Sunteți pe pagina 1din 28

Metabolic Oxidative Stress

and Cancer
Douglas R. Spitz, PhD

Free Radical and Radiation Biology Program


Department of Radiation Oncology
Holden Comprehensive Cancer Center
The University of Iowa, Iowa City, IA
What is life?
“An electron, given off easily, has a high energy level, while
an orbital which tends to take up an electron, has a low
energy level. The transmitted electron thus goes from a high
to a low level, and releases the energy which corresponds to
the difference of the two levels. It is this energy which drives
life.”
Albert Szent-Györgyi, Electronic Biology and Cancer, Marcel Dekker Inc. 1976

 In essence, Szent-Györgyi was one of the first scientists to


recognize that all of the forces necessary for the maintenance
of living systems derive from the ability of complex higher
order biological structures to extract, store, and move
electrons.
Why do we breath oxygen? The Chemiosmotic Hypothesis
Why is it potentially harmful?

Lehninger; Principles in
Biochemistry 3 rd Edition

Chemiosmotic model. In this simple representation of the chemiosmotic theory applied to mitochondria,
electrons from NADH and other oxidizable substrates pass through a chain of carriers arranged
asymmetrically in the inner membrane. Electron flow is accompanied by proton transfer across the
membrane, producing both a chemical gradient (pH) and an electrical gradient (). The inner
mitrochondrial membrane is impermeable to protons; protons can reenter the matrix only through proton
specific channels (F0). The proton-motive force that drives protons back into the matrix provides the energy
for ATP synthesis, catalyzed by the F1 complex associated with F0.
Intermembrane space
Complex IV
NADH
NAD+
e- O2 Cyt c

FMN Cyt c1
O2
Complex I
Fe S Fe S Q
-
Myxothiazol e
Rotenone
Cyt bL
Q QH2 QH2
Q
Succinate Cyt bH
QH2
Fumarate
e- Antimycin A
O2 Q
FAD Fe S Q Complex III

O2 Complex II
O2 O2

Matrix

Ahmad IM, Aykin-Burns N, Sim JE, Walsh SA, Higashikubo R, Buettner GR, Venkataraman S, Mackey MA,
Flanagan S, Oberley LW, and Spitz DR: Mitochondrial O2- and H2O2 mediate glucose deprivation-induced
cytotoxicity and oxidative stress in human cancer cells. J. Biol. Chem. 2005; 280(6):4254-4263.
The Circle of Life and The Circle of Death
What is the
relationship DNA Repair →
between
Replication
metabolic Differentiation
and genetic
Cell Cycle
processes Differentiation
Adaptive Responses
Cell Division
Differentiated Cell Functions

necessary
for life?
Spitz DR, Azzam Reactive  Antioxidants
EI, Li JJ, and Gius Species
D: Metabolic
oxidation/reduction
reactions and
cellular responses
to ionizing
radiation: a unifying
concept in stress
response biology.
Cancer and
Metastasis
Reviews 2004;
23:311–322.
Metabolic Theories of Cancer
Warburg O (1956) Science 123:309-314
 Cancer cells exhibit increased rates of glycolysis and slightly
decreased rates of respiration.
 It was proposed this was the result of “damage” to the respiratory
mechanism and tumor cells increased glycolysis to compensate for
this defect.

Weber G (1977) New Engl. J. Med. 296:541-551


 Cancer cells exhibit increased rates of pentose phosphate cycle
activity characterized by increases in glucose-6-phosphate
dehydrogenase activity.

Oberley LW, Oberley TD, and Buettner GR (1980 and 1981) Med.
Hypoth. 6:49-68; 7:21-42
 Tumor cells have aberrant respiration caused by a decrease in
MnSOD activity leading to increased steady state levels of superoxide
and hydrogen peroxide that causes DNA damage and activates
signaling pathways leading to uncontrolled growth, the inability to
differentiate, and the malignant phenotype.
Genetic Theory of Cancer
Bishop (1987) Science 235:305-311; Varmus (1987)
Science 238:1337-1339
 Cancer is a multi-step genetic disease in which
mutations resulting in the aberrant expression of
cellular homologues of oncogenes (i.e., Ras, c-Fos, c-
Jun, and c-Myc, etc.) associated with growth and
development as well as tumor suppressor genes (i.e.,
p53) gradually accumulate over time, eventually
resulting in immortalization, the loss of control of cell
proliferation, and progression to the malignant
phenotype.

How can we unify metabolic and genetic


theories of cancer?
In the mid-1990’s Dr. Yong J. Lee’s laboratory showed:
 Within minutes - glucose deprivation-induced activation of signal
transduction in (i.e., ERK1, ERK2, Lyn Kinase, JNK, Ras, AP-1, etc.) in
MCF-7/ADR human breast cancer cells.

 Within 2-4 hours - glucose deprivation-induced increases in steady


state levels of mRNA coding for bFGF and c-Myc in MCF-7/ADR.

 Within 4 hours - glucose deprivation-induced clonogenic cell killing


in MCF-7/ADR cells.
Molecular and Cellular Biochemistry, 155:163-171, 1996.
Journal of Cell Science, 110:681-686, 1997.
Molecular and Cellular Biochemistry, 170:23-30, 1997.
Journal of Biological Chemistry, 272:11690-11693, 1997.

 Thiswork supported the existence of a mechanistic link


between glucose metabolism, signal transduction, and
gene expression governing the malignant phenotype that
also governed the survival of cancer cells.
What is this mechanistic link?
 Dr. Lee and his collaborators then discovered
that all the effects of glucose deprivation in MCF-
7 ADR could be suppressed by treatment with
the thiol antioxidant, N-acetylcysteine, indicating
a causal role for oxidative stress in the effects of
glucose deprivation in cancer cells.

 Lee YJ, Galoforo SS, Berns CM, Chen JC, Davis BH, Sim JE, Corry PM, and
Spitz DR: Glucose deprivation-induced cytotoxicity and alterations in mitogen-
activated protein kinase activation are mediated by oxidative stress in multidrug-
resistant human breast carcinoma cells. J. Biol. Chem. 1998; 273:5294-5299.

 Blackburn RV, Spitz DR, Liu X, Galoforo SS, Sim JE, Ridnour LA, Chen JC,
Davis BH, Corry PM, and Lee YJ: Metabolic oxidative stress activates signal
transduction and gene expression during glucose deprivation in human tumor
cells. Free Radic. Biol. Med. 1999; 26:419-430.
 Theoretical model outlining a biochemical rationale for unifying
metabolic and genetic theories of cancer.
Spitz et al. (2000) Ann. N.Y. Acad. Sci. 899:349-362. ? = thioredoxin, glutaredoxin, and direct redox changes on proteins

Kinases (ie, Lyn, PKC, etc.)


G proteins (ie, Ras, etc.)
Kinases (ie, Raf, MEK, ERK, JNK, etc.) DNA
Phosphatases
RNA (ie, c-Fos,
Trxn. factors (ie, AP-1, etc.) c-Jun, c-Myc,
bFGF, etc.)
Glucose ?
NUCLEUS
PPC NADP+ GSH H 2O 2
Glucose 6-phosphate (TrxS2H2)
Glycolysis GR GPX (TrxPx) Proteins
H 2O 2 (TR)
Pyruvate NADPH GSSG H2 O
(TrxSS)
O2• H2 O 2
H2 O Site II 1e-
Site I Site III
Site IV

Fatty/Amino NADH
Acids TCA 4e-
H2O
FADH 2
O2

MITOCHONDRIA PLASMA
MEMBRANE
 Rodent tumor cells had reduced levels of MnSOD activity that were
proportional to increased growth rate and lack of ability to differentiate.

 Rodent Tumor cells demonstrated mitochondrial abnormalities that


were more pronounced in fast growing undifferentiated cancers.
Bize IB, Oberley LW, Morris HP (1980) Superoxide dismutase and superoxide radical in Morris
hepatomas. Cancer Res. 40(10):3686-93.

normal liver well differentiated undifferentiated


mitochondria hepatoma hepatoma
slow growing fast growing
 Human cancer cells demonstrated
mitochondrial abnormalities that are more
pronounced in more malignant tissues.
Springer EL: Comparative study of the cytoplasmic organelles of epithelial cell
lines derived from human carcinomas and nonmalignant tissues. Cancer Res.
1980; 40(3):803-17.

 The cytoplasmic organelles of 16 human cell types derived from normal


as well as from primary and metastatic carcinomas were characterized by
electron microscopy in a blinded fashion.

 Mitochondrial pleomorphisms was expressed slightly by normal cells and


to a much greater extent by all cell types derived from malignant tissues.

 These pleomorphisms were characterized by hypertrophied mitochondria


with longitudinal cristal arrangements in almost all the malignant cells lines,
but not in any lines derived from nonmalignant tissues of cancerous organs
or from normal tissues.
 Increasing mitochondrial superoxide
scavenging by increasing expression of
MnSOD alters the malignant phenotype of
cancer cells
Church SL, Grant JW, Ridnour LA, Oberley LW, Swanson PE, Meltzer PS, Trent JM:
Increased manganese superoxide dismutase expression suppresses the malignant
phenotype of human melanoma cells. Proc Natl Acad Sci U S A. 1993; 90(7):3113-7.

 Melanoma cell lines over expressing MnSOD cDNAs lost their ability to
form colonies in soft agar and tumors in nude mice.

 These findings were the first demonstration that stable over expression
MnSOD suppressed the malignant phenotype in human cancer cells

 These results suggested that increased steady-state levels of


superoxide and/or H2O2 significantly contributed to the maintenance of
the malignant phenotype in human cancer cells.
 Human cancer cells were reported to
produce large amounts of H2O2, relative to
normal cells
Szatrowski TP and Nathan CF: Production of large amounts of hydrogen peroxide
by human tumor cells. Cancer Res. 1991; 51(3):794-8.

 Seven human tumor cell lines demonstrated constitutively


elevated H2O2 such that cumulative amounts were comparable
to the amount of H2O2 produced by phorbol ester-activated
neutrophils.

 Constitutive generation of large amounts of reactive oxygen


intermediates by cancer cells, if it occurs in vivo, might
contribute to genomic instability, tumor heterogeneity, invasion,
and metastasis.
Fig 1
60
#, 
 Human colon carcinoma
Normalized Mean Fluoresscence Intensity
DHE
50 DHE + AntA # ,
cells demonstrate
#, 
(A)
40 increased steady-state
30
levels of superoxide
*
*
relative to normal human
20
colon epithelial cells and
*
10 fibroblasts
#
0
FHC HT29 SW480 HCT116
 Mitochondrial electron
* transport chain blockers
Normalized Mean Fluorescence Intensity

8 DHE
make this difference more
(B)
6
*
pronounced

4 *
 Aykin-Burns N, Ahmad IM, Zhu Y,
Oberley LW, and Spitz DR: Inhibition of
glucose metabolism causes enhanced
2
cytotoxicity in human cancer vs. normal
cells via metabolic oxidative stress.
0 Biochem. J. 2009; 418:29-37.
33Co HT29 SW480 HCT116
250
2.5
Normalized Mean Fluorescence Intensity

* HMEC
* MB231  Human breast
2.0
carcinoma cells
demonstrate
1.5 increased steady-
state levels of
1.0
superoxide and
hydroperoxides,
relative to normal
0.5
human breast
epithelial cells
0.0
DHE CDCFH2 CDCF

 Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, and Spitz DR: Inhibition of glucose
metabolism causes enhanced cytotoxicity in human cancer vs. normal cells via
metabolic oxidative stress. Biochem. J. 2009; 418:29-37.
1
(A)
 Human colon
Normalized Survival Fraction

0.1 *
and breast
carcinoma cells
0.01
* demonstrate
increased
*
0.001
sensitivity to
33Co glucose
HCT116
0.0001
deprivation-
1 (B) induced
Normalized Survival Fraction

cytotoxicity,
# relative to normal
# human cells
0.1
#

Aykin-Burns N, Ahmad IM, Zhu Y,


Oberley LW, and Spitz DR: Inhibition of
HMEC
glucose metabolism causes enhanced
MB231
cytotoxicity in human cancer vs. normal
0.01
0 24 48 72 96 cells via metabolic oxidative stress.
Time (h) Biochem. J. 2009; 418:29-37.
 Mitochondrial O2- and H2O2 mediate glucose deprivation-
induced toxicity and oxidative stress in human cancer cells.
(A)
Normalized Surviving Fraction

p<0.04 p<0.03 p<0.001 0.25 Bgl II / -Glu #

Normalized Surviving Fraction


AdMnSOD/ AdmitCAT/ -Glu
(relative to +Glu from each group)

100
0.20
* (A)

0.15
#
0.10

0.05

0.00
10-1
40
p<0.02 (B) (B)
48 h/ +Glu 30
48 h/ -Glu p<0.2
Bgl II/ +Glu
Bgl II/ -Glu
30 25 *
AdMnSOD/ AdmitCAT/ -Glu
(in GSH equivalents)

p<0.3
(nmole/ mg)
GSSG

20

20
* ,#

%GSSG
**
15

*
10 10

0
AdBgl II AdMnSOD AdBgl II AdMitCat AdBgl II AdMnSOD/ 0
(10 MOI) (10 MOI) (50 MOI) (50 MOI) (60 MOI) AdMitCat
(10/50 MOI) HCT116 MB231

 Ahmad IM, Aykin-Burns N, Sim JE, Walsh SA, Higashikubo R, Buettner GR, Venkataraman S, Mackey MA, Flanagan S, Oberley LW, and Spitz
DR: Mitochondrial O2- and H2O2 mediate glucose deprivation-induced cytotoxicity and oxidative stress in human cancer cells. J. Biol. Chem. 2005;
280(6):4254-4263.
(C) HCT116 MB231
MnSOD activity (U/mg protein)
 Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, and Spitz DR: Inhibition of glucose metabolism causes enhanced cytotoxicity in human cancer vs.
Bgl II 2.5 <2
normal cells via metabolic oxidative stress. Biochem. J. 2009; 418:29-37. mitoCAT/MnSOD 18.3 15.6
(numbe
FHC/ 2DG

100  Human colon carcinoma cells areFHC

Fig 6
(number of cells exclude dye/ total number)

more sensitive to cell killing by 2-


100

(number of cells exclude dye/ total number)


deoxyglucose (2DG), relative to
normal colon epithelial cells
Viability

Viability
 2DG-induced cytotoxicity can be
(B)
(A) inhibited by over expressing
HT29/ 2DG
FHC/ 2DG
FHC
mitochondrially
HT29 targeted catalase
and superoxide dismutase
0 24 48 72
100 Time (h)
(number of cells exclude dye/ total number)

1.4 HT29 (48 h)

Normalized Surviving Fraction


#
(C)
1.2 #
Viability

1.0

0.8 *
(B)
HT29/ 2DG 0.6

HT29
0.4
AdBgl II AdBgl II AdmitCAT AdmitCAT
0 24 48 72 Control 2DG AdMnSOD AdMnSOD
Time (h) Control 2DG

Aykin-Burns
1.4 HT29N, Ahmad IM, Zhu Y, Oberley (D)
(48 h) LW, and Spitz DR: Inhibition of glucose
g Fraction

# Bgl II mitoCAT/MnSO
metabolism
(C)
causes enhanced cytotoxicity inSOD
human cancer
activity (U/mg vs. normal2.0cells via
protein) 15.3
1.2 #
metabolic oxidative stress. Biochem. J. 2009; 418:29-37
CAT activity (mk/mg protein) 13.1 29.9
 Mutations in mtDNA encoded genes (ND6)
can lead to increased ROS and increased
metastatic potential that is suppressed by
ROS scavengers.
Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y,
Hayashi J: ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 2008;
320(5876):661-4.

 Cytoplasmic hybrid (cybrid) technology was used to replace endogenous


mtDNA in a mouse tumor cell line that was poorly metastatic with mtDNA
from a highly metastatic cell line and the recipient tumor cells acquired the
metastatic potential of the transferred mtDNA.

 The mtDNA conferring high metastatic potential contained mutations in the


gene encoding NADH (reduced form of nicotinamide adenine dinucleotide)
dehydrogenase subunit 6 (ND6) that was associated with overproduction of
reactive oxygen species (ROS).

 Pretreatment of the highly metastatic tumor cells with ROS scavengers


suppressed their metastatic potential in mice.
 Mutations in nuclear encoded genes that code for
electron transport chain proteins (SDH subunits B,
C, D) can also lead to increased steady-state levels
of superoxide, increased mutation rates, increased
genomic instability, and tumorigenesis in rodents
as well as being associated with neuro-endocrine
tumor formation in humans.
Ishii T, Yasuda K, Akatsuka A, Hino O, Hartman PS, Ishii N: A mutation in the SDHC gene of
complex II increases oxidative stress, resulting in apoptosis and tumorigenesis. Cancer Res
2005; 65(1):203-9.

Slane BG, Aykin-Burns N, Smith BJ, Kalen AL, Goswami PC, Domann FE, Spitz DR:
Mutation of succinate dehydrogenase subunit C results in increased O2-, oxidative stress, and
genomic instability. Cancer Res 2006; 66(15):7615-20.
Bardella C, Pollard PJ, Tomlinson I: SDH mutations in cancer. Biochim Biophys Acta 2011;
1807(11):1432-43.
Owens KM, Aykin-Burns N, Dayal D, Coleman MC, Domann FE, and Spitz DR: Genomic
instability induced by mutant succinate dehydrogenase subunit D (SDHD) is mediated by O2- and
H2O2. Free Radic Biol Med 2011; [Epub ahead of print] PMID: 22041456
Conclusions and Speculations:
 Cancer cells appear to exist in a condition of metabolic
oxidative stress characterized by increased steady-state
levels of superoxide and hydrogen peroxide.
 This increase in reactive oxygen species (ROS) appears
to be compensated for by increases in glucose and
hydroperoxide metabolism.
 The condition of metabolic oxidative stress in cancer cells
may be the result of alterations in mitochondrial oxidative
metabolism.
 Metabolic oxidative stress appears to contribute to
selective sensitivity of cancer vs. normal cells to glucose
deprivation-induced cytotoxicity and oxidative stress.
Kinases (ie
(ie,, Lyn, PKC, etc.)
G proteins (ie
(ie,, Ras, etc.)
Kinases (ie
(ie,, Raf, MEK, ERK, JNK, etc.) DNA
Phosphatases
RNA (ie
(ie,, c-
c-Fos,
Trxn.
Trxn. factors (ie
(ie,, AP-
AP-1, etc.) c-Jun, c-
c-Myc,
bFGF, etc.)
Glucose ?
NUCLEUS
PPC NADP+ GSH H 2O 2
Glucose 6-
6-phosphate (TrxS2H2)
Glycolysis GR GPX (TrxPx) Proteins
H 2O 2 (TR)
Pyruvate NADPH GSSG H2 O
(TrxSS)
O2• H2 O 2
H2 O Site II 1e-
Site I Site III
Site IV

Fatty/Amino NADH
Acids TCA 4e-
4e-
H2O
FADH 2
O2

MITOCHONDRIA PLASMA
MEMBRANE
SPECULATION
 Cancer might represent a constellation of metabolic and/or genetic diseases where the
common theme is uncoupling of normal cellular processes that govern cell growth and
differentiation caused by the inappropriate flow of electrons from metabolic
oxidation/reduction reactions to redox sensitive proteins governing signal transduction and
gene expression. (Spitz, et al. Ann. N.Y. Acad. Sci. 899:349-362, 2000)
Clinical Implications: Therapy
If glucose metabolism is increased in cancer cells to
compensate for excess hydroperoxide production
from mitochondrial respiration, then inhibiting glucose
and hydroperoxide metabolism while forcing cells to
derive energy from respiration should preferentially
kill cancer cells, relative to normal cells.

Combinations of agents designed to take advantage of


this hypothesis might include:
Inhibitors of Glucose Metabolism (ie., 2-deoxyglucose, etc.)
Inhibitors of the Pentose Phosphate Cycle (ie., DHEA, etc.)
Inhibitors of Hydroperoxide Metabolism (ie., BSO, AUR, BCNU)
Dietary Manipulation (high protein/fats, no carbohydrates)
Agents that Increase the Metabolic Production of Prooxidants
and Increase Oxidative DNA Damage (ie., Radiation, quinones,
Cisplatin, Paclitaxel, inflammatory mediators, etc.)
 Inhibitors of glucose and hydroperoxide metabolism selectively
(relative to normal cells) enhance cancer cell killing as well as
selectively sensitizing cancer cells to therapeutic agents.
 Lin X, Zhang F, Bradbury CW, Kaushal A, Li L, Spitz DR, Aft R, and Gius D: 2-Deoxy-d-glucose-induced cytotoxicity and
radiosensitization in tumor cells is mediated via disruptions in thiol metabolism. Cancer Res 2003; 63:3413–3417.

 Simons AL, Ahmad IM, Mattson DM, Dornfeld KJ, and Spitz DR: 2-Deoxy-D-glucose combined with cisplatin enhances
cytotoxicity via metabolic oxidative stress in human head and neck cancer cells. Cancer Res 2007; 67(7): 3364–70.

 Coleman MC, Asbury CR, Daniels D, Du J, Aykin-Burns N, Smith BJ, Li L, Spitz DR, Cullen JJ: 2-deoxy-D-glucose causes
cytotoxicity, oxidative stress, and radiosensitization in pancreatic cancer. Free Radic Biol Med 2008; 44(3):322-31.

 Fath MA, Diers AR, Aykin-Burns N, Simons AL, Hua L, and Spitz DR: Mitochondrial electron transport chain blockers
enhance 2-deoxy-D-glucose induced oxidative stress and cell killing in human colon carcinoma cells. Cancer Biol Ther 2009;
8(13):1228-36.

 Hadzic T, Aykin-Burns N, Coleman MC, Zhu Y, Jacobson G, and Spitz DR: Paclitaxel combined with inhibitors of glucose
and hydroperoxide metabolism enhances breast cancer cell killing via H2O2-mediated oxidative stress. Free Radic Biol Med 2010;
48(8):1024-33. (in press).

 Sinthupibulyakit C, Ittarat W, St Clair WH, St Clair DK: p53 Protects lung cancer cells against metabolic stress. Int J Oncol.
2010; 37(6):1575-81.

 Shutt DC, O'Dorisio MS, Aykin-Burns N, Spitz DR: 2-deoxy-D-glucose induces oxidative stress and cell killing in human
neuroblastoma cells. Cancer Biol Ther 2010; 9(11):853-61.

 Sharma PK, Bhardwaj R, Dwarakanath BS, Varshney R: Metabolic oxidative stress induced by a combination of 2-DG and 6-
AN enhances radiation damage selectively in malignant cells via non-coordinated expression of antioxidant enzymes. Cancer Lett.
2010; 295(2):154-66.
 Fath M, Ahmad IM, Smith CJ, Spence JM, Spitz DR: Enhancement of carboplatin-mediated lung cancer cell killing by
simultaneous disruption of glutathione and thioredoxin metabolism. Clin Cancer Res. 2011; [Epub ahead of print]

 Scarbrough PM, Mattson DM, Gius D, Watson WH, and Spitz DR: Simultaneous inhibition of glutathione- and thioredoxin-
dependent metabolism is necessary to potentiate 17AAG-induced cancer cell killing via oxidative stress. Free Radic Biol Med
2011; (in press).
Clinical Implications: Imaging
 If glucose metabolism is increased in cancer cells to
compensate for increased production of prooxidants by
mitochondrial metabolism, then alterations in glucose
metabolism and mitochondrial function should be proportional to
susceptibility to therapies based on taking advantage of this
metabolic defect to selectively kill cancer cells.

 Use FDG-PET imaging to correlate changes in glucose


metabolism (FDG uptake) with sensitivity to:
 Radiation + 2DG
 Cisplatin + 2DG
 Simons AL, Fath MA, Mattson DM, Smith BJ, Walsh SA, Graham MM, Hichwa
RD, Buatti JM, Dornfeld KJ, and Spitz DR: Enhanced response of human head and
neck cancer xenograft tumors to Cisplatin combined with 2-deoxy-D-glucose correlates
with increased 18F-FDG uptake as determined by PET imaging. Int J Radiat Oncol
Biol Phys 2007; 69(4):1222-1230.
Conclusions and Speculations:
 Metabolic oxidative stress appears to contribute to the selective
sensitization of cancer vs. normal cells to 2DG and inhibitors of
glutathione and thioredoxin metabolism in a variety of human
cancer cell models.
 Inhibition of glucose and hydroperoxide metabolism may provide
a biochemical target for selectively enhancing cytotoxicity and
oxidative stress in cancer cells treated with conventional
therapeutic agents (radiation and/or chemotherapy) that cause
oxidative stress in clinical trials.
 Taking advantage of fundamental differences in oxidative
metabolism between cancer vs. normal cells to design combined
modality cancer therapies, could provide improve outcomes while
limiting normal tissue injury by allowing for the de-escalation of
doses of highly cytotoxic agents while maintaining or improving
efficacy of cancer cell killing.
Thanks for
your
attention!
Supported by: R01CA133114; R21CA139182;
R21CA161182; T32CA078586; P30CA086862

S-ar putea să vă placă și