Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2020 Apr 23;245(10):851–860. doi: 10.1177/1535370220921547

Role of hyperoxic treatment in cancer

Sei W Kim 1, In K Kim 1,2, Sang H Lee 1,2,
PMCID: PMC7268933  PMID: 32326758

Abstract

The occurrence of hypoxia is common in many solid tumors, and it enhances aggressive features of cancer such as cell survival, angiogenesis, and metastasis while minimizing the efficacies of chemotherapy and radiotherapy. Hypoxia also plays a pivotal role in regulating immune cell function which is important for immunotherapy. Hypoxia-inducible factor has been suggested as a master regulator of tumor cell adaptation to the hypoxic microenvironment. Currently, several approaches have been proposed to eliminate the hypoxic state in tumors for delaying cancer progression and improving therapeutic efficacy. In this review, we summarize current findings on the relevance of hyperoxia-based therapeutics for cancer treatment. Accumulating evidence indicates that hyperoxic therapy inhibits tumor growth and increases treatment efficacy. Primary antitumor effect of hyperoxic therapy may be due to the reversal of tumor hypoxia and the generation of reactive oxygen species. Restoring immune function is also suggested as a potential mechanism. Hyperoxic therapy can also cause cellular injury and organ dysfunction. In conclusion, overcoming tumor hypoxia is a major problem that needs to be solved. Further studies to standardize and personalize hyperoxia therapy according to the type of cancer, stage, and comorbidities are needed.

Impact statement

Tumor hypoxia promotes cancer cell aggressiveness, and is strongly associated with poor prognosis across multiple tumor types. The hypoxic microenvironments inside tumors also limit the effectiveness of radiotherapy, chemotherapy, and immunotherapy. Several approaches to eliminate hypoxic state in tumors have been proposed to delay cancer progression and improve therapeutic efficacies. This review will summarize current knowledge on hyperoxia, used alone or in combination with other therapeutic modalities, in cancer treatment. Molecular mechanisms and undesired side effects of hyperoxia will also be discussed.

Keywords: Neoplasms, hyperoxia, oxygen, reactive oxygen species, oxidative stress, antitumor effect

Introduction

Oxygen is an indispensable element for cells in our body to fulfill energy requirement from aerobic metabolism. Therefore, reduced oxygen levels, hypoxia, may produce cell death and resulting organ dysfunction as occurs in heart attacks or strokes. Hypoxia commonly arises in the core of most solid tumors as a result of an inadequate supply of oxygen from abnormal vasculature, and an increased oxygen demand from changes in tumor metabolism.1Severity of tumor hypoxia varies depending on tissue of origin and tumor size, and median oxygen levels in untreated tumors are frequently less than 2% in comparison to the normal human tissues (5%).2,3The hypoxic regions within the tumor are heterogeneously distributed and may even located near to vessels.4

Traditionally, hypoxia was thought of as a factor limiting the cancer growth by reducing the ability of cells to divide.5However, increasing evidence indicates that tumor hypoxia plays an important role in cancer progression. When tumor cells adapt to the imbalance between oxygen supply and demand, malignant features of solid tumors such as resistance to cell death, angiogenesis, and metastasis were invariably enhanced.6,7Emerging features of cancer development such as genome instability, the enrichment of cancer stem cells, and aberrant exosomal secretion were also suggested as hallmarks of tumor hypoxia.810Overall, hypoxic microenvironment in tumor promotes acquisition of the more aggressive cancer phenotypes and thus is associated with poor prognosis.11,12In addition, tumor hypoxia has been considered as one of the biggest barriers to treating cancer because it renders tumors more resistant to surgery, radiotherapy, chemotherapy, and immunotherapy.1316In this regard, it is widely accepted that hypoxia is a potential cancer-specific target,11,17and several approaches targeting tumor hypoxia for cancer therapy such as erythropoietin, nicotinamide, hypoxia-activated prodrugs, and nanocarriers have been proposed.14,18

Numerous studies also suggest increasing oxygen content using hyperoxia as a promising therapeutic modality to reverse tumor hypoxia as well as resistance to cancer therapeutics.5,1921Recently, we also identified anti-cancer effect of hyperoxia and its mechanism of action in mouse lung cancer.22,23However, despite of growing interest and clinical trials, the application of hyperoxia to cancer treatment is still in its nascent stage. Here, we will focus on the recent insights into the understanding of hyperoxia-mediated cancer treatment.

Hyperoxic therapy in cancer

Oxygen therapy is a type of treatment to enhance the amount of dissolved oxygen in the plasma by breathing supplemental oxygen, thereby increasing O2delivery to body tissue. For hyperoxygenation, 100% oxygen is administered to a patient either at ambient atmospheric pressure (normobaric oxygen, NBO) or under hyperbaric conditions usually in a pressurized chamber (hyperbaric oxygen, HBO). Oxygen therapy has long been regarded as integral to the management of various medical conditions for centuries. For cancer treatment, there is still no evidence that it is effective in treating cancer.24However, despite ongoing controversy, interest in applying hyperoxic therapy as adjuvant for cancer treatment is rapidly growing. For example, in Pubmed search concerning oxygen therapy and cancer, since the first article in 1951,25more than 20,000 were found. A brief summary of hyperoxic therapies on cancer is listed in Table 1.

Table 1.

Summary of studies on cancer with hyperoxic therapies.

References Year Type of tumor Type of hyperpoxic treatment Combination treatment Summary of the study
Moen et al.26 2009 DMBA-induced mammary tumor mouse model 2 bar, pO2 = 2 bar, 4 times (day 1, 4, 7 and 10) exposures of 90 min 5-FU HBO increases the uptake of [3H]-5FU
Kawasoe et al.27 2009 Osteosarcoma LM8 cells mouse model 2.5 bar, pO2 = 2.5 bar, 60 min, 5 times a week until 5 weeks Carboplatin HBO or HBO with carboplatin inhibits tumor growth and lung metastasis, synergistically
Selvendiran et al.28 2010 A2780 ovarian xenograft tumor mouse model 100% O22 atm for 90 min, 21 days Cisplatin Reduction of tumor volume. No significance between HBO and cisplatin-treated group
Sun et al.29 2012 Human glioblastoma multiforme cells (D54, U87) 40%, 80% O2, 72 h Temozolomide NBO enhanced TMZ toxicity in GBM cells
Lee et al.30 2014 Human glioblastoma multiforme cells (D54, U87, U251) 40% O2, 24, 48 and 72 h Temozolomide NBO enhanced the sensitivity to temozolomide in chemosensitive and -resistant GBM cells
Hatfield et al.31 2016 MCA205 fibrosarcoma mouse model 60% O2, 72 h and 11 days NBO reverses the hypoxia–A2-adenosinergic immunosuppression during acute inflammation
Yttersian Sletta et al.21 2017 Breast cancer mouse model 2.5 bar, pO2 = 2.5 bar, 90 min, every third day until day 16 5-FU Suppressed tumor growth and metastatic lesions, HBO does not enhance the 5-FU efficacy
Kim et al.23 2018 Lewis lung carcinoma cell injected mouse model 24 h NBO (95% O2)/normoxia cycle for two weeks NBO inhibits the progression of lung cancer by inducing apoptosis
Lee et al.22 2018 benzo[a]pyrene -induced lung tumorigenesis mouse model 95% O2for 3 h/day, days 21–28 Carboplatin Intermittent NBO with carboplatin displays a synergistic tumoricidal effect
Qian et al.20 2019 triple-negative breast cancer mouse model 60% O2, day 7-28 NBO reverses immunosuppression and control the extend of lung metastases

In addition to oxygen breathing, although relatively uncommon, other forms of oxygen therapy such as oxygen-containing substances (i.e. ozone or hydrogen peroxide), and prodrugs that are activated only in the hypoxic tumor environment also have been tried for cancer treatment.3234

Hyperbaric oxygen therapy

HBO treatment is breathing 100% oxygen at higher atmospheric pressure usually between 1.5 and 3 atm which enable the lungs to receive more oxygen up to three times than under normal air pressure, resulting in immediate saturation of plasma with oxygen. In normal subject, compared to normobaric air, HBO at 3 atm increases both arterial (from 100 to 2000 mmHg) and tissue (from 55 to 500 mmHg) oxygen tensions (PO2).35Similarly, the PO2level was significantly increased in tumor tissue by HBO exposure, demonstrating three to four times greater hyperoxygenation effect in tumor tissue than NBO.36,37Therefore, HBO rather than NBO has been preferentially used to eliminate poorly oxygenated regions of tumor which play a major role in tumor development and resistance to other therapeutic modalities. In addition, HBO therapy is considered safe and well tolerated, and side effects are rare.38Although several studies showed the favorable outcomes of HBO by itself in cancer treatment,5,28,39HBO alone gives a limited curative effects, and it even enhances tumor growth.19,40Accordingly, HBO is preferentially used as an adjuvant treatment for enhancing tumor sensitivity and decreasing complications of other therapies.34

Normobaric oxygen therapy

NBO therapy is a routine adjuvant oxygenation intervention supplied by nasal cannula or facemask under ambient pressure. For cancer treatment, NBO is an attractive alternative to HBO due to its ease of administration and lower complication in actual clinical practice.41However, when compared to HBO, effect of NBO on arterial and tissue oxygenation is much weaker. Arterial PO2in aorta has increased only 4-fold after exposure to NBO (345 vs. 84.1 mmHg).42In animal study, most of tumors were nearly anoxic (PO2<1 mmHg), but NBO immediately increased PO2(mean >25 mmHg) and remained elevated during gas exposure.43Accordingly, NBO treatment significantly retarded tumor growth.39,44We also previously showed the NBO inhibits lung cancer in in vivoand in vitrothrough reactive oxygen species (ROS) generation and apoptosis.23

Molecular mechanisms: Hypoxia-inducible factor 1α

Hypoxia

When oxygen levels in tumor microenvironment are dropping, transcriptional induction of a series of genes necessary for maintaining cell survival/proliferation and promoting more aggressive features occurs in tumor cells. These oxygen-dependent responses are tightly regulated by HIF-1α, the master transcriptional regulator of the hypoxic response as well as a representative endogenous biomarker for hypoxia.45HIF-1α is induced by hypoxia through post-translational modification, and it binds to specific recognition sequences in the genome to increase the expression of HIF-1α target genes.46Since the seminal discovery of HIF in the early 1990s by Gregg Semenza, a Nobel Laureate in Physiology or Medicine for 2019,47thousands of genes are identified as direct targets of HIF-1α,48and therefore a myriad of changes associated with tumor aggravation occur in hypoxic tumor cells including angiogenesis and oxygen supply, stemness/self-renewal, proliferation, epithelial to mesenchymal transition, metastasis and invasion, redox homoestasis, anti-apoptosis, and metabolic reprogramming.49,50Metabolic reprogramming from oxidative phosphorylation to accelerated glycolysis in cancer cells is also known to be mediated via HIF-1α.51In addition, HIF-1α contributes to the development of tumor resistance to therapeutic approaches and serves as a promising biomarker.52,53For example, HIF-1α limits T cell recognition of tumor cells by downregulating MHC class I molecule expression.54It also induces multidrug resistance, the major cause of chemotherapy failure, by inducing multidrug resistance-associated protein 1 in cancer cells.55Accordingly, in cancer patients, protein levels of HIF-1α in solid tumors are considered as a critical prognostic factor.56Based on these findings, HIF-1α has become targets for developing novel cancer therapeutics. However, no agents directly inhibiting HIF-1α have been approved for treating cancer patients.57

Hyperoxia

Many reports suggest various beneficial effects of hyperoxia on hypoxic tumor. Hyperoxic breathing of 60% O2recovers oxygen homeostasis in tumor microenvironment hypoxia to normoxia, inhibits survival/proliferation, stemness, and immune escape of cancer cells, resensitizes chemoresistance, leading to tumor regression.29,58,59We also reported anti-cancer effects of hyperoxia, alone or in combination with a chemotherapeutic drug carboplatin, on mouse lung cancer; normobaric hyperoxia significantly induces oxidative stress and apoptosis in tumor tissue and reduces tumor mass and migration/invasion.22,23Differential response of cancer and normal cells to hyperoxia has also provided a treatment rationale of hyperoxia in cancer treatment. Basal antioxidant defense levels are aberrant in tumor cells, thus an increased oxidative stress has been observed. Since the superoxide dismutase activity in most tumors are lower than normal tissue, tumor cells show higher susceptibility to increased ROS activity induced by hyperoxia.60Synthesis of glutathione (GSH), an antioxidant preventing ROS-induced damage, increases proportional to ambient oxygen tension, but not in cancer cells.61In consistent with these findings, we demonstrated that NBO showed anti-tumor activity in lung tumor cells but not in normal lung cells.23In addition, hyperoxia is angiogenic in normal tissues, but anti-angiogenic in tumor tissues.62

Anti-cancer mechanisms of hyperoxia can also be inferred indirectly from clinical and experimental findings. Roles of HIF-1α in hypoxia-mediated cancer development are widely acknowledged, and hyperoxic treatment has long been used to cure clinical disorders such as hypoxia or ischemic diseases by increasing oxygen delivery to oxygen-deficient tissues. We can also take a hint from well-known mechanisms for the induction and regulation of HIF-1α: HIF-1α protein is subject to degradation through an oxygen-dependent ubiquitination,63and hypoxia-induced HIF-1α protein is rapidly decayed within 5 min upon exposure to normoxia (20% O2).64In addition, transcriptional activity of HIF-1α is enhanced by hypoxia-induced ROS,65and inhibited by oxygen in nonhypoxic cells.66Taken these findings together, it seems reasonable that therapeutic effects of hyperoxia on hypoxic tumor are mediated through not direct regulation of HIF-1α but primarily the reversal of hypoxia and the attenuation of the HIF-mediated effects.

However, despite of accumulating evidence supporting this idea, many reports also indicate HIF-1α–independent effects of hyperoxia on tumor. For example, both normoxia and hyperoxia induces higher levels of HIF-1α than hypoxia in tumors, but tumor grows faster in hypoxia group, suggesting that signaling pathways other than HIF-1α driven response may play important roles for in vivocancer cell proliferation.67Recent report also showed that HIF-1α levels in tumor cells was significantly downregulated in hyperoxia (60% O2) than normoxia (20% O2), indicating that alternative mechanisms other than simple reversal of tumor hypoxia to normoxia underlie anti-tumor effects of hyperoxia.59Coincidentally, Rocco et al.68suggest that relative changes of oxygen availability rather than steady state hypoxic or hyperoxic conditions play an important role in HIF transcriptional effects. In addition, hyperoxia activates HIF-1α overexpression through the activation of Src oncogene, and also inhibits the stability of HIF-1α by reducing ROS formation.65,69,70Despite numerous attempts, mechanisms underlying hyperoxia-mediated anti-cancer activity remain to be elucidated.

Side effects

Hyperoxic therapy is usually well tolerated with an acceptable rate of complications; however, as with all medical treatments, it also includes medical risks. Dependent on the type of hyperoxic therapy, patients may experience two prominent side effects due to exposure to high levels of oxygen (oxygen toxicity) or high atmospheric pressure (barotrauma). As shown above, PO2used in HBO therapy is much higher than that of NBO, thus oxygen toxicity is more common in patients exposed to HBO. In addition, only HBO therapy is carried out in a hyperbaric chamber, thus barotrauma occurs only in patients taking HBO therapy.

Oxygen toxicity

Exposure to high concentration of oxygen is well known cause of cell damage. For example, oxygen, at concentrations of 95% or more, is severely cytotoxic to the pulmonary cells of many animal species, including humans.71Humans appear more resistant to oxygen-induced damage and the risk of hyperoxic acute lung injury is minimal when the FiO2 is 0.672but continuous exposure to elevated levels of oxygen may cause oxygen toxicity. Major organs subject to oxygen toxicity are lungs (hyperoxic acute lung injury), central nervous system (loss of consciousness and oxygen toxicity seizure), and eye (hyperoxic myopia and cataract).7375Oxygen toxicity is believed to be mediated primarily by a production of ROS at levels exceeding the capacity of antioxidant defence mechanisms.76Following HBO treatment, ROS increased to about 2.14–2.44 fold in mitochondria and 1.32–1.42 fold in whole cell.77Reduction of antioxidant enzymes such as superoxide dismutase and glutathione by HBO, but not by NBO, also contributes to increased oxygen toxicity.78Excessive ROS can damage all essential macromolecules, including nucleic acids, lipids and proteins, leading to an overall progressive decline in physiological function.79For example, protein oxidation and nitrosylation can impair a wide variety of enzymatic processes and growth factors that can result in marked cellular dysfunction.80Lipid peroxidation activates apoptosis through activation of sphingomyelinase and release of ceramide.81Nucleic acid oxidation has been linked with aging and DNA strand breaks, leading to necrosis and/or apoptosis.82Therefore, HBO-driven ROS can display many harmful activities including the induction of DNA damage, cell death, cellular senescence, and deleterious inflammatory response which in turn exacerbates oxidative toxicity and tissue damage.83,84Several mechanisms involved in hyperoxia-induced oxygen toxicity have been proposed. For example, ROS stimulates signaling pathways mediated via protein kinases (Akt, MAPK and PKC), resulting in activation of transcription factors (Nrf2, NF-κB, and AP-1) responsible for cell death and inflammation.85In addition, ROS-independent mechanisms such as the induction of apoptosis by direct activation of Bax, Bak, or FAS,86chemokine receptor CXCR2-mediated tissue inflammation,87and toll-like receptor-linked cell damages88also have been suggested.

Barotrauma and other complications

In HBO therapy, unlike to NBO, patients are exposed to the high atmospheric pressure in hyperbaric chamber. Barotrauma, pressure-induced injury, is caused by inability to equalize pressure between the environment and the air-filled space in the body such as lungs, ear, sinuses, eyes, and teeth are concurrently at risk. The most common type of barotrauma (>17%) is middle ear barotrauma which can lead to permanent hearing loss and vertigo.89

Some patients can develop a feeling of claustrophobia, the fear of being enclosed in small spaces with no escape, due to the confined nature of hyperbaric chamber. HBO therapy also causes mild increase in blood pressure in both hypertensive and non-hypertensive patients, and hypoglycemia can occur in diabetic patients.90In addition, NBO increases pulmonary metastasis of tumor91and inhibits glucose-induced insulin release.92

Because hyperoxia, unlike hypoxia, is a man-made condition, specific adaptive response to hyperoxia has not been evolved in humans. To enhance safety and to prevent side-effects of HBO therapy in cancer treatment as well as other clinical trials, further investigation to maximize therapeutic efficacy and minimize complications by standardizing therapy protocol in particular with regard to pressure and duration is required.

Role of hyperoxia treatment in chemotherapy

Hypoxia reduces the sensitivity of cancer to chemotherapy.6In addition, hypoxic cells do not receive sufficient chemotherapeutic agents due to distance from the capillary and because of abnormal vascularization of tumors. Hypoxia remotes resistance through the HIF-1-mediated upregulation of different genes and signaling pathways.93Hypoxia-induced drug resistance is also explained by inhibition of apoptotic pathways23,94and increased intracellular drug efflux.95Currently, respiratory hyperoxia is mainly used for the treatment of hypoxic tissue damage. Also, hyperoxia has also been shown to improve the treatment efficacy of chemotherapy in animal models.20,22,26From our previous study, NBO therapy was found to be tumoricidal and NBO with carboplatin exhibited a synergistic antitumor effect on B[a]P-induced lung cancers in mice.22,23Oxidative stress and its effects on DNA are increased following exposure to hyperoxia and even more with chemotherapy, and this may lead to apoptosis of lung tumors. Lee et al.30showed that NBO treatment resensitizes chemoresistant glioblastoma cells to temozolomide through unfolded protein response.

Other studies were mostly performed under HBO treatment. Moen et al.26showed that HBO treatment increases the uptake of 5-fluorouracil in mammary tumors for the duration of, and immediately after, HBO treatment. Kawasoe et al.27showed significant suppression of osteosarcoma with HBO plus carboplatin compared with monotherapy, both in in vitroand in vivo.Other studies also showed increased efficacy against variety of malignancies with combination of HBO and chemotherapy.28,96,97However, precise mechanisms are not known until now and more standard combination treatment protocols are needed. In addition, a combination of HBO treatment and particular chemotherapeutic agents (doxorubicin, bleomycin, and disulfiram) may cause potential toxicity98because it can potentiate oxygen-related serious organ damage.99101However, studies showing conflicting results also exist.102,103

Role of hyperoxic treatment in radiation therapy

The primary mechanism of radiation therapy is creation of ROS, which in turn induces cell death by the mechanisms including apoptosis, necrosis, autophagy, and senescence.104In hypoxic state, DNA radicals are repaired by abstracting hydrogen from sulfhydryl group present in protein.1Since oxygen is required for ROS generation, hypoxic tumors are resistant to the cytotoxic effects of radiotherapy.105HIF-1 plays a role in radioresistance of a tumor by up-regulating downstream genes, which are involved in apoptosis, metabolism, proliferation, and neovascularization.106In general, cells irradiated under normal oxygenated conditions are two- to three-fold more radiosensitive than cells irradiated under hypoxic or anoxic conditions.107,108Several human studies reported significant improvement of survival and local tumor control in patients with cancer treated with radiotherapy and HBO.109111However, other studies suggest a high rate of complications from combination of HBO and radiotherapy, including severe tissue radiation injury and seizures.34,110Concerning this matter, beneficial effects of NBO on tumor radiosensitivity have been also reported.112,113In addition, both the extent and the timing of this hyperoxic therapy are variable. From systematic reviews in patients with high grade gliomas, radiation therapy after HBO treatment was tolerated and beneficial.111

Role of hyperoxic treatment in immunotherapy

Oxygen tension directly affects immune cell function, and thus hypoxia can cause immunosuppression and/or immune dysfunction.114It is widely appreciated that hypoxic tumor microenvironment negatively affects anti-tumor immune responses, and also is responsible for resistance to immunotherapy.16,115Many mechanisms underlying hypoxia-induced immunomodulation in cancer have been suggested.116Extracellular adenosine, a potent immunosuppressive metabolite, is increased in hypoxic conditions, and controlled by two cell surface nucleotidases; CD39 and CD73 in tumors,117,118providing evidence that the adenosine-dependent immunoregulation is important for hypoxia-mediated immunosuppression. TGF-β, a potent immunosuppressive cytokine, is upregulated in tumor cells after culturing in hypoxic conditions.119Hypoxia also increases the accumulation of intracellular adenosine by HIF-1α-dependent mechanism, resulting in the elevation of extracellular adenosine independent of CD39/CD73.120,121Programmed cell death-1 (PD-1) and programmed death-ligand 1 (PD-L1), important players in immune checkpoint pathways, are also regulated oxygen-dependently in the tumor microenvironment. Hypoxia increases PD-L1 expression which induces cancer cell resistance to T-cell dependent cytotoxicity.122,123Exposure to adenosine or the activation of its receptors in T cells also downregulates T cell activities by inducing PD-1 and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) expression.124,125Activities of myeloid-derived suppressor cells (MDSCs), typical immunosuppressive cells in the tumor microenvironment, are also regulated by hypoxia and extracellular adenosine.126,127

Based on these findings, hyperoxia therapy has been attempted to restore hypoxia-induced impairment of immune function in cancer cells, in particular by downregulation of immune checkpoint pathways.31In mice, 60% oxygen efficiently reduced tumor burden only in wild-type mice, but not in immunocompromised mice, indicating the involvement of immune response in anti-tumor activities of hyperoxia.31Hyperoxia-induced alleviation of hypoxia reduces the levels of immunosuppressive molecules such as adenosine, TGF-β and PD-L1 in the tumor, and enhances anti-tumor immune responses.31,58,59Population of typical immunosuppressive immune cells such as MDSCs and Treg cells in the tumor microenvironment is also decreased by hyperoxia therapy.20,31Recently, Wang et al.59reported that hyperoxia reduces stemness of colorectal cancer cells through the inhibition of hypoxia-mediated production of exosome from granulocytic MDSCs.

The role of cancer immunotherapy has become increasingly important compared to traditional cancer treatments. Accumulating evidence indicates that attenuation of immunosuppressive activity in the tumor microenvironment by regulating immune checkpoints is the key factor for the success of cancer immunotherapy.128In this regard, it is intriguing that the inhibition of immune checkpoints pathways is the main mechanism underlying anti-tumor activity of hyperoxia.114Although several immune regulators and molecular mechanisms involved in hyperoxia-mediated antitumor activities have been identified, precise adjunctive role for hyperoxia in cancer immunotherapy still remains unclear.

Future directions and conclusion

Tumor hypoxia is a major treatment target for effective cancer therapy and inhibition of cancer progression. Hyperoxia therapy has been suggested to reverse cancer hypoxia, and it is more often used as an adjunctive treatment for cancer treatment along with other therapeutic modalities. Currently, no standard protocols for hyperoxic tumor therapy are approved. In case of combining hyperoxia with radiation, hyperoxic periods are relatively short and may not have significant side effects,34but in other cases where oxygen is administered over a long period, hyperoxia can cause cellular injury and organ dysfunction. Further, randomized, large, and well-organized clinical research could reinforce the use of hyperoxia therapy in the clinical setting for cancer treatment with minimal complications. Furthermore, more personalized approach according to the type of cancer and comorbidities are needed. Hypoxia-activated prodrugs or HIF inhibitors are suggested as possible alternatives to hyperoxic cancer therapy. The number of preclinical and clinical trials targeting low-oxygen tumor compartments via hypoxia-activated prodrugs is increasing.129In addition, combinations of clinical immunotherapy and immunomodulation from HIF inhibitor have a possibility of being powerful treatment option.130In conclusion, overcoming tumor hypoxia is an urgent problem to be solved for effective treatment of cancer patients. Further research is required to determine the mechanism for the role of oxygen tension in cancer progress and treatment, and to develop standard protocols for increasing efficacies and safety of hyperoxia therapy in cancer.

Authors’ contributions

All authors contributed to the preparation of this manuscript and approved the final version. SWK and IKK wrote the manuscript. SHL reviewed and edited 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

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2014R1A1A2058026) and by grants Clinical Research Laboratory of The Catholic University of Korea, St. Paul’s Hospital.

ORCID iD

Sei W Kim https://orcid.org/0000-0002-2798-421X

References

  • 1.Brown JM, Wilson WR. Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer 2004; 4:437–47 [DOI] [PubMed] [Google Scholar]
  • 2.Li XF, Carlin S, Urano M, Russell J, Ling CC, O’Donoghue JA. Visualization of hypoxia in microscopic tumors by immunofluorescent microscopy. Cancer Res 2007; 67:7646–53 [DOI] [PubMed] [Google Scholar]
  • 3.McKeown SR. Defining normoxia, physoxia and hypoxia in tumours-implications for treatment response. Br J Radiol 2014; 87:1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 2007; 26:225–39 [DOI] [PubMed] [Google Scholar]
  • 5.Moen I, Stuhr LE. Hyperbaric oxygen therapy and cancer – a review. Target Oncol 2012; 7:233–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.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; 18:157–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Science 2016; 352:175–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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 2019; 51:308–18 [DOI] [PubMed] [Google Scholar]
  • 9.Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 2017; 140:10–22 [DOI] [PubMed] [Google Scholar]
  • 10.Choudhry H, Harris AL. Advances in hypoxia-inducible factor biology. Cell Metab 2018; 27:281–98 [DOI] [PubMed] [Google Scholar]
  • 11.Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer 2011; 11:393–410 [DOI] [PubMed] [Google Scholar]
  • 12.Campbell EJ, Dachs GU, Morrin HR, Davey VC, Robinson BA, Vissers M. Activation of the hypoxia pathway in breast cancer tissue and patient survival are inversely associated with tumor ascorbate levels. BMC Cancer 2019; 19:307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dietl B, Marienhagen J, Schafer C, Kolbl O. The prognostic value of anaemia at different treatment times in patients with locally advanced head and neck cancer treated with surgery and postoperative radiotherapy. Clin Oncol 2007; 19:228–33 [DOI] [PubMed] [Google Scholar]
  • 14.Manoochehri Khoshinani H, Afshar S, Najafi R. Hypoxia: a double-edged sword in cancer therapy. Cancer Invest 2016; 34:536–45 [DOI] [PubMed] [Google Scholar]
  • 15.Hughes VS, Wiggins JM, Siemann DW. Tumor oxygenation and cancer therapy-then and now. Br J Radiol 2019; 92:1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Noman MZ, Hasmim M, Messai Y, Terry S, Kieda C, Janji B, Chouaib S. Hypoxia: a key player in antitumor immune response. A review in the theme: cellular responses to hypoxia. Am J Physiol Cell Physiol 2015; 309:C569–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bosco MC, D’Orazi G, Del Bufalo D. Targeting hypoxia in tumor: a new promising therapeutic strategy. J Exp Clin Cancer Res 2020; 39:8–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Jahanban-Esfahlan R, de la Guardia M, Ahmadi D, Yousefi B. Modulating tumor hypoxia by nanomedicine for effective cancer therapy. J Cell Physiol 2018; 233:2019–31 [DOI] [PubMed] [Google Scholar]
  • 19.Doguchi H, Saio M, Kuniyoshi S, Matsuzaki A, Yoshimi N. The enhancing effects of hyperbaric oxygen on mouse skin carcinogenesis. J Toxicol Pathol 2014; 27:67–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Qian X, Zhang Q, Shao N, Shan Z, Cheang T, Zhang Z, Su Q, Wang S, Lin Y. Respiratory hyperoxia reverses immunosuppression by regulating myeloid-derived suppressor cells and PD-L1 expression in a triple-negative breast cancer mouse model. Am J Cancer Res 2019; 9:529–45 [PMC free article] [PubMed] [Google Scholar]
  • 21.Yttersian Sletta K, Tveitaras MK, Lu N, Engelsen AST, Reed RK, Garmann-Johnsen A, Stuhr L. Oxygen-dependent regulation of tumor growth and metastasis in human breast cancer xenografts. PLoS One 2017; 12:e0183254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lee HY, Kim IK, Lee HI, Lee HY, Kang HS, Yeo CD, Kang HH, Moon HS, Lee SH. Combination of carboplatin and intermittent normobaric hyperoxia synergistically suppresses benzo[a]pyrene-induced lung cancer. Korean J Intern Med 2018; 33:541–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kim SW, Kim IK, Ha JH, Yeo CD, Kang HH, Kim JW, Lee SH. Normobaric hyperoxia inhibits the progression of lung cancer by inducing apoptosis. Exp Biol Med 2018; 243:739–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.FDA. Hyperbaric oxygen therapy: don’t be misled, www.fda.gov/consumers/consumer-updates/hyperbaric-oxygen-therapy-dont-be-misled(2013, accessed 10 April 2020)
  • 25.Sherman LF, Bacon HE. An evaluation of oxygen therapy as an adjunct in the postoperative management of carcinoma of the rectum and sigmoid Colon. Am J Surg 1951; 81:105–10 [DOI] [PubMed] [Google Scholar]
  • 26.Moen I, Tronstad KJ, Kolmannskog O, Salvesen GS, Reed RK, Stuhr LE. Hyperoxia increases the uptake of 5-fluorouracil in mammary tumors independently of changes in interstitial fluid pressure and tumor stroma. BMC Cancer 2009; 9:446–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kawasoe Y, Yokouchi M, Ueno Y, Iwaya H, Yoshida H, Komiya S. Hyperbaric oxygen as a chemotherapy adjuvant in the treatment of osteosarcoma. Oncol Rep 2009; 22:1045–50 [DOI] [PubMed] [Google Scholar]
  • 28.Selvendiran K, Kuppusamy ML, Ahmed S, Bratasz A, Meenakshisundaram G, Rivera BK, Khan M, Kuppusamy P. Oxygenation inhibits ovarian tumor growth by downregulating STAT3 and cyclin-D1 expressions. Cancer Biol Ther 2010; 10:386–90 [DOI] [PubMed] [Google Scholar]
  • 29.Sun S, Lee D, Lee NP, Pu JK, Wong ST, Lui WM, Fung CF, Leung GK. Hyperoxia resensitizes chemoresistant human glioblastoma cells to temozolomide. J Neurooncol 2012; 109:467–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lee D, Sun S, Ho AS, Kiang KM, Zhang XQ, Xu FF, Leung GK. Hyperoxia resensitizes chemoresistant glioblastoma cells to temozolomide through unfolded protein response. Anticancer Res 2014; 34:2957–66 [PubMed] [Google Scholar]
  • 31.Hatfield SM, Kjaergaard J, Lukashev D, Schreiber TH, Belikoff B, Abbott R, Sethumadhavan S, Philbrook P, Ko K, Cannici R, Thayer M, Rodig S, Kutok JL, Jackson EK, Karger B, Podack ER, Ohta A, Sitkovsky MV. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med 2015; 7:1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Benito J, Ramirez MS, Millward NZ, Velez J, Harutyunyan KG, Lu H, Shi YX, Matre P, Jacamo R, Ma H, Konoplev S, McQueen T, Volgin A, Protopopova M, Mu H, Lee J, Bhattacharya PK, Marszalek JR, Davis RE, Bankson JA, Cortes JE, Hart CP, Andreeff M, Konopleva M. Hypoxia-Activated prodrug TH-302 targets hypoxic bone marrow niches in preclinical leukemia models. Clin Cancer Res 2016; 22:1687–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Luongo M, Brigida AL, Mascolo L, Gaudino G. Possible therapeutic effects of ozone mixture on hypoxia in tumor development. Anticancer Res 2017; 37:425–35 [DOI] [PubMed] [Google Scholar]
  • 34.Stepien K, Ostrowski RP, Matyja E. Hyperbaric oxygen as an adjunctive therapy in treatment of malignancies, including brain tumours. Med Oncol 2016; 33:101–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tibbles PM, Edelsberg JS. Hyperbaric-oxygen therapy. N Engl J Med 1996; 334:1642–8 [DOI] [PubMed] [Google Scholar]
  • 36.Weaver LK, Howe S. Normobaric measurement of arterial oxygen tension in subjects exposed to hyperbaric oxygen. Chest 1992; 102:1175–81 [DOI] [PubMed] [Google Scholar]
  • 37.Beppu T, Kamada K, Yoshida Y, Arai H, Ogasawara K, Ogawa A. Change of oxygen pressure in glioblastoma tissue under various conditions. J Neurooncol 2002; 58:47–52 [DOI] [PubMed] [Google Scholar]
  • 38.Heyboer M, 3rd, Sharma D, Santiago W, McCulloch N. Hyperbaric oxygen therapy: side effects defined and quantified. Adv Wound Care 2017; 6:210–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Raa A, Stansberg C, Steen VM, Bjerkvig R, Reed RK, Stuhr LE. Hyperoxia retards growth and induces apoptosis and loss of glands and blood vessels in DMBA-induced rat mammary tumors. BMC Cancer 2007; 7:23–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Braks JA, Spiegelberg L, Koljenovic S, Ridwan Y, Keereweer S, Kanaar R, Wolvius EB, Essers J. Optical imaging of tumor response to hyperbaric oxygen treatment and irradiation in an orthotopic mouse model of head and neck squamous cell carcinoma. Mol Imaging Biol 2015; 17:633–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rockswold SB, Rockswold GL, Zaun DA, Zhang X, Cerra CE, Bergman TA, Liu J. A prospective, randomized clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe traumatic brain injury. J Neurosurg 2010; 112:1080–94 [DOI] [PubMed] [Google Scholar]
  • 42.Ivanov KP, Sokolova IB, Vovenko EP. Oxygen transport in the rat brain cortex at normobaric hyperoxia. Eur J Appl Physiol Occup Physiol 1999; 80:582–7 [DOI] [PubMed] [Google Scholar]
  • 43.Braun RD, Lanzen JL, Snyder SA, Dewhirst MW. Comparison of tumor and normal tissue oxygen tension measurements using OxyLite or microelectrodes in rodents. Am J Physiol Heart Circ Physiol 2001; 280:H2533–44 [DOI] [PubMed] [Google Scholar]
  • 44.Stuhr LE, Raa A, Oyan AM, Kalland KH, Sakariassen PO, Petersen K, Bjerkvig R, Reed RK. Hyperoxia Retards growth and induces apoptosis, changes in vascular density and gene expression in transplanted gliomas in nude rats. J Neurooncol 2007; 85:191–202 [DOI] [PubMed] [Google Scholar]
  • 45.Le QT, Courter D. Clinical biomarkers for hypoxia targeting. Cancer Metastasis Rev 2008; 27:351–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Masoud GN, Li W. HIF-1alpha pathway: role, regulation and intervention for cancer therapy. Acta Pharm Sin B 2015; 5:378–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 1992; 12:5447–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lopez-Barneo J, Simon MC. Cellular adaptation to oxygen deficiency beyond the Nobel award. Nat Commun 2020; 11:607–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell 2010; 40:294–309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dengler VL, Galbraith M, Espinosa JM. Transcriptional regulation by hypoxia inducible factors. Crit Rev Biochem Mol Biol 2014; 49:1–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Koyasu S, Kobayashi M, Goto Y, Hiraoka M, Harada H. Regulatory mechanisms of hypoxia-inducible factor 1 activity: two decades of knowledge. Cancer Sci 2018; 109:560–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Harada H. Hypoxia-inducible factor 1-mediated characteristic features of cancer cells for tumor radioresistance. J Radiat Res 2016; 57:i99–i105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Xia Y, Jiang L, Zhong T. The role of HIF-1alpha in chemo-/radioresistant tumors. Onco Targets Ther 2018; 11:3003–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sethumadhavan S, Silva M, Philbrook P, Nguyen T, Hatfield SM, Ohta A, Sitkovsky MV. Hypoxia and hypoxia-inducible factor (HIF) downregulate antigen-presenting MHC class I molecules limiting tumor cell recognition by T cells. PLoS One 2017; 12:e0187314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lv Y, Zhao S, Han J, Zheng L, Yang Z, Zhao L. Hypoxia-inducible factor-1alpha induces multidrug resistance protein in colon cancer. Onco Targets Ther 2015; 8:1941–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hussein AA, Forouzanfar T, Bloemena E, de Visscher J, Brakenhoff RH, Leemans CR, Helder MN. A review of the most promising biomarkers for early diagnosis and prognosis prediction of tongue squamous cell carcinoma. Br J Cancer 2018; 119:724–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fallah J, Rini BI. HIF inhibitors: status of current clinical development. Curr Oncol Rep 2019; 21:6–15 [DOI] [PubMed] [Google Scholar]
  • 58.Hatfield SM, Kjaergaard J, Lukashev D, Belikoff B, Schreiber TH, Sethumadhavan S, Abbott R, Philbrook P, Thayer M, Shujia D, Rodig S, Kutok JL, Ren J, Ohta A, Podack ER, Karger B, Jackson EK, Sitkovsky M. Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1alpha-dependent and extracellular adenosine-mediated tumor protection. J Mol Med 2014; 92:1283–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Y, Yin K, Tian J, Xia X, Ma J, Tang X, Xu H, Wang S. Granulocytic Myeloid-Derived suppressor cells promote the stemness of colorectal cancer cells through exosomal S100A9. Adv Sci 2019; 6:1901278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Das U. A radical approach to cancer. Med Sci Monit 2002; 8:Ra79–92 [PubMed] [Google Scholar]
  • 61.Allen RG, Balin AK. Effects of oxygen on the antioxidant responses of normal and transformed cells. Exp Cell Res 2003; 289:307–16 [DOI] [PubMed] [Google Scholar]
  • 62.Folkman J. Seminars in medicine of the Beth Israel hospital, boston. Clinical applications of research on angiogenesis. N Engl J Med 1995; 333:1757–63 [DOI] [PubMed] [Google Scholar]
  • 63.Chen S, Sang N. Hypoxia-inducible factor-1: a critical player in the survival strategy of stressed cells. J Cell Biochem 2016; 117:267–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA 1995; 92:5510–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat Rev Cancer 2004; 4:891–9 [DOI] [PubMed] [Google Scholar]
  • 66.Mahon PC, Hirota K, Semenza GL. FIH-1: a novel protein that interacts with HIF-1alpha and VHL to mediate repression of HIF-1 transcriptional activity. Genes Dev 2001; 15:2675–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Terraneo L, Virgili E, Caretti A, Bianciardi P, Samaja M. In vivo hyperoxia induces hypoxia-inducible factor-1alpha overexpression in LNCaP tumors without affecting the tumor growth rate. Int J Biochem Cell Biol 2014; 51:65–74 [DOI] [PubMed] [Google Scholar]
  • 68.Rocco M, D’Itri L, D, Bels D, Corazza F, Balestra C. The “normobaric oxygen paradox”: a new tool for the anesthetist? Minerva Anestesiol 2014; 80:366–72 [PubMed] [Google Scholar]
  • 69.Chowdhury AK, Watkins T, Parinandi NL, Saatian B, Kleinberg ME, Usatyuk PV, Natarajan V. Src-mediated tyrosine phosphorylation of p47phox in hyperoxia-induced activation of NADPH oxidase and generation of reactive oxygen species in lung endothelial cells. J Biol Chem 2005; 280:20700–11 [DOI] [PubMed] [Google Scholar]
  • 70.Kim JY, Lee JY. Targeting tumor adaption to chronic hypoxia: implications for drug resistance, and how it can be overcome. Int J Mol Sci 2017; 18:1–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Clark JM, Lambertsen CJ. Pulmonary oxygen toxicity: a review. Pharmacol Rev 1971; 23:37–133 [PubMed] [Google Scholar]
  • 72.Kallet RH, Matthay MA. Hyperoxic acute lung injury. Respir Care 2013; 58:123–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Clark JM, Lambertsen CJ, Gelfand R, Flores ND, Pisarello JB, Rossman MD, Elias JA. Effects of prolonged oxygen exposure at 1.5, 2.0, or 2.5 ATA on pulmonary function in men (predictive studies V). J Appl Physiol 1999; 86:243–59 [DOI] [PubMed] [Google Scholar]
  • 74.Ciarlone GE, Hinojo CM, Stavitzski NM, Dean JB. CNS function and dysfunction during exposure to hyperbaric oxygen in operational and clinical settings. Redox Biol 2019; 27:101159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.McMonnies CW. Hyperbaric oxygen therapy and the possibility of ocular complications or contraindications. Clin Exp Optom 2015; 98:122–5 [DOI] [PubMed] [Google Scholar]
  • 76.He CH, Waxman AB, Lee CG, Link H, Rabach ME, Ma B, Chen Q, Zhu Z, Zhong M, Nakayama K, Nakayama KI, Homer R, Elias JA. Bcl-2-related protein A1 is an endogenous and cytokine-stimulated mediator of cytoprotection in hyperoxic acute lung injury. J Clin Invest 2005; 115:1039–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zhou Q, Huang G, Yu X, Xu W. A novel approach to estimate ROS origination by hyperbaric oxygen exposure, targeted probes and specific inhibitors. Cell Physiol Biochem 2018; 47:1800–8 [DOI] [PubMed] [Google Scholar]
  • 78.Körpınar Ş, Uzun H. The effects of hyperbaric oxygen at different pressures on oxidative stress and antioxidant status in rats. Medicina 2019; 55:205–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sanders LH, Timothy Greenamyre J. Oxidative damage to macromolecules in human parkinson disease and the rotenone model. Free Radic Biol Med 2013; 62:111–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Stadtman ER, Levine RL. Protein oxidation. Ann N Y Acad Sci 2000; 899:191–208 [DOI] [PubMed] [Google Scholar]
  • 81.Fruhwirth GO, Hermetter A. Mediation of apoptosis by oxidized phospholipids. Subcell Biochem 2008; 49:351–67 [DOI] [PubMed] [Google Scholar]
  • 82.Auten RL, Whorton MH, Nicholas Mason S. Blocking neutrophil influx reduces DNA damage in hyperoxia-exposed newborn rat lung. Am J Respir Cell Mol Biol 2002; 26:391–7 [DOI] [PubMed] [Google Scholar]
  • 83.Barazzone C, White CW. Mechanisms of cell injury and death in hyperoxia: role of cytokines and bcl-2 family proteins. Am J Respir Cell Mol Biol 2000; 22:517–9 [DOI] [PubMed] [Google Scholar]
  • 84.Panayiotidis MI, Rancourt RC, Allen CB, Riddle SR, Schneider BK, Ahmad S, White CW. Hyperoxia-induced DNA damage causes decreased DNA methylation in human lung epithelial-like A549 cells. Antioxid Redox Signal 2004; 6:129–36 [DOI] [PubMed] [Google Scholar]
  • 85.Gore A, Muralidhar M, Espey MG, Degenhardt K, Mantell LL. Hyperoxia sensing: from molecular mechanisms to significance in disease. J Immunotoxicol 2010; 7:239–54 [DOI] [PubMed] [Google Scholar]
  • 86.Budinger GR, Tso M, McClintock DS, Dean DA, Sznajder JI, Chandel NS. Hyperoxia-induced apoptosis does not require mitochondrial reactive oxygen species and is regulated by bcl-2 proteins. J Biol Chem 2002; 277:15654–60 [DOI] [PubMed] [Google Scholar]
  • 87.Sue RD, Belperio JA, Burdick MD, Murray LA, Xue YY, Dy MC, Kwon JJ, Keane MP, Strieter RM. CXCR2 is critical to hyperoxia-induced lung injury. J Immunol 2004; 172:3860–8 [DOI] [PubMed] [Google Scholar]
  • 88.Murray LA, Knight DA, McAlonan L, Argentieri R, Joshi A, Shaheen F, Cunningham M, Alexopolou L, Flavell RA, Sarisky RT, Hogaboam CM. Deleterious role of TLR3 during hyperoxia-induced acute lung injury. Am J Respir Crit Care Med 2008; 178:1227–37 [DOI] [PubMed] [Google Scholar]
  • 89.Plafki C, Peters P, Almeling M, Welslau W, Busch R. Complications and side effects of hyperbaric oxygen therapy. Aviat Space Environ Med 2000; 71:119–24 [PubMed] [Google Scholar]
  • 90.Al-Waili NS, Butler GJ, Beale J, Abdullah MS, Finkelstein M, Merrow M, Rivera R, Petrillo R, Carrey Z, Lee B, Allen M. Influences of hyperbaric oxygen on blood pressure, heart rate and blood glucose levels in patients with diabetes mellitus and hypertension. Arch Med Res 2006; 37:991–7 [DOI] [PubMed] [Google Scholar]
  • 91.Adamson IY, Young L, Orr FW. Tumor metastasis after hyperoxic injury and repair of the pulmonary endothelium. Lab Invest 1987; 57:71–7 [PubMed] [Google Scholar]
  • 92.Ma Z, Moruzzi N, Catrina SB, Grill V, Bjorklund A. Hyperoxia inhibits glucose-induced insulin secretion and mitochondrial metabolism in rat pancreatic islets. Biochem Biophys Res Commun 2014; 443:223–8 [DOI] [PubMed] [Google Scholar]
  • 93.Karakashev SV, Reginato MJ. Progress toward overcoming hypoxia-induced resistance to solid tumor therapy. Cancer Manag Res 2015; 7:253–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Erler JT, Cawthorne CJ, Williams KJ, Koritzinsky M, Wouters BG, Wilson C, Miller C, Demonacos C, Stratford IJ, Dive C. Hypoxia-mediated down-regulation of bid and bax in tumors occurs via hypoxia-inducible factor 1-dependent and -independent mechanisms and contributes to drug resistance. Mol Cell Biol 2004; 24:2875–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ding Z, Yang L, Xie X, Xie F, Pan F, Li J, He J, Liang H. Expression and significance of hypoxia-inducible factor-1 alpha and MDR1/P-glycoprotein in human Colon carcinoma tissue and cells. J Cancer Res Clin Oncol 2010; 136:1697–707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Suzuki Y, Tanaka K, Negishi D, Shimizu M, Yoshida Y, Hashimoto T, Yamazaki H. Pharmacokinetic investigation of increased efficacy against malignant gliomas of carboplatin combined with hyperbaric oxygenation. Neurol Med Chir 2009; 49:193–7; discussion 7 [DOI] [PubMed] [Google Scholar]
  • 97.Stuhr LE, Iversen VV, Straume O, Maehle BO, Reed RK. Hyperbaric oxygen alone or combined with 5-FU attenuates growth of DMBA-induced rat mammary tumors. Cancer Lett 2004; 210:35–40 [DOI] [PubMed] [Google Scholar]
  • 98.Kindwall EP.Contraindications and side effects to hyperbaric oxygen treatment. 2nd ed Flagstaff, AZ: Best Publishing Company, 2002, pp. 83–99 [Google Scholar]
  • 99.Upton PG, Yamaguchi KT, Myers S, Kidwell TP, Anderson RJ. Effects of antioxidants and hyperbaric oxygen in ameliorating experimental doxorubicin skin toxicity in the rat. Cancer Treat Rep 1986; 70:503–7 [PubMed] [Google Scholar]
  • 100.Goldiner PL, Carlon GC, Cvitkovic E, Schweizer O, Howland WS. Factors influencing postoperative morbidity and mortality in patients treated with bleomycin. Br Med J 1978; 1:1664–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Forman HJ, York JL, Fisher AB. Mechanism for the potentiation of oxygen toxicity by disulfiram. J Pharmacol Exp Ther 1980; 212:452–5 [PubMed] [Google Scholar]
  • 102.Karagoz B, Suleymanoglu S, Uzun G, Bilgi O, Aydinoz S, Haholu A, Turken O, Onem Y, Kandemir EG. Hyperbaric oxygen therapy does not potentiate doxorubicin-induced cardiotoxicity in rats. Basic Clin Pharmacol Toxicol 2008; 102:287–92 [DOI] [PubMed] [Google Scholar]
  • 103.Torp KD, Carraway MS, Ott MC, Stolp BW, Moon RE, Piantadosi CA, Freiberger JJ. Safe administration of hyperbaric oxygen after bleomycin: a case series of 15 patients. Undersea Hyperb Med 2012; 39:873–9 [PubMed] [Google Scholar]
  • 104.Sia J, Szmyd R, Hau E, Gee HE. Molecular mechanisms of Radiation-Induced cancer cell death: a primer. Front Cell Dev Biol 2020; 8:41–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ward JF. DNA damage produced by ionizing radiation in mammalian cells: identities, mechanisms of formation, and reparability. Prog Nucleic Acid Res Mol Biol 1988; 35:95–125 [DOI] [PubMed] [Google Scholar]
  • 106.Ruan K, Song G, Ouyang G. Role of hypoxia in the hallmarks of human cancer. J Cell Biochem 2009; 107:1053–62 [DOI] [PubMed] [Google Scholar]
  • 107.Hsia TC, Yang JS, Chen GW, Chiu TH, Lu HF, Yang MD, Yu FS, Liu KC, Lai KC, Lin CC, Chung JG. The roles of endoplasmic reticulum stress and Ca2+ on rhein-induced apoptosis in A-549 human lung cancer cells. Anticancer Res 2009; 29:309–18 [PubMed] [Google Scholar]
  • 108.Gray LH, Conger AD, Ebert M, Hornsey S, Scott OC. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol 1953; 26:638–48 [DOI] [PubMed] [Google Scholar]
  • 109.Watson ER, Halnan KE, Dische S, Saunders MI, Cade IS, McEwen JB, Wiernik G, Perrins DJ, Sutherland I. Hyperbaric oxygen and radiotherapy: a medical research council trial in carcinoma of the cervix. Br J Radiol 1978; 51:879–87 [DOI] [PubMed] [Google Scholar]
  • 110.Bennett MH, Feldmeier J, Smee R, Milross C. Hyperbaric oxygenation for tumour sensitisation to radiotherapy. Cochrane Database Syst Rev 2018; 4:1–115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Chen JR, Xu HZ, Ding JB, Qin ZY. Radiotherapy after hyperbaric oxygenation in malignant gliomas. Curr Med Res Opin 2015; 31:1977–84 [DOI] [PubMed] [Google Scholar]
  • 112.Rojas A, Carl U, Reghebi K. Effect of normobaric oxygen on tumor radiosensitivity: fractionated studies. Int J Radiat Oncol Biol Phys 1990; 18:547–53 [DOI] [PubMed] [Google Scholar]
  • 113.Overgaard J. Hypoxic radiosensitization: adored and ignored. J Clin Oncol 2007; 25:4066–74 [DOI] [PubMed] [Google Scholar]
  • 114.Ohta A. Oxygen-dependent regulation of immune checkpoint mechanisms. Int Immunol 2018; 30:335–43 [DOI] [PubMed] [Google Scholar]
  • 115.Lequeux A, Noman MZ, Xiao M, Sauvage D, Van Moer K, Viry E, Bocci I, Hasmim M, Bosseler M, Berchem G, Janji B. Impact of hypoxic tumor microenvironment and tumor cell plasticity on the expression of immune checkpoints. Cancer Lett 2019; 458:13–20 [DOI] [PubMed] [Google Scholar]
  • 116.Multhoff G, Vaupel P. Hypoxia compromises anti-Cancer immune responses. Adv Exp Med Biol 2020; 1232:131–43 [DOI] [PubMed] [Google Scholar]
  • 117.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]
  • 118.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]
  • 119.Hasmim M, Noman MZ, Messai Y, Bordereaux D, Gros G, Baud V, Chouaib S. Cutting edge: hypoxia-induced nanog favors the intratumoral infiltration of regulatory T cells and macrophages via direct regulation of TGF-beta1. J Immunol 2013; 191:5802–6 [DOI] [PubMed] [Google Scholar]
  • 120.Kunzli BM, Bernlochner MI, Rath S, Kaser S, Csizmadia E, Enjyoji K, Cowan P, d’Apice A, Dwyer K, Rosenberg R, Perren A, Friess H, Maurer CA, Robson SC. Impact of CD39 and purinergic signalling on the growth and metastasis of colorectal cancer. Purinergic Signal 2011; 7:231–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Stagg J, Divisekera U, Duret H, Sparwasser T, Teng MW, Darcy PK, Smyth MJ. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res 2011; 71:2892–900 [DOI] [PubMed] [Google Scholar]
  • 122.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-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 2014; 211:781–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Allard B, Pommey S, Smyth MJ, Stagg J. Targeting CD73 enhances the antitumor activity of anti-PD-1 and anti-CTLA-4 mAbs. Clin Cancer Res 2013; 19:5626–35 [DOI] [PubMed] [Google Scholar]
  • 124.Ohta A, Kini R, Ohta A, Subramanian M, Madasu M, Sitkovsky M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol 2012; 3:190–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, Cho HI, Celis E, Quiceno DG, Padhya T, McCaffrey TV, McCaffrey JC, Gabrilovich DI. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med 2010; 207:2439–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ryzhov S, Novitskiy SV, Goldstein AE, Biktasova A, Blackburn MR, Biaggioni I, Dikov MM, Feoktistov I. Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b+Gr1+ cells. J Immunol 2011; 187:6120–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chiu DK, Tse AP, Xu IM, Di Cui J, Lai RK, Li LL, Koh HY, Tsang FH, Wei LL, Wong CM, Ng IO, Wong CC. Hypoxia inducible factor HIF-1 promotes myeloid-derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nat Commun 2017; 8:517–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Sanina N, Chopenko N, Mazeika A, Davydova L, Leonova G, Stenkova A, Uversky VN, Kostetsky E. Immunogenicity and protective activity of a chimeric protein based on the domain III of the Tick-Borne encephalitis virus E protein and the OmpF porin of Yersinia pseudotuberculosis incorporated into the TI-complex. Int J Mol Sci 2018; 19:2988–3001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Baran N, Konopleva M. Molecular pathways: hypoxia-activated prodrugs in cancer therapy. Clin Cancer Res 2017; 23:2382–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.AiErken N, Shi HJ, Zhou Y, Shao N, Zhang J, Shi Y, Yuan ZY, Lin Y. High PD-L1 expression is closely associated with tumor-infiltrating lymphocytes and leads to good clinical outcomes in chinese triple negative breast cancer patients. Int J Biol Sci 2017; 13:1172–9 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES