF-2α antagonist PT2399 does not prevent all ccRCC tumor growth [29]; some other evidences indicate that VEGF can be induced by K-ras under hypoxia in a HIF-independent manner [30]; multiple pathways and transcription factors (TFs) such as NF-kB, AP-1, and CEBP are known to be induced by hypoxia in a HIF-independent pattern [31]. Elvidge and colleagues demonstrated that some genes regulated by hypoxia were not regulated by HIFs, suggesting that there are other HIF-independent signaling controlled by oxygen sensing pathway [32]. Now, more and more evidence identified non-HIFα substrates of pVHL. In ccRCC, pVHL binds hydroxylated ZHX2 and SFMBT1 for ubiquitinated degradation [26, 33]. In breast cancer, G9a stabilization is mediated through pVHL interaction and subsequent proteasome-mediated degradation, which links G9a as an epigenetic sensor of a hypoxic microenvironment [34]. NDRG3 has been identified as a bona fide substrate of the EglN1/pVHL system, which indicates that the EglN1/VHL system can control both HIF-dependent and HIF-independent hypoxia responses [35]. Also, several other putative pVHL substrates have been identified, including aPKC, RBP1/7, CERKL, TET2 and so on [15, 36]. By performing proteome and ubiquitome co-analysis together with the ccRCC CPTAC data, Wang and colleagues identified 57 proteins including TGFBI and NFKB2 that were ubiquitinated and downregulated by pVHL [37]. Based on these findings, a combinatorial targeting of HIF and other pVHL substrates may prove to be more effective than monotherapy in cancer therapy such as ccRCC. For example, Rouven Hoefflin and colleagues showed that the inhibitor of SPHK1 signaling regulated by pVHL-SFMBT1 axis [33] demonstrated excellent therapeutic activities in several experimental ccRCC models that are resistant to HIF-2α inhibitor PT2399 treatment [38]. These findings suggest that functional prolyl hydroxylation is beyond HIF. Although previous study suggested that other prolyl hydroxylation substrates beside HIF could not be validated through in vitro hydroxylation assays with recombinant prolyl hydroxylases [39], it exists the possibility that there may be some co-factors in vivo required to catalyze robust prolyl hydroxylation reactions for other potential substrates. Nonetheless, it is imperative to study the multi-functionality of prolyl hydroxylation in hypoxia signaling and cancer, which may yield additional therapeutic targets in cancer and other diseases. Undoubtedly, HIF-2α is a key therapeutic target for ccRCC. However, uncovering potentially additional HIFα-independent targets of VHL could provide new therapeutic opportunities and new combination treatment with HIF-2α inhibitor.

However, not all proteins recognized by pVHL are labeled for proteasomal degradation. It has been shown that AKT is hydroxylated by EglN1 on proline residues 125 and 313, and pVHL interacts with hydroxylated AKT and suppresses its activity in an E3 ubiquitin ligase-independent fashion [40]. Roe and colleagues demonstrated that pVHL binds to p53 and blocks the Mdm2-mediated ubiquitination and nuclear export of p53 [41]. EglN1 hydroxylates TBK1 on proline residue 48, resulting in pVHL-TBK1 interaction that helps recruit phosphatase PPM1B, therefore, suppressing TBK1 activity [42]. These findings highlight a new mechanism through pVHL that can affect cancer progression and response to treatment.

3.3. 2-Oxoglutarate-dependent dioxygenases

As cellular oxygen sensors by regulating the hypoxia-inducible transcription factors HIF-1α and HIF-2α, EglNs proteins belong to the class of 2-oxoglutarate-dependent (2-OG) dioxygenases (2OGDDs). 2OGDDs require dioxygen, divalent iron (Fe2+) and 2OG as co-substrates, and produce a hydroxylated product, CO2, and succinate [43]. There are approximately seventy known and putative 2OGDDs in the GenBank DNA database, including Proline/lysine hydroxylases, JmjC domain-containing enzymes, nucleic acid oxygenases, fatty acid and small-molecule oxygenases [44]. The details of 2OGDD enzymes have been reviewed recently in other excellent reviews, which will not be repeated here [43]. Some 2OGDDs have specific role in cancer, but the mechanism of 2OGDDs in cancer has not been fully understood (Table 1).

Table 1.

List of known 2 OG-dependent dioxygenases and their roles in cancer.

2OGDD Cancer types Function
Proline/lysine hydroxylases
EglN1 PG Mutated and inactivated [201, 202]
EglN2 BRCA Overexpressed [203]
EglN3 Glioma CpG sites were methylated, suppressor [204]
P4HA1–3 _ _
P4HB _ _
PLOD1–3 _ _
LEPRE1 Colo, BRCA and Lung Overexpressed [205]
LEPREL1–2 _ _
P4HTM _ _
BBOX2 _ _
DNA/RNA-modifying enzymes
TET1 Many cancer types Mutated and inactivated, silenced [206208]
TET2 Many cancer types Mutated and inactivated [209211]
TET3 Glioma Epigenetically repressed [212]
ABH1,2,3,4,6 _ _
ABH5 AML Overexpressed [213]
FTO AML and CESC Overexpressed [214, 215]
JmjC domain-containing enzymes
KDM2A BRCA and LUAD Amplified and overexpressed [216, 217]
KDM2B PACA Overexpressed [218]
Aggressive brain tumors Silenced [219]
KDM3A _ _
KDM3B AML Suppressor [220]
KDM4A Many cancer types Amplified and overexpressed [221]
KDM4B OV and EGC Overexpressed [222, 223]
KDM4C BRCA, ESCs and ESCA Amplified [224226]
KDM4D ccRCC Overexpressed [227]
KDM5A Glioma Silenced [228]
Many cancer types Amplified and overexpressed [229231]
KDM5B Many cancer types Amplified and overexpressed [232]
KDM5C Many cancer types Mutated and inactivated [233, 234]
KDM5D PCa Copy number loss [235]
KDM6A Many cancer types Mutated and inactivated [236]
BRCA Overexpressed [237]
KDM6B Haematologic cancers Overexpressed [238]
KDM7A _ _
KDM8 BRCA and PCa Overexpressed [239, 240]
HR BRCA Suppressor [241]
JARID2 AML Chromosomal deletion, suppressor [242]
JHDM1C _ _
JMJD1C _ _
JMJD4,7,8 _ _
JMJD6 Many cancer types Overexpressed [234, 243, 244]
MINA53 Many cancer types Overexpressed [245]
PHF2 Many cancer types Suppressor [246]
NO66 PCa Overexpressed [247]
PHF8 HCC Overexpressed [248]
UTY _ _
Other hydroxylases
ASPH Many cancer types Overexpressed [249]
ASPHD1–2 _ _
BBOX1 BRCA and HCC Overexpressed [250, 251], Suppressor [252]
FIH1 _ Regulates HIF [23]
HSPBAP1 PCa Overexpressed [253]
OGFOD1 BRCA Overexpressed [254]
OGFOD2 _ _
PAHX-AP1 _ _
PHYH _ _
PHYHD1 _ _

PG, Paraganglioma; BRCA, Breast invasive carcinoma; Colo, Colorectal Cancer; Lung; Lung Cancer; AML, Acute Myeloid Leukemia; CESC, cervical cancer; LUAD, lung adenocarcinoma; PACA, Prostate cancer; OV, Ovarian cancer; EGC, Gastric cancer; ESCs, Esophageal squamous cell carcinomas; ESCA, Esophageal carcinoma; PCa, Prostate cancer; HCC, Hepatocellular carcinoma.

As major cellular oxygen sensors, EglNs have O2 Km values at/near atmospheric oxygen levels [43]. Notably, Liu et al. show that EglN2 binds with and hydroxylates H3 at proline 16 directly (H3P16oh). This work provides a further intriguing link between oxygen sensor pathway, histone modifications and gene transcription [47]. Besides EglN proteins, some other 2OGDDS such as JmjC domain-containing enzymes can also act as potential oxygen sensors [45]. Batie et al. reported various histone methylation marks were increased in HIF-independent as early as 30 min under acute hypoxia. In these histone markers, H3K4me3 level was induced by the inactivation of KDM5A [46]. This is consistent with the observation that KDM5A has the highest O2 Km of the KDM5 family members (90 μM) [43]. KDM6A is highly oxygen-sensitive and controls cellular differentiation. Two residues (M1190 and E1335) are involved in KDM6A’s oxygen sensor function, which are not conserved in KDM6B [47]. These new studies clarify that histone modifiers can act as new oxygen sensors directly rather than through the HIF pathway and the underlying regulatory mechanisms still need to be investigated.

4. Hypoxia shapes the landscape of the tumor microenvironment

4.1. How does hypoxia affect the TME?

The fast-proliferating rate of tumor cells is often accompanied by poor vasculature formation, therefore leading to the development of intertumoral pathological hypoxia. Although HIF could stimulate the formation of new blood vessels in the tumor region to compensate for the shortage of oxygen, however, the tumor vasculature is featured with poor organization and irregular architecture which is easy to collapse under the pressure of tumor formation and stroma growth [48, 49]. This phenomenon causes an irregular oxygen supply and an overall hypoxic environment in the tumor.

The tumor microenvironment (TME) is the ecosystem that surrounds the tumor cells, which is comprised of the non-cellular and cellular components. The non-cellular component includes the extracellular matrix (ECM), various signaling molecules such as cytokines and chemokines, and a chemical milieu including the pH, pO2, nitric oxide, and metabolites. The cellular component mainly consists of immune cells, stromal cells, and blood vessels [1]. The TME is characterized by several notable features such as low extracellular pH, high level of reactive oxygen species (ROS), hypoxia, and a general immunosuppressive niche in comparison with normal tissue [50]. Crosstalk and mutual causal relationship may exist between each of these characteristics, while hypoxia could stand out as the prerequisite above all in TME.

Overall, as a universal intrinsic hallmark in all solid malignant tumors, hypoxia affects TME in many forms and from different aspects [51, 52] (Fig. 2). For example, hypoxia promotes ECM remodeling by changing the composition of ECM to enhance the invasive potential of tumor cells [53], which could be partially via stimulating the paracrine secretion of soluble factors that increases the fibrotic and stiff content of the ECM [54]. The pro-inflammatory cytokines and chemokines are important signaling molecules secreted by immune cells such as the nature killer (NK) cells, a study reported that hypoxia modifies the transcriptome profiles of NK cells which shows negative effects on generating these immune-modulatory signaling molecules [55]. On the other hand, the hypoxia-driven metabolic programming in the tumor or stromal cells also changes the level and distribution of certain metabolites in TME, especially for some important regulatory metabolites such as lactate, adenosine, and reactive oxygen species that exert inhibitory function to the infiltrating immune cells [56]. Therefore, the impact on tumor progression and immune invasion by hypoxia could be direct or indirect, either by direct inducing changes in cellular metabolism or creating a metabolic crosstalk between tumor and stromal cells via secreting metabolites [57]. From another perspective, hypoxia also shapes the tumor evolutionary landscape through applying a selective pressure for certain molecular aberrations. A recent study by Bhandari et al. suggests hypoxia is associated with elevated genomic instability and unique features of driver-mutation signatures in a large portion of tumor types. Intriguingly, they show that hypoxia could also regulate microRNAs abundance and lead to their dysregulation in tumors [58].

Fig. 2.

Fig. 2.

The relationship between hypoxia and TME.

Increased distance from blood vessels leads to a reduction in the oxygen supply of solid cancer. Upregulated glycolytic metabolism in hypoxic cells contributes to the acidification of the tumor microenvironment. The ECM composition is changed and subsequently enhance the invasive potential of tumor cells under hypoxic condition. Hypoxia modifies the transcriptome profiles of immune cells and suppresses the immune response. Metabolic programming in hypoxic area changes the level and distribution of certain metabolites such as lactate, adenosine, and ROS in TME. In addition, hypoxic conditions can drive genomic instability and mutagenesis, increasing the probability of metastasis and creating chemotherapy/radiation resistant clones.

Additionally, hypoxia changes the TME not only by directly affecting the intertumoral environment, but also through modulating the processes and regulatory molecular machinery in cells. As the predominant hypoxia-responsive molecular determinant, the HIF guides the cellular adaptation to oxygen availability by regulating gene expression. However, the functions of HIF-1α and HIF-2α as oncogenes or tumor suppressors in tumor development are still under debate. This controversial concept comes from a large body of evidence that suggests HIF-1α or HIF-2α play divergent roles either in promoting or suppressing tumor growth in different cancer types under different contexts [59]. In addition, the spatiotemporal distribution of HIF subunits within tumors could also contribute to their complex role in different cancers [59].

As tumor progression is not just a simple proliferation and growth of tumor cells, instead, it is more like a dynamic process engaged by every component of the TME, including tumor cells, stromal cells and other non-cellular components such as ECM [60]. Overall, hypoxia creates a favorable environment for tumor progression by shaping the landscape of TME that promote angiogenesis, metabolic adaptation, immune escape, metastasis, and therapy resistance. In the following parts of this section, more details on how hypoxia affects the TME will be summarized.

4.2. Hypoxia induces angiogenesis in TME

The environment in the TME is highly hypoxic, acidic, and nutrient competitive, and these are the major unfavorable factors for cell survival [61]. To overcome these harsh conditions, cells within the TME coordinate a program to promote angiogenesis in order to restore oxygen level, replenish nutrient supply and remove metabolic waste [12]. The importance of angiogenesis in promoting tumorigenesis and tumor progression is well recognized and documented over the last fifty years [62]. Besides, angiogenesis is necessary for tumor metastases, as the neovascular network allows tumor cells to disseminate and migrate through the blood vessels from the local tumor mass to distant organs [63, 64].

When tumor size is small with diameter less than 1 mm at the tumor initiation stage, tumor cells acquire oxygen mainly through blood perfusion and permeation from the surrounding tissue. However, due to the expansion of the tumor mass, the inefficient tumor vasculature hampers the oxygen supply which leads to hypoxia in the intertumoral region. As the main player involved in the transcriptional program of angiogenesis, the HIF-1α is subsequently stabilized and activated to drive the gene expression of vascular endothelial growth factor (VEGF), one of the major target genes of HIF-1α and a primary regulator of angiogenesis [11, 12]. The hypoxia induced HIF-α also leads to the expression of other pro-angiogenic factors such as platelet derived growth factor (PDGF) [65], nitric oxide synthase [66], adrenomedullin [67], and interleukin 8 [68] to further promote angiogenesis in TME. Besides, Hypoxia may also influence angiogenesis through other processes, for instance, by altering the morphology of fiber in the ECM [69]. It should be noted that the hypoxia-induced and HIF-1α-mediated angiogenesis is not limited to tumor cells, but also other cell types within the TME, including immune cells and cancer-associated fibroblasts (CAFs) [70].

4.3. Hypoxia induced long noncoding RNAs play multiple roles in TME

Emerging reports show that modulation of long noncoding RNAs (lncRNAs) in response to hypoxia could play pivotal roles in biological processes related to tumorigenesis, metastasis, angiogenesis, metabolism, and immune response [7176].

In tumor cells, the hypoxia induced lncRNAs have been shown to participate in regulating gene expression at multiple levels including epigenetic, transcriptional, post-transcriptional regulation and protein levels [71, 7780]. For example, the hypoxia induced WT1-AS promotes oncogenic transcription factor WT-1 expression through modulating histone H3K4 and H3K9 methylation in acute myeloid leukemia [81]. The lncRNA HOTAIR which induced by hypoxia may serve as scaffold by providing binding surfaces for histone modification complex and thus to impact the chromatin modification and transcription [82]. Besides the chromatin structure remodeling, the lncRNAs could modulate transcription by directly regulating specific transcription factors. Hypoxia inducible LncHIFCAR is a co-activator driver of HIF-1α via direct binding and subsequently promotes the recruitment of HIF-1α and p300 cofactor to the target promoters in oral carcinoma [83]. At post-transcription level, multiple hypoxia-induced lncRNAs are involved in mRNA stability regulation and miRNA-mediated gene silencing regulation. The most prominent example of lncRNAs regulating mRNA stability is HIF-1α mRNA is regulated by HIF1A-AS2 DANCR and lncRNA-LET [79, 8487]. In addition, HIF1A-AS2 has been reported to sequester miR153–3p to enhance HIF-1α expression suggesting that lncRNAs involve in miRNA regulating [88]. In TME, lots of existing studies have indicated that lncRNA are involved in the crosstalk between tumor cells and other cell types including stromal cells, endothelial cells, and immune cells [8993]. The exosomes that derived from tumor cells deliver lncRNAs to mediate the communication between tumor cells and other TME components and subsequently regulate tumorigenesis and progression. Although the lncRNAs have been shown to be able to serves as diagnostic and prognostic markers [94, 95], the clinical application of lncRNAs is still required to be further explored.

4.4. Hypoxic TME dampens tumor immunity

Tumor cells escape immunosurveillance by various mechanisms while tumor hypoxia is among the most important factors influencing immune responses [96]. It should be noted that hypoxia affects immunity divergently in an immunological context dependent manner [97]. In contrast to hypoxia that controls proliferation, development and effector function of immune cells in physiological immunological niches, tumor hypoxia typically causes immune cell dysregulation that eventually drives tumor progression and development [97]. Hypoxia restrains the continuous surveillance and attack of immune cells to tumor cells, generating an immunosuppressive environment to facilitate tumor progression. Specifically, hypoxia modulates the tumor immune microenvironment from at least three major aspects by: 1) impeding function or infiltration of immune cells, 2) recruiting immunosuppressive cells to block the immune response, and 3) upregulating regulatory molecules that further blocks the activation of immune effector cells. In the context of antitumor immunotherapy, hypoxia also confer resistance to immunotherapeutic drugs by inducing expression of membrane proteins that increases drug efflux or regulates specific cell signaling pathways that are critical to the response of immunotherapy. Therefore, a fully understanding of these processes is necessary to achieve successful therapeutic efficacy (Fig. 3).

Fig. 3.

Fig. 3.

Hypoxia shapes the metabolic and immune environment of TME.

Hypoxia in the TME changes metabolites profile and promotes secretion of cytokines/chemokines that regulates the function of immune cells, as well as mediates the anti-tumor response of various types of immune cells.

Tumor infiltrating immune cells are critical components of the tumor microenvironment and contain various cell types including T cells, B cells, macrophages, neutrophils, and dendritic cells [1]. In general, as previously mentioned, hypoxia impedes the function of tumor-killing immune cell populations while enhancing the pro-tumorigenic role of regulatory immune cells to achieve an overall favorable niche for tumor growth. However, the effect of hypoxia on immune cells is complicated due to the intricate biology of the immune cell differentiation programs and a variety of types of immune cell populations.

T cells are the predominant immune population for tumor immune surveillance. Hypoxia modulates T cells by first affecting the recruitment of T cells into tumor areas. For example, in some malignancies such as prostate cancer, the hypoxic tumor region lacks substantial infiltration by any type of T cells. Alleviation of tumor hypoxia restores T cell access and leads to increased T cell proliferation within the TME [98]. On the other hand, hypoxia increases T regulatory cells (Treg) in the TME to suppress CD8+ T cell mediated cytotoxic killing of tumor cells [99]. In addition, hypoxia also causes metabolic dysregulation in T cells. Studies have shown that the hypoxia/HIF-1α signaling could increase the expression of pyruvate dehydrogenase kinase (PDK1), thus decreases OXPHOS and increases glycolysis in the T cells [100, 101]. Intriguingly, in some other cases, hypoxia produces favorable roles in T cell function. A study by Palazon et al. found that hypoxia and HIF-1α drive CD8+ T cell migration and effector function, while loss of HIF-1α promotes tumor progression [102]. Another study also found similar results that CD8+ T cells could be activated and survive well under hypoxia condition and kill tumor cells more efficiently than normoxic T cells through increasing granzyme B packaging into granules [103]. Hypoxia affects multiple types of T cells in addition to CD8+ population. Recently, hypoxia has been reported to suppress the antitumor function, but not the infiltrating potential, of γδ T cells in brain tumors [104].

Tumor-associated macrophages (TAMs) are exceptionally plastic cells and a prevalent cell type in a variety of tumors [105]. Two main polarization states exist in macrophages which represent the classical M1 phenotype antitumoral and the alternative M2 immunosuppressive phenotype [106]. Mounting evidence suggests that hypoxia selectively induces specific macrophage phenotypes promoting tumor malignancy while repressing immunity [107]. This is because macrophages are very sensitive to oxygen fluctuation with the M1 polarization preferentially undertake in normoxia, while the M2 phenotype is induced in hypoxic circumstances [108]. The dendritic cells (DCs) play a major role on presenting the tumor antigens to naïve T cells to initiate the immune adaptive response. Hypoxia, although does not hamper DC maturation, inhibits antigen uptake as well as changes the DC chemokine expression profile in the in vitro setting, therefore may have a critical role in DC differentiation, adaptation, and activation [109]. This is in line with the finding that HIF-1α can promote the anti-inflammatory functions of DCs in vivo [110]. Other immune effector or regulatory cells could also be affected by hypoxia including myeloid-derived suppressor cells (MDSCs), whose immune suppressive effects are also enhanced by the hypoxia/HIF signaling [111], and the NK cells as hypoxic TME reduces the cytotoxicity and causes the elimination of NK cells by inducing mitochondrial fragmentation in NK cells [112].

In addition to directly affecting the function of immune cells, hypoxia can also induce several regulatory molecules through HIF-1α to further promote innate immune evasion. These regulatory molecules include the immune checkpoint inhibitor molecules (PD-L1 and CTLA-4) and CD47, which will bind to macrophages/effector T cells and block their activity. Under hypoxia, HIF-1α protein will accumulate and directly induces PD-L1 expression in multiple cell types such as tumor cells, macrophages, dendritic cells and MDSCs [113115]. These cell surface PD-L1 could further bind PD-1 receptor to block T cell function. CTLA-4 receptor is induced and expressed on CD8+ T cells by hypoxia while binding CD80 and CD86 ligands on the surface of antigen-presenting cells (APCs), leading to effector T cell inhibition and regulatory T cell activation [116]. The CD47 “don’t eat me” signal controls immune evasion through interacting with signal regulatory protein α (SIRPα) expressed on TAMs [117] and MDSCs [118] and its expression is also controlled by HIF-1α [119]. Interestingly, HIF-induced VEGF is also an immunosuppressive cytokine in the hypoxic TME, through promoting the CD4+ T cells differentiation to Treg cells [120]. Overall, hypoxia in the TME regulates the function of immune cells, as well as the expression of immune inhibitory molecules, which in turn plays a role in promoting tumor development and immune evasion.

4.5. Hypoxia disturbs metabolism in TME

Tumor hypoxia develops largely because of imbalance between the high demand of oxygen consumption and its insufficient supply in the TME. Furthermore, the immature blood vessel formation directly causes a shortage of various nutrient supplies and the related metabolic activities in the TME. With this premise, hypoxia must reshape the landscape of metabolism in TME, not only by forcing the metabolic reprogramming in cells to adapt to the TME, but also by altering the level and composition of metabolites and nutrients within the hypoxic tumor region.

Unfortunately, tumor cells basically win the battle for oxygen and nutrient utilization and show higher metabolic adaptability and plasticity than the normal cells [121]. This is usually reflected on the tumor cells rewiring the aerobic respiration from the mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis (or known as the Warburg effect [122]), an inefficient but rapid way to ferment glucose, even when oxygen is readily available. Consequently, tumor cells consume more glucose to generate cellular building blocks including nucleic acids and amino acids, and more “waste” products such as lactate which subsequently leads to an acidic environment to furtherly suppress the activity of infiltrating immune cells. The glycolytic shift is largely due to the HIF signaling dependent induction of the glycolysis driver genes under either hypoxia, oncogenic signaling or pseudo-hypoxia conditions due to pVHL loss in kidney cancer [123, 124]. These genes encode transporters and key enzymes involved in glucose uptake and catabolism including the glucose transporter proteins (GLUT1 and GLUT3) [125, 126], hexokinases (HK1 and HK2) [127], phosphoglycerate kinase (PGK) [128], pyruvate kinase M2 [129], and lactate dehydrogenase (LDHA) [130], etc.

The repression of mitochondrial respiration is also mediated by HIF under hypoxia through multiple mechanisms. Firstly, HIF modulates glycolytic and mitochondrial metabolism by directly regulating some key metabolic enzymes. For example, HIF-1α or −2α activates the expression of lactate dehydrogenase A (LDHA), a key metabolic enzyme of glycolysis that converts pyruvate to lactate, in the cancer cells [131]. This leads to the accumulation of lactate and regeneration of NAD+ which is an electron acceptor to maintain the glycolytic flux [132]. On the other hand, HIF-1 also transactivates the gene coding PDK1 which mediates the phosphorylation and inhibition of pyruvate dehydrogenase (PDH), thereby reducing cellular acetyl-CoA production and suppression of TCA activity in mitochondria [100, 101]. Secondly, HIF also could mediate an indirect way since activated HIF-1α suppresses c-MYC transcriptional activity to the mitochondrial genes [133] or transcriptionally represses PINK1 [134]. Moreover, hypoxia also represses mitochondrial biogenesis by controlling Forkhead-box protein O3a (FOXO3a) mediated mitochondrial biogenesis regulation [135]. It is worth mentioning that the hypoxia-induced mitochondrial ROS could also inhibit the turnover of HIF. Inhibition of the activity of PHDs by increased ROS leads to stabilization of HIFα subunits and HIF-mediated transcription [136].

Besides repression of OXPHOS and enhancement of glycolysis, other metabolic pathways are also modulated in cells under hypoxia. For instance, tumor cells activate the pentose phosphate pathway (PPP) and synthesis of amino acid to meet the high demands of biomass synthesis [137, 138]. Hypoxia can also regulate lipid metabolism. A previous study by Qiu et al. demonstrated that HIF-2α, but not HIF-1α, promotes lipid storage to sustain ER homeostasis by transcriptional activating the lipid droplets coat protein gene PLIN2 in ccRCC [139]. Furthermore, HIF can directly suppress fatty-acid oxidation through inhibition of medium/long chain Acyl-CoA dehydrogenase (M/LCAD) [140] or cause a lipid deposition phenotype as most commonly observed in ccRCC by repressing carnitine palmitoyltransferase 1A (CPT1A) and subsequent lipid transportation into mitochondria [141]. Cells under hypoxia can also rely on reductive carboxylation of glutamine for de novo lipogenesis [142]. Therefore, hypoxia has a broad effect on metabolic fate of the cells in TME.

Because of the metabolic reprogramming induced by hypoxia, the alteration of metabolites that present in the TME also has profound impacts on the cell fate and causes symbiotic or competitive interactions between different tumor resident cells (Fig. 3). In the 1920s, Otto Warburg first observed that cancer cells could uptake excessive amount of glucose and preferentially produce lactate [143]. The lactate generated by tumor cells induced by hypoxia is rapidly secreted into the extracellular space by monocarboxylate transporters (MCTs) to prevent intracellular acidification, however, this process could gradually lead to the accumulation of lactate and decrease the pH level of the TME [144]. Elevation of lactate level in the TME is reported to be associated with poor clinical outcome [145], which is largely due the broad effect of lactate on negatively modulating immune response [146] or act as a signaling molecule involved in various mechanisms to directly promote cancer cell proliferation, survival, and metastasis [147]. Accompanying with elevated levels of lactate, extracellular glucose level is dramatically decreased in the TME. It should be noted that not only the cancer cells are highly glycolytic, T cells also highly dependent on glucose and glycolysis to maintain cell proliferation and function. Glucose deficiencies therefore impedes T cell activation and limit antitumor responses [139, 148]. Apart from glucose, glutamine is another critical metabolite readily scavenged by proliferating cells in hypoxic environment [149151]. Similar as glucose oxidation is repressed under hypoxia, glutamine oxidation in mitochondria is also blocked and detoured to lipogenesis instead [152, 153]. Glutamine restriction in the TME could affect tumor cells, but also has a deep impact on immune cells. As several studies reported, limitation of glutamine changes T cell differentiation from CD4+ T cells to FOXP3+ Treg cells [150, 154]. Another important immune regulatory metabolite that is activated by hypoxia is adenosine, which has been well accepted for its immunosuppressive function and as a metabolic checkpoint for immune therapy [155]. Hypoxia activates adenosine pathway not only by inducing the release of ATP, which is the precursor of extracellular adenosine synthesis, but also by upregulating ectonucleotidases including CD39, CD73, as well as adenosinergic receptors like A2BR through a HIF-1α dependent manner [156158]. Upregulation of CD39 and CD73 facilitate the two-step conversions from ATP to AMP and AMP to adenosine, respectively. The increased extracellular adenosine could bind adenosine receptors to execute a broad immune repression to either inhibit effect T cells or induce Treg cells [155].

5. The therapeutic implications of targeting hypoxia in TME

Tumor hypoxia has emerged as an attractive therapeutic area due to its essential role for cancer. Upon discovery of a myriad of mechanisms, hypoxia shapes the TME and promotes tumor initiation, progression, metastasis, immune evasion, and drug resistance. A large opportunity of biomedical interventions could be and/or have been made to tackle the cancer through targeting tumor hypoxia. In general, some various strategies and drugs have been developed at the stage of preclinical or clinical application. Based on the therapeutic mechanism of action, these hypoxia targeting strategies could be classified into several categories, such as 1) modifying tumor oxygenation in TME, 2) targeting HIF and HIF-related hypoxia signaling, 3) developing hypoxia-activated prodrugs, and 4) targeting other hypoxia-associated biological processes and pathways. Here we provide the rationale of each of the strategies, as well as outline some of the representative therapeutic applications that overcome the hypoxic barrier in the preclinical or clinical setting (Table 2).

Table 2.

Summary of the representative therapeutic interventions for tumor hypoxia.

Treatment/Drug Action/Effects Clinical trial Refs
1. Modify tumor oxygenation
Hemoglobin transfusion Deliver oxygen / [159]
Erythropoietin (EPO) Stimulate hemoglobin production / [161]
Perfluorocarbons Deliver oxygen / [162]
Hyperbaric oxygen Deliver oxygen / [163]
Carbogen breathing Promotes oxygen delivery and increases blood flow / [164]
Hyperthermia Increases tumor blood flow and perfusion / [165, 166]
Atovaquone Alleviate tumor hypoxia / [167, 168]
Papaverine Alleviate tumor hypoxia / [169]
Metformin Decreases the rate of tumoral mitochondrial oxygen consumption NCT04275713, Phase II;
NCT02394652, Phase II;
NCT03510390, Phase N/A
[170]
2. Target HIF and HIF-related hypoxia signaling
PT2385 HIF-2α antagonist / [172]
Belzutifan (PT2977) HIF-2α antagonist / [173]
HIF-related pathways inhibitors Inhibit HIF mediated hypoxia regulatory pathway / [174178]
2-OG oxygenase inhibitors Inhibit 2-OG oxygenase / [179]
USP37 HIF-2 deubiquitinase / [181]
3. Hypoxia-activated prodrugs
Evofosfamide (TH-302) Hypoxia specific cytotoxins NCT03098160, Phase II [185]
E09 (Apaziquone) Hypoxia specific cytotoxins NCT00461591, Phase III;
NCT00598806, Phase III
[186]
Others Hypoxia specific cytotoxins NCT02096354, Phase II;
NCT02454842, Phase II;
NCT02449681, Phase II;
NCT00394628, Phase I/II;
NCT01037556, Phase I/II
[187, 188]
4. Target other hypoxia-associated biological processes and pathways
Girentuximab CAIX inhibitor NCT00087022, Phase III [192]
SLC-0111 CAIX inhibitor NCT02215850, Phase I [193]
AZD3965 MCT1 inhibitor NCT01791595, Phase I [194]
AZ93 MCT4 inhibitor / [195]
CB-839 Glutaminase inhibitor / [198]
Aminooxyacetate Aminotransferase inhibitor / [199]
Adenosine pathway inhibitors Inhibitors of the adenosine pathway / [200]

5.1. Modify tumor oxygenation in TME

The most straightforward approach to tackle the tumor hypoxia is to increase the oxygen level in the hypoxic tumor region and this could be achieved from various perspectives. Under physiological conditions, the oxygen supply is positively correlated with concentration of the oxygen carrier hemoglobin. Attempts have been made by performing hemoglobin transfusion to improve radiotherapy outcomes in the hemoglobin low-level patients [159], however, the trials failed probably due to a transient effect of the oxygen elevation and adaptation in the tumors [160]. Other similar approaches were also examined including using erythropoietin (EPO) to stimulate hemoglobin production [161], or using artificial blood substances like perfluorocarbons to deliver oxygen [162]. Increasing oxygen delivery is an actionable strategy to improve tumor hypoxia, which leads to the clinical trials to treat patients with hyperbaric oxygen and achieved significant local tumor control [163]. Besides, carbogen breathing (95% oxygen + 5% carbon dioxide) was used as an alternative of hyperbaric oxygen treatment, as it promotes oxygen delivery via vasodilation and increases blood flow stimulated by carbon dioxide [164]. Several studies suggest that the mild Hyperthermia, a physical therapy by applying heat (40 to 45 °C) to treat tumors, could increase tumor blood flow and perfusion and thus increase oxygen delivery [165, 166]. Due to the technical difficulties to achieve cytotoxic temperatures or overcome tissue thermotolerance, Hyperthermia is mainly used as adjuvant therapy in combination with chemotherapy or radiotherapy. Nevertheless, none of these approaches above has yet reached a controllable clinical outcome so far. Apart from increasing oxygenation, suppressing oxygen consumption is the other side of the coin to eliminate tumor hypoxia. Numerous inhibitors targeting the mitochondrial electron transport chain (ETC) complexes have been developed, including the FDA-approved drugs Atovaquone and Papaverine, which recently have been found to alleviate tumor hypoxia in preclinical models [167169]. Metformin is another widely used drug to decrease the rate of tumoral mitochondrial oxygen consumption, although its original usage is to reduce insulin availability for the treatment of type 2 diabetes [170]. There are several ongoing clinical trials using metformin as a tumor oxygenating agent such as two trials for treating cervical cancer (NCT04275713 and NCT02394652, Phase II) and one for treating head and neck squamous cell carcinoma (NCT03510390).

5.2. Target HIF and HIF-related hypoxia signalling

In the hypoxia signaling cascades, HIF is the central hub that connects the upstream oxygen sensing and downstream cellular adaptation. In addition, expression of HIF is also significantly upregulated after radiation or chemotherapy, suggesting HIF pathways are also actively involved in the treatment resistance [171]. Therefore, HIF is served as a pivotal target for intervention. However, due to the nature of HIF-αs as transcription factors, it has been challenging to develop agents to target this transcriptional factor complex. Great progress has been achieved during that last decade. One of the most successful cases is the development of HIF-2α specific antagonists such as PT2385 [172] and Belzutifan (PT2977) [173], while the latter one has been approved by FDA in 2021 for adult patients with von Hippel-Lindau disease including advanced-stage ccRCC. Beside HIF-2α inhibitors, there are also many HIF-1 inhibitors under development and several novel agents have undergone clinical evaluation [2]. Other approaches targeting HIF signaling, including a variety of inhibitors to interfere with mRNA expression, protein synthesis, DNA binding, or transcriptional activity of HIF, have been summarized previously and will not be discussed here [174].

Apart from targeting HIF directly, intervention of the HIF-related hypoxia signaling is also strongly actionable. Indeed, almost every step in the HIF mediated hypoxia regulatory pathway is targetable. These HIF-related pathways include but not be limited to PI3K/mTOR signaling [175], topoisomerases [176], heat shock proteins [177], and histone deacetylases [178]. Numerous inhibitors have been developed and tested preclinically or clinically for targeting these pathways, with some of them have been reviewed previously [2, 179]. On the other hand, the identification of novel HIF regulators could provide an extra layer to intervene hypoxia. For example, some of the 2-OG oxygenase family such as the well-known HIF hydroxylase EglNs (or PHDs) or perhaps other new members, which regulate HIF post-translational modifications and protein stability, are ideal druggable targets to disturb the hypoxia/HIF signaling. Some PHD inhibitors such as Roxadustat were used for treating patients with chemotherapy-induced anemia [180]. It is worth mentioning that the safety of these PHD inhibitors should be carefully evaluated in patients with cancer since they act as HIF activators and may complicate the treatment outcome. Nevertheless, various 2-OG oxygenase inhibitors in the disease contexts have been highlighted previously [179], and new small compounds are under investigation. Recent studies indicate USP37 is a HIF-2α deubiquitinase mediating its protein level through reverses the degradation process in ccRCC [181], and ZHX2 as a novel HIF-1 regulator controlling tumor progression in triple-negative breast cancer [182]. While how to translate these novel upstream targets in combating the hypoxia within TME is unclear and warrants further investigation.

5.3. Hypoxia-activated prodrugs

It remains attractive to specifically target tumor hypoxia while sparing the normoxic surrounding tissue, which motivates the development of a class of hypoxia specific cytotoxins so called the hypoxia-activated prodrugs (HAPs). These compounds were elegantly designed and believed to precisely target the hypoxic region of tumors, owing to their property of selective reduction by endogenous oxidoreductases to yield active cytotoxic agents only under hypoxic conditions [183]. Several HAPs have been developed and are under extensive preclinical or clinical investigation either alone or in combination with conventional radiotherapy, chemotherapy, and immunotherapy [184]. One example is evofosfamide (TH-302) which has been shown to be non-lymphotoxic and thus well applied in combination with immunotherapy such as the blockade of PD-1 and CTLA-4 [185]. The effectiveness of TH-302 has been documented in preclinical models that can induce CD8+ T cells into hypoxic tumor region, meanwhile decreasing MDSCs population and reducing suppressive myeloid stroma [185]. The therapeutic efficacy of TH-302 in combination with ipilimumab against multiple cancers is under a Phase II clinical evaluation (NCT03098160). The mitomycin C derivative prodrug, E09 (Apaziquone), is another representative HAP which has shown efficacy in bladder cancer with two Phase III clinical trials (NCT00461591 and NCT00598806) ongoing [186]. Other representative HAPs that are under clinical evaluation including RRx-001 (NCT02096354, Phase II), TH-4000 (Tarloxotinib) (NCT02454842/NCT02449681, Phase II), AQ4N (NCT00394628, Phase I/II), and PR-104 (NCT01037556, Phase I/II). HAPs derive from various chemical classes and have diverse mechanisms of action, but the fundamental notion is that the reduction of these agents is typically initiated by donating a single election (1e) which leads to generation of a free radical intermediate as an oxygen sensor. A common challenge in development of HAPs is that many chemical compounds such as quinones, nitro, and N-oxides are also subject to concerted two-electron (2e) reduction thus bypassing the oxygen-sensing radical intermediate and leading to an issue of poor selectivity. It has been observed hypoxia-independent 2e reduction events of HAPs, such as PR-104A by aldo-keto reductase 1C3 and Apaziquone by NADPH: quinone oxidoreductase 1 [187, 188], which may potentially hinder their clinical application.

5.4. Target other hypoxia-associated biological processes and pathways

Given the profound impact of hypoxia that gives rise to tumor cells and TME, a broad range of biological processes and signaling pathways are regarded as the direct or indirect consequence of hypoxia. These elicit further potential therapeutic opportunities for treating hypoxic tumors including targeting hypoxia-associated biological processes like acidosis and metabolic dysregulation in the TME, or inhibiting hypoxic related pathways such as the unfolded protein response (UPR), and the DNA damage response (DDR). Due to the space limitation, the UPR and DDR pathways will not be summarized here, detailed therapeutic intervention can be found in previous review [189].

Acidosis in the TME highly parallels with hypoxia. It is mainly induced by intertumoral high rates of glucose anaerobic catabolism, lactate, H+ and carbon dioxide production and secretion, leading to a hostile acidic environment. While tumor cells are more tolerable to acidosis than non-malignant cells due to high expression of pH-regulating proteins and thus provide selective therapeutic targets. Extracellular- facing carbonic anhydrase 9 (CAIX), which encodes by CA9, is one of the representative pH-regulating proteins and attractive therapeutic targets. Not only does high CA9 expression predict worse patient prognosis [190], but also it is strongly upregulated by HIF-1 [191]. Many CAIX targeting drugs have been developed and some of them were at the clinical stage, including Girentuximab, a monoclonal antibody for treating kidney cancer (NCT00087022, Phase III) [192] and SLC-0111, a small-molecule compound for treating advanced solid tumors (NCT02215850, Phase I) [193]. Other drugs targeting tumor acidosis include the lactate transporter inhibitors, such as MCT1 inhibitor AZD3965 (NCT01791595, Phase I) [194], and MCT4 inhibitor AZ93 [195].

As described previously, metabolic reprogramming is a vital consequence of hypoxia within the TME. Therefore, targeting related metabolic enzymes or intermediates in dysregulated pathways could be a remarkable choice to overcome the hypoxic effect. Glycolytic inhibitors such as 2-deoxy-glucose have been efficacy in tumor cells, while the caveat is that they also abrogate the function of immune cells which will limit their further application [196]. Targeting the MCT1/2 lactate transporter by AZD3965 is an alternative way to neutralize the waste metabolite production of glycolysis, and there are phase I clinical trials undergoing for several cancers [197]. In addition, many cancer cells are highly glutamine-dependent, inhibiting glutamine metabolism by targeting the glutaminase (specific inhibitor such as CB-839) or the aminotransferase (specific inhibitor aminooxyacetate) reveals therapeutic efficacy in tumors with hypoxic traits [198, 199]. Finally, inhibition of the hypoxia induced extracellular adenosine signaling has made remarkable progress and is considered as a ‘next-generation’ therapeutics in immuno-oncology [155]. Several inhibitors of the adenosine pathway have been developed and entered clinical trials over the past few years. The drugs that either target CD73, CD39, or adenosine receptors have been systematically summarized elsewhere [200].

6. Conclusion and perspective

In conclusion, hypoxia related research remains one of most dynamic and exciting directions in cancer research. The 2019 Nobel Prize in Medicine or Physiology on hypoxia and oxygen sensing signaling marks the beginning of new era in this line of research. Targeting hypoxia signaling in cancer will be further developed from several different perspectives: 1) targeting upstream oxygen sensing signaling, including canonical oxygen sensors such as prolyl hydroxylases and other 2-OG dependent dioxygenases; 2) targeting hypoxia/VHL loss induced therapeutic vulnerabilities, including HIF signaling, synthetic lethality targets, hypoxia prodrugs and 3) targeting hypoxia signaling pathways in combination with immunotherapies in tumor microenvironment.

Acknowledgments

We thank all members from Qing Zhang lab for the discussion and suggestions during preparing the manuscript. Due to space limitation, we apologize for many other outstanding studies not summarized or cited in this review. This review is supported by the National Cancer Institute (R01CA211732 and R21CA223675), American Cancer Society (ACS) Research Scholar Award (RSG TBE-132187), Department of Defense Kidney Cancer Research Program (W81XWH1910813) and Cancer Prevention and Research Institute of Texas (CPRIT, RP190058 to Q.Z). Q.Z is an American Cancer Society Research Scholar, CPRIT Scholar in Cancer Research, V Scholar, Kimmel Scholar, Susan G. Komen Career Catalyst awardee and Mary Kay Foundation awardee. Q.Z is also supported by Kidney Cancer Research Alliance (KCCure).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Competing Interest

The authors declare no competing interests.

References

  • [1].Anderson NM, Simon MC, The tumor microenvironment, Curr Biol 30(16) (2020) R921–R925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Liao CH, Zhang Q, ASIP COTRAN EARLY CAREER INVESTIGATOR AWARD LECTURE Understanding the Oxygen-Sensing Pathway and Its Therapeutic Implications in Diseases, Am J Pathol 190(8) (2020) 1584–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Zhang Q, Yan Q, Yang H, Wei W, Oxygen sensing and adaptability won the 2019 Nobel Prize in Physiology or medicine, Genes Dis 6(4) (2019) 328–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Singleton DC, Macann A, Wilson WR, Therapeutic targeting of the hypoxic tumour microenvironment, Nat Rev Clin Oncol 18(12) (2021) 751–772. [DOI] [PubMed] [Google Scholar]
  • [5].Vaupel P, Harrison L, Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response, The oncologist 9 Suppl 5 (2004) 4–9. [DOI] [PubMed] [Google Scholar]
  • [6].Carmeliet P, Jain RK, Angiogenesis in cancer and other diseases, Nature 407(6801) (2000) 249–57. [DOI] [PubMed] [Google Scholar]
  • [7].Brown JM, Wilson WR, Exploiting tumour hypoxia in cancer treatment, Nature reviews. Cancer 4(6) (2004) 437–47. [DOI] [PubMed] [Google Scholar]
  • [8].Vaupel P, Mayer A, Hypoxia in cancer: significance and impact on clinical outcome, Cancer metastasis reviews 26(2) (2007) 225–39. [DOI] [PubMed] [Google Scholar]
  • [9].Conley SJ, Gheordunescu E, Kakarala P, Newman B, Korkaya H, Heath AN, Clouthier SG, Wicha MS, Antiangiogenic agents increase breast cancer stem cells via the generation of tumor hypoxia, Proceedings of the National Academy of Sciences of the United States of America 109(8) (2012) 2784–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Semenza GL, Regulation of the breast cancer stem cell phenotype by hypoxia-inducible factors, Clin Sci (Lond) 129(12) (2015) 1037–45. [DOI] [PubMed] [Google Scholar]
  • [11].Carmeliet P, VEGF as a key mediator of angiogenesis in cancer, Oncology-Basel 69 (2005) 4–10. [DOI] [PubMed] [Google Scholar]
  • [12].Liao D, Johnson RS, Hypoxia: a key regulator of angiogenesis in cancer, Cancer Metastasis Rev 26(2) (2007) 281–90. [DOI] [PubMed] [Google Scholar]
  • [13].Quaegebeur A, Carmeliet P, Oxygen sensing: a common crossroad in cancer and neurodegeneration, Current topics in microbiology and immunology 345 (2010) 71–103. [DOI] [PubMed] [Google Scholar]
  • [14].Walsh JC, Lebedev A, Aten E, Madsen K, Marciano L, Kolb HC, The clinical importance of assessing tumor hypoxia: relationship of tumor hypoxia to prognosis and therapeutic opportunities, Antioxidants & redox signaling 21(10) (2014) 1516–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Liu X, Zurlo G, Zhang Q, The Roles of Cullin-2 E3 Ubiquitin Ligase Complex in Cancer, Advances in experimental medicine and biology 1217 (2020) 173–186. [DOI] [PubMed] [Google Scholar]
  • [16].Mole DR, Blancher C, Copley RR, Pollard PJ, Gleadle JM, Ragoussis J, Ratcliffe PJ, Genome-wide association of hypoxia-inducible factor (HIF)-1alpha and HIF-2alpha DNA binding with expression profiling of hypoxia-inducible transcripts, The Journal of biological chemistry 284(25) (2009) 16767–16775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Xia X, Lemieux ME, Li W, Carroll JS, Brown M, Liu XS, Kung AL, Integrative analysis of HIF binding and transactivation reveals its role in maintaining histone methylation homeostasis, Proceedings of the National Academy of Sciences of the United States of America 106(11) (2009) 4260–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Semenza GL, Targeting HIF-1 for cancer therapy, Nature reviews. Cancer 3(10) (2003) 721–32. [DOI] [PubMed] [Google Scholar]
  • [19].Shen C, Beroukhim R, Schumacher SE, Zhou J, Chang M, Signoretti S, Kaelin WG Jr., Genetic and functional studies implicate HIF1alpha as a 14q kidney cancer suppressor gene, Cancer Discov 1(3) (2011) 222–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Cowman SJ, Fuja DG, Liu XD, Tidwell RSS, Kandula N, Sirohi D, Agarwal AM, Emerson LL, Tripp SR, Mohlman JS, Stonhill M, Garcia G, Conley CJ, Olalde AA, Sargis T, Ramirez-Torres A, Karam JA, Wood CG, Sircar K, Tamboli P, Boucher K, Maughan B, Spike BT, Ho TH, Agarwal N, Jonasch E, Koh MY, Macrophage HIF-1alpha Is an Independent Prognostic Indicator in Kidney Cancer, Clin Cancer Res 26(18) (2020) 4970–4982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Hu CJ, Sataur A, Wang L, Chen H, Simon MC, The N-terminal transactivation domain confers target gene specificity of hypoxia-inducible factors HIF-1alpha and HIF-2alpha, Molecular biology of the cell 18(11) (2007) 4528–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Luo W, Chang R, Zhong J, Pandey A, Semenza GL, Histone demethylase JMJD2C is a coactivator for hypoxia-inducible factor 1 that is required for breast cancer progression, Proceedings of the National Academy of Sciences of the United States of America 109(49) (2012) E3367–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Zurlo G, Guo J, Takada M, Wei W, Zhang Q, New Insights into Protein Hydroxylation and Its Important Role in Human Diseases, Biochimica et biophysica acta 1866(2) (2016) 208–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Liu Q, Geng H, Xue C, Beer TM, Qian DZ, Functional regulation of hypoxia inducible factor-1alpha by SET9 lysine methyltransferase, Biochimica et biophysica acta 1853(5) (2015) 881–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Ortmann BM, Burrows N, Lobb IT, Arnaiz E, Wit N, Bailey PSJ, Jordon LH, Lombardi O, Penalver A, McCaffrey J, Seear R, Mole DR, Ratcliffe PJ, Maxwell PH, Nathan JA, The HIF complex recruits the histone methyltransferase SET1B to activate specific hypoxia-inducible genes, Nature genetics 53(7) (2021) 1022–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Zhang J, Wu T, Simon J, Takada M, Saito R, Fan C, Liu XD, Jonasch E, Xie L, Chen X, Yao X, Teh BT, Tan P, Zheng X, Li M, Lawrence C, Fan J, Geng J, Liu X, Hu L, Wang J, Liao C, Hong K, Zurlo G, Parker JS, Auman JT, Perou CM, Rathmell WK, Kim WY, Kirschner MW, Kaelin WG Jr., Baldwin AS, Zhang Q, VHL substrate transcription factor ZHX2 as an oncogenic driver in clear cell renal cell carcinoma, Science 361(6399) (2018) 290–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Iliopoulos O, Kibel A, Gray S, Kaelin WG Jr., Tumour suppression by the human von Hippel-Lindau gene product, Nature medicine 1(8) (1995) 822–6. [DOI] [PubMed] [Google Scholar]
  • [28].Kibel A, Iliopoulos O, DeCaprio JA, Kaelin WG Jr., Binding of the von Hippel-Lindau tumor suppressor protein to Elongin B and C, Science 269(5229) (1995) 1444–6. [DOI] [PubMed] [Google Scholar]
  • [29].Cho H, Du X, Rizzi JP, Liberzon E, Chakraborty AA, Gao W, Carvo I, Signoretti S, Bruick RK, Josey JA, Wallace EM, Kaelin WG, On-target efficacy of a HIF-2alpha antagonist in preclinical kidney cancer models, Nature 539(7627) (2016) 107–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Mizukami Y, Li J, Zhang X, Zimmer MA, Iliopoulos O, Chung DC, Hypoxia-inducible factor-1-independent regulation of vascular endothelial growth factor by hypoxia in colon cancer, Cancer research 64(5) (2004) 1765–72. [DOI] [PubMed] [Google Scholar]
  • [31].Cummins EP, Taylor CT, Hypoxia-responsive transcription factors, Pflugers Archiv : European journal of physiology 450(6) (2005) 363–71. [DOI] [PubMed] [Google Scholar]
  • [32].Elvidge GP, Glenny L, Appelhoff RJ, Ratcliffe PJ, Ragoussis J, Gleadle JM, Concordant regulation of gene expression by hypoxia and 2-oxoglutarate-dependent dioxygenase inhibition: the role of HIF-1alpha, HIF-2alpha, and other pathways, The Journal of biological chemistry 281(22) (2006) 15215–26. [DOI] [PubMed] [Google Scholar]
  • [33].Liu X, Simon JM, Xie H, Hu L, Wang J, Zurlo G, Fan C, Ptacek TS, Herring L, Tan X, Li M, Baldwin AS, Kim WY, Wu T, Kirschner MW, Gong K, Zhang Q, Genome-wide Screening Identifies SFMBT1 as an Oncogenic Driver in Cancer with VHL Loss, Molecular cell 77(6) (2020) 1294–1306 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Casciello F, Al-Ejeh F, Kelly G, Brennan DJ, Ngiow SF, Young A, Stoll T, Windloch K, Hill MM, Smyth MJ, Gannon F, Lee JS, G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis, Proceedings of the National Academy of Sciences of the United States of America 114(27) (2017) 7077–7082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Lee DC, Sohn HA, Park ZY, Oh S, Kang YK, Lee KM, Kang M, Jang YJ, Yang SJ, Hong YK, Noh H, Kim JA, Kim DJ, Bae KH, Kim DM, Chung SJ, Yoo HS, Yu DY, Park KC, Yeom YI, A lactate-induced response to hypoxia, Cell 161(3) (2015) 595–609. [DOI] [PubMed] [Google Scholar]
  • [36].Fan S, Wang J, Yu G, Rong F, Zhang D, Xu C, Du J, Li Z, Ouyang G, Xiao W, TET is targeted for proteasomal degradation by the PHD-pVHL pathway to reduce DNA hydroxymethylation, The Journal of biological chemistry 295(48) (2020) 16299–16313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Wang X, Hu J, Fang Y, Fu Y, Liu B, Zhang C, Feng S, Lu X, Multi-Omics Profiling to Assess Signaling Changes upon VHL Restoration and Identify Putative VHL Substrates in Clear Cell Renal Cell Carcinoma Cell Lines, Cells 11(3) (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Hoefflin R, Harlander S, Abhari BA, Peighambari A, Adlesic M, Seidel P, Zodel K, Haug S, Gocmen B, Li Y, Lahrmann B, Grabe N, Heide D, Boerries M, Kottgen A, Heikenwalder M, Frew IJ, Therapeutic Effects of Inhibition of Sphingosine-1-Phosphate Signaling in HIF-2alpha Inhibitor-Resistant Clear Cell Renal Cell Carcinoma, Cancers 13(19) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Cockman ME, Lippl K, Tian YM, Pegg HB, Figg WDJ, Abboud MI, Heilig R, Fischer R, Myllyharju J, Schofield CJ, Ratcliffe PJ, Lack of activity of recombinant HIF prolyl hydroxylases (PHDs) on reported non-HIF substrates, Elife 8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Guo J, Chakraborty AA, Liu P, Gan W, Zheng X, Inuzuka H, Wang B, Zhang J, Zhang L, Yuan M, Novak J, Cheng JQ, Toker A, Signoretti S, Zhang Q, Asara JM, Kaelin WG Jr., Wei W, pVHL suppresses kinase activity of Akt in a proline-hydroxylation-dependent manner, Science 353(6302) (2016) 929–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Roe JS, Kim H, Lee SM, Kim ST, Cho EJ, Youn HD, p53 stabilization and transactivation by a von Hippel-Lindau protein, Molecular cell 22(3) (2006) 395–405. [DOI] [PubMed] [Google Scholar]
  • [42].Hu L, Xie H, Liu X, Potjewyd F, James LI, Wilkerson EM, Herring LE, Xie L, Chen X, Cabrera JC, Hong K, Liao C, Tan X, Baldwin AS, Gong K, Zhang Q, TBK1 Is a Synthetic Lethal Target in Cancer with VHL Loss, Cancer discovery 10(3) (2020) 460–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Losman JA, Koivunen P, Kaelin WG Jr., 2-Oxoglutarate-dependent dioxygenases in cancer, Nature reviews. Cancer 20(12) (2020) 710–726. [DOI] [PubMed] [Google Scholar]
  • [44].Losman JA, Kaelin WG Jr., What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer, Genes & development 27(8) (2013) 836–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Gallipoli P, Huntly BJP, Histone modifiers are oxygen sensors, Science 363(6432) (2019) 1148–1149. [DOI] [PubMed] [Google Scholar]
  • [46].Batie M, Frost J, Frost M, Wilson JW, Schofield P, Rocha S, Hypoxia induces rapid changes to histone methylation and reprograms chromatin, Science 363(6432) (2019) 1222–1226. [DOI] [PubMed] [Google Scholar]
  • [47].Chakraborty AA, Laukka T, Myllykoski M, Ringel AE, Booker MA, Tolstorukov MY, Meng YJ, Meier SR, Jennings RB, Creech AL, Herbert ZT, McBrayer SK, Olenchock BA, Jaffe JD, Haigis MC, Beroukhim R, Signoretti S, Koivunen P, Kaelin WG Jr., Histone demethylase KDM6A directly senses oxygen to control chromatin and cell fate, Science 363(6432) (2019) 1217–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].McDonald DM, Choyke PL, Imaging of angiogenesis: from microscope to clinic, Nat Med 9(6) (2003) 713–25. [DOI] [PubMed] [Google Scholar]
  • [49].Nussenbaum F, Herman IM, Tumor angiogenesis: insights and innovations, J Oncol 2010 (2010) 132641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Shi R, Tang YQ, Miao H, Metabolism in tumor microenvironment: Implications for cancer immunotherapy, MedComm (2020) 1(1) (2020) 47–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I, The hypoxic tumour microenvironment, Oncogenesis 7(1) (2018) 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Li Y, Zhao L, Li XF, Hypoxia and the Tumor Microenvironment, Technol Cancer Res Treat 20 (2021) 15330338211036304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Gilkes DM, Semenza GL, Wirtz D, Hypoxia and the extracellular matrix: drivers of tumour metastasis, Nat Rev Cancer 14(6) (2014) 430–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Lu P, Weaver VM, Werb Z, The extracellular matrix: a dynamic niche in cancer progression, J Cell Biol 196(4) (2012) 395–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Parodi M, Raggi F, Cangelosi D, Manzini C, Balsamo M, Blengio F, Eva A, Varesio L, Pietra G, Moretta L, Mingari MC, Vitale M, Bosco MC, Hypoxia Modifies the Transcriptome of Human NK Cells, Modulates Their Immunoregulatory Profile, and Influences NK Cell Subset Migration, Front Immunol 9 (2018) 2358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Roy DG, Kaymak I, Williams KS, Ma EH, Jones RG, Immunometabolism in the Tumor Microenvironment, Annu Rev Canc Biol 5 (2021) 137–159. [Google Scholar]
  • [57].Elia I, Haigis MC, Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism, Nat Metab 3(1) (2021) 21–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Bhandari V, Hoey C, Liu LY, Lalonde E, Ray J, Livingstone J, Lesurf R, Shiah YJ, Vujcic T, Huang X, Espiritu SMG, Heisler LE, Yousif F, Huang V, Yamaguchi TN, Yao CQ, Sabelnykova VY, Fraser M, Chua MLK, van der Kwast T, Liu SK, Boutros PC, Bristow RG, Molecular landmarks of tumor hypoxia across cancer types, Nat Genet 51(2) (2019) 308–318. [DOI] [PubMed] [Google Scholar]
  • [59].Davis L, Recktenwald M, Hutt E, Fuller S, Briggs M, Goel A, Daringer N, Targeting HIF-2 alpha in the Tumor Microenvironment: Redefining the Role of HIF-2 alpha for Solid Cancer Therapy, Cancers 14(5) (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Hanahan D, Coussens LM, Accessories to the crime: functions of cells recruited to the tumor microenvironment, Cancer Cell 21(3) (2012) 309–22. [DOI] [PubMed] [Google Scholar]
  • [61].Parks SK, Chiche J, Pouyssegur J, Disrupting proton dynamics and energy metabolism for cancer therapy, Nat Rev Cancer 13(9) (2013) 611–23. [DOI] [PubMed] [Google Scholar]
  • [62].Maman S, Witz IP, A history of exploring cancer in context, Nat Rev Cancer 18(6) (2018) 359–376. [DOI] [PubMed] [Google Scholar]
  • [63].Folkman J, Role of angiogenesis in tumor growth and metastasis, Semin Oncol 29(6 Suppl 16) (2002) 15–8. [DOI] [PubMed] [Google Scholar]
  • [64].Bielenberg DR, Zetter BR, The Contribution of Angiogenesis to the Process of Metastasis, Cancer J 21(4) (2015) 267–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Yoshida D, Kim K, Noha M, Teramoto A, Hypoxia inducible factor 1-alpha regulates of platelet derived growth factor-B in human glioblastoma cells, J Neurooncol 76(1) (2006) 13–21. [DOI] [PubMed] [Google Scholar]
  • [66].Kimura H, Weisz A, Kurashima Y, Hashimoto K, Ogura T, D’Acquisto F, Addeo R, Makuuchi M, Esumi H, Hypoxia response element of the human vascular endothelial growth factor gene mediates transcriptional regulation by nitric oxide: control of hypoxia-inducible factor-1 activity by nitric oxide, Blood 95(1) (2000) 189–197. [PubMed] [Google Scholar]
  • [67].Garayoa M, Martinez A, Lee S, Pio R, An WG, Neckers L, Trepel J, Montuenga LM, Ryan H, Johnson R, Gassmann M, Cuttitta F, Hypoxia-inducible factor-1 (HIF-1) up-regulates adrenomedullin expression in human tumor cell lines during oxygen deprivation: a possible promotion mechanism of carcinogenesis, Mol Endocrinol 14(6) (2000) 848–62. [DOI] [PubMed] [Google Scholar]
  • [68].Mizukami Y, Jo WS, Duerr EM, Gala M, Li JN, Zhang XB, Zimmer MA, Iliopoulos O, Zukerberg LR, Kohgo Y, Lynch MP, Rueda BR, Chung DC, Induction of interleukin-8 preserves the angiogenic response in HIF-1 alpha-deficient colon cancer cells, Nature Medicine 11(9) (2005) 992–997. [DOI] [PubMed] [Google Scholar]
  • [69].Hielscher A, Qiu C, Porterfield J, Smith Q, Gerecht S, Hypoxia Affects the Structure of Breast Cancer Cell-Derived Matrix to Support Angiogenic Responses of Endothelial Cells, J Carcinog Mutagen Suppl 13 (2013) 005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A, Maggiolini M, HIF-1alpha/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs), Breast Cancer Res 15(4) (2013) R64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Peng PH, Hsu KW, Chieh-Yu Lai J, Wu KJ, The role of hypoxia-induced long noncoding RNAs (lncRNAs) in tumorigenesis and metastasis, Biomed J 44(5) (2021) 521–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Luo Y, Yang J, Yu J, Liu X, Yu C, Hu J, Shi H, Ma X, Long Non-coding RNAs: Emerging Roles in the Immunosuppressive Tumor Microenvironment, Front Oncol 10 (2020) 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Guo Y, Xie Y, Luo Y, The Role of Long Non-Coding RNAs in the Tumor Immune Microenvironment, Front Immunol 13 (2022) 851004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].De Martino M, Esposito F, Pallante P, Long non-coding RNAs regulating multiple proliferative pathways in cancer cell, Transl Cancer Res 10(6) (2021) 3140–3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Liu H, Luo J, Luan S, He C, Li Z, Long non-coding RNAs involved in cancer metabolic reprogramming, Cellular and molecular life sciences : CMLS 76(3) (2019) 495–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Liu LX, Zhang Y, Lu J, The roles of long noncoding RNAs in breast cancer metastasis, Cell Death & Disease 11(9) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Jiang W, Agrawal DK, Boosani CS, Non-coding RNAs as Epigenetic Gene Regulators in Cardiovascular Diseases, Adv Exp Med Biol 1229 (2020) 133–148. [DOI] [PubMed] [Google Scholar]
  • [78].Zhang X, Wang W, Zhu W, Dong J, Cheng Y, Yin Z, Shen F, Mechanisms and Functions of Long Non-Coding RNAs at Multiple Regulatory Levels, Int J Mol Sci 20(22) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Kuo TC, Kung HJ, Shih JW, Signaling in and out: long-noncoding RNAs in tumor hypoxia, J Biomed Sci 27(1) (2020) 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Barreca MM, Zichittella C, Alessandro R, Conigliaro A, Hypoxia-Induced Non-Coding RNAs Controlling Cell Viability in Cancer, Int J Mol Sci 22(4) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].McCarty G, Loeb DM, Hypoxia-sensitive epigenetic regulation of an antisense-oriented lncRNA controls WT1 expression in myeloid leukemia cells, PLoS One 10(3) (2015) e0119837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, Chang HY, Long noncoding RNA as modular scaffold of histone modification complexes, Science 329(5992) (2010) 689–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Shih JW, Chiang WF, Wu ATH, Wu MH, Wang LY, Yu YL, Hung YW, Wang WC, Chu CY, Hung CL, Changou CA, Yen Y, Kung HJ, Long noncoding RNA LncHIFCAR/MIR31HG is a HIF-1alpha co-activator driving oral cancer progression, Nat Commun 8 (2017) 15874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Wen X, Liu X, Mao YP, Yang XJ, Wang YQ, Zhang PP, Lei Y, Hong XH, He QM, Ma J, Liu N, Li YQ, Long non-coding RNA DANCR stabilizes HIF-1alpha and promotes metastasis by interacting with NF90/NF45 complex in nasopharyngeal carcinoma, Theranostics 8(20) (2018) 5676–5689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ma MZ, Kong X, Weng MZ, Zhang MD, Qin YY, Gong W, Zhang WJ, Quan ZW, Long non-coding RNA-LET is a positive prognostic factor and exhibits tumor-suppressive activity in gallbladder cancer, Mol Carcinog 54(11) (2015) 1397–406. [DOI] [PubMed] [Google Scholar]
  • [86].Bertozzi D, Iurlaro R, Sordet O, Marinello J, Zaffaroni N, Capranico G, Characterization of novel antisense HIF-1alpha transcripts in human cancers, Cell Cycle 10(18) (2011) 3189–97. [DOI] [PubMed] [Google Scholar]
  • [87].Chen WM, Huang MD, Kong R, Xu TP, Zhang EB, Xia R, Sun M, De W, Shu YQ, Antisense Long Noncoding RNA HIF1A-AS2 Is Upregulated in Gastric Cancer and Associated with Poor Prognosis, Dig Dis Sci 60(6) (2015) 1655–62. [DOI] [PubMed] [Google Scholar]
  • [88].Li L, Wang M, Mei Z, Cao W, Yang Y, Wang Y, Wen A, lncRNAs HIF1A-AS2 facilitates the up-regulation of HIF-1alpha by sponging to miR-153–3p, whereby promoting angiogenesis in HUVECs in hypoxia, Biomed Pharmacother 96 (2017) 165–172. [DOI] [PubMed] [Google Scholar]
  • [89].Pathania AS, Challagundla KB, Exosomal Long Non-coding RNAs: Emerging Players in the Tumor Microenvironment, Mol Ther Nucleic Acids 23 (2021) 1371–1383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Yang P, Ding J, Bian Y, Ma Z, Wang K, Li J, Long non-coding RNAs and cancer mechanisms: Immune cells and inflammatory cytokines in the tumor microenvironment, Med Oncol 39(7) (2022) 108. [DOI] [PubMed] [Google Scholar]
  • [91].Entezari M, Ghanbarirad M, Taheriazam A, Sadrkhanloo M, Zabolian A, Goharrizi M, Hushmandi K, Aref AR, Ashrafizadeh M, Zarrabi A, Nabavi N, Rabiee N, Hashemi M, Samarghandian S, Long non-coding RNAs and exosomal lncRNAs: Potential functions in lung cancer progression, drug resistance and tumor microenvironment remodeling, Biomed Pharmacother 150 (2022) 112963. [DOI] [PubMed] [Google Scholar]
  • [92].Chen D, Lu T, Tan J, Li H, Wang Q, Wei L, Long Non-coding RNAs as Communicators and Mediators Between the Tumor Microenvironment and Cancer Cells, Front Oncol 9 (2019) 739. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Lin YH, Wu MH, Yeh CT, Lin KH, Long Non-Coding RNAs as Mediators of Tumor Microenvironment and Liver Cancer Cell Communication, Int J Mol Sci 19(12) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Huarte M, The emerging role of lncRNAs in cancer, Nat Med 21(11) (2015) 1253–61. [DOI] [PubMed] [Google Scholar]
  • [95].Wu M, Zhang X, Han X, Pandey V, Lobie PE, Zhu T, The potential of long noncoding RNAs for precision medicine in human cancer, Cancer Lett 501 (2021) 12–19. [DOI] [PubMed] [Google Scholar]
  • [96].Rivadeneira DB, Delgoffe GM, Antitumor T-cell Reconditioning: Improving Metabolic Fitness for Optimal Cancer Immunotherapy, Clin Cancer Res 24(11) (2018) 2473–2481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Taylor CT, Colgan SP, Regulation of immunity and inflammation by hypoxia in immunological niches, Nat Rev Immunol 17(12) (2017) 774–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Jayaprakash P, Ai M, Liu A, Budhani P, Bartkowiak T, Sheng J, Ager C, Nicholas C, Jaiswal AR, Sun Y, Shah K, Balasubramanyam S, Li N, Wang G, Ning J, Zal A, Zal T, Curran MA, Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy, J Clin Invest 128(11) (2018) 5137–5149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Yan M, Jene N, Byrne D, Millar EKA, O’Toole SA, McNeil CM, Bates GJ, Harris AL, Banham AH, Sutherland RL, Fox SB, Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers, Breast Cancer Research 13(2) (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC, HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption, Cell Metab 3(3) (2006) 187–97. [DOI] [PubMed] [Google Scholar]
  • [101].Kim JW, Tchernyshyov I, Semenza GL, Dang CV, HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia, Cell Metab 3(3) (2006) 177–85. [DOI] [PubMed] [Google Scholar]
  • [102].Palazon A, Tyrakis PA, Macias D, Velica P, Rundqvist H, Fitzpatrick S, Vojnovic N, Phan AT, Loman N, Hedenfalk I, Hatschek T, Lovrot J, Foukakis T, Goldrath AW, Bergh J, Johnson RS, An HIF-1alpha/VEGF-A Axis in Cytotoxic T Cells Regulates Tumor Progression, Cancer Cell 32(5) (2017) 669–683 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Gropper Y, Feferman T, Shalit T, Salame TM, Porat Z, Shakhar G, Culturing CTLs under Hypoxic Conditions Enhances Their Cytolysis and Improves Their Anti-tumor Function, Cell Rep 20(11) (2017) 2547–2555. [DOI] [PubMed] [Google Scholar]
  • [104].Park JH, Kim HJ, Kim CW, Kim HC, Jung Y, Lee HS, Lee Y, Ju YS, Oh JE, Park SH, Lee JH, Lee SK, Lee HK, Tumor hypoxia represses gammadelta T cell-mediated antitumor immunity against brain tumors, Nat Immunol 22(3) (2021) 336–346. [DOI] [PubMed] [Google Scholar]
  • [105].Ngambenjawong C, Gustafson HH, Pun SH, Progress in tumor-associated macrophage (TAM)-targeted therapeutics, Adv Drug Deliv Rev 114 (2017) 206–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Pan Y, Yu Y, Wang X, Zhang T, Tumor-Associated Macrophages in Tumor Immunity, Front Immunol 11 (2020) 583084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Henze AT, Mazzone M, The impact of hypoxia on tumor-associated macrophages, J Clin Invest 126(10) (2016) 3672–3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Diaz-Bulnes P, Saiz ML, Lopez-Larrea C, Rodriguez RM, Crosstalk Between Hypoxia and ER Stress Response: A Key Regulator of Macrophage Polarization, Frontiers in Immunology 10 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Elia AR, Cappello P, Puppo M, Fraone T, Vanni C, Eva A, Musso T, Novelli F, Varesio L, Giovarelli M, Human dendritic cells differentiated in hypoxia down-modulate antigen uptake and change their chemokine expression profile, J Leukocyte Biol 84(6) (2008) 1472–1482. [DOI] [PubMed] [Google Scholar]
  • [110].Tran CW, Gold MJ, Garcia-Batres C, Tai K, Elford AR, Himmel ME, Elia AJ, Ohashi PS, Hypoxia-inducible factor 1 alpha limits dendritic cell stimulation of CD8 T cell immunity, PLoS One 15(12) (2020) e0244366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Vetsika EK, Koukos A, Kotsakis A, Myeloid-Derived Suppressor Cells: Major Figures that Shape the Immunosuppressive and Angiogenic Network in Cancer, Cells-Basel 8(12) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Zheng X, Qian Y, Fu B, Jiao D, Jiang Y, Chen P, Shen Y, Zhang H, Sun R, Tian Z, Wei H, Mitochondrial fragmentation limits NK cell-based tumor immunosurveillance, Nat Immunol 20(12) (2019) 1656–1667. [DOI] [PubMed] [Google Scholar]
  • [113].Barsoum IB, Smallwood CA, Siemens DR, Graham CH, A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells, Cancer Res 74(3) (2014) 665–74. [DOI] [PubMed] [Google Scholar]
  • [114].Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, Bronte V, Chouaib S, PD-L1 is a novel direct target of HIF-1 alpha., and its blockade under hypoxia enhanced MDSC-mediated T cell activation, J Exp Med 211(5) (2014) 781–790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Wang SC, Li YH, Piao HL, Hong XW, Zhang D, Xu YY, Tao Y, Wang Y, Yuan MM, Li DJ, Du MR, PD-1 and Tim-3 pathways are associated with regulatory CD8(+) T-cell function in decidua and maintenance of normal pregnancy, Cell Death Dis 6 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Topalian SL, Taube JM, Anders RA, Pardoll DM, Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy, Nat Rev Cancer 16(5) (2016) 275–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Okazawa H, Motegi SI, Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, Oldenborg PA, Ishikawa O, Matozaki T, Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system, J Immunol 174(4) (2005) 2004–2011. [DOI] [PubMed] [Google Scholar]
  • [118].Pengam S, Durand J, Usal C, Gauttier V, Dilek N, Martinet B, Daguin V, Mary C, Thepenier V, Teppaz G, Renaudin K, Blancho G, Vanhove B, Poirier N, SIRPalpha/CD47 axis controls the maintenance of transplant tolerance sustained by myeloid-derived suppressor cells, Am J Transplant 19(12) (2019) 3263–3275. [DOI] [PubMed] [Google Scholar]
  • [119].Zhang HM, Lu HQ, Xiang LS, Bullen JW, Zhang CZ, Samanta D, Gilkes DM, He JJ, Semenza GL, HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells, P Natl Acad Sci USA 112(45) (2015) E6215–E6223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Kaur S, Chang T, Singh SP, Lim L, Mannan P, Garfield SH, Pendrak ML, Soto-Pantoja DR, Rosenberg AZ, Jin S, Roberts DD, CD47 signaling regulates the immunosuppressive activity of VEGF in T cells, J Immunol 193(8) (2014) 3914–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Pavlova NN, Thompson CB, The Emerging Hallmarks of Cancer Metabolism, Cell Metab 23(1) (2016) 27–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Vander Heiden MG, Cantley LC, Thompson CB, Understanding the Warburg effect: the metabolic requirements of cell proliferation, Science 324(5930) (2009) 1029–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Semenza GL, HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations, J Clin Invest 123(9) (2013) 3664–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Bacigalupa ZA, Rathmell WK, Beyond glycolysis: Hypoxia signaling as a master regulator of alternative metabolic pathways and the implications in clear cell renal cell carcinoma, Cancer Lett 489 (2020) 19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Chen CH, Pore N, Behrooz A, Ismail-Beigi F, Maity A, Regulation of glut1 mRNA by hypoxia-inducible factor-1 - Interaction between H-ras and hypoxia, J Biol Chem 276(12) (2001) 9519–9525. [DOI] [PubMed] [Google Scholar]
  • [126].Mimura I, Nangaku M, Kanki Y, Tsutsumi S, Inoue T, Kohro T, Yamamoto S, Fujita T, Shimamura T, Suehiro J, Taguchi A, Kobayashi M, Tanimura K, Inagaki T, Tanaka T, Hamakubo T, Sakai J, Aburatani H, Kodama T, Wada Y, Dynamic change of chromatin conformation in response to hypoxia enhances the expression of GLUT3 (SLC2A3) by cooperative interaction of hypoxia-inducible factor 1 and KDM3A, Mol Cell Biol 32(15) (2012) 3018–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW, Hypoxia-inducible factor (HIF-1)alpha: its protein stability and biological functions, Exp Mol Med 36(1) (2004) 1–12. [DOI] [PubMed] [Google Scholar]
  • [128].Semenza GL, HIF-1: upstream and downstream of cancer metabolism, Curr Opin Genet Dev 20(1) (2010) 51–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Luo WB, Hu HX, Chang R, Zhong J, Knabel M, O’Meally R, Cole RN, Pandey A, Semenza GL, Pyruvate Kinase M2 Is a PHD3-Stimulated Coactivator for Hypoxia-Inducible Factor 1, Cell 145(5) (2011) 732–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Le A, Cooper CR, Gouw AM, Dinavahi R, Maitra A, Deck LM, Royer RE, Vander Jagt DL, Semenza GL, Dang CV, Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression, Proc Natl Acad Sci U S A 107(5) (2010) 2037–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Cui XG, Han ZT, He SH, Wu XD, Chen TR, Shao CH, Chen DL, Su N, Chen YM, Wang T, Wang J, Song DW, Yan WJ, Yang XH, Liu T, Wei HF, Xiao J, HIF1/2alpha mediates hypoxia-induced LDHA expression in human pancreatic cancer cells, Oncotarget 8(15) (2017) 24840–24852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Fan J, Hitosugi T, Chung TW, Xie J, Ge Q, Gu TL, Polakiewicz RD, Chen GZ, Boggon TJ, Lonial S, Khuri FR, Kang S, Chen J, Tyrosine phosphorylation of lactate dehydrogenase A is important for NADH/NAD(+) redox homeostasis in cancer cells, Mol Cell Biol 31(24) (2011) 4938–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Zhang H, Gao P, Fukuda R, Kumar G, Krishnamachary B, Zeller KI, Dang CV, Semenza GL, HIF-1 inhibits mitochondrial biogenesis and cellular respiration in VHL-deficient renal cell carcinoma by repression of C-MYC activity, Cancer Cell 11(5) (2007) 407–20. [DOI] [PubMed] [Google Scholar]
  • [134].Chiu DKC, Tse APW, Law CT, Xu IMJ, Lee D, Chen MN, Lai RKH, Yuen VWH, Cheu JWS, Ho DWH, Wong CM, Zhang HF, Ng IOL, Wong CCL, Hypoxia regulates the mitochondrial activity of hepatocellular carcinoma cells through HIF/HEY1/PINK1 pathway, Cell Death Dis 10 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Peck B, Ferber EC, Schulze A, Antagonism between FOXO and MYC regulates cellular powerhouse, Front Oncol 3 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Hamanaka RB, Chandel NS, Mitochondrial reactive oxygen species regulate hypoxic signaling, Curr Opin Cell Biol 21(6) (2009) 894–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Rao X, Duan X, Mao W, Li X, Li Z, Li Q, Zheng Z, Xu H, Chen M, Wang PG, Wang Y, Shen B, Yi W, O-GlcNAcylation of G6PD promotes the pentose phosphate pathway and tumor growth, Nat Commun 6 (2015) 8468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Mullarky E, Lucki NC, Beheshti Zavareh R, Anglin JL, Gomes AP, Nicolay BN, Wong JC, Christen S, Takahashi H, Singh PK, Blenis J, Warren JD, Fendt SM, Asara JM, DeNicola GM, Lyssiotis CA, Lairson LL, Cantley LC, Identification of a small molecule inhibitor of 3-phosphoglycerate dehydrogenase to target serine biosynthesis in cancers, Proc Natl Acad Sci U S A 113(7) (2016) 1778–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen QY, Gindin M, Gubin MM, van der Windt GJW, Tonc E, Schreiber RD, Pearce EJ, Pearce EL, Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression, Cell 162(6) (2015) 1229–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Huang D, Li TT, Li XH, Zhang L, Sun LC, He XP, Zhong XY, Jia DY, Song LB, Semenza GL, Gao P, Zhang HF, HIF-1-Mediated Suppression of Acyl-CoA Dehydrogenases and Fatty Acid Oxidation Is Critical for Cancer Progression, Cell Rep 8(6) (2014) 1930–1942. [DOI] [PubMed] [Google Scholar]
  • [141].Du W, Zhang L, Brett-Morris A, Aguila B, Kerner J, Hoppel CL, Puchowicz M, Serra D, Herrero L, Rini BI, Campbell S, Welford SM, HIF drives lipid deposition and cancer in ccRCC via repression of fatty acid metabolism, Nat Commun 8(1) (2017) 1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Metallo CM, Gameiro PA, Bell EL, Mattaini KR, Yang JJ, Hiller K, Jewell CM, Johnson ZR, Irvine DJ, Guarente L, Kelleher JK, Vander Heiden MG, Iliopoulos O, Stephanopoulos G, Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia, Nature 481(7381) (2012) 380–U166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Warburg O, On the origin of cancer cells, Science 123(3191) (1956) 309–14. [DOI] [PubMed] [Google Scholar]
  • [144].Parks SK, Pouyssegur J, Targeting pH regulating proteins for cancer therapy-Progress and limitations, Semin Cancer Biol 43 (2017) 66–73. [DOI] [PubMed] [Google Scholar]
  • [145].Sun SR, Li H, Chen JH, Qian Q, Lactic Acid: No Longer an Inert and End-Product of Glycolysis, Physiology 32(6) (2017) 453–463. [DOI] [PubMed] [Google Scholar]
  • [146].Wang ZH, Peng WB, Zhang P, Yang XP, Zhou Q, Lactate in the tumour microenvironment: From immune modulation to therapy, EBioMedicine 73 (2021) 103627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Hirschhaeuser F, Sattler UG, Mueller-Klieser W, Lactate: a metabolic key player in cancer, Cancer Res 71(22) (2011) 6921–5. [DOI] [PubMed] [Google Scholar]
  • [148].Ho PC, Bihuniak JD, Macintyre AN, Staron M, Liu XJ, Amezquita R, Tsui YC, Cui GL, Micevic G, Perales JC, Kleinstein SH, Abel ED, Insogna KL, Feske S, Locasale JW, Bosenberg MW, Rathmell JC, Kaech SM, Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses, Cell 162(6) (2015) 1217–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, Sugiura A, Cohen AS, Ali A, Do BT, Muir A, Lewis CA, Hongo RA, Young KL, Brown RE, Todd VM, Huffstater T, Abraham A, O’Neil RT, Wilson MH, Xin FX, Tantawy MN, Merryman WD, Johnson RW, Williams CS, Mason EF, Mason FM, Beckermann KE, Vander Heiden MG, Manning HC, Rathmell JC, Rathmell WK, Cell-programmed nutrient partitioning in the tumour microenvironment, Nature 593(7858) (2021) 282-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Klysz D, Tai XG, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz C, Floess S, Fritz V, Matias MI, Yong C, Surh N, Marie JC, Huehn J, Zimmermann V, Kinet S, Dardalhon V, Taylor N, Glutamine-dependent alpha-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation, Sci Signal 8(396) (2015). [DOI] [PubMed] [Google Scholar]
  • [151].Leone RD, Zhao L, Englert JM, Sun IM, Oh MH, Sun IH, Arwood ML, Bettencourt IA, Patel CH, Wen JY, Tam A, Blosser RL, Prchalova E, Alt J, Rais R, Slusher BS, Powell JD, Glutamine blockade induces divergent metabolic programs to overcome tumor immune evasion, Science 366(6468) (2019) 1013-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Sun RC, Denko NC, Hypoxic Regulation of Glutamine Metabolism through HIF1 and SIAH2 Supports Lipid Synthesis that Is Necessary for Tumor Growth, Cell Metabolism 19(2) (2014) 285–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Metallo CM, Gameiro PA, Bell EL, Mattaini KR, Yang J, Hiller K, Jewell CM, Johnson ZR, Irvine DJ, Guarente L, Kelleher JK, Vander Heiden MG, Iliopoulos O, Stephanopoulos G, Reductive glutamine metabolism by IDH1 mediates lipogenesis under hypoxia, Nature 481(7381) (2011) 380–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Metzler B, Gfeller P, Guinet E, Restricting Glutamine or Glutamine-Dependent Purine and Pyrimidine Syntheses Promotes Human T Cells with High FOXP3 Expression and Regulatory Properties, J Immunol 196(9) (2016) 3618–3630. [DOI] [PubMed] [Google Scholar]
  • [155].Vijayan D, Young A, Teng MWL, Smyth MJ, Targeting immunosuppressive adenosine in cancer (vol 17, pg 709, 2017), Nature Reviews Cancer 17(12) (2017) 765–765. [DOI] [PubMed] [Google Scholar]
  • [156].Synnestvedt K, Furuta GT, Comerford KM, Louis N, Karhausen J, Eltzschig HK, Hansen KR, Thompson LF, Colgan SP, Ecto-5’-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia, J Clin Invest 110(7) (2002) 993–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Tak E, Jung DH, Kim SH, Park GC, Jun DY, Lee J, Jung BH, Kirchner VA, Hwang S, Song GW, Lee SG, Protective role of hypoxia-inducible factor-1alpha-dependent CD39 and CD73 in fulminant acute liver failure, Toxicol Appl Pharmacol 314 (2017) 72–81. [DOI] [PubMed] [Google Scholar]
  • [158].Kong T, Westerman KA, Faigle M, Eltzschig HK, Colgan SP, HIF-dependent induction of adenosine A2B receptor in hypoxia, FASEB J 20(13) (2006) 2242–50. [DOI] [PubMed] [Google Scholar]
  • [159].Grogan M, Thomas GM, Melamed I, Wong FL, Pearcey RG, Joseph PK, Portelance L, Crook J, Jones KD, The importance of hemoglobin levels during radiotherapy for carcinoma of the cervix, Cancer 86(8) (1999) 1528–36. [DOI] [PubMed] [Google Scholar]
  • [160].Hirst DG, Anemia - a Problem or an Opportunity in Radiotherapy, Int J Radiat Oncol 12(11) (1986) 2009–2017. [DOI] [PubMed] [Google Scholar]
  • [161].Lavey RS, Dempsey WH, Erythropoietin increases hemoglobin in cancer patients during radiation therapy, Int J Radiat Oncol Biol Phys 27(5) (1993) 1147–52. [DOI] [PubMed] [Google Scholar]
  • [162].Rockwell S, Use of a Perfluorochemical Emulsion to Improve Oxygenation in a Solid Tumor, Int J Radiat Oncol 11(1) (1985) 97–103. [DOI] [PubMed] [Google Scholar]
  • [163].Henk JM, Kunkler PB, Smith CW, Radiotherapy and hyperbaric oxygen in head and neck cancer. Final report of first controlled clinical trial, Lancet 2(8029) (1977) 101–3. [DOI] [PubMed] [Google Scholar]
  • [164].Chaplin DJ, Horsman MR, Siemann DW, Further Evaluation of Nicotinamide and Carbogen as a Strategy to Reoxygenate Hypoxic Cells in-Vivo - Importance of Nicotinamide Dose and Preirradation Breathing Time, Brit J Cancer 68(2) (1993) 269–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Song CW, Park H, Griffin RJ, Improvement of tumor oxygenation by mild hyperthermia, Radiat Res 155(4) (2001) 515–528. [DOI] [PubMed] [Google Scholar]
  • [166].Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Dodge RK, Charles HC, Samulski TV, Prosnitz LR, Dewhirst MW, Radiation therapy and hyperthermia improve the oxygenation of human soft tissue sarcomas, Cancer Research 56(23) (1996) 5347–5350. [PubMed] [Google Scholar]
  • [167].Ashton TM, Fokas E, Kunz-Schughart LA, Folkes LK, Anbalagan S, Huether M, Kelly CJ, Pirovano G, Buffa FM, Hammond EM, Stratford M, Muschel RJ, Higgins GS, McKenna WG, The anti-malarial atovaquone increases radiosensitivity by alleviating tumour hypoxia, Nat Commun 7 (2016) 12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Skwarski M, McGowan DR, Belcher E, Di Chiara F, Stavroulias D, McCole M, Derham JL, Chu KY, Teoh E, Chauhan J, O’Reilly D, Harris BHL, Macklin PS, Bull JA, Green M, Rodriguez-Berriguete G, Prevo R, Folkes LK, Campo L, Ferencz P, Croal PL, Flight H, Qi C, Holmes J, O’Connor JPB, Gleeson FV, McKenna WG, Harris AL, Bulte D, Buffa FM, Macpherson RE, Higgins GS, Mitochondrial Inhibitor Atovaquone Increases Tumor Oxygenation and Inhibits Hypoxic Gene Expression in Patients with Non-Small Cell Lung Cancer, Clin Cancer Res 27(9) (2021) 2459–2469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Benej M, Hong XQ, Vibhute S, Scott S, Wu JH, Graves E, Le QT, Koong AC, Giaccia AJ, Yu B, Chen SC, Papandreou I, Denko NC, Papaverine and its derivatives radiosensitize solid tumors by inhibiting mitochondrial metabolism (vol 115, pg 10756, 2018), P Natl Acad Sci USA 115(49) (2018) E11561–E11561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Pernicova I, Korbonits M, Metformin--mode of action and clinical implications for diabetes and cancer, Nat Rev Endocrinol 10(3) (2014) 143–56. [DOI] [PubMed] [Google Scholar]
  • [171].Lee P, Chandel NS, Simon MC, Cellular adaptation to hypoxia through hypoxia inducible factors and beyond, Nat Rev Mol Cell Bio 21(5) (2020) 268–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Courtney KD, Infante JR, Lam ET, Figlin RA, Rini BI, Brugarolas J, Zojwalla NJ, Lowe AM, Wang KS, Wallace EM, Josey JA, Choueiri TK, Phase I Dose-Escalation Trial of PT2385, a First-in-Class Hypoxia-Inducible Factor-2 alpha Antagonist in Patients With Previously Treated Advanced Clear Cell Renal Cell Carcinoma, J Clin Oncol 36(9) (2018) 867-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Choueiri TK, Bauer TM, Papadopoulos KP, Plimack ER, Merchan JR, McDermott DF, Michaelson MD, Appleman LJ, Thamake S, Perini RF, Zojwalla NJ, Jonasch E, Inhibition of hypoxia-inducible factor-2alpha in renal cell carcinoma with belzutifan: a phase 1 trial and biomarker analysis, Nat Med 27(5) (2021) 802–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Fallah J, Rini BI, HIF Inhibitors: Status of Current Clinical Development, Curr Oncol Rep 21(1) (2019). [DOI] [PubMed] [Google Scholar]
  • [175].Mohlin S, Hamidian A, von Stedingk K, Bridges E, Wigerup C, Bexell D, Pahlman S, PI3K-mTORC2 but not PI3K-mTORC1 Regulates Transcription of HIF2A/EPAS1 and Vascularization in Neuroblastoma, Cancer Research 75(21) (2015) 4617–4628. [DOI] [PubMed] [Google Scholar]
  • [176].Rapisarda A, Uranchimeg B, Sordet O, Pommier Y, Shoemaker RH, Melillo G, Topoisomerase I-mediated inhibition of hypoxia-inducible factor 1: Mechanism and therapeutic implications, Cancer Research 64(4) (2004) 1475–1482. [DOI] [PubMed] [Google Scholar]
  • [177].Baird NA, Turnbull DW, Johnson EA, Induction of the heat shock pathway during hypoxia requires regulation of heat shock factor by hypoxia-inducible factor-1, J Biol Chem 281(50) (2006) 38675–38681. [DOI] [PubMed] [Google Scholar]
  • [178].Liang DM, Kong XG, Sang NL, Effects of histone deacetylase inhibitors on HIF-1, Cell Cycle 5(21) (2006) 2430–2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Shi R, Liao CH, Zhang Q, Hypoxia-Driven Effects in Cancer: Characterization, Mechanisms, and Therapeutic Implications, Cells-Basel 10(3) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Glaspy JA, Gabrail NY, Locantore-Ford PA, Saha GC, Hardy E, Lee T, Modelska K, Henry DH, Open-label, phase 2 study of roxadustat for treatment of anemia in patients receiving chemotherapy for non-myeloid malignancies., J Clin Oncol 40(16) (2022). [DOI] [PubMed] [Google Scholar]
  • [181].Hong K, Hu L, Liu X, Simon JM, Ptacek TS, Zheng X, Liao C, Baldwin AS, Zhang Q, USP37 promotes deubiquitination of HIF2alpha in kidney cancer, Proc Natl Acad Sci U S A 117(23) (2020) 13023–13032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Fang WT, Liao CH, Shi R, Simon JM, Ptacek TS, Zurlo G, Ye YQ, Han L, Fan C, Bao L, Ortiz CL, Lin HR, Manocha U, Luo WB, Peng Y, Kim WY, Yang LW, Zhang Q, ZHX2 promotes HIF1 alpha oncogenic signaling in triple-negative breast cancer, Elife 10 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Denny WA, The role of hypoxia-activated prodrugs in cancer therapy, Lancet Oncol 1(1) (2000) 25–9. [DOI] [PubMed] [Google Scholar]
  • [184].Mistry IN, Thomas M, Calder EDD, Conway SJ, Hammond EM, Clinical Advances of Hypoxia-Activated Prodrugs in Combination With Radiation Therapy, Int J Radiat Oncol 98(5) (2017) 1183–1196. [DOI] [PubMed] [Google Scholar]
  • [185].Jayaprakash P, Ai MD, Liu A, Budhani P, Bartkowiak T, Sheng J, Ager C, Nicholas C, Jaiswal AR, Sun YQ, Shah K, Balasubramanyam S, Li N, Wang GC, Ning J, Zal A, Zal T, Curran MA, Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy, Journal of Clinical Investigation 128(11) (2018) 5137–5149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Hendricksen K, Cornel EB, de Reijke TM, Arentsen HC, Chawla S, Witjes JA, Phase 2 Study of Adjuvant Intravesical Instillations of Apaziquone for High Risk Nonmuscle Invasive Bladder Cancer, J Urology 187(4) (2012) 1195–1199. [DOI] [PubMed] [Google Scholar]
  • [187].Guise CP, Abbattista MR, Singleton RS, Holford SD, Connolly J, Dachs GU, Fox SB, Pollock R, Harvey J, Guilford P, Donate F, Wilson WR, Patterson AV, The Bioreductive Prodrug PR-104A Is Activated under Aerobic Conditions by Human Aldo-Keto Reductase 1C3, Cancer Research 70(4) (2010) 1573–1584. [DOI] [PubMed] [Google Scholar]
  • [188].Bailey SM, Lewis AD, Patterson LH, Fisher GR, Knox RJ, Workman P, Involvement of NADPH: cytochrome P450 reductase in the activation of indoloquinone EO9 to free radical and DNA damaging species, Biochem Pharmacol 62(4) (2001) 461–468. [DOI] [PubMed] [Google Scholar]
  • [189].Wilson WR, Hay MP, Targeting hypoxia in cancer therapy, Nature Reviews Cancer 11(6) (2011) 393–410. [DOI] [PubMed] [Google Scholar]
  • [190].van Kuijk SJA, Yaromina A, Houben R, Niemans R, Lambin P, Dubois LJ, Prognostic Significance of Carbonic Anhydrase IX expression in Cancer Patients: A Meta-Analysis, Front Oncol 6 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Pastorekova S, Gillies RJ, The role of carbonic anhydrase IX in cancer development: links to hypoxia, acidosis, and beyond, Cancer Metast Rev 38(1–2) (2019) 65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Chamie K, Donin NM, Klopfer P, Bevan P, Fall B, Wilhelm O, Storkel S, Said J, Gambla M, Hawkins RE, Jankilevich G, Kapoor A, Kopyltsov E, Staehler M, Taari K, Wainstein AJA, Pantuck AJ, Belldegrun AS, Adjuvant Weekly Girentuximab Following Nephrectomy for High-Risk Renal Cell Carcinoma The ARISER Randomized Clinical Trial, Jama Oncol 3(7) (2017) 913–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].McDonald PC, Chia S, Bedard PL, Chu Q, Lyle M, Tang LR, Singh M, Zhang ZH, Supuran CT, Renouf DJ, Dedhar S, A Phase 1 Study of SLC-0111, a Novel Inhibitor of Carbonic Anhydrase IX, in Patients With Advanced Solid Tumors, Am J Clin Oncol-Canc 43(7) (2020) 484–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Bola BM, Chadwick AL, Michopoulos F, Blount KG, Telfer BA, Williams KJ, Smith PD, Critchlow SE, Stratford IJ, Inhibition of Monocarboxylate Transporter-1 (MCT1) by AZD3965 Enhances Radiosensitivity by Reducing Lactate Transport, Mol Cancer Ther 13(12) (2014) 2805–2816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Marchiq I, Pouyssegur J, Hypoxia, cancer metabolism and the therapeutic benefit of targeting lactate/H+ symporters, J Mol Med 94(2) (2016) 155–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Renner K, Geiselhoringer AL, Fante M, Bruss C, Farber S, Schonhammer G, Peter K, Singer K, Andreesen R, Hoffmann P, Oefner P, Herr W, Kreutz M, Metabolic plasticity of human T cells: Preserved cytokine production under glucose deprivation or mitochondrial restriction, but 2-deoxy-glucose affects effector functions, Eur J Immunol 45(9) (2015) 2504–16. [DOI] [PubMed] [Google Scholar]
  • [197].Renner K, Singer K, Koehl GE, Geissler EK, Peter K, Siska PJ, Kreutz M, Metabolic Hallmarks of Tumor and Immune Cells in the Tumor Microenvironment, Front Immunol 8 (2017) 248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, Janes JR, Laidig GJ, Lewis ER, Li J, MacKinnon AL, Parlati F, Rodriguez MLM, Shwonek PJ, Sjogren EB, Stanton TF, Wang TT, Yang JF, Zhao F, Bennett MK, Antitumor Activity of the Glutaminase Inhibitor CB-839 in Triple-Negative Breast Cancer, Mol Cancer Ther 13(4) (2014) 890–901. [DOI] [PubMed] [Google Scholar]
  • [199].Korangath P, Teo WW, Sadik H, Han LF, Mori N, Huijts CM, Wildes F, Bharti S, Zhang Z, Santa-Maria CA, Tsai HL, Dang CV, Stearns V, Bhujwalla ZM, Sukumar S, Targeting Glutamine Metabolism in Breast Cancer with Aminooxyacetate, Clinical Cancer Research 21(14) (2015) 3263–3273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Allard B, Allard D, Buisseret L, Stagg J, The adenosine pathway in immuno-oncology, Nat Rev Clin Oncol 17(10) (2020) 611–629. [DOI] [PubMed] [Google Scholar]
  • [201].Dahia PL, Pheochromocytomas and Paragangliomas, Genetically Diverse and Minimalist, All at Once!, Cancer cell 31(2) (2017) 159–161. [DOI] [PubMed] [Google Scholar]
  • [202].Eltzschig HK, Eckle T, Grenz A, PHD2 mutation and congenital erythrocytosis with paraganglioma, The New England journal of medicine 360(13) (2009) 1361–2; author reply 1362. [DOI] [PubMed] [Google Scholar]
  • [203].Zhang Q, Gu J, Li L, Liu J, Luo B, Cheung HW, Boehm JS, Ni M, Geisen C, Root DE, Polyak K, Brown M, Richardson AL, Hahn WC, Kaelin WG Jr., Bommi-Reddy A, Control of cyclin D1 and breast tumorigenesis by the EglN2 prolyl hydroxylase, Cancer cell 16(5) (2009) 413–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Henze AT, Garvalov BK, Seidel S, Cuesta AM, Ritter M, Filatova A, Foss F, Dopeso H, Essmann CL, Maxwell PH, Reifenberger G, Carmeliet P, Acker-Palmer A, Acker T, Loss of PHD3 allows tumours to overcome hypoxic growth inhibition and sustain proliferation through EGFR, Nature communications 5 (2014) 5582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Lee AR, Lee S, Shin JY, Kim JY, Moon KS, Jung J, Biomarker LEPRE1 induces pelitinib-specific drug responsiveness by regulating ABCG2 expression and tumor transition states in human leukemia and lung cancer, Scientific reports 12(1) (2022) 2928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Cimmino L, Dawlaty MM, Ndiaye-Lobry D, Yap YS, Bakogianni S, Yu Y, Bhattacharyya S, Shaknovich R, Geng H, Lobry C, Mullenders J, King B, Trimarchi T, Aranda-Orgilles B, Liu C, Shen S, Verma AK, Jaenisch R, Aifantis I, TET1 is a tumor suppressor of hematopoietic malignancy, Nature immunology 16(6) (2015) 653–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Neri F, Dettori D, Incarnato D, Krepelova A, Rapelli S, Maldotti M, Parlato C, Paliogiannis P, Oliviero S, TET1 is a tumour suppressor that inhibits colon cancer growth by derepressing inhibitors of the WNT pathway, Oncogene 34(32) (2015) 4168–76. [DOI] [PubMed] [Google Scholar]
  • [208].Wu BK, Brenner C, Suppression of TET1-dependent DNA demethylation is essential for KRAS-mediated transformation, Cell reports 9(5) (2014) 1827–1840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Bowman RL, Levine RL, TET2 in Normal and Malignant Hematopoiesis, Cold Spring Harbor perspectives in medicine 7(8) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Gaidzik VI, Paschka P, Spath D, Habdank M, Kohne CH, Germing U, von Lilienfeld-Toal M, Held G, Horst HA, Haase D, Bentz M, Gotze K, Dohner H, Schlenk RF, Bullinger L, Dohner K, TET2 mutations in acute myeloid leukemia (AML): results from a comprehensive genetic and clinical analysis of the AML study group, Journal of clinical oncology : official journal of the American Society of Clinical Oncology 30(12) (2012) 1350–7. [DOI] [PubMed] [Google Scholar]
  • [211].Nickerson ML, Das S, Im KM, Turan S, Berndt SI, Li H, Lou H, Brodie SA, Billaud JN, Zhang T, Bouk AJ, Butcher D, Wang Z, Sun L, Misner K, Tan W, Esnakula A, Esposito D, Huang WY, Hoover RN, Tucker MA, Keller JR, Boland J, Brown K, Anderson SK, Moore LE, Isaacs WB, Chanock SJ, Yeager M, Dean M, Andresson T, TET2 binds the androgen receptor and loss is associated with prostate cancer, Oncogene 36(15) (2017) 2172–2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Carella A, Tejedor JR, Garcia MG, Urdinguio RG, Bayon GF, Sierra M, Lopez V, Garcia-Torano E, Santamarina-Ojeda P, Perez RF, Bigot T, Mangas C, Corte-Torres MD, Saenz-de-Santa-Maria I, Mollejo M, Melendez B, Astudillo A, Chiara MD, Fernandez AF, Fraga MF, Epigenetic downregulation of TET3 reduces genome-wide 5hmC levels and promotes glioblastoma tumorigenesis, International journal of cancer 146(2) (2020) 373–387. [DOI] [PubMed] [Google Scholar]
  • [213].Shen C, Sheng Y, Zhu AC, Robinson S, Jiang X, Dong L, Chen H, Su R, Yin Z, Li W, Deng X, Chen Y, Hu YC, Weng H, Huang H, Prince E, Cogle CR, Sun M, Zhang B, Chen CW, Marcucci G, He C, Qian Z, Chen J, RNA Demethylase ALKBH5 Selectively Promotes Tumorigenesis and Cancer Stem Cell Self-Renewal in Acute Myeloid Leukemia, Cell stem cell 27(1) (2020) 64–80 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Zou D, Dong L, Li C, Yin Z, Rao S, Zhou Q, The m(6)A eraser FTO facilitates proliferation and migration of human cervical cancer cells, Cancer cell international 19 (2019) 321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Li Z, Weng H, Su R, Weng X, Zuo Z, Li C, Huang H, Nachtergaele S, Dong L, Hu C, Qin X, Tang L, Wang Y, Hong GM, Wang X, Chen P, Gurbuxani S, Arnovitz S, Li Y, Li S, Strong J, Neilly MB, Larson RA, Jiang X, Zhang P, Jin J, He C, Chen J, FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N(6)-Methyladenosine RNA Demethylase, Cancer cell 31(1) (2017) 127–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Liu H, Liu L, Holowatyj A, Jiang Y, Yang ZQ, Integrated genomic and functional analyses of histone demethylases identify oncogenic KDM2A isoform in breast cancer, Molecular carcinogenesis 55(5) (2016) 977–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Wagner KW, Alam H, Dhar SS, Giri U, Li N, Wei Y, Giri D, Cascone T, Kim JH, Ye Y, Multani AS, Chan CH, Erez B, Saigal B, Chung J, Lin HK, Wu X, Hung MC, Heymach JV, Lee MG, KDM2A promotes lung tumorigenesis by epigenetically enhancing ERK1/2 signaling, The Journal of clinical investigation 123(12) (2013) 5231–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Tzatsos A, Paskaleva P, Ferrari F, Deshpande V, Stoykova S, Contino G, Wong KK, Lan F, Trojer P, Park PJ, Bardeesy N, KDM2B promotes pancreatic cancer via Polycomb-dependent and -independent transcriptional programs, The Journal of clinical investigation 123(2) (2013) 727–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Frescas D, Guardavaccaro D, Bassermann F, Koyama-Nasu R, Pagano M, JHDM1B/FBXL10 is a nucleolar protein that represses transcription of ribosomal RNA genes, Nature 450(7167) (2007) 309–13. [DOI] [PubMed] [Google Scholar]
  • [220].Xu X, Nagel S, Quentmeier H, Wang Z, Pommerenke C, Dirks WG, Macleod RAF, Drexler HG, Hu Z, KDM3B shows tumor-suppressive activity and transcriptionally regulates HOXA1 through retinoic acid response elements in acute myeloid leukemia, Leukemia & lymphoma 59(1) (2018) 204–213. [DOI] [PubMed] [Google Scholar]
  • [221].Lee DH, Kim GW, Jeon YH, Yoo J, Lee SW, Kwon SH, Advances in histone demethylase KDM4 as cancer therapeutic targets, FASEB journal : official publication of the Federation of American Societies for Experimental Biology 34(3) (2020) 3461–3484. [DOI] [PubMed] [Google Scholar]
  • [222].Wilson C, Qiu L, Hong Y, Karnik T, Tadros G, Mau B, Ma T, Mu Y, New J, Louie RJ, Gunewardena S, Godwin AK, Tawfik OW, Chien J, Roby KF, Krieg AJ, The histone demethylase KDM4B regulates peritoneal seeding of ovarian cancer, Oncogene 36(18) (2017) 2565–2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Wu MC, Cheng HH, Yeh TS, Li YC, Chen TJ, Sit WY, Chuu CP, Kung HJ, Chien S, Wang WC, KDM4B is a coactivator of c-Jun and involved in gastric carcinogenesis, Cell death & disease 10(2) (2019) 68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Berry WL, Shin S, Lightfoot SA, Janknecht R, Oncogenic features of the JMJD2A histone demethylase in breast cancer, International journal of oncology 41(5) (2012) 1701–6. [DOI] [PubMed] [Google Scholar]
  • [225].Liu G, Bollig-Fischer A, Kreike B, van de Vijver MJ, Abrams J, Ethier SP, Yang ZQ, Genomic amplification and oncogenic properties of the GASC1 histone demethylase gene in breast cancer, Oncogene 28(50) (2009) 4491–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Yang ZQ, Imoto I, Fukuda Y, Pimkhaokham A, Shimada Y, Imamura M, Sugano S, Nakamura Y, Inazawa J, Identification of a novel gene, GASC1, within an amplicon at 9p23–24 frequently detected in esophageal cancer cell lines, Cancer research 60(17) (2000) 4735–9. [PubMed] [Google Scholar]
  • [227].Jonasch E, Donskov F, Iliopoulos O, Rathmell WK, Narayan VK, Maughan BL, Oudard S, Else T, Maranchie JK, Welsh SJ, Thamake S, Park EK, Perini RF, Linehan WM, Srinivasan R, Investigators MK, Belzutifan for Renal Cell Carcinoma in von Hippel-Lindau Disease, N Engl J Med 385(22) (2021) 2036–2046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Dai B, Huang H, Guan F, Zhu G, Xiao Z, Mao B, Su H, Hu Z, Histone demethylase KDM5A inhibits glioma cells migration and invasion by down regulating ZEB1, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 99 (2018) 72–80. [DOI] [PubMed] [Google Scholar]
  • [229].Hinohara K, Wu HJ, Vigneau S, McDonald TO, Igarashi KJ, Yamamoto KN, Madsen T, Fassl A, Egri SB, Papanastasiou M, Ding L, Peluffo G, Cohen O, Kales SC, Lal-Nag M, Rai G, Maloney DJ, Jadhav A, Simeonov A, Wagle N, Brown M, Meissner A, Sicinski P, Jaffe JD, Jeselsohn R, Gimelbrant AA, Michor F, Polyak K, KDM5 Histone Demethylase Activity Links Cellular Transcriptomic Heterogeneity to Therapeutic Resistance, Cancer cell 34(6) (2018) 939–953 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Oser MG, Sabet AH, Gao W, Chakraborty AA, Schinzel AC, Jennings RB, Fonseca R, Bonal DM, Booker MA, Flaifel A, Novak JS, Christensen CL, Zhang H, Herbert ZT, Tolstorukov MY, Buss EJ, Wong KK, Bronson RT, Nguyen QD, Signoretti S, Kaelin WG Jr., The KDM5A/RBP2 histone demethylase represses NOTCH signaling to sustain neuroendocrine differentiation and promote small cell lung cancer tumorigenesis, Genes & development 33(23–24) (2019) 1718–1738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Yang GJ, Zhu MH, Lu XJ, Liu YJ, Lu JF, Leung CH, Ma DL, Chen J, The emerging role of KDM5A in human cancer, Journal of hematology & oncology 14(1) (2021) 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [232].Jose A, Shenoy GG, Sunil Rodrigues G, Kumar NAN, Munisamy M, Thomas L, Kolesar J, Rai G, Rao PPN, Rao M, Histone Demethylase KDM5B as a Therapeutic Target for Cancer Therapy, Cancers 12(8) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Zheng Q, Li P, Zhou X, Qiang Y, Fan J, Lin Y, Chen Y, Guo J, Wang F, Xue H, Xiong J, Li F, Deficiency of the X-inactivation escaping gene KDM5C in clear cell renal cell carcinoma promotes tumorigenicity by reprogramming glycogen metabolism and inhibiting ferroptosis, Theranostics 11(18) (2021) 8674–8691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Zhan D, Zhang Y, Xiao P, Zheng X, Ruan M, Zhang J, Chen A, Zou Y, Chen Y, Huang G, Hu S, Wang QF, Zhu X, Whole exome sequencing identifies novel mutations of epigenetic regulators in chemorefractory pediatric acute myeloid leukemia, Leukemia research 65 (2018) 20–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Komura K, Yoshikawa Y, Shimamura T, Chakraborty G, Gerke TA, Hinohara K, Chadalavada K, Jeong SH, Armenia J, Du SY, Mazzu YZ, Taniguchi K, Ibuki N, Meyer CA, Nanjangud GJ, Inamoto T, Lee GM, Mucci LA, Azuma H, Sweeney CJ, Kantoff PW, ATR inhibition controls aggressive prostate tumors deficient in Y-linked histone demethylase KDM5D, The Journal of clinical investigation 128(7) (2018) 2979–2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Wang L, Shilatifard A, UTX Mutations in Human Cancer, Cancer cell 35(2) (2019) 168–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Xie G, Liu X, Zhang Y, Li W, Liu S, Chen Z, Xu B, Yang J, He L, Zhang Z, Jin T, Yi X, Sun L, Shang Y, Liang J, UTX promotes hormonally responsive breast carcinogenesis through feed-forward transcription regulation with estrogen receptor, Oncogene 36(39) (2017) 5497–5511. [DOI] [PubMed] [Google Scholar]
  • [238].Ntziachristos P, Tsirigos A, Welstead GG, Trimarchi T, Bakogianni S, Xu L, Loizou E, Holmfeldt L, Strikoudis A, King B, Mullenders J, Becksfort J, Nedjic J, Paietta E, Tallman MS, Rowe JM, Tonon G, Satoh T, Kruidenier L, Prinjha R, Akira S, Van Vlierberghe P, Ferrando AA, Jaenisch R, Mullighan CG, Aifantis I, Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia, Nature 514(7523) (2014) 513–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Hsia DA, Tepper CG, Pochampalli MR, Hsia EY, Izumiya C, Huerta SB, Wright ME, Chen HW, Kung HJ, Izumiya Y, KDM8, a H3K36me2 histone demethylase that acts in the cyclin A1 coding region to regulate cancer cell proliferation, Proceedings of the National Academy of Sciences of the United States of America 107(21) (2010) 9671–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Wang HJ, Pochampalli M, Wang LY, Zou JX, Li PS, Hsu SC, Wang BJ, Huang SH, Yang P, Yang JC, Chu CY, Hsieh CL, Sung SY, Li CF, Tepper CG, Ann DK, Gao AC, Evans CP, Izumiya Y, Chuu CP, Wang WC, Chen HW, Kung HJ, KDM8/JMJD5 as a dual coactivator of AR and PKM2 integrates AR/EZH2 network and tumor metabolism in CRPC, Oncogene 38(1) (2019) 17–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Trager MH, Sah B, Chen Z, Liu L, Control of Breast Cancer Pathogenesis by Histone Methylation and the Hairless Histone Demethylase, Endocrinology 162(8) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Celik H, Koh WK, Kramer AC, Ostrander EL, Mallaney C, Fisher DAC, Xiang J, Wilson WC, Martens A, Kothari A, Fishberger G, Tycksen E, Karpova D, Duncavage EJ, Lee Y, Oh ST, Challen GA, JARID2 Functions as a Tumor Suppressor in Myeloid Neoplasms by Repressing Self-Renewal in Hematopoietic Progenitor Cells, Cancer cell 34(5) (2018) 741–756 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Wang K, Yang C, Li H, Liu X, Zheng M, Xuan Z, Mei Z, Wang H, Role of the Epigenetic Modifier JMJD6 in Tumor Development and Regulation of Immune Response, Frontiers in immunology 13 (2022) 859893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Zhou J, Simon JM, Liao C, Zhang C, Hu L, Zurlo G, Liu X, Fan C, Hepperla A, Jia L, Tcheuyap VT, Zhong H, Elias R, Ye J, Henne WM, Kapur P, Nijhawan D, Brugarolas J, Zhang Q, An oncogenic JMJD6-DGAT1 axis tunes the epigenetic regulation of lipid droplet formation in clear cell renal cell carcinoma, Mol Cell 82(16) (2022) 3030–3044 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Xuan F, Huang M, Zhao E, Cui H, MINA53 deficiency leads to glioblastoma cell apoptosis via inducing DNA replication stress and diminishing DNA damage response, Cell death & disease 9(11) (2018) 1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Lee KH, Park JW, Sung HS, Choi YJ, Kim WH, Lee HS, Chung HJ, Shin HW, Cho CH, Kim TY, Li SH, Youn HD, Kim SJ, Chun YS, PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer, Oncogene 34(22) (2015) 2897–909. [DOI] [PubMed] [Google Scholar]
  • [247].Sinha KM, Bagheri-Yarmand R, Lahiri S, Lu Y, Zhang M, Amra S, Rizvi Y, Wan X, Navone N, Ozpolat B, Logothetis C, Gagel RF, Huard J, Oncogenic and osteolytic functions of histone demethylase NO66 in castration-resistant prostate cancer, Oncogene 38(25) (2019) 5038–5049. [DOI] [PubMed] [Google Scholar]
  • [248].Zhou W, Gong L, Wu Q, Xing C, Wei B, Chen T, Zhou Y, Yin S, Jiang B, Xie H, Zhou L, Zheng S, PHF8 upregulation contributes to autophagic degradation of E-cadherin, epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma, Journal of experimental & clinical cancer research : CR 37(1) (2018) 215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Kanwal M, Smahel M, Olsen M, Smahelova J, Tachezy R, Aspartate beta-hydroxylase as a target for cancer therapy, Journal of experimental & clinical cancer research : CR 39(1) (2020) 163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Liao C, Zhang Y, Fan C, Herring LE, Liu J, Locasale JW, Takada M, Zhou J, Zurlo G, Hu L, Simon JM, Ptacek TS, Andrianov VG, Loza E, Peng Y, Yang H, Perou CM, Zhang Q, Identification of BBOX1 as a Therapeutic Target in Triple-Negative Breast Cancer, Cancer discovery 10(11) (2020) 1706–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Liao C, Zhang Q, BBOX1 promotes triple-negative breast cancer progression by controlling IP3R3 stability, Mol Cell Oncol 7(6) (2020) 1813526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Wang J, Zhou Y, Zhang D, Zhao W, Lu Y, Liu C, Lin W, Zhang Y, Chen K, Wang H, Zhao L, CRIP1 suppresses BBOX1-mediated carnitine metabolism to promote stemness in hepatocellular carcinoma, EMBO J 41(15) (2022) e110218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Saeed K, Ostling P, Bjorkman M, Mirtti T, Alanen K, Vesterinen T, Sankila A, Lundin J, Lundin M, Rannikko A, Nordling S, Mpindi JP, Kohonen P, Iljin K, Kallioniemi O, Rantala JK, Androgen receptor-interacting protein HSPBAP1 facilitates growth of prostate cancer cells in androgen-deficient conditions, International journal of cancer 136(11) (2015) 2535–45. [DOI] [PubMed] [Google Scholar]
  • [254].Kim JH, Lee SM, Lee JH, Chun S, Kang BH, Kwak S, Roe JS, Kim TW, Kim H, Kim WH, Cho EJ, Youn HD, OGFOD1 is required for breast cancer cell proliferation and is associated with poor prognosis in breast cancer, Oncotarget 6(23) (2015) 19528–41. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES