Abstract
Hypoxia or low oxygen concentration in tumor microenvironment has widespread effects ranging from altered angiogenesis and lymphangiogenesis, tumor metabolism, growth, and therapeutic resistance in different cancer types. A large number of these effects are mediated by the transcription factor hypoxia inducible factor 1⍺ (HIF-1⍺) which is activated by hypoxia. HIF1⍺ induces glycolytic genes and reduces mitochondrial respiration rate in hypoxic tumoral regions through modulation of various cells in tumor microenvironment like cancer-associated fibroblasts. Immune evasion driven by HIF-1⍺ further contributes to enhanced survival of cancer cells. By altering drug target expression, metabolic regulation, and oxygen consumption, hypoxia leads to enhanced growth and survival of cancer cells. Tumor cells in hypoxic conditions thus attain aggressive phenotypes and become resistant to chemo- and radio- therapies resulting in higher mortality. While a number of new therapeutic strategies have succeeded in targeting hypoxia, a significant improvement of these needs a more detailed understanding of the various effects and molecular mechanisms regulated by hypoxia and its effects on modulation of the tumor vasculature. This review focuses on the chief hypoxia-driven molecular mechanisms and their impact on therapeutic resistance in tumors that drive an aggressive phenotype.
Impact statement
Hypoxia contributes to tumor aggressiveness and promotes growth of many solid tumors that are often resistant to conventional therapies. In order to achieve successful therapeutic strategies targeting different cancer types, it is necessary to understand the molecular mechanisms and signaling pathways that are induced by hypoxia. Aberrant tumor vasculature and alterations in cellular metabolism and drug resistance due to hypoxia further confound this problem. This review focuses on the implications of hypoxia in an inflammatory TME and its impact on the signaling and metabolic pathways regulating growth and progression of cancer, along with changes in lymphangiogenic and angiogenic mechanisms. Finally, the overarching role of hypoxia in mediating therapeutic resistance in cancers is discussed.
Keywords: Hypoxia, cancer, HIF1⍺, tumor microenvironment, inflammation, angiogenesis, lymphangiogenesis, chemoresistance, radioresistance
Introduction
Physiological oxygen concentration in tissues or normoxia is hampered during cancer progression that gives rise to a low oxygen concentration in the TME known as hypoxia. The specific oxygen concentration during tumor development differs depending on the tumor tissue type and is on average 1–2% or even less.1 Hypoxia can arise due to an abnormal rate of cell division, development of solid tumor masses that disrupt blood vessels from penetrating and hence a resulting drop in oxygen diffusion.2 Intratumoral hypoxia is characterized by formation of irregular vascular networks and regions within the solid tumor that have areas of low oxygen.3 Hypoxia can be classified as acute hypoxia where cells are exposed to low oxygen concentration for a brief period of a few minutes and chronic hypoxia typically seen in large tumors where low oxygen concentrations are prolonged in TME.4 Acute as well as chronic hypoxia can lead to a number of cellular changes that ultimately contribute to metastasis and tumor-associated chemoresistance or radioresistance.5 These changes range from a metabolic shift towards glycolysis for faster ATP synthesis, induction of anti-apoptotic and pro-tumorigenic signaling pathways, and decreased drug penetration. In addition to chronic and acute, a third kind of hypoxia known as intermittent or cycling hypoxia has also been seen in certain regions of different solid tumors.6 Cyclic hypoxia, defined by periodic sequence of hypoxia and reoxygenation, (H-R cycles) is involved in upregulating angiogenesis, metastasis, immune evasion, and increased resistance to therapy in cancer.6 The differing ramifications of a hypoxic TME on the growth, spread, and therapeutic resistance of cancer cells are profound and make understanding the different facets crucial.7 This review aims to summarize the role of hypoxia in shaping cancer progression and therapeutic tolerance focusing on specific regulated signaling pathways and drug resistance mechanisms.
Hypoxia and cancer progression: Role of hypoxia inducible factor
The various components of the TME such as cancer-associated fibroblasts (CAFs), stromal cells, lymphatic and blood vessels, and immune cells undergo pro-tumorigenic adaptation predominantly mediated by hypoxia inducible factor (HIF) when exposed to hypoxia. HIF proteins are a family of transcription factors that consist of oxygen sensitive members such as HIF-1, HIF-2, and HIF-3 of which HIF-1⍺ is the most widely characterized and studied.2 The HIF-1 and HIF-2 primarily play a role as transcriptional activators and hence have both unique and overlapping target genes, while the function of HIF-3 is a little less clearly understood.8 The transcriptional complex of HIF-1 consisting of HIF-1ß and HIF-1⍺ is constitutively synthesized in normoxic conditions.5 HIF-1⍺ under normoxia is hydroxylated at proline residues 402 and 577 and marked for ubiquitination by a prolyl hydroxylases (PHDs). Subsequently, HIF-1⍺ interacts with the tumor suppressor von Hippel Lindau protein (pVHL) and finally is degraded by 26S proteasome. Under hypoxia, the activity of PHDs which require O2 as a cofactor is reduced and HIF-1⍺ is stabilized in the cytoplasm. Mitochondrial O2 sensors also stabilize HIF-1⍺ by increasing the production of mitochondrial reactive oxygen species (mtROS) which helps in HIF-1⍺ nuclear translocation. Post nuclear translocation HIF-1⍺ forms a heterodimer with aryl hydrocarbon nuclear receptor (ARNT) and transactivates genes which have hypoxia responsive elements (HREs) in their promoter and enhancer regions. HIF2⍺ acts in a similar manner and shares overall 48% amino acid identity.9 Low O2 conditions or hypoxia stabilize HIF-1⍺ by taking advantage of the low availability of O2, an essential PHD substrate needed for PHD-mediated hydroxylation.10,11
HIF switch in hypoxia
Cancer cells shift their dependence between HIF1⍺ and HIF2⍺ based on the level of oxygen concentration available to them. While HIF2⍺ is more stable at higher oxygen concentration and is active in chronic hypoxia, stable and higher levels of HIF1⍺ are observed during acute hypoxia.12,13 These differences in stabilization patterns arise partly from the ease of hydroxylating HIF2⍺ by PHD and asparagynyl hydroxylases (FIH-1) during acutely low oxygen levels.14 Multiple studies point to the temporal regulation of HIF1⍺ or HIF2⍺ levels during hypoxia. This switch from HIF-1 to HIF-2 and HIF-3 signaling is required in order to adapt the endothelium to prolonged hypoxia.8 HIF1⍺ levels are usually highest at 4–8 h after onset of hypoxia after which they decrease and diminish at 18–24 h, while HIF2⍺ levels stabilize much later around 24–72 h of hypoxia.13,15–17 In addition to the ease of hydroxylation of HIF1⍺, ubiquitination by heat shock protein (HSP)-70 complex and ensuing degradation under chronic hypoxia takes place exclusively with HIF1⍺.18,19 miR-429 has been seen to regulate the switch from HIF1⍺ to HIF3⍺ during chronic hypoxia in endothelial cells indicating a potential involvement of a variety of cell type-specific molecular factors that govern hypoxia-mediated effects.20
Cyclic hypoxia and its impact on HIFs
Cyclic or intermittent hypoxia has recently come under increasing scrutiny because of the high levels of HIF1⍺ found in cancer cells during periodic hypoxia.21,22 Stable and enhanced HIF1⍺ levels of cyclic hypoxia have been attributed to multiple mechanisms such as the post-translational modification of HIF1⍺ or high phosphorylated HIF1⍺ levels during intermittent hypoxia takes place possibly through PKA activation in HAMEC-1 and EAhy926 endothelial cells. This heightened level of HIF1⍺ also leads to increased migration and angiogenesis by endothelial cells.15,21,23 The effects of cyclic hypoxia in induction of angiogenesis and migration become important while considering therapeutic resistance of cancer cells. Verduzco et al.24 have shown that exposure to intermittent hypoxia leads to cancer cells having low levels of p53 and E-cadherin and led to increased survival, metastasis, and drug resistance to etoposide. The genes induced by HIFs activate signaling cascades ultimately contributing towards cancer growth, angiogenesis, lymphangiogenesis, metastasis, and resistance to therapeutic intervention.1,9,25–27 Signaling pathways regulated by HIF-1⍺ and consequently hypoxia can be distinguished under the following major areas:
Role of HIF-1⍺ in mitochondrial biogenesis
HIF-1⍺ preferentially shifts dependence of cellular metabolism from mitochondrial respiration to glycolysis. As shown in Figure 1, HIF⍺ proteins play a major role in mitochondrial biogenesis by regulating expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC 1⍺), that is activated by PGC1β, in turn activated by pro-oncogenic transcription factor MYC.28,29 Activated HIF-1⍺ induces MYC-associated factor X(MAX)-interacting protein 1 (MAXI1) expression that represses MYC activity by binding to MYC target genes’ (such as PGC1⍺/β) promoter sites. Additionally, Forkhead-box protein O3a (FOXO3a), activated by hypoxia transmits this inhibitory effect of HIF-1⍺ on MYC by reducing MYC structural stability and upregulating expression of miRNA which interferes with MYC translation.29,30 In cells expressing HIF-2⍺ an opposite effect of hypoxia on MYC activity is observed, HIF2⍺ binds and stabilizes MAX-MYC heterodimer needed to exert downstream effects of MYC. The consequence of HIF2⍺ regulation is thus the pro-tumorigenic regulation of cell cycle proteins.31,32 However, the inhibitory effect on PGC1⍺/β and in extension on mitochondrial biogenesis is primarily mediated by HIF-1⍺. Hypoxia drives reactive oxygen species (ROS) production in cancer cells which triggers antioxidant enzyme activity through PGC1⍺/β expression that ultimately contributes to cancer cell survival by ROS removal.33
Figure 1.
Hypoxia-mediated mechanisms of mitochondrial biogenesis in tumor cell. Hypoxia activates FOXO3a gene transcription which in turn destabilizes structural integrity of Myc; ROS production is upregulated under hypoxia which stimulates PGC1⍺/β enzymatic activity that ultimately removes ROS thus facilitating cancer cell survival; Hypoxia also activates MAXI1 which interacts with PGC1⍺/β and inhibits mitochondrial biogenesis by PGC1⍺/β. In cancer cells that express HIF2⍺, the MAXI1/MYC dimer is stabilized that leads to higher mitochondrial biogenesis. FOXO3a: Forkhead box protein O3; ROS: reactive oxygen species; HIF1/2⍺: hypoxia inducible factor 1/2⍺; MYC: myelocytomatosis gene; PGC1⍺/β: peroxisome proliferator-activated receptor gamma coactivator 1-alpha/beta; MAXI1: myc-associated factor X interactor 1.
Role of HIF-1⍺ in cellular metabolism
Apart from the role of HIF⍺ proteins in controlling mitochondrial biogenesis, the principal role of hypoxia during tumorigenesis and cancer progression is regulation of cellular metabolism. HIF-1⍺ activates transcription of key glycolytic genes such as glucose transporter1 (GLUT1), GLUT3, phosphoglycerate kinase 1 (PGK), and hexokinase (HK)-1/2. Additionally, lactate dehydrogenase A (LDHA) responsible for NAD+ replenishment and monocarboxylate transporter 4 (MCT4), the transporter responsible for lactate efflux, are also induced by HIF-1⍺. This HIF-1⍺-mediated regulation of glycolytic genes ultimately results in a shift towards dependence of cancer cell metabolism on glycolysis.34,35 In addition to inducing glycolytic gene expression, HIF-1⍺ also reduces the rate of mitochondrial respiration in hypoxic cancer cells. Pyruvate dehydrogenase, a critical enzyme belonging to the pyruvate dehydrogenase complex (PDC) converts pyruvate to acetyl-CoA that then enters tricarboxylic acid cycle (TCA cycle) in mitochondria. Pyruvate dehydrogenase kinase 1 (PDK1) phosphorylates and inactivates pyruvate dehydrogenase and as a result pyruvate is unavailable for mitochondrial respiration. PDK1 is induced by HIF-1⍺ with the net effect of cancer cells utilizing glycolytic ATP. An additional advantage of reducing mitochondrial dependence is the generation of lower levels of mtROS.36,37 It has been shown that HIF-1⍺ also reduces mtROS by inducing two proteins: LONP1, a protease that degrades cytochrome c oxidase subunit 4 isoform 1 (COX 4–1) and COX4-2, a more efficient form of COX4-1.38 These studies show that hypoxia increases glycolysis and reduces mitochondrial respiration and ROS production in tumor cells.
Impact of hypoxia on the tumor microenvironment
Tumor cells along with the local non-malignant cells constitute the TME. It has been well understood now that the interactions of the neighboring stromal cells in the microenvironment are critical for the progression of tumor cells. The TME is primarily comprised of the chemical microenvironment that includes the pH, nitric oxide, and metabolites (like glucose) and the cellular microenvironment that involves tumor cells, stromal cells, and the extracellular matrix (ECM).7 Here, we mainly focus on the influence of hypoxia on stromal cells and ECM and their effects on tumor progression. One of the major cells that constitute the TME are the CAFs.39 CAFs are spindle shaped cells that are actively involved in secreting ECM proteins and are abundantly present in connective tissues.40 During cancer development, majority of CAFs are derived from a mesoderm-derived precursor cell. However, CAFs also originate from trans-differentiation of pericytes, from endothelial cells through endothelial-mesenchymal transition, and from epithelial cells by epithelial-mesenchymal transitions.41 CAFs have also been derived from normal fibroblasts during chemotherapy as a result of the created hypoxic environment.42 CAFs in the TME are large spindle shaped cells that express α-smooth muscle actin and are stiffer than normal fibroblasts.2,43. However, there are additional biomarkers like vimentin (VIM), fibroblast activation protein (FAP), and fibroblast-specific protein-1 (FSP-1) that are used to identify CAFs.44,45 Hypoxia is one of the main driver events that alters the functioning of CAFs in TME and defines its role in tumor progression. CAFs affect both tumor development and progression in various ways both by directly regulating tumor growth factors, EMT mechanisms, and chemokines. Hypoxic conditions increase the expression of the angiogenic factors angiopoietin and vascular endothelial growth factor (VEGF) in CAFs as well as regulate the expression of the chemokine CXCL3 in activated myofibroblasts.25 However, CAFs also provide critical metabolites and undergo metabolic reprogramming to support glycolysis.46,47 HIF-1α also mediates metabolic reprograming in CAFs by upregulating the expression of MCT4 and subsequently downregulating caveolin-1 in order to supply energy to the growing tumor cells.48 Tumor cells switch their metabolic state between glycolysis and oxidative phosphorylation through metabolic interplay with CAFs, and exhibit the Warburg effect under hypoxia and reverse Warburg effect under normoxia.49 This metabolic interplay is critical as it further promotes tumor growth and metastasis.49 Tumor cell-derived ROS decreases the expression of caveolin-1 in CAFs which further stabilizes HIF-1α.50,51 This metabolic symbiosis however does not occur equally in all tumor types and accounts for tumor heterogeneity in the same tumor and thus significantly affects therapeutic strategies and chemoresistance.51
Another major player in TME regulation is the ECM, that maintains cell dynamics and regulates the cellular microenvironment.52 It has been observed that during the initial stages of cancer development, CAFs and other stromal cells secrete factors that modify ECM composition and dynamics.53 The altered ECM then maintains the behavior of stromal cells, thereby influencing the development and maintenance of TME.52 Hypoxia affects ECM through fibrosis, regulating collagen deposition and by mediating ECM degradation.54 HIF-1α-mediated fibrosis enhances cell invasion and migration by increased ECM deposition.55–57 In adipose tissue, kidney, and liver cells, it has been observed that HIF-1α also influences ECM remodeling.57–59 ECM stiffness is regulated by hypoxia-mediated collagen deposition. Specifically, HIFs regulate prolyl 4-hydroxylase subunit alpha (P4HA) and procollagen-lysine 2-oxyglutarate 5-dioxygenase (PLOD) enzyme family genes which play a vital role in collagen synthesis.59 P4HA2 plays a role in breast cancer progression and enhanced expression of PLOD2 by HIF-1α in primary tumors is implicated in sarcoma metastasis and increased mortality in breast cancers.60–62 PLOD2 also promotes alignment of cancer cells along collagen fibers which in turn enhances invasion and metastasis to lungs and lymph nodes.63 Apart from P4HA and PLOD genes, HIF also influences collagen deposition through upregulation of lysyl oxidase (LOX) enzymes that regulate collagen crosslinking in the ECM.64 HIF-increased LOX expression has been associated with breast and head and neck cancer metastasis.65 LOX expression inhibits E-cadherin and promotes epithelial-mesenchymal transition (EMT), which drives invasion and metastasis.66 Finally, hypoxia regulates ECM by promoting ECM degradation via expression of matrix metalloproteinases like MMP2, MMP9, and MMP15 and thus significantly influences the tumor microenvironment.2
Hypoxia-mediated regulation of cancer immune evasion
Hypoxia can also negatively regulate the antitumor immune responses in cancer and some of the key mechanisms by which it mediates immune evasion in cancers are illustrated in Figure 2. Among the target genes whose expression is regulated by HIF-1⍺ is Cluster of differentiation 47 (CD47). CD47, a membrane protein is responsible for regulating a number of cellular responses such as cell migration, T cell activation, cytokine secretion, and metastasis.67 Zhang et al. showed that breast cancer cells were protected from bone marrow-derived macrophage-mediated phagocytosis by expressing a stem cell phenotype. They also showed that HIF-1⍺ activates CD47 transcription in these breast cancer cells.68 CD47 controls immune evasion through CD47-signal regulatory protein⍺ (SIRP⍺) axis where SIRP⍺ is expressed on TAMs (tumor-associated macrophages) and myeloid-derived suppressor cells (MDSCs).69,70 Hypoxia has been shown to stimulate VEGF secretion by M2 macrophages in the TME.71 TAMs residing in hypoxic areas of TME, achieve a M2-inducing phenotype through increased secretion of molecules like TGFβ.72 Hypoxic tumor microenvironment has been shown to promote anti-tumor immunity through increased expression of the master regulator of T regulatory cell differentiation, FoxP3.73 Increased number of T regulatory cells in the tumor microenvironment suppresses CD8+ T cell-mediated cytotoxic killing of tumor cells.74 Programmed death ligand 1 (PDL1) expressed on the surface of cancer cells binds to its receptor PD1 on the surface of CD8+ T cells and inactivates CD8+ T cell antitumor response. HIF-1⍺-mediated PDL1 expression was recorded on tumor cells, dendritic cells, as well as macrophages.75,76 Hypoxia, in addition to having direct effects on the survival, differentiation, and activation of key immune effectors also activates an immune metabolism modulating molecule, adenosine. A hypoxic TME upregulates the expression of CD39, an ectonucleoside triphosphate dihydrophosphohydrolase (ENTPD1) which converts ATP/ADP to AMP and CD73 a 5ʹ-ectonucleotidase that finally converts AMP into adenosine.77,78 Moreover, hypoxia induces increased expression of the cognate receptor of adenosine, A2A adenosine receptor (A2AR).79 Notably, the adenosine-A2AR binding causes T cell apoptosis, negatively affects T cell activation and effector responses.80 Thus, hypoxia negatively regulates anti-tumor immune responses by facilitating immune evasion and promoting T cell apoptosis through adenosine production.
Figure 2.
Hypoxia-driven tumor immune evasion pathways. Hypoxia induces CD73 and CD39 gene expression in cancer cell. CD39 converts ATP/ADP to AMP and CD73 converts AMP to adenosine which leads to T cell apoptosis. Under hypoxia, HIF1⍺ translocates to the nucleus in the tumor cell and induces expression of CD47. CD47 then engages with SIRP⍺ on TAMs and inhibits TAM-based tumor phagocytosis. Under hypoxia, cancer cells also express PDL1 which binds to PD1 on CD8+ T cell leading to CD8+ T cell apoptosis. In a hypoxic TME, induced FoxP3 expression leads to T regulatory cell proliferation that then inhibits cytotoxic T cell death. TME: tumor microenvironment; Treg cell: T regulatory cell; FoxP3: forkhead box protein P3; TAMs: tumor-associated macrophages; SIRP⍺: signal regulatory protein alpha; CD47: cluster of differentiation 47; CD73: cluster of differentiation 73; CD39: cluster of differentiation 39; PD1: programmed death 1(receptor); PDL1: programmed death ligand 1; HIF1⍺: hypoxia inducible factor 1⍺; CD8 T cell: cluster of differentiation 8 expressing T cell. Figure was created with Biorender.com.
Hypoxia and lymphangiogenesis
It has been very clearly demonstrated that lymphatic vessels play a critical role in promoting tumor progression and metastasis. Expression of lymphangiogenic growth factor, VEGF-C, high lymphatic vessel density, and high incidence of lymphatic invasion have been associated with metastasis and reduced survival in cancer patients.81–84 However, the impact of a low oxygen environment on the lymphatic vasculature and effect on cancer metastasis has not been clearly elucidated although multiple pathways have been implicated in hypoxia-mediated regulation of the lymphatic-cancer crosstalk.85–87 Tumor-induced lymphangiogenesis is promoted by secretion of growth factors such as VEGF-C, VEGF-D, PDGF-B, IGF1 and -2, FGF2, HGF, and other mediators such as angiopoietin-2, sphingosine-1- phosphate, adrenomedullin, and IL-7 by the cancer cells and other cells within the TME.85,88–90 Hypoxia has been shown to favor the processes of lymphangiogenesis and metastasis of cancer cells via lymphatic vessels predominantly through activation of HIF-1-α.91 HIF-1α regulates the expression of growth factors, tumor-promoting pathways and cytokines that are involved in lymphatic endothelial cell (LEC) proliferation, migration, and subsequent lymph node metastasis.86 HIF-1α levels are shown to correlate with the expression of VEGF-C and peritumoral lymphangiogenesis in breast cancer.87 Further, HIF-1⍺ expression also correlates with increased VEGF-C and lymphatic vessel density in biopsies from oral squamous cell carcinoma patients.92 Interestingly, in contrast, Schito et al. have shown that in MDA-MB-231 breast cancer cells, hypoxia decreased the mRNA levels of VEGF-D and did not increase the VEGF-C levels. However, it was shown that hypoxia induces LEC proliferation and migration via HIF-1⍺→PDGF-B→PDGFRβ signaling.93 HIF-1α expressed during tumor hypoxia influences peritumoral lymphangiogenesis.87 Hypoxia increases apelin expression in cancer cells.94 The apelin/APJ pathway has been implicated in progression of lymphangiogenesis and lymph node metastasis.95 Hypoxia also increases expression of the β1 integrin subunit, PI3K/Akt and mTOR pathways, mechanisms that have been widely implicated in promoting lymphangiogenesis and lymph node metastasis.81 Inhibition of mTOR with rapamycin reduces lymphangiogenesis in primary tumors and prevents spread of cancer cells to the cervical lymph nodes.98 Hypoxia activates transcription factors, activator-protein-1 (AP-1), and Prox-1 that are closely associated with the lymphatic vasculature.97 AP-1 activation co-operates with HIF-1α for upregulation of VEGF gene expression in hypoxic conditions.96 AP-1-inducible genes are triggered in LECs due to hypoxia, including ET-1, MMP9, and c-jun.97 The AP-1 complex exemplifies a network of transcription factors that coordinately function in lymphangiogenesis and promote tumor development and progression.96,97 The expression of Prox-1, a transcription factor essential for embryonic lymphangiogenesis, is increased under hypoxia or induction of HIF-1α. Importantly, both Prox-1 and NFκB activate the VEGFR-3 promoter in LECs, that enhances the responsiveness of LECs to VEGFR-3 ligands VEGF-C and VEGF-D, ultimately resulting in robust lymphangiogenesis.25 The ET-1/ETBR signaling, that activates HIF-1α, induces LEC proliferation and branching morphogenesis concomitantly with activation of p42/44 mitogen-activated protein kinases (MAPK) and Akt pathways. HIF-1α silencing desensitizes VEGF-A and VEGF-C production in response to ET-1 or hypoxia, implicating HIF-1α/VEGF as downstream signaling molecules of ET-1 axis.46 The ET-1/ETBR axis and hypoxia may thus act through a HIF-1α-dependent mechanism to promote lymphangiogenesis via VEGF family members. Hypoxia has been shown to directly promote the attraction and adhesion of cancer cells to the lymphatic endothelium by inducing CXCR4 levels in cancer cells and its ligand CXCL12 in LECs.51 Further, hypoxia induces NOTCH signaling that downregulates VEGFR2 in LECs and decreases migration of lymphatic vessels which might be critical for maintaining the balance between lymphangiogenesis and angiogenesis.53
Hypoxia and targeting angiogenesis for cancer therapy
Tumors require nutrients and oxygen from the surrounding tissue to survive. After reaching a certain size,99 enlarging tumors must obtain their own blood supply to meet increasing metabolic needs. When tumors become hypoxic and continue to grow, HIFs secrete various factors that influence angiogenesis.100 One of the major angiogenic genes regulated by HIF-1α is the VEGF, a primary regulator of angiogenesis1,100 and highly induced in several cancers.1,101 Hypoxic stabilization of HIFα results in expression of angiogenic factors like nitric oxide synthase, adrenomedullin, and interleukin 8.102–104 Suppression of HIF-1α in endothelial cells alters NO levels, suppresses tumor cell migration, and inhibits tumor metastasis. But loss of HIF-2α was observed to have the opposite effect.46 Hypoxia also influences angiogenesis by altering the fiber morphology in the ECM.105 As discussed above, tumors secrete proangiogenic factors, VEGF, basic fibroblast growth factor (bFGF), and placental growth factor (PIGF), that activates neighboring endothelial cells and induces proliferation and migration toward the tumor mass.106 As tumor angiogenesis serves as a rate limiting step to continued growth, it has emerged as a promising potential therapeutic target.107 Several drugs targeting VEGF or its receptors have been developed and approved for cancer therapy. While there are some clinical examples of success using anti-angiogenesis drugs in combination with chemotherapeutics,108 the use of anti-angiogenic drugs as a monotherapy has not significantly improved patient survival rates.109–110 After treatment, the tumor can upregulate other angiogenic stimulators, such as bFGF, PlGF, TGFβ, and angiopoietins, circumventing the loss of VEGF signaling.112–114 This acquired resistance is possibly a response to the increase in tumor hypoxia induced by the anti-angiogenic therapy, that affects multiple cellular signaling pathways promoting tumor progression, invasiveness, and metastasis.112,115–117 Further, the ineffectiveness of the combination therapy may be attributed to the architecture of the tumor vasculature. The tumor vessels are highly tortuous and heterogeneous, with thin-walled, leaky large vessels, devoid of pericyte coverage, and irregularly branched smaller vessels that connect randomly. As a result, oxygen distribution to the tumor cells through these irregular blood vessels causes hypoxic regions within tumor cells. These tumor cells, while adapting to the hypoxic microenvironment, become more aggressive and attain therapeutically resistant tumor phenotypes.118 Endothelial cells associated with the tumor in many cases have lost polarity, detached from the basement membrane, and contain wide junctions, causing the vessels to be leaky.119–122 Because the vascular endothelial cells no longer serve as a functional barrier, tumor vessels enable transendothelial tumor cell migration.120 The irregular vessels and loss of pericyte coverage cause non-uniform blood flow within the tumor which further prevents uniform delivery and uptake of co-administered chemotherapeutic agents and impedes immune cell infiltration.122 Additionally, tumor hypoxia caused by anti-angiogenic therapies can hinder the efficacy of irradiation and chemotherapies that function through the formation of ROS to kill tumor cells.123,124 As the tumor vasculature serves as an impediment to the efficacy of chemotherapy, improving vessel function through increasing and normalizing blood flow throughout the tumor, enhancing pericyte coverage, and decreasing vessel permeability, can improve chemotherapeutic delivery throughout the tumor. Further, by reducing tumor hypoxia, clinicians can prevent development of a more aggressive and metastatic phenotype and enhance radiation therapy.123,125 Intriguingly, anti-angiogenic drugs, such as bevacizumab, when used at low doses can normalize the tumor vasculature. The vessels become less tortuous with improved blood perfusion, decreased interstitial pressure, and increased tumor oxygenation, preventing tumor cell diapedesis due to enhanced endothelial cell barrier function.123,125–130 In additional studies, tumors implanted in mice heterozygous for PHD2, which acts as an oxygen sensor to promote the HIF-driven hypoxic response, showed increased perfusion and oxygenation. PHD2 haplodeficiency promoted tumor vessel maturity and normalization and prevented tumor cell invasion, diapedesis, and metastasis.120 These data support that normalization of the tumor vasculature through modulation of HIF-mediated signaling is likely to benefit treatment of solid tumors.
Hypoxia and tumor therapy outcome
The role of drug resistance in cancer metastasis
Resistance of cancer cells to chemotherapy/radiotherapy can be broadly divided into two classes: intrinsic and acquired resistance. Factors conferring resistance to tumor cells prior to therapy result in development of intrinsic drug resistance, while acquired resistance refers to the development of resistance in cancer cells during the course of drug administration/treatment. Both of these resistance modes give rise to a population of tumor cells that do not respond to chemotherapeutic treatments and continue to proliferate rendering treatment ineffective.131 A number of factors described below results in drug resistance of cancer cells:
Drug efflux pathways are well-recognized to confer drug resistance in various cancers
Three most extensively studied members of the well-known ATP-binding cassette (ABC) transporter family that regulates the bulk of the chemotherapeutic efflux across the plasma membrane are related to multi-drug resistance (MDR). These three proteins are, MD1(aka P-glycoprotein/ABCB1), MRP1(MDR-associated protein 1/ABCC1), and BCRP (breast cancer resistance protein/ABCG2). These three proteins play critical roles in eliminating a broad range of widely used chemotherapeutic agents. MDR1 overexpression (both before and after chemotherapy) is associated with intrinsic as well as acquired drug resistance in hepatic and renal cancers along with leukemia and lymphoma.132,133 MDR1 is found to be highly expressed on the surface of epithelial cells forming the lining of excretory organs like colon, renal proximal tubules, bile ductules, etc. compared to other tissues under normal conditions which highlights the role of MDR1 in drug excretion.134,135 Similarly, BCRP has been correlated with drug resistance in breast cancer and leukemia.136,137 The role of ABC transporter overexpression in cancer cells is further underlined by the finding that cancer stem cells also show high MDR transporter overexpression levels, particularly for BCRP.138
Hypoxia is thus a critical element of the TME contributing to drug resistance. A number of reasons such as low drug bioavailability in hypoxic tumors, HIF-1⍺-mediated MDR1 overexpression and decreased expression of drug targets such as topoisomerase II have been reported to be responsible for hypoxia-mediated therapeutic resistance in cancer.139
Hypoxia and tumor chemoresistance
The most prominent player in hypoxia-induced chemoresistance is HIF-1⍺ gene identified through a number of studies where poor patient prognosis has been correlated with HIF-1⍺ overexpression in a variety of cancer types such as neuroblastoma and non-small-cell lung cancer (NSCLC).5 A number of HIF-1⍺ regulated genes such as Bcl-2, Glut-1, VEGF, and MDR1 induces chemoresistance.140 Thus, HIF-1⍺ inhibition-based treatment regimens have been shown to rescue hypoxia-mediated chemoresistance.141 In neuroblastoma, hypoxia-mediated chemoresistance is seen for drugs such as doxorubicin, cisplatin, etoposide, etc.,142 while head and neck squamous cancer and gastric cancer acquire resistance towards cisplatin, 5-fluorouracil.143,144 Different mechanisms have been identified by which this chemoresistance is induced, including upregulation of P-glycoprotein expression, downregulation of topoisomerase, blockade of apoptosis, induction of autophagy, and upregulation of telomerase.
Induction of P-glycoprotein expression
Under hypoxic conditions, HIF-1⍺ binds to the promoter of MDR1 gene and induces overexpression of membrane transporter P-gp that then mediates chemoresistance. P-gp is an important efflux transporter belonging to ABC family of transporters which transports hydrophobic/amphiphilic drugs out of the cell when overexpressed in chemoresistant cells thus causing increased cell proliferation.26 A large number of chemotherapeutic drugs have been identified as substrates of P-gp all of whom are amphiphilic in nature.134,145 Since low solubility of P-gp substrates has been identified, treatment strategies such as the usage of liposome nanoparticle mediated drug delivery have been suggested to prevent P-gp-mediated drug efflux and increase intracellular drug availability.146
Altered drug target expression
Resistance to known topoisomerase II inhibitors such as etoposide has been observed in breast cancer cells as a response to hypoxia in the TME. Such resistance is believed to be caused by HIF-1⍺-mediated low nuclear levels of topo II⍺.147
Hypoxia-mediated anti-apoptosis
Mitochondria-based cell apoptosis through Bcl family proteins Bax and Bad are direct targets of hypoxia-mediated microRNA, miR-26a overexpression. In a study published in 2018, miR26a overexpression by HIF-1⍺ in hypoxic conditions was identified which was inversely related to Bax and Bad expression leading to resistance to the drug temozolamide.148
Autophagy induction and metabolic regulation
HIF-1⍺ caused protective autophagy in cancer cells exposed to fenretinide leading to drug resistance by HeLa cells. Inhibition of HIF-1⍺ led to enhanced cancer cell death by fenretinide.149 Colorectal cancer cells with high ATP levels were found to be resistant to 5-fluorouracil and oxaliplatin which points to the metabolic regulation by HIF-1⍺. HIF-1⍺ is known to upregulate PDK1 protein which reduces mitochondrial respiration by phosphorylating and inactivating pyruvate dehydrogenase (PDH). As a result of this, hypoxic cancer cells become dependent on glycolysis as a fast and cheap source of ATP which is thus produced in high levels.27 Inhibiting glycolysis in colorectal cancer cells resensitized them to 5-fluoruracil and cisplatin treatment.37
Upregulation of telomerase
A number of studies have shown that HIF-1⍺ causes upregulation of telomerase gene expression by binding to telomerase promoter site and by causing alternative splicing. As a result of this, active telomerase variants are produced and cancer cells are protected from replicative senescence that is also believed to play a role in ultimate cancer cell chemoresistance.150
In addition, a number of factors other than HIF-1⍺ have been identified to cause hypoxia-based chemoresistance. Such evidence has come from studies demonstrating that inhibiting HIF-1⍺ only partially reverses chemoresistance.151 Overexpression of anti-apoptotic members of Bcl-2 family and IAP3 in response to hypoxia is one such example of HIF-1⍺-independent chemoresistance in cancer cells.152 This has been clearly demonstrated in studies with different tumor cells, where blocking the activity of HIF-1⍺ did not affect the increased apoptotic susceptibility of the tumors subjected to hypoxia.152 In addition, nuclear factor kappa-B (NFκB), cyclooxygenase-2 (COX2), STAT3, etc. when inhibited also decrease chemoresistance in hypoxic cancer cells.153
Hypoxia and tumor radioresistance
Radiotherapy kills cancer by producing ROS that induces DNA damage in recipient cells. Hypoxia plays a major role and contributes almost three times to radioresistance compared to other factors. This is because the presence of oxygen is critical for radiotherapy-induced generation of ROS. As one of the most electronegative elements inside cells, oxygen respond to ionizing radiation by accepting free radicals generated due to radiotherapy.154 These free radicals further induce DNA damage by strand breaks and kill cancer cells. Under hypoxia, as cancer cells are deprived of oxygen, free radicals generated from DNA under radiotherapy undergo reduction by molecules containing sulfhydryl (SH) groups that lead to DNA repair.154 The ability of oxygen to bolster radiotherapy-induced DNA damage is popularly known as oxygen fixation hypothesis.155 Hypoxic status in cancer types such as cervix carcinoma, sarcoma, head and neck cancer has emerged to be a major prognostic factor for poor radiotherapy results.155–157 Radiotherapy treatment under hypoxia has shown a number of mechanisms through which ROS adaptation by cancer cells takes place. At the helm of most of these mechanisms is HIF-1⍺. HIF-1⍺ transcriptionally activates antioxidant gene expression as well as ROS efflux.35,158 Hypoxia can also cause increase in ROS during its initial phase (ischemia) which instead of inducing cell death acts a signaling molecules for HIF-1⍺ stabilization.159 This hypoxic ROS-based HIF-1⍺ activity takes place through PI3K/AKT pathway activation and ERK phosphorylation that ultimately increases HIF-1⍺ expression.160,161 A number of strategies have been implemented to re-sensitize radioresistant hypoxic tumor cells to radiotherapy by using drug therapy which are briefly summarized as follows:
NO donors
NO homeostasis is critical to prevent hypoxia-mediated therapeutic resistance by cancer cells. Low intracellular NO levels destabilize HIF-1⍺ during hypoxia due to which a number of NO donors such as sodium nitroprusside,162 nitroglycerin,163 insulin,164 spermin nonoate165 have sensitized hypoxic cancer cells to radiotherapy.
Enzymatic inhibition
Drugs such as diethylmaleate,166 diethylfumarate167 (both deplete glutathione which is an antioxidant for ROS), auranofin (inhibits thioredoxin reductase activity),168 auranofin + Buthionine sulphoximine (thioredoxin reductase inhibition and glutathione depletion)169 that inhibit the activity of antioxidant enzymes radiosensitize the hypoxic cancer cells by increasing the availability of ROS inside the hypoxic cancer cells.
Metabolic inhibitors
Inhibiting tumor cell metabolism by agents such as dichloroacetate (glycolytic inhibitor),170 ritonavir (glucose transporter inhibitor)171 that target the metabolic mechanisms regulated by hypoxic conditions, has been shown to be a potent radiosensitizing technique.
Regulation of oxygen consumption
Drugs like metformin that inhibits mitochondrial complex I as well as glucocorticoids has been shown to decrease oxygen consumption thus increasing oxygen availability for radiotherapy in cancer cells under hypoxia.172,173
HIF-1⍺ inhibition
Since HIF-1⍺ is the master regulator for mediating hypoxia-related effects, a number of radiosensitizing agents inhibiting HIF-1⍺ expression, translation and downstream activation have been developed such as atorvastatin,174 berberine, YC-1 (degrades HIF-1⍺ in addition to inhibiting HIF-1⍺ translation).175 Gold nanoparticles augmenting ROS production by electron donation have also been shown to be effective in radiosensitization in a variety of tumor types.176
Conclusion and future directions
Hypoxia is the key regulator for chemo-/radio resistance in different cancer types that account for increased cancer morbidity. As depicted in Figure 3, a variety of mechanisms both HIF-1 and HIF-2 dependent and independent are adopted by hypoxic cancer cells as a means to bypass therapy and account for tumor heterogeneity that promotes survival in unfavorable conditions. The metabolic interplay between tumor cells and components of a hypoxic TME coupled with activation of lymphangiogenic and angiogenic mechanisms further complicates the response to specific clinical interventions. Thus, the need to identify new drug targets of a hypoxic TME as well as studies combining drug therapy with radiotherapy are needed to better understand and directly target mechanisms contributing to therapeutic resistance of cancer cells. In addition, the specific signaling pathways that are regulated by HIF-1⍺ can be studied to identify prognostic factors under hypoxia which will help to identify radiotherapeutic outcomes. Homeostasis and not a mere absence/presence of molecular species such as NO and ROS have emerged to be important in shaping cellular response to therapy. Thus, in conjunction with identifying and targeting specifically overexpressed downstream targets of HIF-1⍺ targeting antioxidant enzyme production, decreasing oxygen demand, reducing ATP availability by inhibiting glycolysis have emerged to some of the more novel strategies for combating hypoxia-mediated therapeutic resistance. In summary, understanding key mechanisms of hypoxia-based cancer cell growth and progression along with novel therapeutic strategies is required to ameliorate the pro-tumorigenic effects of hypoxia.
Figure 3.
Hypoxia-associated tumor microenvironment of a cancer cell and related effects: Hypoxia causes nuclear translocation of HIF-1⍺ which binds at various sites in the DNA and upregulates expression of MDR1, VEGF, NO, etc. PI3K/Akt/ERK signaling is activated, drug availability of topoisomerase II is reduced, intracellular ATP levels are upregulated due to high rate of glycolysis. CXCR4 and CXCL12 expression is upregulated in presence of hypoxic conditions from both cancer cell and LECs. Tortuous abnormal blood vessel growth further contributes to the alterations in the TME under hypoxia. High efflux transporter (P-gp, MRP1, BCRP) expression leads to drug efflux from cancer cell. Figure was created with Biorender.com.
Authors’ contributions
SR, SK, AS, CD, KB and SC have written, proofread, and approved the manuscript.
DECLARATION OF CONFLICTING INTERESTS
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article
FUNDING
The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by Auf-X-Grant Award from Texas A&M University Health Science, Research Enhancement Grant from department of Medical Physiology and American Heart Association grant 17SDG33670306 to S.C.
ORCID iDs
Ankith Sharma https://orcid.org/0000-0002-0724-5444
Camille L Duran https://orcid.org/0000-0002-7614-3265
Sanjukta Chakraborty https://orcid.org/0000-0002-4869-3198
References
- 1.Büchler P, Reber HA, Büchler M, Shrinkante S, Büchler MW, Friess H, Semenza GL, Hines OJ. Hypoxia-inducible factor 1 regulates vascular endothelial growth factor expression in human pancreatic cancer. Pancreas 2003; 26:56–64 [DOI] [PubMed] [Google Scholar]
- 2.Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I. The hypoxic tumour microenvironment. Oncogenesis 2018; 7:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Semenza GL. Intratumoral hypoxia, radiation resistance, and HIF-1. Cancer Cell 2004; 5:405–6 [DOI] [PubMed] [Google Scholar]
- 4.Bayer C, Shi K, Astner ST, Maftei C-A, Vaupel P. Acute versus chronic hypoxia: why a simplified classification is simply not enough. Int J Radiat Oncol Biol Phys 2011; 80:965–8 [DOI] [PubMed] [Google Scholar]
- 5.Semenza GL. HIF-1 and tumor progression: pathophysiology and therapeutics. Trends Mol Med 2002; 8:S62–7 [DOI] [PubMed] [Google Scholar]
- 6.Saxena K, Jolly MK. Acute vs. chronic vs. cyclic hypoxia: their differential dynamics, molecular mechanisms, and effects on tumor progression. Biomolecules 2019; 9:339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Semenza GL. The hypoxic tumor microenvironment: a driving force for breast cancer progression. Biochim Biophys Acta 2016; 1863:382–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Serocki M, Bartoszewska S, Janaszak-Jasiecka A, Ochocka RJ, Collawn JF, Bartoszewski R. miRNAs regulate the HIF switch during hypoxia: a novel therapeutic target. Angiogenesis 2018; 21:183–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hu CJ, Wang LY, Chodosh LA, Keith B, Simon MC. Differential roles of hypoxia-inducible factor 1alpha (HIF-1alpha) and HIF-2alpha in hypoxic gene regulation. Mol Cell Biol 2003; 23:9361–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Park BK, Paik YH, Park JY, Park KH, Bang S, Park SW, Chung JB, Park YN, Song SY. The clinicopathologic significance of the expression of vascular endothelial growth factor-C in intrahepatic cholangiocarcinoma. Am J Clin Oncol 2006; 29:138–42 [DOI] [PubMed] [Google Scholar]
- 11.Rosmorduc O, Housset C. Hypoxia: a link between fibrogenesis, angiogenesis, and carcinogenesis in liver disease. Semin Liver Dis 2010; 30:258–70 [DOI] [PubMed] [Google Scholar]
- 12.Holmquist L, Jogi A, Pahlman S. Phenotypic persistence after reoxygenation of hypoxic neuroblastoma cells. Int J Cancer 2005; 116:218–25 [DOI] [PubMed] [Google Scholar]
- 13.Holmquist-Mengelbier L, Fredlund E, Lofstedt T, Noguera R, Navarro S, Nilsson H, Pietras A, Vallon-Christersson J, Borg A, Gradin K, Poellinger L, Pahlman S. Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell 2006; 10:413–23 [DOI] [PubMed] [Google Scholar]
- 14.Koivunen P, Hirsila M, Gunzler V, Kivirikko KI, Myllyharju J. Catalytic properties of the asparaginyl hydroxylase (FIH) in the oxygen sensing pathway are distinct from those of its prolyl 4-hydroxylases. J Biol Chem 2004; 279:9899–904 [DOI] [PubMed] [Google Scholar]
- 15.Bartoszewski R, Moszynska A, Serocki M, Cabaj A, Polten A, Ochocka R, Dell’Italia L, Bartoszewska S, Kroliczewski J, Dabrowski M, Collawn JF. Primary endothelial cell-specific regulation of hypoxia-inducible factor (HIF)-1 and HIF-2 and their target gene expression profiles during hypoxia. FASEB J 2019; 33:7929–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Koh MY, Lemos R, Jr., Liu X, Powis G. The hypoxia-associated factor switches cells from HIF-1alpha- to HIF-2alpha-dependent signaling promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res 2011; 71:4015–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lin Q, Cong X, Yun Z. Differential hypoxic regulation of hypoxia-inducible factors 1alpha and 2alpha. Mol Cancer Res 2011; 9:757–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Luo W, Zhong J, Chang R, Hu H, Pandey A, Semenza GL. Hsp70 and CHIP selectively mediate ubiquitination and degradation of hypoxia-inducible factor (HIF)-1alpha but not HIF-2alpha. J Biol Chem 2010; 285:3651–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bento CF, Fernandes R, Ramalho J, Marques C, Shang F, Taylor A, Pereira P. The chaperone-dependent ubiquitin ligase CHIP targets HIF-1alpha for degradation in the presence of methylglyoxal. PLoS One 2010; 5:e15062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kochan-Jamrozy K, Kroliczewski J, Moszynska A, Collawn JF, Bartoszewski R. miRNA networks modulate human endothelial cell adaptation to cyclic hypoxia. Cell Signal 2019; 54:150–60 [DOI] [PubMed] [Google Scholar]
- 21.Toffoli S, Roegiers A, Feron O, Van Steenbrugge M, Ninane N, Raes M, Michiels C. Intermittent hypoxia is an angiogenic inducer for endothelial cells: role of HIF-1. Angiogenesis 2009; 12:47–67 [DOI] [PubMed] [Google Scholar]
- 22.Martinive P, Defresne F, Quaghebeur E, Daneau G, Crokart N, Gregoire V, Gallez B, Dessy C, Feron O. Impact of cyclic hypoxia on HIF-1alpha regulation in endothelial cells – new insights for anti-tumor treatments. FEBS J 2009; 276:509–18 [DOI] [PubMed] [Google Scholar]
- 23.Toffoli S, Feron O, Raes M, Michiels C. Intermittent hypoxia changes HIF-1alpha phosphorylation pattern in endothelial cells: unravelling of a new PKA-dependent regulation of HIF-1alpha. Biochim Biophys Acta 2007; 1773:1558–71 [DOI] [PubMed] [Google Scholar]
- 24.Verduzco D, Lloyd M, Xu L, Ibrahim-Hashim A, Balagurunathan Y, Gatenby RA, Gillies RJ. Intermittent hypoxia selects for genotypes and phenotypes that increase survival, invasion, and therapy resistance. PLoS One 2015; 10:e0120958–e58 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ammirante M, Shalapour S, Kang Y, Jamieson CA, Karin M. Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc Natl Acad Sci U S A 2014; 111:14776–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Comerford KM, Wallace TJ, Karhausen J, Louis NA, Montalto MC, Colgan SP. Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res 2002; 62:3387–94 [PubMed] [Google Scholar]
- 27.Frezza C, Zheng L, Tennant DA, Papkovsky DB, Hedley BA, Kalna G, Watson DG, Gottlieb E. Metabolic profiling of hypoxic cells revealed a catabolic signature required for cell survival. PLoS One 2011; 6:e24411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Li F, Wang Y, Zeller KI, Potter JJ, Wonsey DR, O’Donnell KA, Kim JW, Yustein JT, Lee LA, Dang CV. Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol Cell Biol 2005; 25:6225–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.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 2007; 11:407–20 [DOI] [PubMed] [Google Scholar]
- 30.Peck B, Ferber EC, Schulze A. Antagonism between FOXO and MYC regulates cellular powerhouse. Front Oncol 2013; 3:96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gordan JD, Bertout JA, Hu CJ, Diehl JA, Simon MC. HIF-2alpha promotes hypoxic cell proliferation by enhancing c-Myc transcriptional activity. Cancer Cell 2007; 11:335–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gordan JD, Lal P, Dondeti VR, Letrero R, Parekh KN, Oquendo CE, Greenberg RA, Flaherty KT, Rathmell WK, Keith B, Simon MC, Nathanson KL. HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma. Cancer Cell 2008; 14:435–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Austin S, St-Pierre J. PGC1alpha and mitochondrial metabolism–emerging concepts and relevance in ageing and neurodegenerative disorders. J Cell Sci 2012; 125:4963–71 [DOI] [PubMed] [Google Scholar]
- 34.Semenza GL. HIF-1: upstream and downstream of cancer metabolism. Curr Opin Genet Dev 2010; 20:51–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Semenza GL. Hypoxia-inducible factors: coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J 2017; 36:252–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.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 2006; 3:177–85 [DOI] [PubMed] [Google Scholar]
- 37.Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab 2006; 3:187–97 [DOI] [PubMed] [Google Scholar]
- 38.Fukuda R, Zhang H, Kim JW, Shimoda L, Dang CV, Semenza GL. HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells. Cell 2007; 129:111–22 [DOI] [PubMed] [Google Scholar]
- 39.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med 2013; 19:1423–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Erdogan B, Webb DJ. Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans 2017; 45:229–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.LeBleu VS, Kalluri R. A peek into cancer-associated fibroblasts: origins, functions and translational impact. Dis Model Mech 2018; 11.doi:10.1242/dmm.029447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Peiris-Pagès M, Sotgia F, Lisanti MP. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget 2015; 6:10728–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Xing F, Saidou J, Watabe K. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front Biosci 2010; 15:166–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Berdiel-Acer M, Sanz-Pamplona R, Calon A, Cuadras D, Berenguer A, Sanjuan X, Paules MJ, Salazar R, Moreno V, Batlle E, Villanueva A, Molleví DG. Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information. Mol Oncol 2014; 8:1290–305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li J, Jia Z, Kong J, Zhang F, Fang S, Li X, Li W, Yang X, Luo Y, Lin B, Liu T. Carcinoma-associated fibroblasts lead the invasion of salivary gland adenoid cystic carcinoma cells by creating an invasive track. PLoS One 2016; 11:e0150247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Branco-Price C, Zhang N, Schnelle M, Evans C, Katschinski DM, Liao D, Ellies L, Johnson RS. Endothelial cell HIF-1α and HIF-2α differentially regulate metastatic success. Cancer Cell 2012; 21:52–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang D, Wang Y, Shi Z, Liu J, Sun P, Hou X, Zhang J, Zhao S, Zhou BP, Mi J. Metabolic reprogramming of cancer-associated fibroblasts by IDH3alpha downregulation. Cell Rep 2015; 10:1335–48 [DOI] [PubMed] [Google Scholar]
- 48.Yan Y, Wang LF, Wang RF. Role of cancer-associated fibroblasts in invasion and metastasis of gastric cancer. World J Gastroenterol 2015; 21:9717–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wanandi SI, Ningsih SS, Asikin H, Hosea R, Neolaka G. Metabolic interplay between tumour cells and cancer-associated fibroblasts (CAFs) under hypoxia versus normoxia. Malays J Med Sci 2018; 25:7–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pavlides S, Tsirigos A, Vera I, Flomenberg N, Frank PG, Casimiro MC, Wang C, Fortina P, Addya S, Pestell RG, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Loss of stromal caveolin-1 leads to oxidative stress, mimics hypoxia and drives inflammation in the tumor microenvironment, conferring the “reverse warburg effect”: a transcriptional informatics analysis with validation. Cell Cycle 2010; 9:2201–19 [DOI] [PubMed] [Google Scholar]
- 51.Yoshida GJ. Metabolic reprogramming: the emerging concept and associated therapeutic strategies. J Exp Clin Cancer Res 2015; 34:111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol 2012; 196:395–406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature 2004; 432:332–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer 2014; 14:430–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kai F, Laklai H, Weaver VM. Force matters: biomechanical regulation of cell invasion and migration in disease. Trends Cell Biol 2016; 26:486–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wolf K, Te Lindert M, Krause M, Alexander S, Te Riet J, Willis AL, Hoffman RM, Figdor CG, Weiss SJ, Friedl P. Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol 2013; 201:1069–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Halberg N, Khan T, Trujillo ME, Wernstedt-Asterholm I, Attie AD, Sherwani S, Wang ZV, Landskroner-Eiger S, Dineen S, Magalang UJ, Brekken RA, Scherer PE. Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 2009; 29:4467–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Moon JO, Welch TP, Gonzalez FJ, Copple BL. Reduced liver fibrosis in hypoxia-inducible factor-1alpha-deficient mice. Am J Physiol Gastrointest Liver Physiol 2009; 296:G582–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gilkes DM, Bajpai S, Chaturvedi P, Wirtz D, Semenza GL. Hypoxia-inducible factor 1 (HIF-1) promotes extracellular matrix remodeling under hypoxic conditions by inducing P4HA1, P4HA2, and PLOD2 expression in fibroblasts. J Biol Chem 2013; 288:10819–29 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 60.Xiong G, Deng L, Zhu J, Rychahou PG, Xu R. Prolyl-4-hydroxylase α subunit 2 promotes breast cancer progression and metastasis by regulating collagen deposition. BMC Cancer 2014; 14:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Eisinger-Mathason TS, Zhang M, Qiu Q, Skuli N, Nakazawa MS, Karakasheva T, Mucaj V, Shay JE, Stangenberg L, Sadri N, Pure E, Yoon SS, Kirsch DG, Simon MC. Hypoxia-dependent modification of collagen networks promotes sarcoma metastasis. Cancer Discov 2013; 3:1190–205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gilkes DM, Bajpai S, Wong CC, Chaturvedi P, Hubbi ME, Wirtz D, Semenza GL. Procollagen lysyl hydroxylase 2 is essential for hypoxia-induced breast cancer metastasis. Mol Cancer Res 2013; 11:456–66 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 63.Gilkes DM, Chaturvedi P, Bajpai S, Wong CC, Wei H, Pitcairn S, Hubbi ME, Wirtz D, Semenza GL. Collagen prolyl hydroxylases are essential for breast cancer metastasis. Cancer Res 2013; 73:3285–96 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 64.Barker HE, Cox TR, Erler JT. The rationale for targeting the LOX family in cancer. Nat Rev Cancer 2012; 12:540. [DOI] [PubMed] [Google Scholar]
- 65.Erler JT, Bennewith KL, Nicolau M, Dornhöfer N, Kong C, Le Q-T, Chi J-T, Jeffrey SS, Giaccia AJ. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 2006; 440:1222. [DOI] [PubMed] [Google Scholar]
- 66.Schietke R, Warnecke C, Wacker I, Schodel J, Mole DR, Campean V, Amann K, Goppelt-Struebe M, Behrens J, Eckardt KU, Wiesener MS. The lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: insights into cellular transformation processes mediated by HIF-1. J Biol Chem 2010; 285:6658–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zhao H, Wang J, Kong X, Li E, Liu Y, Du X, Kang Z, Tang Y, Kuang Y, Yang Z, Zhou Y, Wang Q. CD47 promotes tumor invasion and metastasis in non-small cell lung cancer. Sci Rep 2016; 6:29719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhang H, Lu H, Xiang L, Bullen JW, Zhang C, Samanta D, Gilkes DM, He J, Semenza GL. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc Natl Acad Sci U S A 2015; 112:E6215–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Okazawa H, Motegi S, 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 2005; 174:2004–11 [DOI] [PubMed] [Google Scholar]
- 70.Liu X, Kwon H, Li Z, Fu YX. Is CD47 an innate immune checkpoint for tumor evasion? J Hematol Oncol 2017; 10:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lewis JS, Landers RJ, Underwood JC, Harris AL, Lewis CE. Expression of vascular endothelial growth factor by macrophages is up-regulated in poorly vascularized areas of breast carcinomas. J Pathol 2000; 192:150–8 [DOI] [PubMed] [Google Scholar]
- 72.Huang Y, Yuan J, Righi E, Kamoun WS, Ancukiewicz M, Nezivar J, Santosuosso M, Martin JD, Martin MR, Vianello F, Leblanc P, Munn LL, Huang P, Duda DG, Fukumura D, Jain RK, Poznansky MC. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A 2012; 109:17561–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, de Zoeten EF, Cambier JC, Stenmark KR, Colgan SP, Eltzschig HK. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA 2012; 109:E2784–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J Clin Oncol 2006; 24:5373–80 [DOI] [PubMed] [Google Scholar]
- 75.Barsoum IB, Smallwood CA, Siemens DR, Graham CH. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 2014; 74:665–74 [DOI] [PubMed] [Google Scholar]
- 76.Palsson-McDermott EM, Dyck L, Zaslona Z, Menon D, McGettrick AF, Mills KHG, O’Neill LA. Pyruvate kinase M2 is required for the expression of the immune checkpoint PD-L1 in immune cells and tumors. Front Immunol 2017; 8:1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Decking UK, Schlieper G, Kroll K, Schrader J. Hypoxia-induced inhibition of adenosine kinase potentiates cardiac adenosine release. Circ Res 1997; 81:154–64 [DOI] [PubMed] [Google Scholar]
- 78.Eltzschig HK, Thompson LF, Karhausen J, Cotta RJ, Ibla JC, Robson SC, Colgan SP. Endogenous adenosine produced during hypoxia attenuates neutrophil accumulation: coordination by extracellular nucleotide metabolism. Blood 2004; 104:3986–92 [DOI] [PubMed] [Google Scholar]
- 79.Morote-Garcia JC, Rosenberger P, Kuhlicke J, Eltzschig HK. HIF-1-dependent repression of adenosine kinase attenuates hypoxia-induced vascular leak. Blood 2008; 111:5571–80 [DOI] [PubMed] [Google Scholar]
- 80.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 2002; 110:993–1002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Dadras SS, Paul T, Bertoncini J, Brown LF, Muzikansky A, Jackson DG, Ellwanger U, Garbe C, Mihm MC, Detmar M. Tumor lymphangiogenesis: a novel prognostic indicator for cutaneous melanoma metastasis and survival. Am J Pathol 2003; 162:1951–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Miyahara M, Tanuma J, Sugihara K, Semba I. Tumor lymphangiogenesis correlates with lymph node metastasis and clinicopathologic parameters in oral squamous cell carcinoma. Cancer 2007; 110:1287–94 [DOI] [PubMed] [Google Scholar]
- 83.Skobe M, Hawighorst T, Jackson DG, Prevo R, Janes L, Velasco P, Riccardi L, Alitalo K, Claffey K, Detmar M. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med 2001; 7:192–8 [DOI] [PubMed] [Google Scholar]
- 84.Ohta Y, Shridhar V, Bright RK, Kalemkerian GP, Du W, Carbone M, Watanabe Y, Pass HI. VEGF and VEGF type C play an important role in angiogenesis and lymphangiogenesis in human malignant mesothelioma tumours. Br J Cancer 1999; 81:54–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wei R, Lv M, Li F, Cheng T, Zhang Z, Jiang G, Zhou Y, Gao R, Wei X, Lou J, Wu X, Luo D, Ma X, Jiang J, Ma D, Xi L. Human CAFs promote lymphangiogenesis in ovarian cancer via the Hh-VEGF-C signaling axis. Oncotarget 2017; 8:67315–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ji RC. Hypoxia and lymphangiogenesis in tumor microenvironment and metastasis. Cancer Lett 2014; 346:6–16 [DOI] [PubMed] [Google Scholar]
- 87.Schoppmann SF, Fenzl A, Schindl M, Bachleitner-Hofmann T, Nagy K, Gnant M, Horvat R, Jakesz R, Birner P. Hypoxia inducible factor-1alpha correlates with VEGF-C expression and lymphangiogenesis in breast cancer. Breast Cancer Res Treat 2006; 99:135–41 [DOI] [PubMed] [Google Scholar]
- 88.Karaman S, Detmar M. Mechanisms of lymphatic metastasis. J Clin Invest 2014; 124:922–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Weichand B, Popp R, Dziumbla S, Mora J, Strack E, Elwakeel E, Frank AC, Scholich K, Pierre S, Syed SN, Olesch C, Ringleb J, Oren B, Doring C, Savai R, Jung M, von Knethen A, Levkau B, Fleming I, Weigert A, Brune B. S1PR1 on tumor-associated macrophages promotes lymphangiogenesis and metastasis via NLRP3/IL-1beta. J Exp Med 2017; 214:2695–713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Li S, Li Q. Cancer stem cells, lymphangiogenesis, and lymphatic metastasis. Cancer Lett 2015; 357:438–47 [DOI] [PubMed] [Google Scholar]
- 91.Semenza GL. Cancer-stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis. Oncogene 2013; 32:4057–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Liang X, Yang D, Hu J, Hao X, Gao J, Mao Z. Hypoxia inducible factor-alpha expression correlates with vascular endothelial growth factor-C expression and lymphangiogenesis/angiogenesis in oral squamous cell carcinoma. Anticancer Res 2008; 28:1659–66 [PubMed] [Google Scholar]
- 93.Schito L, Rey S, Tafani M, Zhang H, Wong CC, Russo A, Russo MA, Semenza GL. Hypoxia-inducible factor 1-dependent expression of platelet-derived growth factor B promotes lymphatic metastasis of hypoxic breast cancer cells. Proc Natl Acad Sci U S A 2012; 109:E2707–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Heo K, Kim YH, Sung HJ, Li HY, Yoo CW, Kim JY, Park JY, Lee UL, Nam BH, Kim EO, Kim SY, Lee SH, Park JB, Choi SW. Hypoxia-induced up-regulation of apelin is associated with a poor prognosis in oral squamous cell carcinoma patients. Oral Oncol 2012; 48:500–6 [DOI] [PubMed] [Google Scholar]
- 95.Berta J, Hoda MA, Laszlo V, Rozsas A, Garay T, Torok S, Grusch M, Berger W, Paku S, Renyi-Vamos F, Masri B, Tovari J, Groger M, Klepetko W, Hegedus B, Dome B. Apelin promotes lymphangiogenesis and lymph node metastasis. Oncotarget 2014; 5:4426–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Michiels C, Minet E, Michel G, Mottet D, Piret JP and Raes M. HIF-1 and AP-1 Cooperate to Increase Gene Expression in Hypoxia: Role of MAP Kinases. IUBMB Life 2001;52(1-2):49–53 [DOI] [PubMed]
- 97.Bandyopadhyay RS, Phelan M, Faller DV. Hypoxia induces AP-1-regulated genes and AP-1 transcription factor binding in human endothelial and other cell types. Biochimica et Biophysica Acta 1995;1264(1):72–78 [DOI] [PubMed]
- 98.Patel V, Marsh CA, Dorsam RT, Mikelis CM, Masedunskas A, Amornphimoltham P, Nathan CA, Singh B, Weigert R, Molinolo AA, Gutkind JS. Decreased lymphangiogenesis and lymph node metastasis by mTOR inhibition in head and neck cancer. Cancer Res 2011; 71:7103–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gimbrone MA, Jr., Leapman SB, Cotran RS, Folkman J. Tumor dormancy in vivo by prevention of neovascularization. J Exp Med 1972; 136:261–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes Cancer 2011; 2:1117–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD, Semenza GL. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol 1996; 16:4604–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lin C, McGough R, Aswad B, Block JA, Terek R. Hypoxia induces HIF-1α and VEGF expression in chondrosarcoma cells and chondrocytes. 2004; 22:1175–81 [DOI] [PubMed] [Google Scholar]
- 103.Cejudo-Martin P, Morales-Ruiz M, Ros J, Navasa M, Fernandez-Varo G, Fuster J, Rivera F, Arroyo V, Rodes J, Jimenez W. Hypoxia is an inducer of vasodilator agents in peritoneal macrophages of cirrhotic patients. Hepatology 2002; 36:1172–9 [DOI] [PubMed] [Google Scholar]
- 104.Desbaillets I, Diserens AC, Tribolet N, Hamou MF, Van Meir EG. Upregulation of interleukin 8 by oxygen-deprived cells in glioblastoma suggests a role in leukocyte activation, chemotaxis, and angiogenesis. J Exp Med 1997; 186:1201–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.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 Carcinogen Mutagene 2013;005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chung AS, Ferrara N. Targeting the tumor microenvironment with SRC kinase inhibition. Clin Cancer Res 2010; 16:775–7 [DOI] [PubMed] [Google Scholar]
- 107.Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971; 285:1182–6 [DOI] [PubMed] [Google Scholar]
- 108.Giantonio BJ, Catalano PJ, Meropol NJ, O’Dwyer PJ, Mitchell EP, Alberts SR, Schwartz MA, Benson AB. Eastern cooperative oncology group study E. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern cooperative oncology group study E3200. J Clin Oncol 2007; 25:1539–44 [DOI] [PubMed] [Google Scholar]
- 109.Okines A, Cunningham D. Current perspective: bevacizumab in colorectal cancer – a time for reappraisal? Eur J Cancer 2009; 45:2452–61 [DOI] [PubMed] [Google Scholar]
- 110.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 2008; 8:592–603 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Vasudev NS, Reynolds AR. Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 2014; 17:471–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Casanovas O, Hicklin DJ, Bergers G, Hanahan D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 2005; 8:299–309 [DOI] [PubMed] [Google Scholar]
- 113.Fernando NT, Koch M, Rothrock C, Gollogly LK, D’Amore PA, Ryeom S, Yoon SS. Tumor escape from endogenous, extracellular matrix-associated angiogenesis inhibitors by up-regulation of multiple proangiogenic factors. Clin Cancer Res 2008; 14:1529–39 [DOI] [PubMed] [Google Scholar]
- 114.Bertolini F, Mancuso P, Shaked Y, Kerbel RS. Molecular and cellular biomarkers for angiogenesis in clinical oncology. Drug Discov Today 2007; 12:806–12 [DOI] [PubMed] [Google Scholar]
- 115.Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009; 139:871–90 [DOI] [PubMed] [Google Scholar]
- 116.Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 2009; 15:232–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009; 15:220–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, Shu Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol Cancer 2019; 18:157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Baluk P, Hashizume H, McDonald DM. Cellular abnormalities of blood vessels as targets in cancer. Curr Opin Genet Dev 2005; 15:102–11 [DOI] [PubMed] [Google Scholar]
- 120.Mazzone M, Dettori D, Leite de Oliveira R, Loges S, Schmidt T, Jonckx B, Tian YM, Lanahan AA, Pollard P, Ruiz de Almodovar C, De Smet F, Vinckier S, Aragones J, Debackere K, Luttun A, Wyns S, Jordan B, Pisacane A, Gallez B, Lampugnani MG, Dejana E, Simons M, Ratcliffe P, Maxwell P, Carmeliet P. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 2009; 136:839–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Ozawa MG, Yao VJ, Chanthery YH, Troncoso P, Uemura A, Varner AS, Kasman IM, Pasqualini R, Arap W, McDonald DM. Angiogenesis with pericyte abnormalities in a transgenic model of prostate carcinoma. Cancer 2005; 104:2104–15 [DOI] [PubMed] [Google Scholar]
- 122.Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH, Rabie T, Kaden S, Grone HJ, Hammerling GJ, Arnold B, Ganss R. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature 2008; 453:410–4 [DOI] [PubMed] [Google Scholar]
- 123.Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell 2014; 26:605–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Moeller BJ, Richardson RA, Dewhirst MW. Hypoxia and radiotherapy: opportunities for improved outcomes in cancer treatment. Cancer Metastas Rev 2007; 26:241–8 [DOI] [PubMed] [Google Scholar]
- 125.Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov 2011; 10:417–27 [DOI] [PubMed] [Google Scholar]
- 126.Jain RK, Duda DG, Clark JW, Loeffler JS. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol 2006; 3:24–40 [DOI] [PubMed] [Google Scholar]
- 127.Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med 2001; 7:987–9 [DOI] [PubMed] [Google Scholar]
- 128.Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 2005; 307:58–62 [DOI] [PubMed] [Google Scholar]
- 129.Yuan F, Chen Y, Dellian M, Safabakhsh N, Ferrara N, Jain RK. Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody. Proc Natl Acad Sci U S A 1996; 93:14765–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumour biology: herceptin acts as an anti-angiogenic cocktail. Nature 2002; 416:279–80 [DOI] [PubMed] [Google Scholar]
- 131.Longley DB, Johnston PG. Molecular mechanisms of drug resistance. J Pathol 2005; 205:275–92 [DOI] [PubMed] [Google Scholar]
- 132.Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002; 2:48–58 [DOI] [PubMed] [Google Scholar]
- 133.Thomas H, Coley HM. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control 2003; 10:159–65 [DOI] [PubMed] [Google Scholar]
- 134.Ambudkar SV, Dey S, Hrycyna CA, Ramachandra M, Pastan I, Gottesman MM. Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol 1999; 39:361–98 [DOI] [PubMed] [Google Scholar]
- 135.Choi CH. ABC transporters as multidrug resistance mechanisms and the development of chemosensitizers for their reversal. Cancer Cell Int 2005; 5:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 1998; 95:15665–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Robey RW, Shukla S, Steadman K, Obrzut T, Finley EM, Ambudkar SV, Bates SE. Inhibition of ABCG2-mediated transport by protein kinase inhibitors with a bisindolylmaleimide or indolocarbazole structure. Mol Cancer Ther 2007; 6:1877–85 [DOI] [PubMed] [Google Scholar]
- 138.Bhatavdekar JM, Patel DD, Chikhlikar PR, Trivedi TI, Gosalia NM, Ghosh N, Shah NG, Vora HH, Suthar TP. Overexpression of CD44: a useful independent predictor of prognosis in patients with colorectal carcinomas. Ann Surg Oncol 1998; 5:495–501 [DOI] [PubMed] [Google Scholar]
- 139.Doktorova H, Hrabeta J, Khalil MA, Eckschlager T. Hypoxia-induced chemoresistance in cancer cells: the role of not only HIF-1. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:166–77 [DOI] [PubMed] [Google Scholar]
- 140.Liu L, Ning X, Sun L, Zhang H, Shi Y, Guo C, Han S, Liu J, Sun S, Han Z, Wu K, Fan D. Hypoxia-inducible factor-1 alpha contributes to hypoxia-induced chemoresistance in gastric cancer. Cancer Sci 2008; 99:121–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Song X, Liu X, Chi W, Liu Y, Wei L, Wang X, Yu J. Hypoxia-induced resistance to cisplatin and doxorubicin in non-small cell lung cancer is inhibited by silencing of HIF-1alpha gene. Cancer Chemother Pharmacol 2006; 58:776–84 [DOI] [PubMed] [Google Scholar]
- 142.Daijo H, Kai S, Tanaka T, Wakamatsu T, Kishimoto S, Suzuki K, Harada H, Takabuchi S, Adachi T, Fukuda K, Hirota K. Fentanyl activates hypoxia-inducible factor 1 in neuronal SH-SY5Y cells and mice under non-hypoxic conditions in a mu-opioid receptor-dependent manner. Eur J Pharmacol 2011; 667:144–52 [DOI] [PubMed] [Google Scholar]
- 143.Li DW, Dong P, Wang F, Chen XW, Xu CZ, Zhou L. Hypoxia induced multidrug resistance of laryngeal cancer cells via hypoxia-inducible factor-1alpha. Asian Pacific J Cancer Prevent 2013; 14:4853–8 [DOI] [PubMed] [Google Scholar]
- 144.Xuan Y, Hur H, Ham IH, Yun J, Lee JY, Shim W, Kim YB, Lee G, Han SU, Cho YK. Dichloroacetate attenuates hypoxia-induced resistance to 5-fluorouracil in gastric cancer through the regulation of glucose metabolism. Exp Cell Res 2014; 321:219–30 [DOI] [PubMed] [Google Scholar]
- 145.Zhou SF. Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition. Xenobiotica 2008; 38:802–32 [DOI] [PubMed] [Google Scholar]
- 146.Cavaco MC, Pereira C, Kreutzer B, Gouveia LF, Silva-Lima B, Brito AM, Videira M. Evading P-glycoprotein mediated-efflux chemoresistance using solid lipid nanoparticles. Eur J Pharm Biopharm 2017; 110:76–84 [DOI] [PubMed] [Google Scholar]
- 147.Sullivan R, Graham CH. Hypoxia prevents etoposide-induced DNA damage in cancer cells through a mechanism involving hypoxia-inducible factor 1. Mol Cancer Ther 2009; 8:1702–13 [DOI] [PubMed] [Google Scholar]
- 148.Ge X, Pan MH, Wang L, Li W, Jiang C, He J, Abouzid K, Liu LZ, Shi Z, Jiang BH. Hypoxia-mediated mitochondria apoptosis inhibition induces temozolomide treatment resistance through miR-26a/bad/bax axis. Cell Death Dis 2018; 9:1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Liu XW, Su Y, Zhu H, Cao J, Ding WJ, Zhao YC, He QJ, Yang B. HIF-1alpha-dependent autophagy protects HeLa cells from fenretinide (4-HPR)-induced apoptosis in hypoxia. Pharmacol Res 2010; 62:416–25 [DOI] [PubMed] [Google Scholar]
- 150.Nishi H, Nakada T, Kyo S, Inoue M, Shay JW, Isaka K. Hypoxia-inducible factor 1 mediates upregulation of telomerase (hTERT). Mol Cell Biol 2004; 24:6076–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Schnitzer SE, Schmid T, Zhou J, Brune B. Hypoxia and HIF-1alpha protect A549 cells from drug-induced apoptosis. Cell Death Differ 2006; 13:1611–3 [DOI] [PubMed] [Google Scholar]
- 152.Coffey RN, Morrissey C, Taylor CT, Fitzpatrick JM, Watson RW. Resistance to caspase-dependent, hypoxia-induced apoptosis is not hypoxia-inducible factor-1 alpha mediated in prostate carcinoma cells. Cancer 2005; 103:1363–74 [DOI] [PubMed] [Google Scholar]
- 153.Rohwer N, Dame C, Haugstetter A, Wiedenmann B, Detjen K, Schmitt CA, Cramer T. Hypoxia-inducible factor 1alpha determines gastric cancer chemosensitivity via modulation of p53 and NF-kappaB. PLoS One 2010; 5:e12038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Ohno T. Particle radiotherapy with carbon ion beams. EPMA J 2013; 4:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Grimes DR, Partridge M. A mechanistic investigation of the oxygen fixation hypothesis and oxygen enhancement ratio. Biomed Phys Eng Exp 2015; 1:045209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Stadler P, Becker A, Feldmann HJ, Hansgen G, Dunst J, Wurschmidt F, Molls M. Influence of the hypoxic subvolume on the survival of patients with head and neck cancer. Int J Radiat Oncol Biol Phys 1999; 44:749–54 [DOI] [PubMed] [Google Scholar]
- 157.Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Bean JM, Prosnitz LR, Dewhirst MW. Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Res 1996; 56:941–3 [Database [PubMed] [Google Scholar]
- 158.Lu H, Samanta D, Xiang L, Zhang H, Hu H, Chen I, Bullen JW, Semenza GL. Chemotherapy triggers HIF-1-dependent glutathione synthesis and copper chelation that induces the breast cancer stem cell phenotype. Proc Natl Acad Sci U S A 2015; 112:E4600–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Chandel NS, Maltepe E, Goldwasser E, Mathieu CE, Simon MC, Schumacker PT. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc Natl Acad Sci USA 1998; 95:11715–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Koshikawa N, Hayashi J, Nakagawara A, Takenaga K. Reactive oxygen species-generating mitochondrial DNA mutation up-regulates hypoxia-inducible factor-1alpha gene transcription via phosphatidylinositol 3-kinase-Akt/protein kinase C/histone deacetylase pathway. J Biol Chem 2009; 284:33185–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Palmer LA, Gaston B, Johns RA. Normoxic stabilization of hypoxia-inducible factor-1 expression and activity: redox-dependent effect of nitrogen oxides. Mol Pharmacol 2000; 58:1197–203 [DOI] [PubMed] [Google Scholar]
- 162.Verovski VN, Van den Berge DL, Soete GA, Bols BL, Storme GA. Intrinsic radiosensitivity of human pancreatic tumour cells and the radiosensitising potency of the nitric oxide donor sodium nitroprusside. Br J Cancer 1996; 74:1734–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Illum H, Wang DH, Dowell JE, Hittson WJ, Torrisi JR, Meyer J, Huerta S. Phase I dose escalation trial of nitroglycerin in addition to 5-fluorouracil and radiation therapy for neoadjuvant treatment of operable rectal cancer. Surgery 2015; 158:460–5 [DOI] [PubMed] [Google Scholar]
- 164.Janssens MY, Van den Berge DL, Verovski VN, Monsaert C, Storme GA. Activation of inducible nitric oxide synthase results in nitric oxide-mediated radiosensitization of hypoxic EMT-6 tumor cells. Cancer Res 1998; 58:5646–8 [PubMed] [Google Scholar]
- 165.Griffin RJ, Makepeace CM, Hur WJ, Song CW. Radiosensitization of hypoxic tumor cells in vitro by nitric oxide. Int J Radiat Oncol Biol Phys 1996; 36:377–83 [DOI] [PubMed] [Google Scholar]
- 166.Bump EA, Yu NY, Brown JM. Radiosensitization of hypoxic tumor cells by depletion of intracellular glutathione. Science 1982; 217:544–5 [DOI] [PubMed] [Google Scholar]
- 167.Held KD, Epp ER, Clark EP, Biaglow JE. Effect of dimethyl fumarate on the radiation sensitivity of mammalian cells in vitro. Radiat Res 1988; 115:495–502 [PubMed] [Google Scholar]
- 168.Rodman SN, Spence JM, Ronnfeldt TJ, Zhu Y, Solst SR, O’Neill RA, Allen BG, Guan X, Spitz DR, Fath MA. Enhancement of radiation response in breast cancer stem cells by inhibition of thioredoxin- and glutathione-dependent metabolism. Radiat Res 2016; 186:385–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Wang H, Bouzakoura S, de Mey S, Jiang H, Law K, Dufait I, Corbet C, Verovski V, Gevaert T, Feron O, Van den Berge D, Storme G, De Ridder M. Auranofin radiosensitizes tumor cells through targeting thioredoxin reductase and resulting overproduction of reactive oxygen species. Oncotarget 2017; 8:35728–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Sun L, Moritake T, Ito K, Matsumoto Y, Yasui H, Nakagawa H, Hirayama A, Inanami O, Tsuboi K. Metabolic analysis of radioresistant medulloblastoma stem-like clones and potential therapeutic targets. PLoS One 2017; 12:e0176162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Maggiorella L, Wen B, Frascogna V, Opolon P, Bourhis J, Deutsch E. Combined radiation sensitizing and anti-angiogenic effects of ionizing radiation and the protease inhibitor ritonavir in a head and neck carcinoma model. Anticancer Res 2005; 25:4357–62 [PubMed] [Google Scholar]
- 172.Zannella VE, Dal Pra A, Muaddi H, McKee TD, Stapleton S, Sykes J, Glicksman R, Chaib S, Zamiara P, Milosevic M, Wouters BG, Bristow RG, Koritzinsky M. Reprogramming metabolism with metformin improves tumor oxygenation and radiotherapy response. Clin Cancer Res 2013; 19:6741–50 [DOI] [PubMed] [Google Scholar]
- 173.Zhang XD, Chen J, Luo Z, Wu D, Shen X, Song SS, Sun YM, Liu PX, Zhao J, Huo S, Fan S, Fan F, Liang XJ, Xie J. Enhanced tumor accumulation of Sub-2 nm gold nanoclusters for cancer radiation therapy. Adv Healthcare Mater 2014; 3:133–41 [DOI] [PubMed] [Google Scholar]
- 174.Chen B, Zhang M, Xing D, Feng Y. Atorvastatin enhances radiosensitivity in hypoxia-induced prostate cancer cells related with HIF-1alpha inhibition. Biosci Rep 2017; 37. doi:10.1042/BSR20170340 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Lee DE, Alhallak K, Jenkins SV, Vargas I, Greene NP, Quinn KP, Griffin RJ, Dings RPM, Rajaram N. A radiosensitizing inhibitor of HIF-1 alters the optical redox state of human lung cancer cells in vitro. Sci Rep 2018; 8:8815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Wang H, Mu X, He H, Zhang XD. Cancer radiosensitizers. Trends Pharmacol Sci 2018; 39:24–48 [DOI] [PubMed] [Google Scholar]



