Sie sind auf Seite 1von 20

Ó 2006 by the Société Internationale de Chirurgie World J Surg (2006) 30: 2112–2131

Published Online: 7 November 2006 DOI: 10.1007/s00268-006-0190-6

Hyperbaric Oxygen Therapy for Malignancy:


A Review
Jurstine Daruwalla, B.Sc, Chris Christophi, MD
Department of Surgery, University of Melbourne, Austin Hospital, Level 8 Lance Townsend Building, Austin Health,
Studley Road, Heidelberg, Victoria, 3084 Australia

Abstract
One unique feature of tumors is the presence of hypoxic regions, which occur predominantly at the
tumor center. Hypoxia has a major impact on various aspects of tumor cell function and prolif-
eration. Hypoxic tumor cells are relatively insensitive to conventional therapy owing to cellular
adaptations effected by the hypoxic microenvironment. Recent efforts have aimed to alter the
hypoxic state and to reverse these adaptations to improve treatment outcome. One way to
increase tumor oxygen tensions is by hyperbaric oxygen (HBO) therapy. HBO therapy can
influence the tumor microenvironment at several levels. It can alter tumor hypoxia, a potent
stimulus that drives angiogenesis. Hyperoxia as a result of HBO also produces reactive oxygen
species, which can damage tumors by inducing excessive oxidative stress. This review outlines
the importance of oxygen to tumors and the mechanisms by which tumors survive under hypoxic
conditions. It also presents data from both experimental and clinical studies for the effect of HBO
on malignancy.

H yperbaric oxygen (HBO) therapy involves the


intermittent administration of 100% oxygen at high
pressure. HBO increases oxygen tensions and oxygen
adjuvant setting with certain types of malignancy. HBO
therefore remains ineffective as a stand-alone therapy or
even as a reliable adjuvant. However, HBO may enhance
delivery to tissues independent of hemoglobin. HBO the efficacy of certain therapies that are limited because
promotes new vessel growth in poorly perfused areas of the hypoxic tumor microenvironment. Further research
and has been used to treat a variety of conditions should also consider treatment feasibility and economic
including wounds,2–4 carbon monoxide poisoning,5–9 and expenditure. These factors should be weighed against
necrotizing soft tissue infections.10 HBO has also been potential therapeutic benefit before HBO can be given
used in combination with drugs for the treatment of credit in the treatment of malignancy.
malignancy.11–16 This is based on the rationale that
tumors may become sensitized to irradiation and other
forms of therapy by increasing the intratumoral oxygen TUMOR BIOLOGY
tension.
Although most of the experimental and clinical studies Tumor Hypoxia
suggest that HBO has no direct effect on tumors, there is
Growth of tumors is limited by the delivery of oxygen
a considerable amount of conflicting evidence to support
and nutrients and the removal of waste products. Oxygen
the idea that HBO has an effect. The most convincing
and nutrients are initially delivered to tumor cells by
effects of HBO are observed when it has been used in an
diffusion from the surrounding microenvironment. As a
Correspondence to: Jurstine Daruwalla, B.Sc, e-mail: jurstine@
tumor grows, cells undergo nutrient deprivation and aci-
pgrad.unimelb.edu.au dosis, and they become hypoxic. Moderately sized
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2113

experimental tumors, 4 to 10 mm in diameter, exhibit cells produce proangiogenic growth factors to initiate the
large regions of hypoxia.17,18 This results in decreased formation of new blood vessels. This is known as the
oxygen tensions (< 20 mmHg) that approach 0 mmHg at angiogenic switch involving up-regulation of proangio-
the tumor center, leading to central necrosis.19 Oxygen genic factors and down-regulation of angiogenic inhibi-
tension in tumors ranges from 2.5 to 30.0 mmHg. This tors. The resultant tumor microvasculature is highly
is in contrast to normal tissue and the tumor periph- disorganized and contains many tortuous vessels that are
ery where oxygen tensions are between 30 and irregular in diameter. These vessels have heterogeneous
60 mmHg.19,20 Three levels of oxygenation coexist in permeability and are functionally abnormal.31,32 Large
tumors: normoxic (tumor periphery and groups of cells in pools of coalesced vessels interspersed with avascular
a tumor mass), hypoxic (adjacent to regions of necrosis areas lead to regions of stagnant or intermittent blood
distant from blood vessels), and anoxic (tumor center). flow.33 This results in a highly inefficient, variable, and
This makes the tumor microenvironment toxic. greatly reduced blood supply compared to normal vas-
Hypoxic tumor cells survive by adapting to the adverse culature, which leads to further hypoxia.
conditions, and are a potential source of tumor recur- The most potent stimulus for angiogenesis is metabolic
rence and treatment failure in several forms of malig- stress induced during hypoxia. Proangiogenic factors,
nancy.21–25 Tumor hypoxia can limit the efficacy of which are secreted by tumor cells, surrounding endo-
therapy in several ways. Malignant cells in hypoxic re- thelial cells, or infiltrating inflammatory cells, are involved
gions are exposed to lower drug concentrations owing to in endothelial cell invasion, migration, and survival.
the limited access of intravenously administered drugs to Vascular endothelial growth factor (VEGF), also known
avascular regions. Hypoxic cells undergo arrest and enter as vascular permeability factor (VPF), and the angio-
a nonproliferating state. A study conducted by Cuisnier poietin families are involved in the development and dif-
et al. on squamous cell carcinoma cells in culture showed ferentiation of the vascular system.34 High levels of
a 20% increase in the number of cells in G0/G1 arrest circulating plasma VEGF have been correlated with a
when exposed to chronic hypoxia compared to normoxic poor prognosis in several cancers including breast,35
cells.26 These cells are less susceptible to chemotherapy prostate,24,36 pancreatic,37 renal,38 head and neck,22 and
and radiotherapy, which target rapidly proliferating cells. colorectal cancer.39
Furthermore, hypoxia directly affects the expression of The most commonly expressed cytokine, VEGF is
many gene products that are involved in angiogenesis, induced by hypoxia.40 It is a multifunctional cytokine that
apoptosis, and glycolysis. Surviving malignant cells are increases microvascular permeability and induces
preferentially selected and undergo clonal expansion, endothelial cell migration by promoting the invasion of
giving rise to a highly malignant cell line. This culminates collagen by vascular endothelial cells, resulting in the
in a more aggressive phenotype associated with poor formation of tube-like structures.41,42 The resultant ves-
patient survival as seen in patients with cancer of sels are abnormal with chaotic blood flow, resulting in
the uterine cervix,23,27–29 squamous cell carcinoma of the hypoxia, which in turn drives further angiogenesis. This
head and neck21 and renal and bladder cancer.30 acts as a positive feedback mechanism (Fig.1, A).
Tumor cells adapt to the ischemic and low nutrient Hypoxia is thus both a cause and a consequence of
microenvironment by three main adaptations. First is the angiogenesis.
angiogenic switch, which results in a shift in balance of In vitro43,44 and in vivo24,45 evidence supports hypoxia
proangiogenic versus antiangiogenic factors leading to as a potent stimulator of VEGF expression.46 VEGF
the formation of an aberrant vascular network. Second is mRNA has been shown to either co-localize with hypoxic
deregulation of apoptosis, where critical components of cells or localize adjacent to hypoxic cells.47 The VEGF
the apoptotic cascade are altered allowing tumor cells to gene has been shown to be particularly active under
evade apoptotic destruction. Third is the glycolytic shift, hypoxic conditions at the level of transcription, increased
where tumor cells preferentially switch to anaerobic gly- stability of VEGF mRNA, and preferential transla-
colysis. All three mechanisms are driven by the hypoxic tion.40,48–50 A study conducted in human tumors found
tumor microenvironment. that in all tumors highly expressing VEGF the mRNA
signal pattern is highly correlated with hypoxia as deter-
Angiogenic Switch mined by binding of the hypoxia marker EF5.45 Increased
VEGF mRNA and protein levels were also found in
Successful growth and metastases of tumors requires hypoxic brain tissue compared with normoxic tissue.51
the establishment of an efficient blood supply. Tumor A study conducted on ovarian cancer cells showed
2114 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

Figure 1. Tumors survive under hypoxic conditions via adaptations that are regulated by the hypoxic microenvironment. These
adaptations facilitate tumor progression by re-regulating molecular mechanisms involved in angiogenesis and survival. HBO may
interfere with each adaptation (*) by altering the hypoxic state of tumors. #Increased glycolysis promotes expression of hypoxia
inducible factor-1 via stabilization of hypoxia inducible factor-1a (HIF-1). VEGF: vascular endothelial growth factor; ROS: reactive
oxygen species; ATP: adenosine triphosphate; NADPH: reduced nicotinamide adenine dinucleotide phosphate.

increased VEGF expression as a consequence of factor (FGF),57 angiogenin,58 epidermal growth factor
hypoxia via up-regulation of hypoxia inducible factor (EGF), nitric oxide synthase, transforming growth factor-b
(HIF)-1a.43,45 (TGF-b). The proangiogenic cytokines also play a role
Transcriptional activation of VEGF is achieved by the including granulocyte/macrophage colony-stimulating
transcription factors HIF-1 and HIF-2. HIF-1 is a hetero- factor (GM-CSF) and interleukin-8 (IL-8). All of these
dimeric transcription factor that regulates oxygen factors are elevated in the presence of hypoxia.
homeostasis and physiological responses to oxygen Aberrant IL-8 expression has been reported in several
deprivation. HIF-1 consists of two subunits (HIF-1a and solid malignancies including breast,59,60 colorectal, and
HIF-1b) and is up-regulated as a result of decreased pancreatic cancers.25 Oxidative stress induced by the
cellular oxygen tensions via stabilization of the HIF-1a oxygen radical generating sugar thymidine phosphory-
protein.43,52 Under normoxia HIF-1a is degraded by lase resulted in induction of IL-8 along with increased
hydroxylases, but under hypoxia HIF-1a evades degra- VEGF and matrix metalloproteinase-1 (MMP-1).61
dation and dimerizes with HIF-1b. Reexposure to a Hypoxia also induced IL-8 mRNA and protein expression
normoxic environment has been shown to result in rapid in the most aggressive human melanoma cells in vitro.62
decay of HIF-1 activity.53 Similarly, loss of HIF-1a in The presence of IL-8 has also been implicated in the
endothelial cells results in profound inhibition of blood production of FGF-2.
vessel growth in solid tumors54 and causes vascular FGF-2 is a potent growth factor in prostatic epithelial
regression in HIF-1a-deficient mouse embryos.52 This and stromal tissue as has been shown in prostatic cells.63
shows that HIF-1 is a key transcriptional mediator of Kuwabara and colleagues showed that macrophages ex-
VEGF-induced angiogenesis in response to hypoxia55,56 posed to low oxygen tensions secreted PDGF and FGF,
(Fig. 1, A). HIF-2 is responsible for vascular remodeling which further stimulated proliferation of hypoxic endothelial
following angiogenesis and also regulates the expression cells.57 Angiogenin, when bound to its receptor, facilitates
of VEGFR-2 and the VEGF receptor Flk-1. endothelial cell digestion of the extracellular matrix and
There are a number of other proangiogenic factors that degradation of the basement membrane, promoting
are up-regulated during the angiogenic switch including endothelial cell migration and angiogenesis. High levels of
platelet-derived growth factor (PDGF), fibroblast growth the angiogenin protein and mRNA have been found in
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2115

highly aggressive human melanoma cells in vitro and in using its own RNA as a template to add telomeric repeats
vivo under hypoxic conditions.58 The same group showed onto the ends of chromosomes. Telomerase is expressed
that only angiogenin and VEGF were up-regulated, and in tumor cells of patients with colorectal,74 ovarian,75
other growth factors tested (e.g., bFGF, PDGF, TGFb) in gastric,76 and lung77,78 cancer. This results in the
parallel showed minimal elevation. Epidermal growth fac- uncontrolled replication of malignant cells with acquired
tor (EGF), when bound to its receptor EFGR, results in genomic instability.68,71–73 Minamino et al. showed that
increased cell proliferation. Increased expression of EFGR telomerase is particularly up-regulated under chronic
has been associated with a worse prognosis and reduced hypoxia.79 Hypoxia also disrupts the regulation of other
response to chemotherapy in gastric,64 colorectal,65 cer- genes associated with apoptosis.68,80
vical66 cancer and has been used as a prognostic marker Members of the Bcl-2 family act as inhibitors (Bcl-2,
for patients with bladder cancer.67 In addition to angio- Bcl-XI, Bcl-W) and promoters (Bax, Bad, Bak, Bcl-Xs) of
genesis promoting tumor growth and progression, malig- apoptosis. Alterations in the ratio of these protein
nant cells also acquire the potential to evade apoptotic expressions may attenuate an antiapoptotic effect. Bcl-2
destruction. is a potent inhibitor of cell death that is particularly up-
regulated in some tumors, especially in the presence of
hypoxia.81 This allows Bcl-2 to promote tumor cell sur-
Deregulation of Apoptosis vival by blocking programmed cell death.82 Conversely,
To cope with the hypoxic microenvironment, tumor Bax, a death promoter, is inactivated in certain types of
cells increase their metabolic rate, which often leads to colon cancer.83 The Bax gene, known to promote apop-
DNA damage.68 Under physiological conditions, when tosis is mutated in several forms of cancer. Overexpres-
cellular repair enzymes cannot correct the DNA damage, sion of Bcl-2 combined with loss or mutation of Bax and
the apoptotic cascade is activated, resulting in cell death. p53 causes a significant reduction in the apoptotic
Tumors, on the other hand, possess cellular mechanisms capacity of cells (Fig. 1, B).
(particularly active under hypoxic conditions) that allow These adaptations are integrated to some extent.
them to evade apoptosis despite the extent of DNA Deregulation of apoptosis can influence angiogenesis.
damage. Spontaneously regressing tumors69 and tumors A study conducted on colorectal tumor xenografts in nude
responding to cytotoxic therapy exhibit a high degree of mice found that deletion of p53 promoted neovascular-
apoptosis. These mechanisms involve deregulating cel- ization of tumors through enhanced HIF-1 levels, which
lular components and genes critical to cell replication and augmented the expression of VEGF.80
apoptosis, such as the p53 tumor suppressor gene. The Chronic hypoxia induced neither apoptosis nor necro-
p53 protein plays a pivotal role in cell cycle regulation and sis of the KB-3-1 head and neck squamous cell carci-
is a promoter of apoptosis.68,69 It is the most commonly noma cell line due to an imbalance in the ratio of Bcl-2/
mutated gene in human cancer and correlates with ad- Bax.26 Hypoxic regions have also been correlated with
vanced tumor stage and indicates a poor patient prog- reduced apoptotic potential of tumors with a highly
nosis.70–73 Deregulation of p53 in cancer occurs through malignant phenotype.23,68 The cellular responses
both inactivation of wild-type p53 and accumulation of described above are adaptations tumor cells make as a
mutated p53. Loss of p53 function increases tumor cell result of oxygen deficiency. Hypoxia hereby acts as a
viability, chromosomal instability, and cellular life-span. physiological selective agent promoting the clonal
Telomere length limits the replicating ability of cells, expansion of a highly aggressive lineage of cells with a
resulting in cell senescence. Developing cells undergo range of cytogenetic abnormalities that are resistant to
telomere erosion as a result of rapid division. Once the apoptosis.
telomeres have eroded, the cell becomes senescent or Cells in a hypoxic microenvironment are deprived not
undergoes apoptosis via activation of p53. Replicative only of oxygen but also of nutrients. The third adaptation
senescence or irreversible cell cycle arrest limits the tumor cells make in response to hypoxia and nutrient
proliferation of damaged cells and is an important tumor deprivation is to attain energy from an alternate pathway,
suppression mechanism. When DNA is damaged, wild- known as the glycolytic shift.
type p53 has the ability to induce cell cycle arrest and, if
irreversible damage has occurred, induce apoptosis. Glycolytic Shift
Mutated p53 results in reduced telomere erosion via
activation of the enzyme telomerase (Fig. 1, B). Telo- As a result of increased energy demands amidst a
merase enhances the proliferative capacity of cells by diminished oxygen supply, tumors depend on anaerobic
2116 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

glycolysis. The switch to anaerobic glycolysis is an lytic capacity of tumor cells. Increased glycolysis
important adaptation facilitating rapid tumor progres- places tumors under constant oxidative stress for several
sion and is known as the glycolytic shift. This was first reasons.
proposed by Otto Warburg and is termed the ‘‘Warburg Chronic exposure of cells to high glucose levels has
effect’’; it results in a shift in energy production from oxi- been shown to increase intracellular reactive oxygen
dative phosphorylation to anaerobic glycolysis.84 species (ROS) production.94 Due to the low ATP yield
Glycolysis is a universal metabolic pathway for the from glycolysis, ATP is generated from alternate sources
catabolism of pyruvate accompanied by the formation of such as the degradation of cellular proteins and amino
adenosine triphosphate (ATP), which is the main source of acids. This degradation results in production of ROS,
energy for cells. The glycolytic pathway is regulated by key which induces further oxidative stress (Fig. 1, D). In-
enzymes beginning with glucose entering the cell bound to creased glycolysis results in the production of nicotin-
a glucose transporter, either GLUT-1 or GLUT-3. The first amide adenine dinucleotide phosphate (NADPH)
phosphorylation is catalyzed by the enzyme hexokinase. oxidase, a primary inducer of the superoxide and cata-
Under aerobic conditions pyruvate is metabolized to form lase radicals.95 To ensure that the ROS content does not
carbon dioxide and water, resulting in a high ATP yield. reach toxic levels, antioxidant defenses are switched on.
Under hypoxia, insufficient oxygen is available to support The antioxidant glutathione peroxidase is produced. It
the aerobic oxidation of pyruvate. Instead, anaerobic gly- has been shown that production of this antioxidant is
colysis occurs where pyruvate is reduced to lactate sustained by NADPH produced as a result of increased
resulting in a low ATP yield. To compensate for the low glycolysis.96,97
ATP yield, tumors increase their glycolytic rate.1 In- The glycolytic shift provides a pathway for tumors to
creased glucose metabolism produces nicotinamide sustain an increased metabolic rate under hypoxia. More
adenine dinucleotide (NADH), which is constantly reoxi- importantly, this adaptation induces stabilization of HIF-1,
dized to sustain continual anaerobic glycolysis. which regulates processes that not only maintain glycol-
The glycolytic shift is beneficial for tumors at several ysis but facilitate tumor progression. Transcription of
levels and primarily occurs under the influence of the VEGF and IL-8 are up-regulated in tumor cells in re-
transcriptional factor HIF-1 and other cell signaling sponse to glucose deprivation via HIF-1-dependent
mechanisms driven by hypoxia. As discussed earlier, mechanisms. This is an example of how one adaptive
HIF-1 regulates numerous genes involved in angiogene- mechanism (glycolytic shift) sustains another mechanism
sis,55 cell cycle control,43 and glycolysis,52 including of adaptation (angiogenesis) to promote tumor growth.
GLUT-1 and hexokinase.85,86
Hypoxia elevates the expression of many key glycolytic ROS Production
enzymes, as shown by Webster et al. in partially differ-
entiated mammalian myotubes in vitro under normoxic Reactive oxygen species, or free radicals, are a
and hypoxic conditions. Under hypoxic conditions, the by-product of aerobic respiration and cellular metabolism
glycolytic enzyme mRNAs increase and the respiratory and are produced by all eukaryotic cells. Low levels of
mRNAs (involved in oxidative phosphorylation) decrease. ROS are generated in the mitochondria and are important
The inverse occurred under normoxic conditions.87 for regulating signal transduction and normal cell prolif-
Overexpression of the glucose transporter genes GLUT-1 eration and function.98,99 ROS include the superoxide
and GLUT-3 has been observed in human tumors.85,88 anion (.O2)), hydroxyl radical (.OH), hydrogen peroxide
HIF-1 also up-regulates mitochondria-bound hexoki- (H2O2), and singlet oxygen (1O2). The main cellular
nase.89,90 This enzyme is involved in the first phosphor- components susceptible to damage by ROS are lipids,
ylation of glucose during glycolysis and is unresponsive to proteins, carbohydrates, and nucleic acids.100 In excess,
feedback inhibition. This commits the tumor cell to con- ROS cause lipid peroxidation, compromise cell mem-
tinued glycolysis (Fig. 1, D). There is also evidence of the brane integrity, and lead to cell death. Excess ROS also
end-products of glycolysis, such as pyruvate promote cause DNA strand breaks, resulting in mutations or
stabilization of the HIF-1a protein and activated HIF-1 deletions of various genes.101,102
gene expression,91 thereby facilitating further glycolysis There is substantial evidence for the involvement of
and tumor progression (Fig. 1, #). ROS in carcinogenesis.99,102,103 ROS accumulation has
In addition to HIF-1, the mutated oncogenes p53 and been implicated in the initiation and progression of
myc92 and defects in cell signaling such as the Akt kinase tumors.101,104 ROS are induced by oxidative stress during
pathway93 have also been shown to increase the glyco- oxygen deficiency, reoxygenation (reperfusion), or
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2117

excess oxygen (hyperoxia). Levels of ROS are tightly SOD,114 glutathione peroxidase, catalase,115 and biliru-
regulated by antioxidant defenses, which prevent oxida- bin,116 which have all shown increased expression and
tive damage. During oxidative stress, the antioxidants activity in tumors compared to normal tissue. Evidence
superoxide dismutase (SOD), catalase, glutathione per- supporting the threshold effect is provided by clinical
oxidase, and bilirubin are up-regulated in tumors103 studies where increased H2O2 levels result in proliferation
compared to normal tissue.105 of normal cells and destruction of tumor cells, whereas
The production of ROS in normal and tumor tissue is when H2O2 is decreased, the reverse occurs. This sup-
fundamentally different. In nonmalignant cells, ROS lev- ports the idea that ROS levels in tumors are normally
els are relatively low and tightly regulated by antioxi- at sublethal doses, and any increase would induce cyto-
dants.99,106 In contrast, tumors are under constant toxicity.106,111–113
oxidative stress due to increased glycolysis, transcription The concept of the threshold effect may be an attrac-
factor activation, and vascular architecture. The chaotic tive therapeutic approach to destroying tumors with per-
and erratic blood flow of tumors results in intermittent sistent ROS production.103,117–119 The nature of ROS in
periods of hypoxia followed by reperfusion. Reperfusion tumors is therefore paradoxical. Although their accumu-
following myocardial infarction or cerebral ischemia is lation leads to cancer initiation and sustained progres-
known to cause generation of ROS. Based on the same sion, they may also serve as a target for therapy.
concept, oxidative stress induced by reperfusion is a Therapies that induce ROS production include the che-
major source of ROS production in tumors.70 ROS pro- motherapeutic agent doxorubicin, an O2)-generating
duction also mitigates cell signaling, which has been agent, and bleomycin. Radiotherapy and photodynamic
shown to promote oncogenic transformation and uncon- therapy also induce ROS generation in tumors.
trolled proliferation.107 Markers of oxidative stress have Oxygen deficiency limits treatment efficacy on several
been detected in samples from in vivo breast carcinomas, fronts. First, chemotherapeutic drugs are unable to
and human tumor cell lines in vitro have been shown to reach all tumor cells in a poorly perfused microenvi-
produce more ROS than nonmalignant cell lines.70 ronment. Administration of a higher dose is not an op-
Elevated ROS offer a selective growth advantage to tion because of the severe dose-limiting side effects.
tumor cells in several ways. ROS are responsible for DNA Radiotherapy destroys tumor cells only in well oxygen-
strand breaks, leading to mutations in tumor cells. These ated regions. Second, hypoxia induces cell cycle arrest
mutations may affect the genes responsible for apoptosis so some tumor cells become trapped in the G0/G1
or induce oncogenic transformation of cells.108,109 ROS phase and remain noncycling.120 In addition to this,
promote the constant activation of transcription factors, hypoxia drives angiogenesis-promoting tumor growth
leading to increased proliferation of cells with acquired and metastases under oxidative stress. Tumor cells
DNA damage. These cells are genomically unstable ow- under oxidative stress produce ROS, which result in
ing to intrinsic mutations and contribute to a more mutations. Deregulation of the apoptotic cascade in the
aggressive phenotype (Fig.1, C).102 presence of hypoxia prevents malignant cell destruction.
Upon exposure to some anticancer agents, intratu- The selective replication of these defective cells leads to
moral ROS become greatly elevated. Initially this oxida- genomic instability. All of these events culminate in a
tive stress triggers apoptosis. In advanced cancer highly aggressive tumor in which regions of cells are
however, adaptive mechanisms such as evasion of resistant to destruction and can cause tumor recurrence.
apoptosis or up-regulation of antioxidants prevent Novel strategies target tumors by altering the hypoxic
destruction of these cells which then undergo clonal state through improved oxygenation to possibly reverse
expansion99 (Fig. 1, C). Antioxidants such as SOD and or remove the adaptive defenses of tumors or induce
glutathione peroxidase are markedly up-regulated in oxidative stress to promote tumor destruction. The latter
several forms of cancer.110 strategy has been investigated on human colon and liver
Prolonged exposure to anticancer agents leads to carcinoma cell lines in vitro.106 Improving tumor oxy-
apoptosis via excessive ROS generation, which occurs genation and vascularization may increase drug deliv-
when the antioxidant system is overwhelmed. This can be ery. This has been shown experimentally121 and in nude
explained by the ‘‘threshold effect’’ whereby ROS reach a mice with human epithelial ovarian cancer treated with
level beyond which the antioxidant capacity is inundated, cisplatin.122 It may also reduce or remove the hypoxic
resulting in irreversible damage and apoptosis.106,111–113 stimulus that triggers the adaptive mechanisms of tu-
To ensure ROS do not exceed the threshold level, tu- mors. One way to improve tumor oxygenation is to
mors induce rapid up-regulation of antioxidants such as administer hyperbaric oxygen.
2118 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

Table 1.
Animal studies for the effect of HBO on malignancy

Study Year Animal model Animal tumor HBO regimen


De Cosse14 1966 Syrian hamsters Melanoma 2.0 atm 7–12 exposures, 6 days
(n = 160)

McCredie143 1966 C3H mice (n = 282) C3HBA murine tumor 3.0 atm 12 exposures 30 minutes

Suit144 1966 BDF mice Mammary tumor 3.0 atm 30 exposures 60 minutes
Johnson145 1967 CDBA (F1) mice Melanoma and leukemia 3.0 atm 20 exposures 30 minutes
(n = 350)
Dettmer149 1968 CFN albino rats Walker carcinosarcoma 1.0 and 3.0 atm8–15 exposures
(n = 60)
Feder146 1968 C3H mice (n = 418) C3H rhabdomyo-sarcoma 3.0 atm 20 exposures 20 minutes
Valaitis139 1968 Swiss mice (n = 600) Ehrlich ascites tumor 2.0 atm 7 exposures 120 minutes

Evans161 1969 CBA mice (n = 245) Squamous skin carcinoma 2.0 atm 1 exposure

Johnson147 1971 DBA/2 mice (n = 92) Lymphoblastic leukemia 3.0 atm 11 exposures 90 minutes

Shewell137 1980 C3H/Bts mice (n = 44) Transplanted and 3.0 atm 6 exposures 20 minutes
spontaneous
mammary tumors

Martin162 1987 WAG/rij-Y rats BA1112 3.0 atm 30 minutes


rhabdomyosarcoma
Marx131 1987 Hamster DMBA-induced SCC 2.4 atm 20 exposures
Frid163 1989 SHR miceC57/B1 mice Transplanted sarcoma Not reported
37 Melanoma B16
McMillan138 1989 Syrian hamsters DMBA-induced oral 2.5 atm 85 exposures 99 minutes
(n = 30) mucosal SCC
Granstrom164 1990 C-57 mice Sarcoma 2.8 atm 9 exposures 120 minutes
Mestrovic148 1990 Y59 rats (n = 38) Anaplastic CA-induced lung 1.0 or 3.0 atm 16
(n = 16) metastases Anaplastic exposures 90 minutes
CA in hind foot

Headley165 1991 Nude mice Human SCC xenografts 2.4 atm 15 exposures
Sklizovic166 1993 Nude mice (n = 40) Human head and neck SCC 2.0 atm 21–28 exposures
Lian135 1995 ICR mice (n = 120) S-180 murine sarcoma 2.5 atm 18 exposures 90 minutes

McDonald167 Syrian hamsters DMBA-induced tumors 2.8 atm 30 exposures 60 minutes


(n = 40)
Takiguchi15 2001 DDY mice (n = 41) Sarcoma 180 2.0 atm 17 exposures 90 minutes

Huang6 2003 C3H mice C3H tumors 3.0 atm 15 minutes

Petre12 2003 Sprague-Dawley rats MCA2 sarcoma 2.0 atm 7 exposures 30 minutes
(n = 24)

Shi168 2005 Ncr-nu/nu mice Head and neck SCCA 2.4 atm 13-28 exposures 90 minutes
HBO: hyperbaric oxygen; atm: atmospheres; PDT: photodynamic therapy; SCC: squamous cell carcinoma; CA: cancer;
DMBA: dimethylbenzanthracene; N/A: survival not assessed.
+: HBO had a tumor stimulatory/adverse effect; –: HBO had a tumor inhibitory effect; 0: HBO had no effect on tumors.
If two symbols are given, the effect was mixed.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2119

Additional
therapy Outcome Comment Survival
Mechlorethamine, – HBO alone decreased pulmonary metastases Increased survival
cyclophosphamide HBO had no synergistic effect (P < 0.001)
+ amethopterin on primary tumor growth
None 0 No effect on primary tumor No effect N/A
on lung metastases
50 days after excising primary tumor
None 0 No effect on primary or metastases N/A
None 0 No effect on primary tumor size or Leukemia No
number of metastases effect on survival
None – HBO significantly reduced distribution and number N/A
of distant metastases
None 0 No effect on metastases N/A
Nitrogen + Increased tumor growth and metastases HBO HBO alone
mustard (HN2) appeared to act synergistically with HN2 decreased survival
Radiotherapy 0 No effect on lung metastases with N/A
radiotherapy and HBO (P > 0.1)
None 0 No effect on tumor weight, growth rate, No effect on
or metastases (P = 0.5). survival (P = 0.05)
None 0 No effect on transplanted primary tumors N/A
+ Higher incidence of lung metastases N/A
in spontaneous tumors in HBO group
(88.8%) vs. control (66%)
Fluosol-DA and – Pretreatment with HBO and Fluosol N/A
radiotherapy significantly reduced tumor cell survival
None – HBO delayed tumor growth N/A
None 0 No effect on growth or metastases of N/A
None transplanted tumors
None – HBO reduced number of tumors (P < 0.01). N/A
HBO increase tumor size (P < 0.02)
None 0 No effect on tumor growth No effect on survival
None – 1 atm HBO had no effect on tumor growth3 Increased
atm HBO strongly suppressed lung survival (P < 0.01)
metastases (P < 0.001)
No effect on tumor growth.
None 0 No effect on tumor growth N/A
None 0 No effect on tumor growth, volume ,or histology N/A
None – Reduced tumor volume in HBO group HBO benefited survival
compared to control (P < 0.01) Increased 26.7%, vs. control 6.7%.
necrosis in HBO group (33%) compared
to control group (17%)
None – HBO significantly reduced tumor size (P < 0.05) N/A

5-Fluorouracil – Combined HBO + 5-FU resulted in greatest N/A


(5-FU) tumor reduction (P < 0.01).
PDT – HBO significantly improved tumor oxygenation N/A
and tumor cell kill of PDT
Doxorubicin – Combined therapy significantly reduced lung N/A
metastases (P < 0.01) and overall lung
weight (P < 0.01) compared to doxorubicin alone
None 0 HBO had no effect on tumors N/A
2120 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

Table 2. Table 2.
Clinical trials for the effect of HBO on malignancy Clinical tria
Patients
Study Year (no.) Trial type Tumor HBO regimen
Johnson136 1966 25 Uncontrolled Advanced cervical CA 3.0 atm 30 exposures

Cade140 1967 49 Randomized Bronchogenic 3.0 atm 40 exposures,


40 control trial CA Bladder CA < 40 minutes

Van den Brenk151 1967 85 Controlled Advanced head 3.0 atm 2–6 exposures
Radiotherapy 51 trial and neck Misc.
CA (breast, bladder
bowel, uterus)
Johnson169 1974 64 Controlled Cervical CA 3.0 atm 25–30 exposures
trial
Bennett170 1977 213 Controlled Cervical SCC 3.0 atm 10 exposures
trial

Henk11 1977 276 First controlled Head and neck CA 3.0 atm 10 exposures
trial

Henk171 1977 104 Second Head and neck 3.0 atm 10 exposures
controlled trial

Dische152 1978 1500 Controlled trial Head and neck, bladder, 3.0 atm 6–12 exposures
bronchus, or cervical CA
Perrins78 1978 236 Controlled Bladder CA 3.0 atm 6–40 exposures

Watson172 1978 320 Controlled trial Cervical CA 3.0 atm6–27 exposures

Brady173 1981 65 Controlled trial Cervical SCC 3.0 atm 10–12 exposures

Henk154 1986 104 Prospective Head and neck SCC 4.0 atm 10 exposures
et al. [154]. controlled trial
Sealy155 1986 130 Prospective Head and neck SCC 3.0 atm 6 exposures
randomized trial

Eltorai141 1987 3 Anecdotal report 2 Bladder and 2.0 atm 10–20 exposures
1 urothelial CA
Bradfield150 1996 4 Anecdotal report Head and neck SCC Pressure not reported
8–14 exposures
Granstrom174 1996 123 Prospective trial Head and neck 2.5 atm 30–90 exposures

Dische175 1999 335 Randomized Advanced cervical SCC 3.0 atm 10 exposures
controlled trial

Haffty176 1999 48 Randomized trial Head and neck SCC 4 atm 2 exposures

Haffty177 1999 45 Retrospective trial Laryngeal CA 4 atm 2 exposures

Kohshi178 1999 29 Nonrandomized trial Glioblastoma 2.5 atm 20–30 exposures

Maier157 2000 75 Prospective Advanced 2.0 atm 1–3 exposures


nonrandomized trial esophageal CA
MCA: methylcholanthrene; Gy: Gray (1 gy = 100 rad); fx: fractions; N/A – survival not assessed
+: HBO had a tumor stimulatory/adverse effect. –: HBO had a tumor inhibitory effect; 0: HBO had no effect on tumors.
If two symbols are given, the effect was mixed.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2121

Additional therapy Outcome Comments Survival


Radiotherapy + HBO group had early appearance and N/A
unusual frequency and pattern of metastases
Radiotherapy 0 No effect on primary tumor growth or metastases No effect on survival
+ Enhanced tumor development HBO decreased survival
Radiotherapy and doubled metastases
Radiotherapy – Fewer metastases with HBO (41%) vs. No effect on survival
Radiotherapy control (68%) (P < 0.05). Significantly
decreased metastases (P < 0.014)

None 0 No effect on metastases HBO improved 5-year


survival (44% vs. control 16%)
Radiotherapy 0 Combined treatment with HBO increased No effect on survival
local clearance rate but had no effect
on metastases
Radiotherapy 35 Gy/10 fx 0 Improved local tumor control in combined No effect on survival
group Reduced need for salvage
surgery (P < 0.01)
Radiotherapy – Local recurrence free rate better in HBO group Statistically improved
disease-free survival in
HBO group
Radiotherapy 0 HBO reduced recurrence (P < 0.001) Improved 5-year survival
but had no effect on metastases (P = 0.97) (P < 0.001)
Radiotherapy 0 HBO had no effect on hypoxic No effect on survival at
tumor cells or metastases 4 years (P = 0.68)
Radiotherapy 0 No effect on metastases Increased No effect on survival
recurrence-free rate with combined
therapy only in patients
< 55 years (P < 0.001)
Radiotherapy 0 Distant failure higher in control group No effect on survival
34% vs. HBO group (16%)
Radiotherapy 35 Gy/10 fx – Improved local control of tumors 5-Year survival 60% for combined
and less advanced tumors therapy vs. 30% for control
Radiotherapy 36 Gy/6 fx – Combined therapy improved N/A
+ misonidazole 63 Gy/30 fx local tumor control by 15%
in air
– + Aggressive tumor growth after HBO therapy N/A

Radiotherapy + Rapid progression of tumors and increased HBO did not improve survival
tumor recurrence after HBO therapy
Radiotherapy – Recurrence rate 16% lower in N/A
combined therapy group
Radiotherapy 0 Combined therapy did not improve local No effect on survival
tumor control. Some late morbidity
with HBO observed
Radiotherapy 23 Gy/2 fx with – Significantly improved 5-year local tumor No effect on survival
HBO 25 Gy/ 2 fx in air control at both radiotherapy doses
Radiotherapy 22 Gy/2 fx – Complete response in 87% of casesImproved N/A
with or without HBO local 10-year control in most responders
Radiotherapy 57.8 Gy – 73% Tumor regression in half of responders Median survival 24 months in
with or without HBO combined group vs.
12 months (P < 0.05)
PDT – Combined therapy reduced HBO improved survival (P = 0.0098)
tumor length (P = 0.0002)
2122 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

MODIFICATION OF TUMOR HYPOXIA WITH Hypoxia is essential for stabilization of HIF-1a and sub-
HYPERBARIC OXYGEN THERAPY sequent VEGF expression. Reoxygenation of hypoxic
cells induces rapid degradation of HIF-1a degradation53
Hyperbaric oxygen (HBO) therapy involves the admin- and subsequent VEGF production and angiogenesis in
istration of pure oxygen at a pressure greater than 1 vitro.54 ROS, at low levels, assist tumor growth but
atmosphere (atm).13,123 At normal atmospheric pressure become toxic at high levels. This has been shown in vitro
(1 atm), hemoglobin is approximately 97% saturated with on human colon, liver,106 leukemic, and ovarian114 cell
oxygen. This is equivalent to 19.5 volume percent (vol%) lines. HBO may increase intratumoral ROS levels past
oxygen. Approximately 0.32 vol% of oxygen is dissolved the threshold and induce tumor cell destruction, as has
in plasma. Any further increase in oxygen pressure or been shown in vitro in mouse fibroblast cells134 and in
concentration has minimal impact on total hemoglobin vivo in mice with S-180 sarcoma.135 Current opinion on
oxygen saturation. Most HBO treatments are performed the effect of HBO therapy on tumors remains controver-
at a pressure of 2 to 3 atm. The additional pressure when sial. Despite theoretical considerations of tumor stimula-
coupled with inspiration of 100% oxygen substantially tion, to date there is enough evidence to preclude any
increases the amount of oxygen dissolved in blood tumor stimulatory effects of HBO.
plasma. At a pressure of 3 atm, the amount of plasma Johnson and Lauchlan first reported a tumor-stimula-
oxygen increases from 0.32 vol% (at 1 atm) to 6 vol%. tory effect with HBO, demonstrating increased metasta-
This is a 95% increase in plasma oxygen concentration ses in patients with cervical cancer.136 This has been
compared to atmospheric conditions. supported by animal studies137–139 and other clinical tri-
The short-term effects of hyperoxia include enhanced als.136,140,141 However, in a review of animal and clinical
oxygen delivery to ischemic tissues,124 vasoconstriction, studies conducted by Feldmeier et al. it was concluded
reduction of edema, and immunomodulatory properties that intermittent HBO exposure had no stimulatory effect
such as activation of phagocytosis.125,126 Long-term on primary or metastatic tumors.129
effects include neovascularization122,127,128 and stimula- In vitro studies have shown that 6 atm of absolute
tion of collagen formation by fibroblasts.127 HBO can oxygen inhibits the growth of Erlich ascites tumor
thereby be applied clinically to heal hypoxic and ische- cells.142 However, in vivo animal studies have produced
mic wounds and to the recovery of radiation-injured varying results, with reports of both minimal and no ef-
tissue.127,129–133 fect.143-147 It may be speculated that the absence of
During a standard HBO treatment, the rise in oxygen effect may be a result of tumor cell compensatory
partial pressure of arterial blood can cause up to a four- mechanisms. Such mechanisms include antioxidant de-
fold increase in the distance that oxygen diffuses through fenses that would override the potential adverse effects
normal tissue. There is concern that increased oxygen of oxidative damage induced by ROS production during
may stimulate tumor growth via reoxygenation of hypoxic HBO treatment. Kaelin et al. showed a significant in-
tumor cells and increased angiogenesis as observed crease in the activity of SOD and improved survival of
during wound healing. Although HBO promotes angio- the skin flaps of rats exposed to HBO.112 The time
genesis in healing wounds, this does not mean that it schedule of HBO exposure may also influence its ef-
would induce tumor growth via the same mechanism. fects. Mestrovic et al. showed significantly improved
There are several rationales for the use of HBO as an survival and reduced lung metastatic deposits in rats
adjuvant therapy. HBO, in theory, has the potential to after HBO (3 atm) administered on days 1 to 6 or 7 to
intercept each of the adaptations tumor cells make under 12. However, no effect was seen in rats exposed to
hypoxic conditions (asterisks in Fig. 1). HBO greatly HBO on days 13 to 18.148
improves oxygen perfusion in tumors, thus altering the Experimental and clinical evidence for the effect of
hypoxic microenvironment. This may have implications HBO on tumors have been varied. The data presented in
for angiogenesis and apoptosis and push ROS levels the tables summarizes animal (Table 1) and clinical
past the threshold level. Altering hypoxia may remove the (Table 2) studies over the past 50 years. The outcome of
stimulus for the angiogenic switch. HBO may promote each study is reported where HBO had a tumor stimula-
apoptosis via the production of ROS, which can over- tory (+), inhibitory ()), or no (0) effect. In the literature
whelm the tumor’s antioxidant defenses. Improving the reviewed over the past 50 years, only 10% of studies
oxygenation of hypoxic tumor cells may remove the (both experimental and clinical) reported that HBO has a
hypoxic stimulus that initiates the angiogenic switch. tumor-stimulatory effect.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2123

Among animal studies (Table 1), there is evidence both or 3 atm,149 Mestrovic et al. demonstrated suppression
supporting an effect with HBO and negating an effect. of lung metastases with improved survival in Y59 rats
Studies that combined HBO with other therapies were exposed to 3 atm but no effect at 1 atm.148
more successful in achieving tumor control. Takiguchi et Among the research studies conducted in experimental
al. had more favorable results when HBO was combined models, a desirable effect was observed when HBO was
with 5-fluorouracil (5-FU) than with drug treatment alone. combined with doxorubicin12 and in another study with
Similarly, Petre et al. reported significantly improved photodynamic therapy.16 One of the mechanisms of ac-
tumor control with combined HBO and doxorubicin com- tion of doxorubicin is production of ROS. Photodynamic
pared to drug therapy alone. It is important to note that therapy is dependent on the presence of oxygen to
positive effects may be tumor-specific as better tumor destroy cells. Perhaps HBO had a positive effect in these
control was always achieved in animal studies investi- studies because it can directly influence both ROS pro-
gating the effect of HBO on sarcomas. Huang et al. duction and improve tumor oxygenation.
showed enhanced tumor oxygenation and improved It is more difficult to draw conclusions from clinical
subsequent tumor cell kill using HBO in combination with studies due to variability in investigation techniques and
photodynamic therapy on mice with subcutaneous patients. To date, experimental and clinical evidence of
implantation of mammary adenocarcinoma.16 the effect HBO combined with therapies other than
In studies where HBO had a tumor-inhibitory effect, a radiotherapy is limited. The lack of effect of HBO in
common finding in addition to overall tumor reduc- experimental models as a stand-alone therapy may
tion was a reduction in the distribution149 and occur- explain why it has not been investigated extensively in a
rence12,14,148,149 of distal metastases. Again, half of these clinical setting. Nevertheless, by altering oxygen levels in
studies were conducted on sarcomas. A less desirable vivo, HBO can improve the radiosensitivity of tumors,158
effect was observed by Valaitis et al. on Erlich ascites enhance photodynamic therapy,16,81,159 or enhance oxi-
tumors.139 The remaining studies (46% of total animal dative stress and tumor cell kill of certain chemother-
studies) reported no effect with HBO. All models of apy.127,160 This has been investigated in clinical studies.
human head and neck squamous cell carcinoma con-
ducted in nude mice reported no effect with HBO therapy.
A similar trend was observed among clinical studies HBO AS AN ADJUVANT THERAPY
(Table 2) where a small number of researchers reported
tumor stimulation as a consequence of HBO exposure. Clinically, HBO has been investigated when combined
Most of the clinical studies investigated the use of HBO with chemotherapy, photodynamic therapy or radiother-
as an adjuvant to radiotherapy. apy. Radiotherapy induces DNA damage through the
In some cases of advanced cervical136 and blad- ionization of oxygen to produce ROS. Intratumoral oxy-
der140,141 cancer and one case of head and neck can- gen tension therefore determine the effectiveness of
cer,150 HBO increased tumor aggressiveness when radiotherapy. Hypoxia reduces the radiosensitivity of cells
administered alongside radiotherapy. Studies incorpo- as they require three times as much radiation to become
rating a larger cohort of patients with bladder cancer did sensitized as cells with normal oxygen tension. HBO can
not find that HBO influenced tumor progression in com- be administered simultaneously with or prior to irradia-
bination with radiotherapy.151–153 Patients with head and tion to increase the oxygen tension of hypoxic tumor
neck cancer undergoing radiotherapy were most cells.155,162 Alternatively, HBO can be applied after irra-
responsive to HBO, but it improved survival in only just diation to reduce radiation-induced tissue injury once
over 40% of cases.151,152,154–157 normal tissue side effects manifest.156,179 The objective
Although both animal and clinical studies have reported of this is to extend the oxygen diffusion gradient to
varied results, some deductions can be made. First, HBO reoxygenate previously hypoxic cells and thereby radio-
does not overtly contribute to increased tumor growth, nor sensitize them. It has been shown that these intercellular
is it effective as a stand-alone treatment. Second, the conditions persist for some time after leaving the cham-
effect of HBO is dependent on multiple factors including ber.
tumor type and stage as well as the timing, duration, Patients are ideally irradiated prior to or while inside a
atmospheric pressure, and number of HBO exposures. pressure chamber. After numerous clinical trials this
Regarding atmospheric pressure, whereas Dettmer et al. approach has been shown to be of benefit in squa-
found no significant difference in tumor volume of trans- mous cell carcinoma (SCC) of the head and
planted Walker carcinosarcoma in rats following HBO at 1 neck.151,152,154,155,157,174,176 HBO significantly reduced
2124 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

metastatic spread 3 months after irradiation of head and cancer.180,181 Again, the limitations in many of these trials
neck tumors and other primary tumors (breast, bowel, include the practicality of placing patients in HBO cham-
bladder, uterus) treated with radiation.151 Henk et al. bers while simultaneously administering radiation ther-
reported no survival difference in patients receiving apy, as reported in earlier trials.154,155 In addition, one
radiotherapy with or without HBO. However, better local study reporting tumor stimulation with combined HBO/
tumor control was observed in the HBO group with min- radiotherapy recruited patients with varying tumor
imal salvage surgery compared to the non-HBO group.11 grades.140 Grade and stage of tumors are important
In most of the cases, however, HBO has provided no determinants of treatment outcome and should therefore
added benefit during radiotherapy especially in the be evenly distributed among the various treatment
treatment of patients with cervical cancer.169,170,172,175 groups.
Therefore, the application of HBO therapy on certain In 2001, the European Society for Therapeutic Radiol-
patients and tissues may be justified. However, the gen- ogy and Oncology (ESTRO) concluded that the effect of
eral consensus is that HBO does not offer any significant HBO on neoangiogenesis and osteogenesis was graded
clinical benefits or improvement in survival. On review, a level 1 according to evidence-based medicine criteria.182
limited number of hyperbaric facilities are located in the The COST (Cooperation in the field of Science and
proximity of radiation oncology departments. Although Technology) B14 initiative was established in 1999 to
intratumoral oxygen tension persists after HBO exposure attain clinical data based on this level 1 evidence. In the
it is nevertheless temporary. To overcome this problem, most recent review of combined HBO and radiotherapy,
HBO can be administered while patients are irradiated, Mayer et al. presented four randomized clinical trials that
but it is difficult and costly. outlined the activities of the B14 working group.13 Given
Clinical data obtained by the British Medical Council in the variability in past clinical trials, the four proposals
a clinical trial with HBO and radiotherapy found signifi- presented by the COST Action B14 Committee, which
cantly better local tumor control and survival for carci- have been amended and peer-reviewed, are regarded as
noma of the cervix.172 In another randomized control consistent with the ‘‘best practice’’ in the field of hyper-
trial conducted in that same year, HBO therapy gave no baric medicine. At present the trials are open for enroll-
additional therapeutic benefit and in fact increased the ment of patients.
occurrence of late morbidity.152 This may be due to the The first of four initiatives aims to determine whether
combined HBO group being given more fractions of HBO enhances tumor radiosensitivity in patients with
radiation compared to the control group, where patients previously irradiated histologically confirmed recurrent
received standard fractions. This is one of the major head and neck carcinoma. The second is to determine if
obstacles when trying to combine HBO and radiother- HBO improves median survival when applied in combi-
apy. Generally, larger radiation doses are administered nation with conventional fractionation in patients with
in fewer fractions to minimize the number of times pa- glioblastoma multiforme. The last two proposals investi-
tients need to enter the pressure chamber. This can gate the effects of HBO on postradiation injury. All as-
lead to considerable postradiation injury to normal tis- pects of the trials are kept consistent including patient
sue. recruitment, HBO regimen, radiation fractions, and out-
A large multicenter trial conducted by Perrins et al. come measures. Furthermore, it is stipulated that all
found no additional benefit of HBO therapy with irradia- irradiation fractions should precede HBO treatment, and
tion of carcinoma of the bladder and speculated that each fraction must be given within a specified time after
either HBO does not alter the hypoxic state or failure of HBO exposure. This is the first multicenter initiative to
radiotherapy to cure bladder cancer is not due to hypoxic evaluate HBO under controlled settings; and, pending
tumor cells.78 Four other trials investigating the effect of results, more concrete conclusions will be possible.
radiotherapy and bladder cancer reported varied results, Resistance to chemotherapy is common in hypoxic
two of which suggested that combined treatment pro- tumors. HBO may help overcome chemotherapy resis-
moted tumor growth.140,141 However, both of these trials tance by increasing both tumor perfusion and cellular
had a small cohort of fewer than 40 patients. A meta- sensitivity. HBO therapy in combination with chemother-
analysis again investigating HBO combined with radio- apy increases cellular uptake of certain anticancer agents
therapy reviewed 19 trials of tumors at various sites. and the susceptibility of cells to these agents. HBO has
Locoregional control with the combined modality was been shown experimentally to increase the susceptibility
significantly greater than radiotherapy alone. The great- of malignant cells to destruction with taxol,183 doxorubi-
est effect was observed in patients with head and neck cin,12,183 and 5-FU15,184 (Table 1).
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2125

Combined administration of 5-FU and HBO significantly group was significantly lower than that observed with
increased intratumoral drug concentrations in mice PDT alone. Mean survival was 12 months versus 7
implanted with sarcoma15 and limited angiogenesis and months, in favor of the combined therapy.190 Similar
tumor size of 7,12-dimethylbenz[a]anthracene (DMBA)- success was reported a year later on 30 patients, this
induced mammary tumors in rats.184 By increasing ROS time with inoperable non-small-cell bronchogenic carci-
levels,185,186 HBO enhanced the ROS-localized effects of noma.191 A major drawback of PDT is that administering
bleomycin and doxorubicin.20,138 In an experimental laser therapy to the tumor site involves surgery that is
model of pulmonary sarcoma, the chemotherapeutic more invasive than other therapies. This may explain the
effects of doxorubicin were enhanced following HBO limited number of combined PDT/HBO trials.
exposure at 2 atm for 7 days12 HBO stimulated prolifer- Although clinical experience with HBO is generally
ation of an MCA-2 metastatic lung tumor cell line and associated with relatively few side effects, the heteroge-
induced cells to enter the replicating cycle compared to neity of the investigation techniques makes it difficult to
cells left at ambient pressure.12 Another study found that draw conclusions. These variations include the patients’
HBO increased the percentage of prostate cancer cells in tumor type, stage, and baseline levels. There is also
vitro accumulating in G2/M phases from the G0 arrest variation among studies regarding the total radiation
phase.187 There was, however, only one clinical trial that dose, number of fractions, overall time, and the irradiated
evaluated HBO in combination with chemotherapy. The volume. Furthermore, the number of trials is small with
study reported a modest 15% improvement in local tumor modest sample sizes (most had fewer than 200 patients).
control at 1 year when HBO was combined with misoni- Future trials should be reported with a minimal number
dazole compared to drug therapy alone.155 This study of variables to determine the true effect of HBO therapy.
was conducted 20 years ago, and since then there A sham therapy should be used to mask both the subjects
appears to be no further clinical evidence of improved and the assessors to HBO therapy. Employment of a
outcome of HBO with chemotherapy. Rather, HBO has double-blind trial in which patients are placed in an HBO
been used to reduce the side effects associated with chamber with normobaric pressure oxygen as a control
chemotherapy. Chronic arm lymphedema is a common compared with a chamber exposed to hyperbaric oxygen.
problem in women who have undergone radiation therapy Economic considerations should also be factored along
for breast cancer. HBO has been shown to reduce with the practicality of the treatment in a clinical setting.
localized edema in a cohort of 10 women suffering from Given the variation in pathology, it is not surprising that
this condition.188 Based on this evidence, patients are there is considerable variation in patient baseline char-
being recruited to determine if a more aggressive HBO acteristics at the time of recruitment as well as treatment
regimen can further reduce the volume of edema.189 outcome. Moreover, publication bias may also play a role,
Photodynamic therapy (PDT) utilizes a specific wave- where results from more favorable trials may be more
length of light to activate intravenously preadministered likely to reach completion and subsequent publication.
light-sensitive drugs (photosensitizing agents such as
porphyrins) that are taken up by target cells. Light can then
be targeted to the tumor site. Photochemical activation of CONCLUSIONS
the photosensitizer generates highly toxic singlet oxygen
and other ROS. The response to PDT depends on ade- Tumors are initially susceptible to chemotherapy and
quate tumor oxygenation as well as sufficient intratumoral radiotherapy. Advanced cancers sustain growth in the
accumulation of the photosensitizing agent.16,159 The hypoxic microenvironment by adapting. ROS play a duel
effectiveness of PDT is limited by insufficient photosensi- role in tumor growth. Initially ROS aid tumor progression
tizer reaching poorly perfused tumors. HBO may improve via DNA damage and uncontrolled proliferation of a
the effects of PDT by improving both tumor perfusion and genomically unstable and highly aggressive cell line. In
increasing the amount of singlet oxygen. excess however, ROS are toxic to tumor cells. The
Significantly improved oxygen tension and tumor cell effectiveness of conventional therapies is limited by the
kill was observed with combined HBO/PDT therapy in two presence of hypoxia.
studies conducted on C3H mice implanted with mammary In theory, the use of HBO in an adjuvant setting is
adenocarcinoma.16,159 Promising results were found in a justified by the following: Improved oxygenation im-
pilot study investigating the combined effect of HBO and proves drug delivery to hypoxic regions in the tumor.
PDT on patients with advanced inoperable esophageal HBO may remove the hypoxic stimulus that drives
carcinoma.157 Tumor load in the combined PDT/HBO angiogenesis. Improved oxygenation may also cause
2126 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

cells to enter a proliferative stage, thus sensitizing them 11. Henk JM, et al. Radiotherapy and hyperbaric oxygen in
to radiotherapy and certain chemotherapy. Increasing head and neck cancer: final report of first controlled clinical
intratumoral ROS levels beyond the threshold may trial. Lancet 1977;2:101–103.
induce tumor destruction. It is apparent that the effect of 12. Petre PM, et al. Hyperbaric oxygen as a chemotherapy
adjuvant in the treatment of metastatic lung tumors in a rat
HBO is dependent on the tumor’s type and stage and
model. J Thorac Cardiovasc Surg 2003;125:85–95.
the HBO treatment regimen. Most of the literature indi-
13. Mayer R, et al. Hyperbaric oxygen and radiotherapy.
cates that HBO has no impact on tumor growth—be it
Strahlenther Onkol. 2005;181:113–123.
stimulatory or inhibitory. The most convincing effects are 14. DeCosse JJ, et al. Influence of high-pressure oxygen and
observed when HBO is used in an adjuvant setting, but chemotherapy on the AMel 4 hamster melanoma. Cancer
this is specific to the tumor’s type and stage. HBO Res 1966;26:287–292.
therefore remains ineffective as a stand-alone therapy or 15. Takiguchi N, et al. Use of 5-FU plus hyperbaric oxygen for
even as a reliable adjuvant. Variability among investi- treating malignant tumors: evaluation of antitumor effect
gation techniques at various centers makes it difficult to and measurement of 5-FU in individual organs. Cancer
completely write off HBO as a potential therapeutic Chemother Pharmacol 2001;47:11–14.
adjuvant. Further research may be warranted pending 16. Huang Z, et al. Hyperoxygenation enhances the tumor cell
outcomes of the B14 Committee in evaluating the killing of photofrin-mediated photodynamic therapy. Pho-
tochem Photobiol 2003;78:496–502.
adjunctive potential of HBO with radiotherapy. Further-
17. Moulder JE, et al. Tumor hypoxia: its impact on cancer
more, consideration should be given as to the cost
therapy. Cancer Metastasis Rev 1987;5:313–341.
involved in such combined therapy against the extent of
18. Knisely JP, et al. Importance of hypoxia in the biology and
benefit that can be achieved. treatment of brain tumors. Neuroimaging Clin N Am
2002;12:525–536.
19. Puffer HW, et al. Preliminary observations of oxygen levels
REFERENCES in microcirculation of tumors in C3H mice. Adv Exp Med
Biol 1976;75:605–610.
1. Vaupel P, et al. Blood flow, oxygen consumption and tis- 20. Kizaka-Kondoh S, et al. Tumor hypoxia: a target for
sue oxygenation of human tumors. Adv Exp Med Biol selective cancer therapy. Cancer Sci 2003;94:1021–1028.
1990;277:895–905. 21. Brizel DM, et al. Tumor hypoxia adversely affects the
2. Tandara AA, et al. Oxygen in wound healing—more than a prognosis of carcinoma of the head and neck. Int J Radiat
nutrient. World J Surg 2004;28:294–300. Oncol Biol Physics 1997;38:285–289.
3. Grolman RE, et al. Transcutaneous oxygen measure- 22. Dunst J, et al. Tumor hypoxia and systemic levels of
ments predict a beneficial response to hyperbaric oxygen vascular endothelial growth factor (VEGF) in head and
therapy in patients with nonhealing wounds and critical neck cancers. Strahlenther Onkol 2001;177:469–473.
limb ischemia. Am Surg 2001;67:1072–1080. 23. Hockel M, et al. Hypoxic cervical cancers with low apop-
4. Hammarlund C, et al. Hyperbaric oxygen treatment of totic index are highly aggressive. Cancer Res 1999;
healthy volunteers with U.V.-irradiated blister wounds. 59:4525–4528.
Burns 1991;17:296–301. 24. Cvetkovic D, et al. Increased hypoxia correlates with in-
5. Thom SR, et al. Delayed neuropsychologic sequelae after creased expression of the angiogenesis marker vascular
carbon monoxide poisoning: prevention by treatment with endothelial growth factor in human prostate cancer. Urol-
hyperbaric oxygen. Ann Emerg Med 1995;25:474–480. ogy 2001;57:821–825.
6. Weaver LK, et al. Hyperbaric oxygen for acute carbon 25. Shi Q, et al. Constitutive and inducible interleukin 8
monoxide poisoning. N Engl J Med 2002;347:1057–1067. expression by hypoxia and acidosis renders human pan-
7. Dean BS, et al. Coma reversal with cerebral dysfunction creatic cancer cells more tumorigenic and metastatic. Clin
recovery after repetitive hyperbaric oxygen therapy for Cancer Res 1999;37113721.
severe carbon monoxide poisoning. Am J Emerg Med 26. Cuisnier O, et al. Chronic hypoxia protects against gam-
1993;11:616–618. ma-irradiation-induced apoptosis by inducing bcl-2 up-
8. Hawkins M, et al. Severe carbon monoxide poisoning: regulation and inhibiting mitochondrial translocation and
outcome after hyperbaric oxygen therapy. Br J Anaesth conformational change of bax protein. Int J Oncol
2000;84:584–586. 2003;23:1033–1041.
9. Durmaz E, et al. Carbon monoxide poisoning and hyper- 27. Vaupel P, et al. Tumor oxygenation and its relevance to
baric oxygen therapy. Br J Nurs 1999;8:1067–1072. tumor physiology and treatment. Adv Exp Med Biol
10. Wilkinson D, et al. Hyperbaric oxygen treatment and sur- 2003;510:45–49.
vival from necrotizing soft tissue infection. Arch Surg 28. Vaupel P, et al. Tumor hypoxia and malignant progression.
2004;139:1339–1345. Methods Enzymol 2004;381:335–354.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2127

29. Hockel M, et al. Association between tumor hypoxia and 46. Shweiki D, et al. Vascular endothelial growth factor
malignant progression in advanced cancer of the uterine induced by hypoxia may mediate hypoxia-initiated angio-
cervix. Cancer Res 1996;56:4509–4515. genesis. Nature 1992;359:843–845.
30. Bachtiary B, et al. Overexpression of hypoxia-inducible 47. Plate KH, et al. Vascular endothelial growth factor is a
factor 1alpha indicates diminished response to radiother- potential tumour angiogenesis factor in human gliomas in
apy and unfavorable prognosis in patients receiving radical vivo. Nature 1992;359:845–848.
radiotherapy for cervical cancer. Clin Cancer Res 48. White FC, et al. VEGF mRNA is reversibly stabilized by
2003;9:2234–2240. hypoxia and persistently stabilized in VEGF-overexpress-
31. Hashizume H, et al. Openings between defective endo- ing human tumor cell lines. Growth Factors 1995;12:289–
thelial cells explain tumor vessel leakiness. Am J Pathol 301.
2000;156:1363–1380. 49. Levy AP, et al. Transcriptional regulation of the rat vas-
32. Galmarini CM, et al. Multidrug resistance in cancer ther- cular endothelial growth factor gene by hypoxia. J Biol
apy: role of the microenvironment. Curr Opin Invest Drugs Chem 1995;270:13333–13340.
2003;4:1416–1421. 50. Stein I, et al. Translation of vascular endothelial growth
33. Chaplin DJ, et al. Intermittent blood flow in a murine tumor: factor mRNA by internal ribosome entry: implications for
radiobiological effects. Cancer Res 1987;47:597–601. translation under hypoxia. Mol Cell Biol 1998;18:3112–
34. Sakamoto M, et al. Phenotype changes in tumor vessels 3119.
associated with the progression of hepatocellular carci- 51. Schoch HJ, et al. Hypoxia-induced vascular endothelial
noma. Jpn J Clin Oncol 1993;23:98–104. growth factor expression causes vascular leakage in the
35. Toi M, et al. Association of vascular endothelial growth brain. Brain 2002;125:2549–2557.
factor expression with tumor angiogenesis and with early 52. Iyer NV, et al. Cellular and developmental control of O2
relapse in primary breast cancer. Jpn J Cancer Res homeostasis by hypoxia-inducible factor 1 alpha. Genes
1994;85:1045–1049. Dev 1998;12:149–162.
36. Duque JL, et al. Plasma levels of vascular endothelial 53. Wang GL, et al. Hypoxia-inducible factor 1 is a basic-helix-
growth factor are increased in patients with metastatic loop-helix-PAS heterodimer regulated by cellular O2 ten-
prostate cancer. Urology 1999;54:523–527. sion. Proc Natl Acad Sci USA 1995;92:5510–5514.
37. Karayiannakis AJ, et al. Serum vascular endothelial 54. Tang N, et al. Loss of HIF-1a in endothelial cells disrupts a
growth factor levels in pancreatic cancer patients correlate hypoxia-driven VEGF autocrine loop necessary for
with advanced and metastatic disease and poor prognosis. tumorigenesis. Cancer Cell 2004;6:485–495.
Cancer Lett 2003;194:119–124. 55. Forsythe JA, et al. Activation of vascular endothelial
38. Jacobsen J, et al. Vascular endothelial growth factor as growth factor gene transcription by hypoxia-inducible fac-
prognostic factor in renal cell carcinoma. J Urol 2000;163: tor 1. Mol Cell Biol 1996;16:4604–4613.
343–347. 56. Semenza GL, et al. ‘The metabolism of tumours’: 70 years
39. Akbulut H, et al. Prognostic role of serum vascular endo- later. Novartis Found Symp 2001;240:251–264.
thelial growth factor, basic fibroblast growth factor and 57. Kuwabara K, et al. Hypoxia-mediated induction of acidic/
nitric oxide in patients with colorectal carcinoma. Cytokine basic fibroblast growth factor and platelet-derived growth
2002;20:184–190. factor in mononuclear phagocytes stimulates growth of
40. Ikeda E, et al. Hypoxia-induced transcriptional activation hypoxic endothelial cells. Proc Natl Acad Sci USA
and increased mRNA stability of vascular endothelial 1995;92:4606–4610.
growth factor in C6 glioma cells. J Biol Chem 1995; 58. Hartmann A, et al. Hypoxia-induced up-regulation of an-
270:19761–19766. giogenin in human malignant melanoma. Cancer Res
41. Ferrara N, et al. The biology of vascular endothelial growth 1999;59:1578–1583.
factor. Endocr Rev 1997;18:4–25. 59. Chavey C, et al. IL-8 is a novel marker for breast cancer.
42. Leung DW, et al. Vascular endothelial growth factor is a Third Int Symp Mol Biol Breast Cancer 2005;7(Suppl 2).
secreted angiogenic mitogen. Science 1989;246:1306– 60. Benoy IH, et al. Increased serum interleukin-8 in patients
1309. with early and metastatic breast cancer correlates with
43. Horiuchi A, et al. Hypoxia-induced changes in the early dissemination and survival. Clin Cancer Res
expression of VEGF, HIF-1 alpha and cell cycle-related 2004;10:7157–7162.
molecules in ovarian cancer cells. Anticancer Res 61. Brown NS, et al. Thymidine phosphorylase induces car-
2002;22:2697–2702. cinoma cell oxidative stress and promotes secretion of
44. Rofstad EK, et al. Hypoxia-induced angiogenesis and angiogenic factors. Cancer Res 2000;60:6298–6302.
vascular endothelial growth factor secretion in human 62. Kunz M, et al. Anoxia-induced up-regulation of interleukin-
melanoma. Br J Cancer 1998;77:897–902. 8 in human malignant melanoma: a potential mechanism
45. Ziemer LS, et al. Hypoxia and VEGF mRNA expression for high tumor aggressiveness. Am J Pathol 1999;155:
in human tumors. Neoplasia 2001;3:500–508. 753–763.
2128 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

63. Giri D, et al. Interleukin-8 is a paracrine inducer of fibro- 79. Minamino T, et al. Hypoxia extends the life span of
blast growth factor 2, a stromal and epithelial growth factor vascular smooth muscle cells through telomerase activa-
in benign prostatic hyperplasia. Am J Pathol 2001;159: tion. Mol Cell Biol 2001;3336–3342.
139–147. 80. Ravi R, et al. Regulation of tumor angiogenesis by p53-
64. D’Agnano I, et al. DNA ploidy, proliferative index, and induced degradation of hypoxia-inducible factor 1alpha.
epidermal growth factor receptor: expression and prog- Genes Dev 2000;14:34–44.
nosis in patients with gastric cancers. Lab Invest 1995;72: 81. Koukourakis MI, et al. Hypoxia inducible factor (HIF-1a
432–438. and HIF-2a) expression in early esophageal cancer and
65. Van Schaeybroeck S, et al. Epidermal growth factor response to photodynamic therapy and radiotherapy.
receptor activity determines response of colorectal cancer Cancer Res 2001;61:1830–1832.
cells to gefitinib alone and in combination with chemo- 82. Hockenbery D, et al. Bcl-2 is an inner mitochondrial
therapy. Clin Cancer Res 2005;11:7480–7489. membrane protein that blocks programmed cell death.
66. Oh MJ, et al. Detection of epidermal growth factor receptor Nature 1990;348:334–336.
in the serum of patients with cervical carcinoma. Clin 83. Lowe SW, et al. Apoptosis in cancer. Carcinogenesis
Cancer Res 2000;6:4760–4763. 2000;21:485–495.
67. Gazzaniga P, et al. Detection of epidermal growth factor 84. Warburg O, et al. On growth of cancer cells in media in
receptor mRNA in peripheral blood: a new marker of cir- which glucose is replaced by galactose. Hoppe Seylers Z
culating neoplastic cells in bladder cancer patients. clin Physiol Chem 1967;348:1686–1687.
Cancer Res 2001;7:577–583. 85. Kondo Y, et al. Over expression of hypoxia-inducible
68. Graeber TG, et al. Hypoxia-mediated selection of cells factor-1alpha in renal and bladder cancer cells in-
with diminished apoptotic potential in solid tumours. Nat- creases tumorigenic potency. J Urol 2005;173:1762–
ure 1996;379:88–91. 1766.
69. Shaw P, et al. Induction of apoptosis by wild-type p53 in a 86. Semenza GL, et al. Transcriptional regulation of genes
human colon tumor-derived cell line. Proc Natl Acad Sci encoding glycolytic enzymes by hypoxia-inducible factor 1.
USA 1992;89:4495–4499. J Biol Chem 1994;269:23757–23763.
70. Brown NS, et al. Hypoxia and oxidative stress in breast 87. Webster KA, et al. Coordinate reciprocal trends in glyco-
cancer; oxidative stress: its effects on the growth, meta- lytic and mitochondrial transcript accumulations during the
static potential and response to therapy of breast cancer. in vitro differentiation of human myoblasts. J Cell Physiol
Breast Cancer Res 2001;3:323–327. 1990;142:566–573.
71. Bergh J, et al. Complete sequencing of the p53 gene 88. Maxwell PH, et al. Hypoxia-inducible factor-1 modulates
provides prognostic information in breast cancer patients, gene expression in solid tumors and influences both
particularly in relation to adjuvant systemic therapy and angiogenesis and tumor growth. Proc Natl Acad Sci USA
radiotherapy. Nat Med 1995;1:1029–1034. 1997;94:8104–8109.
72. Havrilesky L, et al. Prognostic significance of p53 mutation 89. Bustamante E, et al. High aerobic glycolysis of rat hepa-
and p53 overexpression in advanced epithelial ovarian toma cells in culture: role of mitochondrial hexokinase.
cancer: a Gynecologic Oncology Group study. J Clin Proc Natl Acad Sci USA 1977;74:3735–3739.
Oncol 2003;21:3814–3825. 90. Mathupala SP, et al. Glucose catabolism in cancer cells.
73. Baker SJ, et al. Chromosome 17 deletions and p53 gene Isolation, sequence, and activity of the promoter for type II
mutations in colorectal carcinomas. Science 1989; hexokinase. J Biol Chem 1995;270:16918–16925.
244:217–221. 91. Lu H, et al. Hypoxia-inducible factor 1 activation by aerobic
74. Chadeneau C, et al. Telomerase activity associated with glycolysis implicates the Warburg effect in carcinogene-
acquisition of malignancy in human colorectal cancer. sis: oncogenes in tumor metabolism, tumorigenesis, and
Camcer Res 1995;55:2533–2536. apoptosis. J Biol Chem 2002;277:23111–23115.
75. Oishi T, et al. Alteration of telomerase activity associated 92. Dang CV, et al. Oncogenes in tumor metabolism, tumori-
with development and extension of epithelial ovarian genesis, and apoptosis. J Bioenerg Biomembr 1997;29:
cancer. Obstet Gynecol 1998;91:568–571. 345–354.
76. Kuniyasu H, et al. Expression of human telomerase RNA 93. Elstrom RL, et al. Akt stimulates aerobic glycolysis in
is an early event of stomach carcinogenesis. Jpn J Cancer cancer cells. Cancer Res 2004;64:3892–3899.
Res 1997;88:103–107. 94. Cohen G, et al. Glutathione peroxidase: the primary agent
77. Kumaki F, et al. Telomerase activity and expression of for the elimination of hydrogen peroxide in erythrocytes.
human telomerase RNA component and human telomer- Biochemistry 1963;2:1420–1428.
ase reverse transcriptase in lung carcinomas. Hum Pathol 95. Talior I, et al. Increased glucose uptake promotes oxida-
2001;188–195. tive stress and PKC-delta activation in adipocytes of ob-
78. Hiyama K, et al. Telomerase activity in small-cell and non- ese, insulin-resistant mice. Am J Physiol Endocrinol Metab
small-cell lung cancers. J Natl Cancer Inst 1995;895–902. 2003;285:E295–E302.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2129

96. McMullin MF. The molecular basis of disorders of red cell 118. Zhou S, et al. Doxorubicin-induced persistent oxidative
enzymes. J Clin Pathol 1999;52:241–244. stress to cardiac myocytes. Toxicol Lett 2001;121:151–
97. Aw TY. Cellular redox: a modulator of intestinal epithelial 157.
cell proliferation. News Physiol Sci 2003;18:201–204. 119. Gackowski D, et al. Persistent oxidative stress in colo-
98. Kamata H, et al. Redox regulation of cellular signalling. rectal carcinoma patients. Int J Cancer 2002;101:395–
Cell Signal 1999;11:1–14. 397.
99. Benhar M, et al. ROS, stress-activated kinases and stress 120. Goda N, et al. Hypoxia-inducible factor 1alpha is essential
signaling in cancer. EMBO Rep 2002;3:420–425. for cell cycle arrest during hypoxia. Mol Cell Biol 2003;23:
100. Blokhina O, et al. Antioxidants, oxidative damage and 359–369.
oxygen deprivation stress: a review. Ann bot 2003; 121. Baish JW, et al. Role of tumor vascular architecture in
91(Spec No):179–194. nutrient and drug delivery: an invasion percolation-based
101. Sharkey S. Current indications for hyperbaric oxygen network model. Microvasc Res 1996;51:327–346.
therapy. Austr Defence Force Health 2000;1:64–72. 122. Alagoz T, et al. Evaluation of hyperbaric oxygen as a
102. Jackson AL, et al. The contribution of endogenous sources chemosensitizer in the treatment of epithelial ovarian
of DNA damage to the multiple mutations in cancer. Mutat cancer in xenografts in mice. Cancer 1995;75:2313–
Res 2001;477:7–21. 2322.
103. Szatrowski TP, et al. Production of large amounts of 123. Thom SR. Hyperbaric Oxygen Therapy: A Committee
hydrogen peroxide by human tumor cells. Cancer Res Report. Bethesda, Undersea and Hyperbaric Medical
1991;51:794–798. Society, 1992;20814.
104. Wiseman H, et al. Damage to DNA by reactive oxygen and 124. Zamboni WA, et al. The effect of acute hyperbaric oxygen
nitrogen species: role in inflammatory disease and pro- therapy on axial pattern skin flap survival when adminis-
gression to cancer. Biochem J. 1996;313( Pt 1):17–29. tered during and after total ischemia. J Reconstr Microsurg
105. Kuhn MA. Oxygen free radicals and antioxidants. AJN Am 1989;5:343–350.
J Nurs 2003;103:58–62. 125. Erdmann D, et al. Skin allograft rejection and hyperbaric
106. Laurent A, et al. Controlling tumor growth by modulating oxygen treatment in immune-histoincompatible mice.
endogenous production of reactive oxygen species. Can- Undersea Hyperb Med 1995;22:395–399.
cer Res 2005;65:948–956. 126. Granowitz EV, et al. Exposure to increased pressure or
107. Behrend L, et al. Reactive oxygen species in oncogenic hyperbaric oxygen suppresses interferon-gamma secre-
transformation. Biochem Soc Trans 2003;31:1441–1444. tion in whole blood cultures of healthy humans. Undersea
108. Arnold RS, et al. Hydrogen peroxide mediates the cell Hyperb Med 2002;29:216–225.
growth and transformation caused by the mitogenic oxi- 127. Marx RE, et al. Relationship of oxygen dose to angio-
dase Nox1. Proc Natl Acad Sci USA 2001;98:5550–5555. genesis induction in irradiated tissue. Am J Surg 1990;
109. Suh YA, et al. Cell transformation by the superoxide- 160:519–524.
generating oxidase Mox1. Nature 1999;401:79–82. 128. Meltzer T, et al. The effect of hyperbaric oxygen on the
110. Shen H, et al. Importance of glutathione and associated bursting strength and rate of vascularization of skin
enzymes in drug response. Oncol Res 1997;9:295–302. wounds in the rat. Am Surg 1986;52:659–662.
111. Harrison LB, et al. Impact of tumor hypoxia and anemia on 129. Feldmeier J, et al. Hyperbaric oxygen: does it promote
radiation therapy outcomes. Oncologist 2002;7:492–508. growth or recurrence of malignancy? Undersea Hyperb
112. Kaelin CM, et al. The effects of hyperbaric oxygen on free Med 2003;30:1–18.
flaps in rats. Arch Surg 1990;125:607–609. 130. Hunt TK. The physiology of wound healing. Ann Emerg
113. Kong Q, et al. A threshold concept for cancer therapy. Med Med 1988;17:1265–1273.
Hypotheses 2000;55:29–35. 131. Marx RE, et al. Studies in the radiobiology of osteoradi-
114. Hileman EO, et al. Intrinsic oxidative stress in cancer cells: onecrosis and their clinical significance. Oral Surg Oral
a biochemical basis for therapeutic selectivity. Cancer Med Oral Pathol 1987;64:379–390.
Chemother Pharmacol 2004;53:209–219. 132. Marx RE, Johnson RP. Problem wounds in oral and
115. Portakal O, et al. Coenzyme Q10 concentrations and maxillofacial surgery: the role of hyperbaric oxygen. In:
antioxidant status in tissues of breast cancer patients. Clin Davis IC, Hunt TK, editors, Problem Wounds: the Role of
Biochem 2000;33:279–284. Oxygen. New York, Elsevier, 1988; 65–123.
116. Choi AM, et al. Heme oxygenase-1: function, regulation, 133. Phillips SJ.. Physiology of wound healing and surgical
and implication of a novel stress-inducible protein in oxi- wound care. ASAIO J 2000;46:S2–S5.
dant-induced lung injury. Am J Respir Cell Mol Biol 134. Conconi MT, et al. Effects of hyperbaric oxygen on pro-
1996;15:9–19. liferative and apoptotic activities and reactive oxygen
117. Toyokuni S, et al. Persistent oxidative stress in cancer. species generation in mouse fibroblast 3T3/J2 cell line.
FEBS Lett 1995;358:1–3. J Invest Med 2003;51:227–232.
2130 Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy

135. Lian QL, et al. Effects of hyperbaric oxygen on S-180 International Congress on Hyperbaric Medicine. Aber-
sarcoma in mice. Undersea Hyperb Med 1995;22:153– deen, Scotland, University Press, 1977;253–258.
160. 154. Henk JM. Late results of a trial of hyperbaric oxygen and
136. Johnson RJR, Lauchlan SC. Epidermoid carcinoma of the radiotherapy in head and neck cancer: a rationale for
cervix treated by 60Co therapy and hyperbaric oxygen. In: hypoxic cell sensitizers? Int J Radiat Oncol Biol Phys
Proceedings of the third International Congress on 1986;12:1339–1341.
Hyperbaric Medicine, 1966;648–652. 155. Sealy R, et al. Irradiation with misonidazole and hyperbaric
137. Shewell J, et al. The effect of hyperbaric oxygen treatment oxygen: final report on a randomized trial in advanced
on pulmonary metastasis in the C3H mouse. Eur J Cancer head and neck cancer. Int J Radiat Oncol Biol Phys
(Oxf) 1980;16:253–259. 1986;12:1343–1346.
138. McMillan T, et al. The effect of hyperbaric oxygen on oral 156. Granstrom G. Hyperbaric oxygen therapy decreases the
mucosal carcinoma. Laryngoscope 1989;99:241–244. rejection rate of osseointegrated implants after radiother-
139. Valaitis J, et al. Effect of hyperbaric oxygen and nitrogen apy. Strahlenther Onkol 1996;172:20–21.
mustard (NSC-762) on Ehrlich ascites tumor. Cancer 157. Maier A, et al. Combined photodynamic therapy and
Chemother Rep 1968;52(Pt 1):405–412. hyperbaric oxygenation in carcinoma of the esophagus
140. Cade IS, et al. Megavoltage radiotherapy in hyperbaric and the esophago-gastric junction. Eur J Cardiothorac
oxygen: a controlled trial. Cancer 1967;20:817–821. Surg 2000;18:649–655.
141. Eltorai I, et al. Does hyperbaric oxygenation provoke an 158. Gray LH, et al. The concentration of oxygen dissolved in
occult carcinoma in man? In: Proceedings of the VIII tissue at the time or irradiation as factor in radiotherapy.
International Conference on Hyperbaric Medicine, North Br J Radiol 1953;26:638.
Carolina, 1987;18–27. 159. Chen Q, et al. Improvement of tumor response by
142. Bean JW, et al. Reaction of Ehrlich ascites cells in expo- manipulation of tumor oxygenation during photodynamic
sure to oxygen at high pressure. Cancer Res 1966; therapy. Photochem Photobiol 2002;76:197–203.
26:2380–2385. 160. Teicher BA, et al. Classification of antineoplastic agents by
143. McCredie JA, et al. Effects of hyperbaric oxygen on growth their selective toxicities toward oxygenated and hypoxic
and metastases of the C3HBA tumor in the mouse. Can- tumor cells. Cancer Res 1981;41:73–81.
cer 1966;19:1537–1542. 161. Evans JC. Metastasis following radiotherapy in hyperbaric
144. Suit HD, et al. Effect of daily exposure to high pressure oxygen. Radiology 1969;93:1155–1157.
oxygen on tumor growth. Am J Roentgenol Radium Ther 162. Martin DF, et al. Enhancement of tumor radiation
Nucl Med 1966;97:1019–1022. response by the combination of a perfluorochemical
145. Johnson RE, et al. Hyperbaric oxygen effect on experi- emulsion and hyperbaric oxygen. Int J Radiat Oncol Biol
mental tumor growth. Radiology 1967;88:775–777. Phys 1987;747–751.
146. Feder BH, et al. The effect of hyperbaric oxygen on pul- 163. Frid IA, et al. Effects of hyperbaric oxygenation on tumor
monary metastases in C3H mice. Radiology 1968; growth. Vopr Onkol 1989;35:970–973.
90:1181–1184. 164. Granstrom G, Westin T, Lyden E, Bengt C, Magnusson BG,
147. Johnson RJR, et al. The effect of hyperbaric oxygen on Edstrom S. Hyperbaric oxygenation does not stimulate
tumor metastases in mice. Clin Radiol 1971;22:538–540. experimental tumour growth. In: Proceedings from the
148. Mestrovic J, et al. Suppression of rat tumor colonies in the XVIth EUBS meeting, Amsterdam, 1990;121–129.
lung by oxygen at high pressure is a local effect. Clin Exp 165. Headley DB, et al. The effect of hyperbaric oxygen on
Metastasis 1990;8:113–119. growth of human squamous cell carcinoma xenografts.
149. Dettmer CM, et al. The effect of increased oxygen ten- Arch Otolaryngol Head Neck Surg 1991;117:1269–1272.
sions upon animal tumor growth. Am J Roentgenolo 166. Sklizovic D, et al. Hyperbaric oxygen therapy and squa-
Radium Ther Nucl Med 1968;102:804–810. mous cell carcinoma cell line growth. Head Neck
150. Bradfield JJ, et al. Rapid progression of head and neck 1993;15:236–240.
squamous carcinoma after hyperbaric oxygenation. 167. McDonald KR, et al. Effect of hyperbaric oxygenation on
Otolaryngol Head Neck Surg 1996;114:793–797. existing oral mucosal carcinoma. Laryngoscope 1996;106:
151. Van den Brenk HA, et al. An analysis of the progression 957–959.
and development of metastases in patients receiving 168. Shi Y, et al. Effects of hyperbaric oxygen exposure on
x-radiation in hyperbaric oxygen. Clin Radiol 1967;18:54– experimental head and neck tumor growth, oxygenation,
61. and vasculature. Head Neck 2005;27:362–369.
152. Dische S. Hyperbaric oxygen: the Medical Research 169. Johnson RJ, et al. Sequential study on the effect of the
Council trials and their clinical significance. Br J Radiol addition of hyperbaric oxygen on the 5 year survival rates
1978;51:888–894. of carcinoma of the cervix treated with conventional frac-
153. Perrins DJD, Wiernik G. Controlled trials in carcinoma of tional irradiations. Am J Roentgenol Radium Ther Nucl
the bladder. In: Smith G, editor, Proceedings of the Sixth Med 1974;120:111–117.
Daruwalla and Christophi: Hyperbaric Oxygen Therapy for Malignancy 2131

170. Bennett MB, Sealy R, Hockly J. The treatment of stage III 180. Horsman M, et al. The Oxygen Effect: Basic Clinical
squamous cell carcinoma of the cervix in air and in Radiobiology, 2nd edition. London, Arnold, 1997.
hyperbaric oxygen. In: Smith G, editor, Proceedings of the 181. Overgaard J, et al. Modification of hypoxia-induced ra-
Sixth International Congress on Hyperbaric Oxygen. dioresistance in tumors by the use of oxygen and sensi-
Aberdeen, Scotland, University press, 1977;247–252. tizers. Semin Radiat Oncol 1996;6:10–21.
171. Henk JM, et al. Radiotherapy and hyperbaric oxygen in 182. Lartigau E, at al. Hyperbaric oxygen in the treatment of
head and neck cancer: interim report of second clinical radio-induced lesions in normal tissues. Presented to the
trial. Lancet 1977;2:104–105. European Society for Therapuetic Radiology and Oncol-
172. Watson ER, et al. Hyperbaric oxygen and radiotherapy: a ogy and European Committee for Hyperbaric Medicine,
Medical Research Council trial in carcinoma of the cervix. Lisbon, 2001.
Br J Radiol 1978;51:879–887. 183. Kalns J, et al. The effect of hyperbaric oxygen on growth
173. Brady LW, et al. Hyperbaric oxygen therapy for carcinoma and chemosensitivity of metastatic prostate cancer. Anti-
of the cervix—stages IIB, IIIA, IIIB and IVA: results of a cancer Res 1998;18:363–367.
randomized study by the Radiation Therapy Oncology 184. Stuhr LE, et al. Hyperbaric oxygen alone or combined with
Group. Int J Radiat Oncol Biol Phys 1981;7:991–998. 5-FU attenuates growth of DMBA-induced rat mammary
174. Granstrom G. Tumor recurrence and development of new tumors. Cancer Lett 2004;210:35–40.
head and neck cancers after HDO2—treatment a pro- 185. Narkowicz CK, et al. Hyperbaric oxygen therapy increases
spective clinical study. In: Proceedings: International Joint free radical levels in the blood of humans. Free Radic Res
Meeting on Hyperbaric and Underwater Medicine, Milan, Commun 1993;19:71–80.
1996;47–60. 186. Dennog C, et al. Analysis of oxidative DNA damage and
175. Dische S, et al. Carcinoma of the cervix and the use of HPRT mutations in humans after hyperbaric oxygen
hyperbaric oxygen with radiotherapy: a report of a rando- treatment. Mutat Res 1999;431:351–359.
mised controlled trial. Radiother Oncol 1999;53:93–98. 187. Kalns JE, et al. Exposure to hyperbaric oxygen induces
176. Haffty BG, et al. Radiation therapy with hyperbaric oxygen cell cycle perturbation in prostate cancer cells. In Vitro Cell
at 4 atmospheres pressure in the management of squa- Dev Biol Anim 1999;35:98–101.
mous cell carcinoma of the head and neck: results of a 188. Teas J, et al. Can hyperbaric oxygen therapy reduce
randomized clinical trial: carcinoma of the larynx treated breast cancer treatment-related lymphedema? A pilot
with hypofractionated radiation and hyperbaric oxygen: study. J Womens Health (Larchmt) 2004;13:1008–1018.
long-term tumor control and complications. Cancer J Sci 189. Yarnold J. Phase II randomized study of hyperbaric oxy-
Am 1999;5:341–347. gen therapy versus standard management in women with
177. Haffty BG, et al. Carcinoma of the larynx treated with chronic arm lymphedema after radiotherapy for early
hypofractionated radiation and hyperbaric oxygen: long- breast cancer. Clinical trial in progress, UK, 2004.
term tumor control and complications. Int J Radiat Oncol 190. Maier A, et al. Does hyperbaric oxygen enhance the effect
Biol Phys 1999;45:13–20. of photodynamic therapy in patients with advanced
178. Kohshi K, et al. Effects of radiotherapy after hyperbaric esophageal carcinoma? A clinical pilot study. Endoscopy
oxygenation on malignant gliomas. Br J Cancer 1999; 2000;32:42–48.
80:236–241. 191. Tomaselli , et al. Acute effects of combined photodynamic
179. Feldmeier JJ, et al. Hyperbaric oxygen an adjunctive therapy and hyperbaric oxygenation in lung cancer—a
treatment for delayed radiation injuries of the abdomen clinical pilot study. Lasers Surg Med 2001;28:399–403.
and pelvis. Undersea Hyperb Med 1996;23:205–213.

Das könnte Ihnen auch gefallen