Next Article in Journal
Chronic Alcoholism and HHV-6 Infection Synergistically Promote Neuroinflammatory Microglial Phenotypes in the Substantia Nigra of the Adult Human Brain
Next Article in Special Issue
Stationed or Relocating: The Seesawing EMT/MET Determinants from Embryonic Development to Cancer Metastasis
Previous Article in Journal
Macrophages in Atherosclerosis, First or Second Row Players?
Previous Article in Special Issue
Role of Calcium Homeostasis in Modulating EMT in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Hyperoxia Effects in Cancer—From Perioperative Clinical Data to Potential Molecular Mechanisms

by
Anca Irina Ristescu
1,2,†,
Crina Elena Tiron
3,†,
Adrian Tiron
3,* and
Ioana Grigoras
1,2
1
Department of Anaesthesia and Intensive Care, School of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
2
Department of Anaesthesia and Intensive Care, Regional Institute of Oncology, 700483 Iasi, Romania
3
TRANSCEND Research Centre, Regional Institute of Oncology, 700483 Iasi, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2021, 9(9), 1213; https://doi.org/10.3390/biomedicines9091213
Submission received: 22 July 2021 / Revised: 6 September 2021 / Accepted: 10 September 2021 / Published: 13 September 2021
(This article belongs to the Special Issue Epithelial-to-Mesenchymal Transition (EMT) in Cancer)

Abstract

:
Increased inspiratory oxygen concentration is constantly used during the perioperative period of cancer patients to prevent the potential development of hypoxemia and to provide an adequate oxygen transport to the organs, tissues and cells. Although the primary tumours are surgically removed, the effects of perioperative hyperoxia exposure on distal micro-metastases and on circulating cancer cells can potentially play a role in cancer progression or recurrence. In clinical trials, hyperoxia seems to increase the rate of postoperative complications and, by delaying postoperative recovery, it can alter the return to intended oncological treatment. The effects of supplemental oxygen on the long-term mortality of surgical cancer patients offer, at this point, conflicting results. In experimental studies, hyperoxia effects on cancer biology were explored following multiple pathways. In cancer cell cultures and animal models, hyperoxia increases the production of reactive oxygen species (ROS) and increases the oxidative stress. These can be followed by the induction of the expression of Brain-derived neurotrophic factor (BDNF) and other molecules involved in angiogenesis and by the promotion of various degrees of epithelial mesenchymal transition (EMT).

1. Background

The incidence of the main types of solid tumours is continuously rising around the world, despite increasing public awareness on modifiable risk factors and the implementation of preventive strategies, but this is also related to advances in screening programs and diagnostic tools. At the same time, the number of oncologic patients undergoing diagnostic, staging, curative, reconstructive or palliative surgical interventions is following the same demographic. It was estimated that, in 2030, there will be 45 million cancer surgeries and, from 22 million new cancer cases, about 17 million patients will need surgical interventions [1].
Despite the development of a more targeted and personalised multimodal treatment, cancer mortality remains high. In the USA, during 2020, a COVID-19 pandemic year, cancer was responsible for almost 600,000 casualties, representing the second leading cause of death after heart disease [2]. The main cause of death is cancer progression, the occurrence of distal metastasis or cancer recurrence, as the vast majority of patients are now successfully treated for primary tumours, with 64% of them having more than 5 years survival [3].
Most cancer patients will need surgery at some time during the cancer care continuum. The perioperative period is currently considered a highly vulnerable time frame in all oncologic patients. Multiple surgical and anaesthetic factors, acting at the systemic level, on the tumoral microenvironment and/or directly on cancer cells, can increase the risk of both postoperative complications and cancer progression [4,5,6]. Onco-anaesthesia, a recently emerged sub-speciality, aims to improve both short-term surgical outcome and also long-term oncological outcome, through the identification of the anaesthetic related factors that can protect from, or can be related to, cancer recurrence and metastasis [7]. While anaesthetic techniques (general versus loco-regional anaesthesia and inhalation versus total intravenous anaesthesia) and drugs (mainly propofol, sevoflurane, lidocaine, opioids) were broadly studied during the last few years [8,9,10,11,12,13], the role of perioperative high inspiratory oxygen concentration exposure is currently not well defined.
In cancer patients receiving general anaesthesia for surgery, increased inspiratory oxygen concentration is commonly used both intraoperatively and, for a variable time period, postoperatively, in order to prevent the potential development of hypoxemia and to ensure an adequate oxygen transport to the organs, tissues and cells. For a long time considered as a useful and harmless intervention, oxygen therapy has been under scrutiny in recent years due to the accumulation of data regarding its deleterious effects in critically ill patients [14] and many areas of acute medicine (stroke, acute myocardial infarction, cardiac arrest) [15,16,17,18,19]. In critically ill patient care, current data does not support a benefit from supranormal oxygen delivery. A meta-analysis of 19 clinical studies showed that hyperoxaemia was associated with increased in-hospital mortality [20]. A large observational multicentric study, collecting almost 300,000 arterial blood gas analysis from more than 14,000 critically ill patients, demonstrated a linear relationship between hyperoxemia and mortality, with both duration and severity adversely affecting the outcome [21]. A recent systematic review of 16,000 critically ill patients suggested that hyperoxia exposure might be associated with increased hospital mortality [22]. The British clinical practice guideline for oxygen therapy in acutely ill medical patients recommends to stop supplemental oxygen therapy when SpO2 reaches 96% and not to start oxygen therapy at or above 93% oxygen saturation in patients with acute stroke or myocardial infarction (strong recommendations) [23].
During the perioperative care of an oncologic patient, the anaesthetist may currently choose between different types of anaesthesia techniques (general anaesthesia versus regional or neuraxial anaesthesia) and can also choose between various anaesthetic and analgesic agents (inhalation versus intravenous anaesthesia). However, oxygen, the most commonly used perioperative drug, cannot be replaced. The optimal dose and duration for oxygen therapy in surgical cancer patients is not clearly established.
The purpose of this narrative review is to summarize and to critically review the currently available clinical and experimental evidence regarding the potential effects of hyperoxia exposure on cancer progression. We further explored the relationship between hyperoxia and reactive oxygen species (ROS) production, the consecutive ROS signalling molecular pathways and the connection with angiogenesis and epithelial mesenchymal transition (EMT) as key biological processes that may foster and sustain cancer growth.

2. Hyperoxia Effects on Surgical Cancer Patients—Clinical and Experimental Data

There is an ongoing debate in the literature regarding the potential benefits or harms associated with the use of increased inspiratory oxygen concentration during the perioperative period. The usefulness claimed by hyperoxia supporters consists of the reduction of surgical site infections (SSI) in intubated surgical patients [24], the antiemetic effects and a theoretical margin of safety in case of intraoperative emergencies [25]. On the other hand, harmful effects of hyperoxia can occur at multiple levels.

2.1. Short-Term Effects of Hyperoxia in Surgical Patients

Hyperoxia exposure in surgical patients may have potentially deleterious respiratory, cardiovascular and cerebral effects and was correlated in clinical and experimental studies with an increased risk of postoperative pulmonary complications and major adverse cardiac events (MACE).

2.1.1. Respiratory Effects

As the lung is the first and the most exposed organ to high inspiratory oxygen concentration, multiple respiratory effects of hyperoxia have been described—Hyperoxic Acute Lung Injury (HALI), characterized by biochemical damage of the tracheobronchial mucosa and alveoli, decreased surfactant production, capillary endothelium alteration and increased production of inflammatory cytokines [26,27]; absorption atelectasis and the inhibition of hypoxic pulmonary vasoconstriction [28].
Hyperoxia exposure effects postoperative pulmonary complications. Multiple clinical and experimental studies have tried to evaluate the consequences of hyperoxia on the postoperative pulmonary function and/or complications. A post-hoc analysis [29] of nearly 5000 surgical patients enrolled in a cohort study [30] showed that perioperative exposure to 80% oxygen does not worsen pulmonary outcome. Additionally, a recent meta-analysis [31] showed that the administration of 80% vs. 30% oxygen was not associated with increased postoperative pulmonary complications. However, this meta-analysis does not include one important retrospective study analysing data of almost 74,000 surgical patients submitted to non-cardiothoracic interventions [32]. The authors of this study found that high intraoperative oxygen concentration was associated, in a dose-dependent manner, with major respiratory complications and with increased 30-day mortality [32].

2.1.2. Cardiovascular Effects

Hyperoxia exposure cardiovascular consequences include systemic vasoconstriction with increased systemic vascular resistance, reduced cerebral and coronary blood flow, reduced cardiac output and pulmonary vasodilation with decrease pulmonary vascular resistance [33,34,35].
The clinical relevance of the cardiovascular effects of perioperative hyperoxia exposure was explored on the postoperative risk of myocardial ischemia. Myocardial injury after noncardiac surgery (MINS) is a frequent postoperative complication, resulting from an imbalance between oxygen myocardial supply and demand. A secondary analysis of data from a large, multicentre, randomized controlled trial, including abdominal surgical patients exposed to either 30% or 80% oxygen intra and postoperatively (PROXI trial), concluded that perioperative hyperoxia might increase the risk of myocardial infarction and cardiovascular diseases [36]. In acute myocardial infarction patients, hyperoxia can increase the production of reactive oxygen species and can aggravate myocardial dysfunction by reducing the myocardial perfusion [37]. Therefore, for the management of acute myocardial infarction patients, current recommendations include tailoring oxygen therapy in order to avoid hyperoxemia [23,38].

2.1.3. Cerebral Effects

The reduction of the cerebral flow and neurotoxicity were described after hyperoxia exposure [39]. The clinical relevance of these effects was not clearly established, as a recent post hoc follow-up study including abdominal surgical patients exposed to 30% vs. 80% oxygen, found no difference in the incidence of stroke or transient cerebral ischemia [40].
Any postoperative complication in patients submitted to major oncologic surgery may have important consequences. On the one hand, adverse postoperative events are associated with decreased overall or decreased recurrence-free survival, as has been demonstrated in recent publications for different types of cancers [41,42]. It has also been shown that the postoperative period is a more important determinant of survival after major surgery than the preoperative risk factors [43]. A possible explanation relating the occurrence of the postoperative complications with cancer progression and outcome could be the amplification of surgical-induced systemic inflammation followed by immune suppression. On the other hand, the development of major complications after cancer surgery can delay or contraindicate subsequent oncological treatments (chemo-, radio-, immune-therapy) [44]. The concept of return to intended oncologic treatment (RIOT) was recently proposed [45], as a novel quality metric for oncological anaesthesia and surgery. Both anaesthetic and surgical strategies, developed in order to reduce potentially avoidable postoperative complications, to enhance recovery and to improve RIOT in oncologic patients with different types of solid tumours, were investigated and proposed [46,47,48].

2.2. Long-Term Effects of Hyperoxia on Surgical Cancer Patients

Data regarding the effects of increased inspiratory oxygen concentration exposure and consecutive hyperoxemia on cancer patient outcome come from the secondary analysis of three clinical trials, primarily investigating hyperoxia exposure effects on the development of surgical site infections.
In the first large, multicentre, randomized controlled trial, Supplemental Oxygen and Complications After Abdominal Surgery, the PROXI trial, including 1400 abdominal surgical patients, the authors found that 80% versus 30% oxygen exposure did not decrease the incidence of surgical site infections (SSI) [49]. In a follow-up analysis of this study [50], the authors reported, in a subgroup of 352 surgical cancer patients, an increased long-term mortality (HR 1.45, 95% CI, 1.10–1.90) with 45% increased hazard ratio over a median of 2.3 years. The authors found that the increased long-term mortality in the 80% oxygen group appeared to be statistically significant in patients undergoing cancer surgery but not in non-cancer patients. Among the possible mechanisms involved, the authors proposed hyperoxia induced angiogenesis that can favour tumoral growth, increased erythropoietin release and DNA damage by reactive oxygen species. The risk of new or recurrent cancer after 80% versus 30% perioperative inspiratory oxygen concentration during abdominal surgery was further assessed in 1377 patients, who completed a median 3.9 year follow-up period [51]. This analysis showed that new cancers occurred at a similar rate, but the cancer-free survival time was significantly shorter in the 80% oxygen group. However, earlier recurrence was insufficient to explain the excess mortality in cancer patients.
The second published study [52] consisted of a long-term mortality analysis of a subset of patients previously included in two trials investigating supplemental oxygen effects on surgical wound infection [53,54]. In 927 patients scheduled for elective colorectal surgery and randomly assigned to receive 30% or 80% oxygen perioperatively (during general anaesthesia and for up to 2 h postoperative), the mortality was no different. Importantly, for the included 451 colorectal cancer patients, the outcome was similar in both 30% and 80% oxygen groups, with a hazard ratio of 0.85 (95% CI, 0.64–1.1) [52].
The third published study is a large, single-centre, multiple crossover cluster trial including 5000 colorectal surgeries performed on 4088 adults patients. The authors tested the effects of 80% versus 30% oxygen concentration on a composite of 30-day deep and organ-space SSIs, healing-related wound complications and mortality, and found that supplemental oxygen does not prevent major infections and healing-related complications after major intestinal surgery [30]. A recent post-hoc analysis of this study, including 3400 operations performed on 2800 patients, investigated patient long-term mortality [55]. After a median of 3 years, the incidence of death was no different in the 80% versus the 30% oxygen group (13% vs. 14%, 95% CI = 0.78–1.13; p = 0.493). The analysis of 995 colorectal cancer patients also showed that 80% inspired oxygen did not influence long-term mortality [55].
Thus, as shown in Table 1, clinical studies testing supplemental oxygen effects on the long-term mortality of surgical cancer patients currently offer conflicting results. These can be related to the heterogeneity of the patients’ demographics, of the surgical interventions (emergency versus elective, major, intermediate or minor surgery) and of the hyperoxia exposure period.

3. Potential Molecular Mechanisms Exploring Hyperoxia Effects on Cancer Progression

Hyperoxia effects on cancer biology were explored following multiple pathways, using both in vitro cancer cell cultures and in vivo tumoral animal models. Experimental data support clinical evidences demonstrating that hyperoxia, mainly if prolonged, can induce lung injury and cerebral damage [56,57,58,59,60,61,62], and this can be counteracted by down-modulation of Akt [63] or by low-dose vitamin D [64] or aspirin [65]. Moreover, hyperoxia diminishes protein synthesis [66], and high levels of reactive oxygen species trigger expression of several microRNAs in cardiac and pulmonary diseases, as recently reviewed [67,68,69].

3.1. ROS Production and Oxidative Stress

Hyperoxia never occurs during natural circumstances. Thus, while normal tissues and cells have extensive adaptive mechanisms to hypoxemia, they have limited protection against hyperoxia. In order to maintain normal cellular functions, a highly regulated balance between oxidant and antioxidant molecular activity emerged during evolution. High concentration oxygen exposure results in increased ROS formation and oxidative stress, overwhelming the antioxidant mechanisms capacity and producing DNA damage, protein damage and lipid peroxidation [70]. ROS are constantly generated and eliminated in biological systems via several regulatory pathways. ROS production is modulated by endogenous and exogenous factors, including oxygen, environmental stressors or ionizing radiation in a dose dependent manner. While intrinsic levels of ROS are involved in the maintenance of cellular homeostasis, high levels of ROS can be toxic to cells. Thus, oxidative stress may lead to cell damage or cell death by apoptosis or necrosis. The severity of cell disturbances depends on the cumulative oxygen dose (concentration and duration of exposure) and on cell susceptibility. At the same time, when a cell experiences high stress levels, it activates autocrine and paracrine mechanisms to protect itself and other cells. In clinical practice, these subtle effects are almost silent and undetected during the postsurgical recovery but may have long-term outcome consequences.
In surgical patients, a recent study confirmed that intraoperative and postoperative hyperoxia exposure (administration of FiO2 = 0.80 in anesthetized patients undergoing abdominal surgery) alters the redox equilibrium at 24 h after surgery, demonstrated by increased lipid peroxidation and decreased antioxidant barrier strength [71].
In many human solid cancers, an important difference in oxygen concentration between the peripheral and the central tumoral cells has been described [72,73]. This variation, mainly explained by the rapid tumoral growth related to the peripheral distribution of blood vessels and abnormal angiogenesis, leads to a hypoxic environment inside the tumoral core. The molecular mechanisms triggered by tumoral hypoxia have been widely investigated. The decreased oxygen availability (intra-tumour hypoxia) will trigger an adaptive response, initially represented by up-regulation and increased levels of the Hypoxia Inducible Factor-1α (HIF-1α) protein. In addition, major genetic and epigenetic alterations already present on the cancer cells can further increase HIF-1α activity [72]. HIF-1α accumulates and induces, at the nuclear level, the transcription of multiple target genes. This process is followed by the synthesis of different proteins involved in angiogenesis, such as Vascular endothelial growth factor A (VEGF-A), in metabolic shift, such as glucose transporters GLUT-1, GLUT-3, in pH adaptation, such as carbonic anhydrase enzymes CA-9, CA-12, and also in migration, invasion, proliferation and metastasis, such as Insulin-like growth factor (IGF-2) and E-Cadherin [73].
Various regimens of normobaric or hyperbaric high oxygen concentrations are used to re-sensitize chemoresistant cancer cells [74,75], as a treatment of various cancer types [76,77,78], as a modulator of immune system anti-cancer responses [79] or in wound healing and neuroprotection [80,81,82].

3.2. Hyperoxia and the Immune System

Hypoxia has the ability to regulate immunosuppressive mechanisms that involve myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAMs), regulatory T-cells (Treg cells) and immune checkpoint pathways, such as the programmed cell death-1 (PD-L1). Using a murine model of triple negative breast cancer, Qian et al. demonstrated that long-time respiratory hyperoxia exposure, 60% O2 concentration administered continuously for 21 days can reverse immunosuppression by regulating MDSCs and PD-L1 expression [79]. Moreover, Hatfield et al. demonstrated, in a murine model of lung cancer, that increased inspiratory oxygen concentrations decreased immunosuppressive molecules, such as transforming growth factor–β (TGF-β), weakened immunosuppression by regulatory T cells and improved lung tumour regression and long-term survival in mice [83]. In a non-cancer model, Kumar et al. demonstrated that neonatal hyperoxia alters the adaptive immune response in adult mice, a mechanism which increase the risk for susceptibility to infection in premature infants [84].
The interaction of hyperoxia with the immune system will require additional investigations regarding the oxygen concentration, exposure length and patient status as there is another piece of evidence that indicates that hyperoxia does not exert immunologic effects in murine and human experimental setups [85]. The exposure of mice for 2.5 h or of healthy volunteers for 3.5 h to 100% oxygen does not affect the inflammatory response induced by administration of endotoxin. The data suggest that short-term hyperoxia has different effects on cancer patients compared with healthy volunteers.

3.3. Angiogenesis and Epithelial Mesenchymal Transition (EMT)

In an in vitro experimental study testing xenon- and sevoflurane-mediated effects on the migration and expression of angiogenesis biomarkers in human breast adenocarcinoma cells, Ash et al. have also documented increased migration of breast cancer cells, when exposed to 65% oxygen as compared to 25% oxygen [86]. Crowley et al. tested the effects of four oxygen concentrations, 21%, 30%, 60% and 80%, O2, on both ER+ and ER− breast cancer cell lines, and found that, short-term, 3 h of 60% oxygen exposure enhances migration and also promotes the secretion of several pro-metastatic angiogenesis factors such as VEGF, IL-8 and angiogenin [87].
Epithelial mesenchymal transition (EMT) is an essential cellular program, normally active during embryogenesis and wound healing. During this biological process, a normal epithelial cell, in contact with the basement membrane, turns into a mesenchymal phenotype and migrates from the original epithelial layer. The onset of EMT in cancer was associated with an increased risk of metastatic disease and cancer progression [88,89]. EMT is accompanied by increasing the expression of the mesenchymal markers (e.g., vimentin) and by decreasing the expression of the epithelial markers, such us E-Cadherin. However, in partial EMT, cells may co-express both mesenchymal and epithelial markers or may lose epithelial markers without gaining mesenchymal markers. Partial EMT plays an important role in metastasis by enhancing tumour cell plasticity [89]. The increased expression of Vimentin, a cytoskeleton protein associated with a migration phenotype, was found to be a poor prognostic marker in cancer [90,91]. The reduced expression of E-Cadherin, a cytoplasmic protein present in the epithelial cells, correlates, in an experimental setting, with an invasive phenotype [92]. Tiron et al. demonstrated that in vitro exposure of ER-breast cancer cell lines to 80% O2 for 6 h increases ROS and induces BDNF, VEGF-R2 and vimentin expression and thus promotes EMT and angiogenesis [93]. Tested in vivo, in a murine model of ER-breast cancer, the perioperative exposure to 80% oxygen for 6 h was associated with an increased number and size of hepatic metastasis, evaluated at 4 weeks after the surgical excision of the tumour [93].
On the other hand, Kim et al. evaluated the effects of hyperoxia exposure on a non-surgical murine model of lung cancer. The authors used 85% oxygen concentration for 24 h, a longer period than used in previous studies, and reported increased ROS activity followed by cellular apoptosis via the MAPK pathway. The exposure has been associated with decreased size and number of lung tumours [94].
The controversial results of these studies, summarized in Table 2, may be explained by the heterogeneity of the experimental designs, including the use of different cancer cell lines (types of tumour, human versus murine cancers), oxygen concentrations and exposure durations.

3.4. Brain-Derived Neurotrophic Factor (BDNF)

Hyperoxia increases ROS and activates cell defence systems in order to keep ROS within physiological range, as previously discussed. BDNF is one of the molecules that are upregulated in response to hyperoxia exposure, as shown in various experimental settings, including the peribronchial smooth muscle of neonatal rats [95], the Alzheimer mouse model [96], the breast cancer mouse model [93] and healthy volunteers [97]. Interestingly, some healthy volunteers enrolled in the study [97] resigned due to the side effects, although the tested oxygen concentration was only 37% O2. BDNF and its main receptor, tropomyosin receptor kinase B (TrkB) have been reported to promote alveolar epithelial regeneration after lung injury [98] or to exert neuroprotective effects [99,100,101,102,103,104]. However, TrkB can be activated by cyclic adenosine monophosphate [105] in order to exert the neuroprotective effects. These data suggest that TrkB plays a pivotal role in response to ROS. However, the activation signals can come either from BDNF or from other agonists, depending on cell type and ROS sub-type. The interaction BDNF-ROS does not consist only in BDNF overexpression in response to increasing levels of ROS, as it has been reported that BDNF exerts various biological effects by the generation of oxidative stress in human vascular endothelial cells [106,107].
BDNF-TrkB axis exerts various effects depending on the cell types. In normal cells, it protects cells from high levels of ROS or induces moderate ROS levels in other circumstances. In cancer cells, in addition to pro-survival signalling, the BDNF-TrkB molecular pathway induces EMT, which is associated with poor prognostic in various cancer types [108,109], may increase migration and invasion [110,111], may promote cancer cell survival and proliferation [112,113] and may increase neo-angiogenesis through increasing vascular endothelial growth factor (VEGF) expression [114,115]. The authors from [116] reported that BDNF-TrkB axis induces VEGF expression via HIF-1α. HIF-1α is generally accepted to have an increased expression in hypoxia and a decreased expression in hyperoxia conditions. Tiron et al. identified an upregulation of HIF-1α protein expression after short hyperoxic exposure of triple negative breast cancer cells in an experimental mouse model of perioperative care [93]. These data join the new sparse data that support HIF-1α increased protein expression after the exposure to hyperoxic episodes [80,117] and suggest that, in certain circumstances (e.g., mild hyperoxia), HIF-1α protein expression is not impaired as in prolonged high grade hyperoxia. In non-cancer patients, HIF-1α may have beneficial effects (e.g., muscle regeneration), but in oncologic patients it may promote cancer progression and recurrence.
Cells that are detaching by the extracellular matrix will undergo a process of cell death called anoikis. There are data [118,119,120,121] demonstrating that BDNF-TrkB axis induce anoikis resistance in various cancer types, in addition to EMT. BDNF synthesized as the result of perioperative oxygen exposure in oncological patients may promote survival of the circulating tumour cells and formation of new metastases; in addition it can decrease the time to relapse by increasing cancer cell proliferation and EMT at the level of existing micrometastases.
Chemoresistance occurs in many types of cancer in clinical setting. In breast cancer, it has been reported that multi-nucleated cells generated by chemotherapy drugs are oxidatively stressed, and this induces chemoresistance, as shown both in vitro and in vivo [122]. Moreover, the multi-nucleated cells induce chemoresistance by secreting VEGF, activating the RAS/MAPK pathway and, thus, ROS—HIF-1α signalling plays a crucial role. It could be possible that oxidatively stressed multi-nucleated cells to increase the expression of BDNF in order to counteract ROS, and BDNF would be responsible for activating the HIF-1α-VEGF pathway, which further induces chemoresistance.
Another source of ROS production in cancer is represented by ionizing radiation. Cancer radiotherapy changes the tumour microenvironment, which in some circumstances induces resistance and recurrence, as already published [123,124]. BDNF does not emerge as a key molecule in radio-resistance; however, the classical downstream signalling pathway of the receptor tyrosine kinase family is involved, as inhibition of PI3K/mTOR induced radio-sensitization in pediatric and adult glioblastoma [125]. However, it has been shown that low-level laser therapy can rescue dendrite atrophy by upregulating BDNF expression via ROS generation [126]. These data suggest that, in certain cancer types, subtypes and radiation clinical models, cancer cells are able to express enough BDNF to protect themselves against the increased ROS levels generated by radiation, the mechanism responsible in many cases for radio-resistance phenomena. On the other hand, it has been demonstrated that total abdominal irradiation causes cognitive deficits in mice models [127]. Abdominal irradiation shifted gut bacterial composition, which increased the expression of miR-34a-5p in small intestine tissue and also in the peripheral blood. Expressed miR-34a-5p targeted the 3′UTR of BDNF mRNA in hippocampus to mediate cognitive dysfunction. This mechanism was further prevented by tail intravenous injection of a miR-34a-5p antagomir. All together, these data may explain why the irradiation of some colon cancers is associated with good prognosis while, in other cases, radio-resistance occurs. Increased BDNF levels may favour radio-resistance by its pro-survival role and also by the induction of the epithelial to mesenchymal transition (EMT) process. On the other hand, a decreased BDNF level induced by irradiation and the shift in the bacterial composition of gut microbiota may sensitize the cancer cells.

3.5. Hyperoxic-Hypoxic Paradox

In clinical circumstances, fluctuations in oxygen levels due to hyperoxic episodes may induce the expression of many mediators usually induced by hypoxia. This phenomenon, known as the “hyperoxic-hypoxic paradox” has been recently reviewed by Hadanny and Efrati [128]. Although the hyperoxic-hypoxic paradox was identified after repeated intermittent hyperoxia, Tiron et al. [93] demonstrates the expression of HIF-1α and VGFR after a single hyperoxia exposure, factors which are normally expressed under hypoxic conditions.
While immunosuppression mediated by tumour hypoxia has been extensively investigated, the data regarding the interaction of perioperative hyperoxia with immune system is lacking. HIF-1α plays an important role in immunosuppression, mediated by chronic hypoxia. We speculate that HIF-1α expressed after acute perioperative hyperoxia may interact with the immune system, probably in a lower extend compared with chronic hypoxia.
Figure 1 describes the link between perioperative hyperoxia exposure and cancer progression in oncologic patients.

4. Conclusions

Hyperoxia exposure, mostly in critically ill patients but also during the perioperative period, was shown to be associated with oxidative stress, organ-specific side effects and increased mortality.
In surgical oncologic patients, increased inspiratory oxygen concentration exposure during the perioperative period can lead to increased production of the reactive oxygen species and oxidative stress, and can induce various grades of partial to full epithelial to mesenchymal transition in cancer cells. Although the primary tumours are surgically removed, the effects of hyperoxia on distal micrometastases and/or on circulating cancer cells can promote cancer progression or recurrence. The molecules that mediate the reactive oxygen species effects on cancer progression may be dependent on cancer cell type. These molecules, such as BDNF, must be upregulated in response to transient increased ROS and, at the same time, be able to induce cell survival, cell proliferation and EMT of cancer cells. EMT generates cancer stem cells, enhances cell migration and invasion and promotes cancer recurrence.
At this moment, the adequate or the safe dose of oxygen therapy in surgical cancer patients, in order to maximize beneficial and minimize harmful effects, is not clearly defined. The available evidence does not support the use of perioperative hyperoxia in major oncological surgery. The oxygen dose during the perioperative period must be tailored in order to avoid both hypoxemia and hyperoxemia.
It is important to produce new experimental and clinical data that may guide practitioners to properly use the most commonly prescribed drug in hospitalized patients.

Funding

This work was funded by “Grigore T Popa” University of Medicine and Pharmacy, Iasi, Romania, Grant number 30335/2017, grant director A.I.R.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data will be freely available by open access.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Sullivan, R.; Alatise, O.I.; Anderson, B.O.; Audisio, R.; Autier, P.; Aggarwal, A.; Balch, C.; Brennan, M.; Dare, A.; D’Cruz, A.; et al. Global cancer surgery: Delivering safe, affordable, and timely cancer surgery. Lancet Oncol. 2015, 16, 1193–1224. [Google Scholar] [CrossRef]
  2. Ahmad, F.B.; Anderson, R.N. The Leading Causes of Death in the US for 2020. JAMA 2021, 325, 1829. [Google Scholar] [CrossRef] [PubMed]
  3. American Cancer Society. Cancer Treatment & Survivorship, Facts & Figures 2019–2021; American Cancer Society: Atlanta, GA, USA, 2019. [Google Scholar]
  4. Horowitz, M.; Neeman, E.; Sharon, E.; Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 2015, 12, 213–226. [Google Scholar] [CrossRef] [Green Version]
  5. Hiller, J.G.; Perry, N.J.; Poulogiannis, G.; Riedel, B.; Sloan, E. Perioperative events influence cancer recurrence risk after surgery. Nat. Rev. Clin. Oncol. 2017, 15, 205–218. [Google Scholar] [CrossRef] [PubMed]
  6. Ristescu, I.; Grigoraș, I.; Dumitras, E.; Dimofte, G. Perioperative immune response alteration. can it influence cancer recurrence? Rev. Med. Chir. Soc. Med. Nat. Iasi 2016, 120, 861–865. [Google Scholar]
  7. Wigmore, T.; Gottumukkala, V.; Riedel, B. Making the Case for the Subspecialty of Onco-Anesthesia. Int. Anesthesiol. Clin. 2016, 54, 19–28. [Google Scholar] [CrossRef] [PubMed]
  8. Wigmore, T.J.; Mohammed, K.; Jhanji, S. Long-term Survival for Patients Undergoing Volatile versus IV Anesthesia for Cancer Surgery. Anesthesiology 2016, 124, 69–79. [Google Scholar] [CrossRef] [PubMed]
  9. Yap, A.; Lopez-Olivo, M.A.; Dubowitz, J.; Hiller, J.; Riedel, B.; Wigmore, T.; Ferguson, M.; Shan, D.; Yee, K.; Meyer, I.; et al. Anesthetic technique and cancer outcomes: A meta-analysis of total intravenous versus volatile anesthesia. Can. J. Anesth. 2019, 66, 546–561. [Google Scholar] [CrossRef] [Green Version]
  10. I Sessler, D.; Pei, L.; Huang, Y.; Fleischmann, E.; Marhofer, P.; Kurz, A.; Mayers, D.B.; A Meyer-Treschan, T.; Grady, M.; Tan, E.Y.; et al. Recurrence of breast cancer after regional or general anaesthesia: A randomised controlled trial. Lancet 2019, 394, 1807–1815. [Google Scholar] [CrossRef]
  11. Wall, T.P.; Crowley, P.D.; Sherwin, A.; Foley, A.G.; Buggy, D.J. Effects of Lidocaine and Src Inhibition on Metastasis in a Murine Model of Breast Cancer Surgery. Cancers 2019, 11, 1414. [Google Scholar] [CrossRef] [Green Version]
  12. Forget, P.; Aguirre, J.A.; Bencic, I.; Borgeat, A.; Cama, A.; Condron, C.; Eintrei, C.; Eroles, P.; Gupta, A.; Hales, T.G.; et al. How Anesthetic, Analgesic and Other Non-Surgical Techniques during Cancer Surgery Might Affect Postoperative Oncologic Outcomes: A Summary of Current State of Evidence. Cancers 2019, 11, 592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Tiron, C.; Patrașcanu, E.; Postu, P.; Trandafir, I.V.; Tiron, A.; Grigoras, I. Sevoflurane Modulates AKT Isoforms in Triple Negative Breast Cancer Cells. An Experimental Study. Curr. Issues Mol. Biol. 2021, 43, 264–275. [Google Scholar] [CrossRef] [PubMed]
  14. Damiani, E.; Adrario, E.; Girardis, M.; Romano, R.; Pelaia, P.; Singer, M.; Donati, A. Arterial hyperoxia and mortality in critically ill patients: A systematic review and meta-analysis. Crit. Care 2014, 18, 711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ibanez, B.; James, S.; Agewall, S.; Antunes, M.J.; Bucciarelli-Ducci, C.; Bueno, H.; Caforio, A.L.P.; Crea, F.; Goudevenos, J.A.; Halvorsen, S.; et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation. Eur. Heart J. 2017, 39, 119–177. [Google Scholar] [CrossRef] [Green Version]
  16. Writing Committee Members; O’Gara, P.T.; Kushner, F.G.; Ascheim, D.D.; Casey, D.E.; Chung, M.K.; De Lemos, J.A.; Ettinger, S.M.; Fang, J.C.; Fesmire, F.M.; et al. 2013 ACCF/AHA Guideline for the Management of ST-Elevation Myocardial Infarction. Circulation 2013, 127, e362–e425. [Google Scholar] [CrossRef] [Green Version]
  17. Powers, W.J.; Rabinstein, A.A.; Ackerson, T.; Adeoye, O.M.; Bambakidis, N.C.; Becker, K.; Biller, J.; Brown, M.; Demaerschalk, B.M.; Hoh, B.; et al. 2018 Guidelines for the Early Management of Patients with Acute Ischemic Stroke: A Guideline for Healthcare Professionals from the American Heart Association/American Stroke Association. Stroke 2018, 49, e46–e99. [Google Scholar] [CrossRef] [PubMed]
  18. Kobayashi, A.; Czlonkowska, A.; Ford, G.A.; Fonseca, A.C.; Luijckx, G.J.; Korv, J.; de la Ossa, N.P.; Price, C.; Russell, D.; Tsiskaridze, A.; et al. European Academy of Neurology and European Stroke Organization consensus statement and practical guidance for pre-hospital management of stroke. Eur. J. Neurol. 2017, 25, 425–433. [Google Scholar] [CrossRef] [Green Version]
  19. Elmer, J.; Scutella, M.; Pullalarevu, R.; Wang, B.; Vaghasia, N.; Trzeciak, S.; Rosario-Rivera, B.L.; Guyette, F.; Rittenberger, J.C.; Dezfulian, C.; et al. The association between hyperoxia and patient outcomes after cardiac arrest: Analysis of a high-resolution database. Intensive Care Med. 2014, 41, 49–57. [Google Scholar] [CrossRef] [Green Version]
  20. Helmerhorst, H.J.; Roos-Blom, M.J.; van Westerloo, D.J.; de Jonge, E. Association between arterial hyperoxia and outcome in subsets of critical illness: A systematic review, meta- analysis, and meta-regression of cohort studies. Crit. Care Med. 2015, 43, 1508–1519. [Google Scholar] [CrossRef] [PubMed]
  21. Helmerhorst, H.J.F.; Arts, D.; Schultz, M.J.; Van Der Voort, P.H.J.; Abu-Hanna, A.; De Jonge, E.; Van Westerloo, D.J. Metrics of Arterial Hyperoxia and Associated Outcomes in Critical Care. Crit. Care Med. 2017, 45, 187–195. [Google Scholar] [CrossRef]
  22. Chu, D.; Kim, L.H.-Y.; Young, P.; Zamiri, N.; A Almenawer, S.; Jaeschke, R.; Szczeklik, W.; Schünemann, H.J.; Neary, J.D.; Alhazzani, W. Mortality and morbidity in acutely ill adults treated with liberal versus conservative oxygen therapy (IOTA): A systematic review and meta-analysis. Lancet 2018, 391, 1693–1705. [Google Scholar] [CrossRef]
  23. Siemieniuk, R.A.C.; Chu, D.; Kim, L.H.-Y.; Güell-Rous, M.-R.; Alhazzani, W.; Soccal, P.M.; Karanicolas, P.J.; Farhoumand, P.D.; Siemieniuk, J.L.K.; Satia, I.; et al. Oxygen therapy for acutely ill medical patients: A clinical practice guideline. BMJ 2018, 363, k4169. [Google Scholar] [CrossRef]
  24. de Jonge, S.; Egger, M.; Latif, A.; Loke, Y.K.; Berenholtz, S.; Boermeester, M.; Allegranzi, B.; Solomkin, J. Effectiveness of 80% vs. 30–35% fraction of inspired oxygen in patients undergoing surgery: An updated systematic review and meta-analysis. Br. J. Anaesth. 2019, 122, 325–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Weenink, R.P.; de Jonge, S.W.; van Hulst, R.A.; Wingelaar, T.T.; van Ooij, P.J.A.; Immink, R.V.; Hollmann, M.W. Perioperative hyperoxyphobia: Justified or not? Benefits and harms of hyperoxia during surgery. J. Clin. Med. 2020, 9, 642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kallet, R.H.; Matthay, M.A. Hyperoxic Acute Lung Injury. Respir. Care 2012, 58, 123–141. [Google Scholar] [CrossRef] [Green Version]
  27. Huang, Z.; Zhang, W.; Yang, J.; Sun, F.; Zhou, H. Interleukin-3 plays a vital role in hyperoxic acute lung injury in mice via mediating inflammation. BMC Pulm. Med. 2018, 18, 164. [Google Scholar] [CrossRef]
  28. Hedenstierna, G.; Edmark, L. Effects of anesthesia on the respiratory system. Best Pract. Res. Clin. Anaesthesiol. 2015, 29, 273–284. [Google Scholar] [CrossRef]
  29. Cohen, B.; Ruetzler, K.; Kurz, A.; Leung, S.; Rivas, E.; Ezell, J.; Mao, G.; Sessler, D.I.; Turan, A. Intra-operative high inspired oxygen fraction does not increase the risk of postoperative respiratory complications: Alternating intervention clinical trial. Eur. J. Anaesthesiol. 2019, 36, 1–7. [Google Scholar] [CrossRef] [PubMed]
  30. Kurz, A.; Kopyeva, T.; Suliman, I.; Podolyak, A.; You, J.; Lewis, B.; Vlah, C.; Khatib, R.; Keebler, A.; Reigert, R.; et al. Supplemental oxygen and surgical-site infections: An alternating intervention controlled trial. Br. J. Anaesth. 2018, 120, 117–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Mattishent, K.; Thavarajah, M.; Sinha, A.; Peel, A.; Egger, M.; Solomkin, J.; de Jonge, S.; Latif, A.; Berenholtz, S.; Allegranzi, B.; et al. Safety of 80% vs. 30–35% fraction of inspired oxygen in patients undergoing surgery: A systematic review and meta-analysis. Br. J. Anaesth. 2019, 122, 311–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Staehr-Rye, A.K.; Meyhoff, C.; Scheffenbichler, F.T.; Melo, M.F.V.; Gätke, M.R.; Walsh, J.L.; Ladha, K.S.; Grabitz, S.D.; Nikolov, M.I.; Kurth, T.; et al. High intraoperative inspiratory oxygen fraction and risk of major respiratory complications. Br. J. Anaesth. 2017, 119, 140–149. [Google Scholar] [CrossRef] [Green Version]
  33. Smit, B.; Smulders, Y.M.; Van Der Wouden, J.C.; Straaten, H.M.O.-V.; Man, A.M.E.S.-D. Hemodynamic effects of acute hyperoxia: Systematic review and meta-analysis. Crit. Care 2018, 22, 1–10. [Google Scholar] [CrossRef] [Green Version]
  34. Smit, B.; Smulders, Y.M.; De Waard, M.C.; Straaten, H.M.O.; Girbes, A.R.J.; Eringa, E.; De Man, A.M.E.S. Hyperoxia does not directly affect vascular tone in isolated arteries from mice. PLoS ONE 2017, 12, e0182637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Damiani, E.; Casarotta, E.; Orlando, F.; Carsetti, A.; Scorcella, C.; Domizi, R.; Adrario, E.; Ciucani, S.; Provinciali, M.; Donati, A. Effects of Normoxia, Hyperoxia, and Mild Hypoxia on Macro-Hemodynamics and the Skeletal Muscle Microcirculation in Anesthetised Rats. Front. Med. 2021, 8, 672257. [Google Scholar] [CrossRef] [PubMed]
  36. Fonnes, S.; Gögenur, I.; Søndergaard, E.S.; Siersma, V.D.; Jorgensen, L.N.; Wetterslev, J.; Meyhoff, C.S. Perioperative hyperoxia—Long-term impact on cardiovascular complications after abdominal surgery, a post hoc analysis of the PROXI trial. Int. J. Cardiol. 2016, 215, 238–243. [Google Scholar] [CrossRef]
  37. Peng, Y.-W.; Mohammed, A.; Deatrick, K.B.; Major, T.; Cheng, D.; Charpie, I.; Charpie, J.R. Differential Effects of Normoxic and Hyperoxic Reperfusion on Global Myocardial Ischemia-Reperfusion Injury. Semin. Thorac. Cardiovasc. Surg. 2019, 31, 188–198. [Google Scholar] [CrossRef]
  38. Kim, T.Y.; Kim, D.H.; Kim, S.C.; Kang, C.; Lee, S.H.; Jeong, J.H.; Lee, S.B.; Park, Y.J.; Lim, D. Impact of early hyperoxia on 28-day in-hospital mortality in patients with myocardial injury. PLoS ONE 2018, 13, e0201286. [Google Scholar] [CrossRef] [Green Version]
  39. López, H.V.; Vivas, M.F.; Ruiz, R.N.; Martínez, J.R.; Navaridas, B.G.-V.; Villa, M.G.; Lázaro, C.L.; Rubio, R.J.; Ortiz, A.M.; Lacal, L.A.; et al. Association between post-procedural hyperoxia and poor functional outcome after mechanical thrombectomy for ischemic stroke: An observational study. Ann. Intensive Care 2019, 9, 1–10. [Google Scholar] [CrossRef] [Green Version]
  40. Kongebro, E.K.; Jorgensen, L.N.; Siersma, V.D.; Meyhoff, C.S. Association between perioperative hyperoxia and cerebrovascular complications after laparotomy-A post-hoc follow-up study. Acta Anaesthesiol. Scand. 2018, 63, 164–170. [Google Scholar] [CrossRef] [PubMed]
  41. Gamboa, A.C.; Lee, R.M.; Turgeon, M.K.; Bs, C.V.; Regenbogen, S.E.; Hrebinko, K.A.; Holder-Murray, J.; Wiseman, J.T.; Ejaz, A.; Ba, M.P.F.; et al. Impact of Postoperative Complications on Oncologic Outcomes After Rectal Cancer Surgery: An Analysis of the US Rectal Cancer Consortium. Ann. Surg. Oncol. 2020, 28, 1712–1721. [Google Scholar] [CrossRef]
  42. Aoyama, T.; Murakawa, M.; Katayama, Y.; Yamaoku, K.; Kanazawa, A.; Higuchi, A.; Shiozawa, M.; Morimoto, M.; Yoshikawa, T.; Morinaga, S.; et al. Impact of post-operative complications on survival and recurrence after resection of colorectal liver metastases: Systematic review and meta-analysis. Ann. Surg. 2019, 270, 1018–1027. [Google Scholar]
  43. Khuri, S.F.; Henderson, W.G.; DePalma, R.G.; Mosca, C.; Healey, N.A.; Kumbhani, D.J. Determinants of Long-Term Survival after Major Surgery and the Adverse Effect of Postoperative Complications. Ann. Surg. 2005, 242, 326–343. [Google Scholar] [CrossRef]
  44. Wu, W.; He, J.; Cameron, J.L.; Makary, M.; Soares, K.; Ahuja, N.; Rezaee, N.; Herman, J.; Zheng, L.; Laheru, D.; et al. The impact of postoperative complications on the administration of adjuvant therapy following pancreaticoduodenectomy for adenocarcinoma. Ann. Surg. Oncol. 2014, 21, 2873–2881. [Google Scholar] [CrossRef] [Green Version]
  45. Aloia, T.A.; Zimmitti, G.; Conrad, C.; Gottumukalla, V.; Kopetz, S.; Vauthey, J.-N. Return to intended oncologic treatment (RIOT): A novel metric for evaluating the quality of oncosurgical therapy for malignancy. J. Surg. Oncol. 2014, 110, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Finnerty, D.T.; Buggy, D.J. Return to intended oncologic therapy: A potentially valuable endpoint for perioperative research in cancer patients? Br. J. Anaesth. 2020, 124, 508–510. [Google Scholar] [CrossRef]
  47. Lillemoe, H.A.; Marcus, R.K.; Kim, B.; Narula, N.; Davis, C.; Aloia, T.A. Detours on the Road to Recovery: What Factors Delay Readiness to Return to Intended Oncologic Therapy (RIOT) after Liver Resection for Malignancy? J. Gastrointest. Surg. 2019, 23, 2362–2371. [Google Scholar] [CrossRef]
  48. Ramos, M.F.K.P.; De Castria, T.B.; Pereira, M.A.; Dias, A.R.; Antonacio, F.F.; Zilberstein, B.; Hoff, P.M.G.; Ribeiro, U.; Cecconello, I. Return to Intended Oncologic Treatment (RIOT) in Resected Gastric Cancer Patients. J. Gastrointest. Surg. 2019, 24, 19–27. [Google Scholar] [CrossRef]
  49. Meyhoff, C.S.; Wetterslev, J.; Jorgensen, L.N.; Henneberg, S.W.; Hogdall, C.; Lundvall, L.; Svendsen, P.E.; Mollerup, H.; Lunn, T.H.; Simonsen, I.; et al. Effect of high perioperative oxygen fraction on surgical site infection and pulmonary complications after abdominal surgery: The PROXI randomized clinical trial. JAMA 2009, 302, 1543–1550. [Google Scholar] [CrossRef] [PubMed]
  50. Meyhoff, C.S.; Jorgensen, L.N.; Wetterslev, J.; Christensen, K.B.; Rasmussen, L.S.; PROXI Trial Group. Increased long-term mortality after a high perioperative inspiratory oxygen fraction during abdominal surgery: Follow-up of a randomized clinical trial. Anesth. Analg. 2012, 115, 849–854. [Google Scholar] [CrossRef] [PubMed]
  51. Meyhoff, C.S.; Jorgensen, L.N.; Wetterslev, J.; Siersma, V.D.; Rasmussen, L.S. Risk of new or recurrent cancer after a high perioperative inspiratory oxygen fraction during abdominal surgery. Br. J. Anaesth. 2014, 113 (Suppl. 1), i74–i81. [Google Scholar] [CrossRef] [Green Version]
  52. Podolyak, A.; Sessler, D.I.; Reiterer, C.; Fleischmann, E.; Akça, O.; Mascha, E.J.; Greif, R.; Kurz, A. Perioperative Supplemental Oxygen Does Not Worsen Long-Term Mortality of Colorectal Surgery Patients. Anesth. Analg. 2016, 122, 1907–1911. [Google Scholar] [CrossRef]
  53. Greif, R.; Akça, O.; Horn, E.-P.; Kurz, A.; Sessler, D.I. Supplemental Perioperative Oxygen to Reduce the Incidence of Surgical-Wound Infection. N. Engl. J. Med. 2000, 342, 161–167. [Google Scholar] [CrossRef] [PubMed]
  54. Kurz, A.; Fleischmann, E.; Sessler, D.; Buggy, D.; Apfel, C.; Akça, O.; Erdik, E.; Eredics, K.; Kabon, B.; Herbst, F.; et al. Effects of supplemental oxygen and dexamethasone on surgical site infection: A factorial randomized trial. Br. J. Anaesth. 2015, 115, 434–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jiang, Q.; Kurz, A.; Zhang, X.; Liu, L.; Yang, D.; Sessler, D.I. Supplemental Intraoperative Oxygen and Long-term Mortality: Subanalysis of a Multiple Crossover Cluster Trial. Anesthesiology 2021, 134, 709–721. [Google Scholar] [CrossRef]
  56. Li, L.-F.; Liao, S.-K.; Ko, Y.-S.; Lee, C.-H.; Quinn, D.A. Hyperoxia increases ventilator-induced lung injury via mitogen-activated protein kinases: A prospective, controlled animal experiment. Crit. Care 2007, 11, R25. [Google Scholar] [CrossRef] [Green Version]
  57. Thiel, M.; Chouker, A.; Ohta, A.; Jackson, E.; Caldwell, C.; Smith, P.; Lukashev, D.; Bittmann, I.; Sitkovsky, M.V. Oxygenation Inhibits the Physiological Tissue-Protecting Mechanism and Thereby Exacerbates Acute Inflammatory Lung Injury. PLoS Biol. 2005, 3, e174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Nagato, A.; Bezerra, F.; Lanzetti, M.; Lopes, A.D.A.; Silva, M.A.D.S.; Porto, L.C.; Valença, S.S. Time course of inflammation, oxidative stress and tissue damage induced by hyperoxia in mouse lungs. Int. J. Exp. Pathol. 2012, 93, 269–278. [Google Scholar] [CrossRef]
  59. Potteti, H.R.; Rajasekaran, S.; Rajamohan, S.B.; Tamatam, C.R.; Reddy, N.M.; Reddy, S.P. Sirtuin 1 Promotes Hyperoxia-Induced Lung Epithelial Cell Death Independent of NF-E2–Related Factor 2 Activation. Am. J. Respir. Cell Mol. Biol. 2016, 54, 697–706. [Google Scholar] [CrossRef] [Green Version]
  60. Terraneo, L.; Paroni, R.; Bianciardi, P.; Giallongo, T.; Carelli, S.; Gorio, A.; Samaja, M. Brain adaptation to hypoxia and hyperoxia in mice. Redox Biol. 2016, 11, 12–20. [Google Scholar] [CrossRef] [Green Version]
  61. Kupiec, A.; Adamik, B.; Forkasiewicz-Gardynik, K.; Goździk, W. Intra-operative hyperoxia and the risk of delirium in elderly patients after cardiac surgery. Aging 2020, 12, 7006–7014. [Google Scholar] [CrossRef]
  62. Li, K.C.; Tam, C.W.Y.; Shum, H.-P.; Yan, W.W. Impact of Hyperoxia and Hypocapnia on Neurological Outcomes in Patients with Aneurysmal Subarachnoid Hemorrhage: A Retrospective Study. Crit. Care Res. Pract. 2019, 2019, 7584573. [Google Scholar] [CrossRef]
  63. Li, L.-F.; Yang, C.-T.; Huang, C.-C.; Liu, Y.-Y.; Kao, K.-C.; Lin, H.-C. Low-molecular-weight heparin reduces hyperoxia-augmented ventilator-induced lung injury via serine/threonine kinase-protein kinase B. Respir. Res. 2011, 12, 90. [Google Scholar] [CrossRef]
  64. Chen, C.; Weng, H.; Zhang, X.; Wang, S.; Lu, C.; Jin, H.; Chen, S.; Liu, Y.; Sheng, A.; Sun, Y. Low-Dose Vitamin D Protects Hyperoxia-Induced Bronchopulmonary Dysplasia by Inhibiting Neutrophil Extracellular Traps. Front. Pediatr. 2020, 8, 335. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, C.M.; Tung, Y.T.; Wei, C.H.; Lee, P.Y.; Chen, W. Anti-inflammatory and reactive oxygen species suppression through aspirin pretreatment to treat hyperoxia-induced acute lung injury in NF-κB–luciferase inducible transgenic mice. Antioxidants 2020, 9, 429. [Google Scholar] [CrossRef] [PubMed]
  66. Shenberger, J.S.; Myers, J.L.; Zimmer, S.G.; Powell, R.J.; Barchowsky, A. Hyperoxia alters the expression and phosphorylation of multiple factors regulating translation initiation. Am. J. Physiol. Cell. Mol. Physiol. 2005, 288, L442–L449. [Google Scholar] [CrossRef] [Green Version]
  67. Climent, M.; Viggiani, G.; Chen, Y.-W.; Coulis, G.; Castaldi, A. MicroRNA and ROS Crosstalk in Cardiac and Pulmonary Diseases. Int. J. Mol. Sci. 2020, 21, 4370. [Google Scholar] [CrossRef]
  68. Zhang, X.; Chu, X.; Gong, X.; Zhou, H.; Cai, C. The expression of miR-125b in Nrf2-silenced A549 cells exposed to hyperoxia and its relationship with apoptosis. J. Cell. Mol. Med. 2019, 24, 965–972. [Google Scholar] [CrossRef] [Green Version]
  69. Cifci, S.P.; Tapan, Y.U.; Erkul, B.T.; Savran, Y.; Comert, B. The Impact of Hyperoxia on Outcome of Patients Treated with Noninvasive Respiratory Support. Can. Respir. J. 2020, 2020, 3953280. [Google Scholar] [CrossRef]
  70. Helmerhorst, H.J.F.; Schultz, M.J.; van der Voort, P.H.J.; de Jonge, E.; van Westerloo, D.J. Bench-to-bedside review: The effects of hyperoxia during critical illness. Crit. Care 2015, 19, 284. [Google Scholar] [CrossRef] [Green Version]
  71. Ottolenghi, S.; Rubino, F.M.; Sabbatini, G.; Coppola, S.; Veronese, A.; Chiumello, D.; Paroni, R. Oxidative Stress Markers to Investigate the Effects of Hyperoxia in Anesthesia. Int. J. Mol. Sci. 2019, 20, 5492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Semenza, G.L. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 2009, 29, 625–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Tavare, A.N.; Perry, N.J.; Benzonana, L.L.; Takata, M.; Ma, D. Cancer recurrence after surgery: Direct and indirect effects of anesthetic agents. Int. J. Cancer 2011, 130, 1237–1250. [Google Scholar] [CrossRef]
  74. Sun, S.; Lee, D.; Lee, N.P.; Pu, J.K.S.; Wong, T.S.; Lui, W.M.; Fung, C.F.; Leung, G.K.K. Hyperoxia resensitizes chemoresistant human glioblastoma cells to temozolomide. J. Neuro-Oncol. 2012, 109, 467–475. [Google Scholar] [CrossRef] [Green Version]
  75. Wang, P.; Wan, W.; Xiong, S.; Wang, J.; Zou, D.; Lan, C.; Yu, S.; Liao, B.; Feng, H.; Wu, N. HIF1α regulates glioma chemosensitivity through the transformation between differentiation and dedifferentiation in various oxygen levels. Sci. Rep. 2017, 7, 7965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. De Bels, D.; Tillmans, F.; Corazza, F.; Bizzarri, M.; Germonpre, P.; Radermacher, P.; Orman, K.G.; Balestra, C. Hyperoxia Alters Ultrastructure and Induces Apoptosis in Leukemia Cell Lines. Biomolecules 2020, 10, 282. [Google Scholar] [CrossRef] [Green Version]
  77. Raa, A.; Stansberg, C.; Steen, V.M.; Bjerkvig, R.; Reed, R.K.; Stuhr, L.E.B. Hyperoxia retards growth and induces apoptosis and loss of glands and blood vessels in DMBA-induced rat mammary tumors. BMC Cancer 2007, 7, 23. [Google Scholar] [CrossRef] [Green Version]
  78. Moen, I.; Øyan, A.M.; Kalland, K.-H.; Tronstad, K.J.; Akslen, L.A.; Chekenya, M.; Sakariassen, P.; Reed, R.K.; Stuhr, L.E.B. Hyperoxic Treatment Induces Mesenchymal-to-Epithelial Transition in a Rat Adenocarcinoma Model. PLoS ONE 2009, 4, e6381. [Google Scholar] [CrossRef] [Green Version]
  79. 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, 529–545. [Google Scholar] [PubMed]
  80. Yamamoto, N.; Oyaizu, T.; Enomoto, M.; Horie, M.; Yuasa, M.; Okawa, A.; Yagishita, K. VEGF and bFGF induction by nitric oxide is associated with hyperbaric oxygen-induced angiogenesis and muscle regeneration. Sci. Rep. 2020, 10, 2744. [Google Scholar] [CrossRef]
  81. Huang, X.; Liang, P.; Jiang, B.; Zhang, P.; Yu, W.; Duan, M.; Guo, L.; Cui, X.; Huang, M.; Huang, X. Hyperbaric oxygen potentiates diabetic wound healing by promoting fibroblast cell proliferation and endothelial cell angiogenesis. Life Sci. 2020, 259, 118246. [Google Scholar] [CrossRef]
  82. Chang, H.-C.; Yang, Y.-R.; Wang, R.-Y. Effects of repetitive hyperbaric oxygen therapy on neuroprotection in middle cerebral artery occlusion rats. Brain Res. 2020, 1748, 147097. [Google Scholar] [CrossRef]
  83. Hatfield, S.M.; Kjaergaard, J.; Lukashev, D.; Schreiber, T.; Belikoff, B.; Abbott, R.; Sethumadhavan, S.; Philbrook, P.; Ko, K.; Cannici, R.; et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci. Transl. Med. 2015, 7, 277ra30. [Google Scholar] [CrossRef] [Green Version]
  84. Kumar, V.H.S.; Wang, H.; Nielsen, L. Adaptive immune responses are altered in adult mice following neonatal hyperoxia. Physiol. Rep. 2018, 6, e13577. [Google Scholar] [CrossRef] [Green Version]
  85. Kiers, D.; Gerretsen, J.; Janssen, E.; John, A.; Groeneveld, R.; Van Der Hoeven, J.G.; Scheffer, G.-J.; Pickkers, P.; Kox, M. Short-term hyperoxia does not exert immunologic effects during experimental murine and human endotoxemia. Sci. Rep. 2015, 5, 17441. [Google Scholar] [CrossRef] [Green Version]
  86. Ash, S.A.; Valchev, G.I.; Looney, M.; Ni Mhathuna, A.; Crowley, P.D.; Gallagher, H.C.; Buggy, D.J. Xenon decreases cell migration and secretion of a pro-angiogenesis factor in breast adenocarcinoma cells: Comparison with sevoflurane. Br. J. Anaesth. 2014, 113, i14–i21. [Google Scholar] [CrossRef] [Green Version]
  87. Crowley, P.D.; Stuttgen, V.; O’Carroll, E.; A Ash, S.; Buggy, D.J.; Gallagher, H.C. Exposure to 60% oxygen promotes migration and upregulates angiogenesis factor secretion in breast cancer cells. Med. Gas Res. 2017, 7, 226–235. [Google Scholar] [CrossRef] [Green Version]
  88. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2018, 20, 69–84. [Google Scholar] [CrossRef] [PubMed]
  89. Aiello, N.; Kang, Y. Context-dependent EMT programs in cancer metastasis. J. Exp. Med. 2019, 216, 1016–1026. [Google Scholar] [CrossRef] [Green Version]
  90. Tadokoro, A.; Kanaji, N.; Liu, D.; Yokomise, H.; Haba, R.; Ishii, T.; Takagi, T.; Watanabe, N.; Kita, N.; Kadowaki, N.; et al. Vimentin Regulates Invasiveness and Is a Poor Prognostic Marker in Non-small Cell Lung Cancer. Anticancer Res. 2016, 36, 1545–1551. [Google Scholar]
  91. Du, L.; Li, J.; Lei, L.; He, H.; Chen, E.; Dong, J.; Yang, J. High Vimentin Expression Predicts a Poor Prognosis and Progression in Colorectal Cancer: A Study with Meta-Analysis and TCGA Database. BioMed Res. Int. 2018, 2018, 6387810. [Google Scholar] [CrossRef]
  92. Chaw, S.Y.; Majeed, A.A.; Dalley, A.J.; Chan, A.; Stein, S.; Farah, C.S. Epithelial to mesenchymal transition EMT) biomarkers–E-cadherin, beta-catenin, APC and Vimentin–in oral squamous cell carcinogenesis and transformation. Oral Oncol. 2012, 48, 997–1006. [Google Scholar] [CrossRef]
  93. Tiron, A.; Ristescu, I.; Postu, P.A.; Tiron, C.E.; Zugun-Eloae, F.; Grigoras, I. Long-Term Deleterious Effects of Short-term Hyperoxia on Cancer Progression—Is Brain-Derived Neurotrophic Factor an Important Mediator? An Experimental Study. Cancers 2020, 12, 688. [Google Scholar] [CrossRef] [Green Version]
  94. Kim, S.W.; Kim, I.K.; Ha, J.H.; Yeo, C.D.; Kang, H.H.; Kim, J.W.; Lee, S.H. Normobaric hyperoxia inhibits the progression of lung cancer by inducing apoptosis. Exp. Biol. Med. 2018, 243, 739–748. [Google Scholar] [CrossRef]
  95. Yao, Q.; Haxhiu, M.A.; Zaidi, S.I.; Liu, S.; Jafri, A.; Martin, R.J. Hyperoxia enhances brain-derived neurotrophic factor and tyrosine kinase B receptor expression in peribronchial smooth muscle of neonatal rats. Am. J. Physiol. Cell. Mol. Physiol. 2005, 289, L307–L314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Choi, J.; Kwon, H.-J.; Lee, J.-E.; Lee, Y.; Seoh, J.-Y.; Han, P.-L. Hyperoxygenation revitalizes Alzheimer’s disease pathology through the upregulation of neurotrophic factors. Aging Cell 2019, 18, e12888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kujawski, S.; Słomko, J.; Morten, K.J.; Murovska, M.; Buszko, K.; Newton, J.L.; Zalewski, P. Autonomic and Cognitive Function Response to Normobaric Hyperoxia Exposure in Healthy Subjects. Preliminary Study. Medicina 2020, 56, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Paris, A.J.; Hayer, K.E.; Oved, J.H.; Avgousti, D.C.; Toulmin, S.A.; Zepp, J.A.; Zacharias, W.J.; Katzen, J.B.; Basil, M.C.; Kremp, M.M.; et al. STAT3–BDNF–TrkB signalling promotes alveolar epithelial regeneration after lung injury. Nat. Cell Biol. 2020, 22, 1197–1210. [Google Scholar] [CrossRef]
  99. Lee, B.; Cao, R.; Choi, Y.-S.; Cho, H.-Y.; Rhee, A.D.; Hah, C.K.; Hoyt, K.; Obrietan, K. The CREB/CRE transcriptional pathway: Protection against oxidative stress-mediated neuronal cell death. J. Neurochem. 2009, 108, 1251–1265. [Google Scholar] [CrossRef] [Green Version]
  100. Yoo, J.-M.; Lee, B.D.; Sok, D.-E.; Ma, J.Y.; Kim, M.R. Neuroprotective action of N-acetyl serotonin in oxidative stress-induced apoptosis through the activation of both TrkB/CREB/BDNF pathway and Akt/Nrf2/Antioxidant enzyme in neuronal cells. Redox Biol. 2017, 11, 592–599. [Google Scholar] [CrossRef] [PubMed]
  101. Hacioglu, G.; Senturk, A.; Ince, I.; Alver, A. Assessment of oxidative stress parameters of brain-derived neurotrophic factor heterozygous mice in acute stress model. Iran. J. Basic Med. Sci. 2016, 19, 388–393. [Google Scholar]
  102. Jiang, J.M.; Zhou, C.F.; Gao, S.L.; Tian, Y.; Wang, C.Y.; Wang, L.; Gu, H.F.; Tang, X.Q. BDNF-TrkB pathway mediates neuroprotection of hydrogen sulfide against formaldehyde-induced toxicity to PC12 cells. PLoS ONE 2015, 10, e0119478. [Google Scholar]
  103. Wu, C.L.; Chen, S.D.; Yin, J.H.; Hwang, C.S.; Yang, D.I. Nuclear factor-kappaB-dependent sestrin2 induction mediates the antioxidant effects of BDNF against mitochondrial inhibition in rat cortical neurons. Mol. Neurobiol. 2016, 53, 4126–4142. [Google Scholar] [CrossRef]
  104. Boyadjieva, N.I.; Sarkar, D.K. Cyclic Adenosine Monophosphate and Brain-Derived Neurotrophic Factor Decreased Oxidative Stress and Apoptosis in Developing Hypothalamic Neuronal Cells: Role of Microglia. Alcohol. Clin. Exp. Res. 2013, 37, 1370–1379. [Google Scholar] [CrossRef] [Green Version]
  105. Huang, Y.Z.; McNamara, J.O. Neuroprotective Effects of Reactive Oxygen Species Mediated by BDNF-Independent Activation of TrkB. J. Neurosci. 2012, 32, 15521–15532. [Google Scholar] [CrossRef] [PubMed]
  106. Usui, T.; Naruo, A.; Okada, M.; Hayabe, Y.; Yamawaki, H. Brain-derived neurotrophic factor promotes angiogenic tube formation through generation of oxidative stress in human vascular endothelial cells. Acta Physiol. 2014, 211, 385–394. [Google Scholar] [CrossRef] [PubMed]
  107. Song, X.; Zhou, B.; Zhang, P.; Lei, D.; Wang, Y.; Yao, G.; Hayashi, T.; Xia, M.; Tashiro, S.-I.; Onodera, S.; et al. Protective Effect of Silibinin on Learning and Memory Impairment in LPS-Treated Rats via ROS–BDNF–TrkB Pathway. Neurochem. Res. 2016, 41, 1662–1672. [Google Scholar] [CrossRef]
  108. Jia, S.; Wang, W.; Hu, Z.; Shan, C.; Wang, L.; Wu, B.; Yang, Z.; Yang, X.; Lei, D. BDNF mediated TrkB activation contributes to the EMT progression and the poor prognosis in human salivary adenoid cystic carcinoma. Oral Oncol. 2015, 51, 64–70. [Google Scholar] [CrossRef]
  109. Kim, M.S.; Lee, W.S.; Jeong, J.; Kim, S.J.; Jin, W. Induction of metastatic potential by TrkB via activation of IL6/JAK2/STAT3 and PI3K/AKT signaling in breast cancer. Oncotarget 2015, 6, 40158. [Google Scholar] [CrossRef] [Green Version]
  110. Kawamura, K.; Kawamura, N.; Okamoto, N.; Manabe, M. Suppression of choriocarcinoma invasion and metastasis following blockade of BDNF /TrkB signaling. Cancer Med. 2013, 2, 849–861. [Google Scholar] [CrossRef]
  111. Götz, R.; Sendtner, M. Cooperation of Tyrosine Kinase Receptor TrkB and Epidermal Growth Factor Receptor Signaling Enhances Migration and Dispersal of Lung Tumor Cells. PLoS ONE 2014, 9, e100944. [Google Scholar] [CrossRef]
  112. Tanaka, K.; Okugawa, Y.; Toiyama, Y.; Inoue, Y.; Saigusa, S.; Kawamura, M.; Araki, T.; Uchida, K.; Mohri, Y.; Kusunoki, M. Brain-Derived Neurotrophic Factor (BDNF)-Induced Tropomyosin-Related Kinase B (Trk B) Signaling Is a Potential Therapeutic Target for Peritoneal Carcinomatosis Arising from Colorectal Cancer. PLoS ONE 2014, 9, e96410. [Google Scholar] [CrossRef]
  113. Polakowski, N.; Terol, M.J.; Hoang, K.; Nash, I.; Laverdure, S.; Gazon, H.; Belrose, G.; Mesnard, J.-M.; Césaire, R.; Peloponese, J.-M.; et al. HBZ Stimulates Brain-Derived Neurotrophic Factor/TrkB Autocrine/Paracrine Signaling To Promote Survival of Human T-Cell Leukemia Virus Type 1-Infected T Cells. J. Virol. 2014, 88, 13482–13494. [Google Scholar] [CrossRef] [Green Version]
  114. Lin, C.-Y.; Hung, S.-Y.; Chen, H.-T.; Tsou, H.-K.; Fong, Y.-C.; Wang, S.-W.; Tang, C.-H. Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem. Pharmacol. 2014, 91, 522–533. [Google Scholar] [CrossRef]
  115. Huang, S.-M.; Lin, C.; Lin, H.-Y.; Chiu, C.-M.; Fang, C.-W.; Liao, K.-F.; Chen, D.-R.; Yeh, W.-L. Brain-derived neurotrophic factor regulates cell motility in human colon cancer. Endocr.-Relat. Cancer 2015, 22, 455–464. [Google Scholar] [CrossRef] [Green Version]
  116. Nakamura, K.; Martin, K.C.; Jackson, J.K.; Beppu, K.; Woo, C.-W.; Thiele, C.J. Brain-Derived Neurotrophic Factor Activation of TrkB Induces Vascular Endothelial Growth Factor Expression via Hypoxia-Inducible Factor-1α in Neuroblastoma Cells. Cancer Res. 2006, 66, 4249–4255. [Google Scholar] [CrossRef] [Green Version]
  117. Terraneo, L.; Virgili, E.; Caretti, A.; Bianciardi, P.; Samaja, M. In vivo hyperoxia induces hypoxia-inducible factor-1α overexpression in LNCaP tumors without affecting the tumor growth rate. Int. J. Biochem. Cell Biol. 2014, 51, 65–74. [Google Scholar] [CrossRef] [PubMed]
  118. Smit, M.A.; Geiger, T.R.; Song, J.Y.; Gitelman, I.; Peeper, D.S. A Twist-Snail axis critical for TrkB-induced epithelial-mesenchymal transition-like transformation, anoikis resistance, and metastasis. Mol. Cell Biol. 2009, 29, 3722–3737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Bao, W.; Qiu, H.; Yang, T.; Luo, X.; Zhang, H.; Wan, X. Upregulation of TrkB Promotes Epithelial-Mesenchymal Transition and Anoikis Resistance in Endometrial Carcinoma. PLoS ONE 2013, 8, e70616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Yuan, Y.; Ye, H.; Ren, Q. Proliferative role of BDNF/TrkB signaling is associated with anoikis resistance in cervical cancer. Oncol. Rep. 2018, 40, 621–634. [Google Scholar] [CrossRef] [PubMed]
  121. Li, T.; Yu, Y.; Song, Y.; Li, X.; Lan, D.; Zhang, P.; Xiao, Y.; Xing, Y. Activation of BDNF/TrkB pathway promotes prostate cancer progression via induction of epithelial-mesenchymal transition and anoikis resistance. FASEB J. 2020, 34, 9087–9101. [Google Scholar] [CrossRef] [PubMed]
  122. Parekh, A.; Das, S.; Parida, S.; Das, C.K.; Dutta, D.; Mallick, S.K.; Wu, P.-H.; Kumar, B.N.P.; Bharti, R.; Dey, G.; et al. Multi-nucleated cells use ROS to induce breast cancer chemo-resistance in vitro and in vivo. Oncogene 2018, 37, 4546–4561. [Google Scholar] [CrossRef] [PubMed]
  123. Barker, H.E.; Paget, J.T.E.; Khan, A.; Harrington, K. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
  124. Jarosz-Biej, M.; Smolarczyk, R.; Cichoń, T.; Kułach, N. Tumor microenvironment as a “game changer” in cancer radiotherapy. Int. J. Mol. Sci. 2019, 20, 3212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Agliano, A.; Balarajah, G.; Ciobota, D.M.; Sidhu, J.; Clarke, P.; Jones, C.; Workman, P.; Leach, M.O.; Al-Saffar, N.M. Pediatric and adult glioblastoma radiosensitization induced by PI3K/mTOR inhibition causes early metabolic alterations detected by nuclear magnetic resonance spectroscopy. Oncotarget 2017, 8, 47969–47983. [Google Scholar] [CrossRef] [PubMed]
  126. Meng, C.; He, Z.; Xing, D. Low-level laser therapy rescues dendrite atrophy via upregulating BDNF expression: Implications for Alzheimer’s disease. J. Neurosci. 2013, 33, 13505–13517. [Google Scholar] [CrossRef]
  127. Cui, M.; Xiao, H.; Li, Y.; Dong, J.; Luo, D.; Li, H.; Feng, G.; Wang, H.; Fan, S. Total abdominal irradiation exposure impairs cognitive function involving miR-34a-5p/BDNF axis. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2017, 1863, 2333–2341. [Google Scholar] [CrossRef]
  128. Amir, H.; Shai, E. The Hyperoxic-Hypoxic Paradox. Biomolecules 2020, 10, 958. [Google Scholar] [CrossRef]
Figure 1. Perioperative hyperoxia may induce cancer progression in oncological patients by increasing cancer cell survival and EMT. (RIOT—return to the intended oncological treatment, EMT—epithelial mesenchymal transition).
Figure 1. Perioperative hyperoxia may induce cancer progression in oncological patients by increasing cancer cell survival and EMT. (RIOT—return to the intended oncological treatment, EMT—epithelial mesenchymal transition).
Biomedicines 09 01213 g001
Table 1. Clinical studies exploring hyperoxia exposure effects during the perioperative period in surgical patients.
Table 1. Clinical studies exploring hyperoxia exposure effects during the perioperative period in surgical patients.
Author, Year, ReferenceStudy TypePatients NumberType of PatientsHyperoxia ExposurePrimary OutcomeResults
Cohen, B. et al., 2019 [29]Post-hoc analysis of [30]5056Colorectal surgeryIntraoperative, 39 vs. 80% O2Postoperative pulmonary complicationsNo difference
Mattishent, K. et al., 2019 [31]Meta-analysis3839,
3458
Surgical patientsPerioperative, 30 vs. 80% O2Postoperative atelectasis, pneumoniaNo difference
Staehr-Rye, A.K. et al., 2017 [32]Retrospective registry study73,922Non-cardiotoracic surgery Intraoperative, 30%, 40%, 51%, 58%, 79% O2Major respiratory complication (re-intubation, respiratory failure, pulmonary oedema, pneumonia) Hyperoxia increased risk, in a dose dependent manner
Smit, B. et al., 2018 [33]Meta-analysis408
392
Healthy volunteers, medical and surgical patientsPaO2 = 234–617 mmHgHemodynamic effectsHyperoxia decreased cardiac output, increased systemic vascular resistance
Fonnes, S. et al., 2016 [37]Post-hoc analysis of [49]1386Abdominal surgeryIntraoperative, 2 h postoperative 30 vs. 80% O2Long-term major cardiovascular complicationHyperoxia (80% O2) increased acute coronary syndrome
Meyhoff, C. et al., 2009 [49]RCT1386Abdominal surgeryIntraoperative, 2 h postoperative 30 vs. 80% O2Surgical site infection within 14 daysNo difference
Meyhoff, C. et al., 2012 [50]2.3 years follow-up1386
352
Abdominal surgery
Cancer patients
Intraoperative, 2 h postoperative 30 vs. 80% O2Long-term mortality Hyperoxia increased long term mortality in cancer patients
Meyhoff, C. et al., 2014 [51]3.9 year follow-up1377Abdominal surgery
Cancer patients
Intraoperative, 2 h postoperative 30% vs. 80% O2Risk of new or recurrent cancer at 3.9 years follow-upShorter cancer-free survival time
Podolyak, A et al., 2016 [52]Follow-up [53,54]927 (432 + 495)Colorectal surgeryIntraoperative, 2 h postoperative 30% vs. 80% O2Long-term mortality analysisNo difference
Greif, R. et al., 2000 [53]RCT500Colorectal surgeryIntraoperative, 2 h postoperative 30% vs. 80% O2Surgical site infection within 30 daysDecreased in hyperoxia group
Kurz, A. et al., 2015 [54]RCT585Colorectal surgeryIntraoperative, 2 h postoperative 30% vs. 80% O2Surgical site infection within 30 daysNo difference
Kurz, A. et al., 2018 [30]Alternating intervention controlled trial5749Major intestinal surgeryIntraoperative 30% and 80% O2, alternating at 2-week intervals for 39 months30-day composite of deep tissue or organ-space SSI, healing-related wound complications, mortalityNo difference
Jiang, Q. et al., 2021 [55]Post-hoc analysis of [30], 3 years follow-up2800
995
Colorectal surgery
Cancer patients
Intraoperative 30% and 80% O2, alternating at 2-week intervals for 39 monthsLong term mortalityNo difference
Table 2. Experimental in vitro and in vivo studies exploring hyperoxia exposure effects in cancer.
Table 2. Experimental in vitro and in vivo studies exploring hyperoxia exposure effects in cancer.
Author, Year, ReferencesStudy TypeExperimental DesignHyperoxia ExposurePrimary OutcomeResults
Li, L. et al., 2007 [56]In vivoC57BL/6 mice exposed to high-VT mechanical ventilation21 vs. 95% O2 for 1–5 hVentilator-induced lung injuryHyperoxia increases lung and inflammation ventilator-induced lung injury
Tiron, A. et al., 2020 [93]In vitro,
In vivo
MCF10A, MDA-MB-231, 4T1 breast cancer cells
4T1 TNBC
Murine model
21%, 40%, 60%, 80% O2 for 6 h 21%, 40%, 60%, 80% O2 for 6 h perioperativeEffects on breast cancer growthHyperoxia (80%) increases ROS, BDNF, EMT and angiogenesis molecules Increases size and number of lung metastasis
Crowley et al., 2018 [87]In vitroMDA-MB-231 MCF-7 breast cancer cells 21%, 30%, 60%, 80% O2 for 3 hEffects on breast cancer cell migration and angiogenesis Hyperoxia (60%) promotes migration and upregulates angiogenesis factor secretion
Ash et al., 2014 [86]In vitroMDA-MB-231 and MCF-7 breast cancer cellsXenon 70% + O2 25%, sevoflurane 2.5% + 65% O2 for 5 hEffects on breast cancer cells migration and angiogenesisHyperoxia (65%) promotes breast cancer cell migration
Kim et al., 2018 [94]In vitro
In vivo
A549 lung cancer cells
Murine model of lung cancer
85% O2 for 24 hMorphological changes in lung cancerHyperoxia increased ROS, apoptosis
Decreases size and number of lung tumors
Qian et al., 2018 [79]In vivo4T1 TNBC murine model21%, vs. 65% O2 for 21 daysEffects on tumor microenvironmentHyperoxia reverses immunosuppression by regulating myeloid-derived suppressor cells and PD-L1 expression
Hatfield et al., 2015 [83]In vitro
In vivo
MCA205 tumor cell line
4T1 TNBC Murine model
40%, 60% O2 for 2 daysEffects on tumor microenvironmentHyperoxia increases tumoral infiltration with CD8+ T cells, proinflammatory cytokines, decreased TGF-β and immunosuppression by T-reg cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ristescu, A.I.; Tiron, C.E.; Tiron, A.; Grigoras, I. Exploring Hyperoxia Effects in Cancer—From Perioperative Clinical Data to Potential Molecular Mechanisms. Biomedicines 2021, 9, 1213. https://doi.org/10.3390/biomedicines9091213

AMA Style

Ristescu AI, Tiron CE, Tiron A, Grigoras I. Exploring Hyperoxia Effects in Cancer—From Perioperative Clinical Data to Potential Molecular Mechanisms. Biomedicines. 2021; 9(9):1213. https://doi.org/10.3390/biomedicines9091213

Chicago/Turabian Style

Ristescu, Anca Irina, Crina Elena Tiron, Adrian Tiron, and Ioana Grigoras. 2021. "Exploring Hyperoxia Effects in Cancer—From Perioperative Clinical Data to Potential Molecular Mechanisms" Biomedicines 9, no. 9: 1213. https://doi.org/10.3390/biomedicines9091213

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop