Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Altering the Response to Radiation: Sensitizers and

Protectors
Deborah E. Citrin and James B. Mitchell

A number of agents are used clinically to enhance the efficacy of radiotherapy today, many of
which are cytotoxic chemotherapies. Agents that enhance radiation induced tumor cell killing
or protect normal tissues from the deleterious effects of ionizing radiation are collectively
termed radiation modifiers. A significant effort in radiobiological research is geared towards
describing and testing radiation modifiers with the intent of enhancing the therapeutic effects
of radiation while minimizing normal tissue toxicity. In this review, we discuss the character-
istics of these agents, the testing required to translate these agents into clinical trials, and
highlight some challenges in these efforts.
Semin Oncol 41:848-859 Published by Elsevier Inc.

R adiation is a local and regional therapeutic


modality that is used in the palliative care
and definitive management of approximately
two thirds of cancer patients in the United States.
Radiation therapy was one of the first effective
efficacy of radiation treatment and a major reduction
in normal tissue exposure and injury.
Radiation is a unique modality in that the dose
delivered to tumor is relatively uniform, typically
within a few percent of the prescription dose. This is
therapeutic modalities for lymphomas and solid in contrast to chemotherapies and other medications
tumors.1–3 Although it is one of the oldest therapies prescribed for cancer, for which the dose delivered
for malignancies, the use of radiation has changed a to tumor can be heterogeneous as a result of the
great deal in regards to indications, dose, and unique aspects of tumor physiology, such as varia-
delivery over the past several decades. Imaging tions in the degree and functionality of tumor
modalities such as magnetic resonance imaging and vasculature, elevated interstitial pressure, altered
positron emission tomography have improved the permeability, physiochemical properties of the drug,
ability to delineate tumor within the body. Linear and target expression.4 One obvious limitation is
accelerators have allowed high-energy beams with that radiation is only effective when targeted to the
better penetration in tissue and thus improved appropriate location. Thus, the benefit of radiother-
homogeneity in tumor and reduced normal tissue apy in that it is selectively targeted also may be seen
doses. Novel radiation delivery techniques, such as as a negative.
intensity modulated radiation therapy, have allowed Despite the ability of radiation to treat a tumor
more conformal therapy. The use of daily imaging homogenously and conformally, the tolerance of
during treatment has improved the accuracy of normal tissues continues to limit the maximum dose
treatment and has reduced the margins of normal that can be delivered safely. For some tumor types,
tissue that must be treated to adequately dose tumor. there is evidence that radiation dose escalation can
Collectively, these advances have led to an improved improve the efficacy of radiotherapy5–7 but at a
potential cost of increased toxicity of normal tissues.
Thus, in many circumstances, the efficacy of radia-
Radiation Oncology Branch and Radiation Biology Branch of the
tion continues to be limited by the tolerance of
National Cancer Institute, Bethesda, MD.
This research was supported by the Intramural Research Program of normal tissues adjacent to the tumor.
the National Institutes of Health, National Cancer Institute.
Disclosures and conflicts of interests: The authors have no conflicts of
interests or relationships to disclose. THE IMPORTANCE OF LOCAL CONTROL
Address correspondence to Deborah E. Citrin, MD, Section of
Translational Radiation Oncology, Radiation Oncology Branch, In cancer therapy, the primary endpoint for many
National Cancer Institute, Bethesda, MD 20892. E-mail:
trials is overall survival. Although local therapies,
citrind@mail.nih.gov
0093-7754/ - see front matter
such as surgery and radiotherapy, are capable in
Published by Elsevier Inc. some instances of improving overall survival,8–10 the
http://dx.doi.org/10.1053/j.seminoncol.2014.09.013 primary rational for their use in the definitive

848 Seminars in Oncology, Vol 41, No 6, December 2014, pp 848-859


Altering the response to radiation 849

treatment of cancer is often to enhance the control such as prevention of a morbid local recurrence can
of disease at the site of the primary. Frequently, a be factored into therapeutic decisions.
debate arises over the appropriateness of recom- Finally, there are cancers for which metastasis is
mending local therapies that do not extend survival, rare, such as glioblastoma multiforme, which more
especially when those therapies have real and meas- typically recurs locally despite aggressive local ther-
ureable toxicities. The primary rationale for the apy. For diseases with a low rate of metastatic failure
development of radiation modifiers is the belief that and a high rate of local failure, local control is of
local and regional control is an important oncologic utmost importance. In these diseases, local failure
endpoint. Perhaps the most important question to often leads to death. Improving the local efficacy of
ask in this regard is “What is the benefit of local and radiotherapy in this setting would provide the
regional control on survival?” opportunity to extend survival to a greater degree
An excellent example to explore this question is than in other diseases dominated by metastatic fail-
breast cancer. In the 1880s, Dr William Halstead ure patterns.
developed the radical mastectomy, guided by his
belief that breast cancer could be cured by an
THE THERAPEUTIC RATIO
aggressive regional treatment. Indeed, the radical
surgery was effective in rendering a number of The therapeutic ratio is a concept well known to
women disease-free and became the standard treat- physicians who prescribe treatment. The therapeu-
ment. In the 1950s, this dogma was questioned, tic ratio is a comparison of the amount of a
eventually leading to the comparison of radical therapeutic agent that causes the desired therapeutic
mastectomy with a more limited surgery and post- effect, in this case tumor control, to that amount
mastectomy radiotherapy by the National Surgical which causes toxicity (Figure 1A). In radiation
Adjuvant Breast and Bowel Project (NSABP).11 oncology as in other disciplines, treatment is pre-
Indeed, the outcomes were similar for these scribed at a dose that will limit moderate to severe
approaches, but distant failure was the most com- toxicity to a tolerable level, typically less than a 5%
mon site of failure, occurring in nearly 30% of these risk of major toxicity, while achieving local control
women. As chemotherapy regimens advanced and in a substantial percentage. For some more locally
were able to sterilize distant disease more effectively, aggressive diseases, a higher rate of toxicity may be
local control took on increasing importance.12 The acceptable to obtain local control.
importance of local control became evident when For many cancers, the potential toxicity of normal
the Early Breast Cancer Trialists’ Collaborative Group tissue limits the dose of radiotherapy that can safely
published updated data regarding breast conserving be delivered to tumor. As a result, the rate of local
therapy and radiotherapy in patients with early-stage control is considerably less than 100%. The explo-
breast cancer.8 This meta-analysis further showed an sion of new technologies and techniques that allow
improvement in breast cancer-specific survival at more conformal therapies and more accurate deliv-
late time points after adjuvant locoregional radio- ery have allowed dose escalation to tumor while
therapy in patients with positive or negative lymph minimizing normal tissue toxicity. However, the
nodes. These data suggested that avoidance of four need to include regions of possible microscopic
local failures at 5 years resulted in prevention of one disease and the need for additional margin to
breast cancer death at 15 years. Thus, these data account for setup error and motion of the target
support the concept that local and regional disease during the treatment limit the ability to completely
may act as a reservoir for distant metastases. Further, exclude surrounding normal tissues from the pres-
these data suggest that in the setting of systemic cription dose.
therapy that is increasingly capable of controlling
distant disease, durable local control becomes
RADIATION MODIFIERS
increasingly important in the eventual survival of
the patient. The paradox of local control is simply An alternative method to widen the therapeutic
that, as systemic treatments improve gradually, local ratio for radiotherapy is the use of agents that
control will become increasingly more important to selectively sensitize tumor to radiation or selectively
curative strategies. protect normal tissues from the effects of radiation.
A benefit in terms of overall survival is not the Collectively, these agents are described as radiation
only reason to deliver a local therapy. Local recur- modifiers, as they are capable of modifying tumor or
rence may in some instances result in severe pain, normal tissue response to irradiation. Importantly,
obstruction, and decrement in quality of life without these agents must exhibit selectivity to be clinically
a loss in survival. Thus, although survival is often useful as modifying the response of both tumor
chosen as the most appropriate endpoint to deter- and normal tissue to the same degree offers no
mine if a therapy is needed, other considerations therapeutic gain.
850 D.E. Citrin and J.B. Mitchell

a higher chance of cure for an equivalent degree of


Probability of cure or toxicity

tumor cure
normal toxicity. Depending on the degree of sensitization of
tissue
toxicity
tumor, dose reduction could be considered to min-
imize normal tissue toxicity while reducing normal
tissue injury.
The term “radiation sensitizer” is used to describe
agents that selectively enhance the cell killing from
irradiation in tumor cells while exhibiting no single
Radiation Dose agent toxicity on tumor or normal tissues.13 In
practice, few agents have been described that meet
these criteria, as most agents used in combination
Probability of cure or toxicity

tumor cure
normal
with radiotherapy in the clinic or in the laboratory
tissue setting have some toxicity as a single agent. Thus,
radiosensitizer toxicity the majority of agents used in this context clinically,
such as cisplatin and 5-fluorouracil (5-FU), are more
accurately described as “radiation modifiers,” despite
the common usage of the term “sensitizer.” A true
sensitizer is synergistic with radiation: the combina-
tion has greater impact on tumor cell killing than the
Radiation Dose
sum of its component parts.
Conversely, agents that protect normal tissues
from the deleterious effects of radiation are
Probability of cure or toxicity

tumor cure normal


tissue described as radioprotectors (Figure 1C). By protect-
toxicity ing the normal tissue from the deleterious effects of
radiation, these agents broaden the therapeutic
window.14 In the presence of a radioprotector, the
radiation dose may be escalated to provide enhance-
radioprotector
ment in local control, or a similar dose may be used
with a reduction in the risk of normal tissue injury.
Radiation Dose Radiation protection can be achieved by “chemical
radiation protectors,” which require high tissue
Figure 1. Therapeutic ratio and radiation modifiers. concentrations of the agent immediately before and
(A) The probability of tumor cure (red) and normal tissue during the radiation exposure. For post-radiation
toxicity (blue) can be plotted as a factor of radiation
exposure, a variety of radiation mitigators are under
dose. Note that these are parallel idealized sigmoidal
curves which may not be representative of the clinical
evaluation to lessen radiation-induced normal tissue
condition. A therapeutic dose may be selected that damage.
delivers a high chance of tumor cure with an acceptable The selectivity of radiation modifiers can be a
rate of toxicity (black dashed line). (B) A tumor selective result of a number of characteristics of the agent or
radiation sensitizer increases the probability of tumor its pharmacology. One possible method of selectivity
cure for a given dose (red dashed line), hence the tumor is the rate or degree of uptake of the agent into
control curve is shifted to the left. This results in a higher tumor cells or normal tissues. This may be related to
chance of cure for the same radiation dose as delivered in selective uptake, selective retention, or more rapid
A) (black dashed line). Alternatively, a lower dose may be uptake, metabolism, or clearance in tumor versus
delivered to yield the same rate of cure with less normal normal tissue. Another possible mechanism of selec-
tissue toxicity. (C) A normal tissue radioprotector
tivity is the presence of the target of the modifier
decreases the probability of normal tissue toxicity for a
given dose of radiation, thus pushing the toxicity curve
only on or at a higher level in tumor cells (for
to the right (blue dashed line). This allows a higher dose sensitizers) or normal tissues (for protectors).
to be given to obtain a higher rate of cure with less or
equivalent normal tissue injury. Refer to online version for RADIATION SENSITIZERS
color reproduction.
The vast majority of agents used clinically and
Agents that enhance the ability of radiation to kill tested in the laboratory are more appropriately
tumor cells are frequently described as radiation termed “radiation modifiers”; however, in clinical
sensitizers (Figure 1B). By enhancing the ability of practice, they are often described as “radiosensitiz-
radiation to kill tumor cells, while not altering the ing.” These agents may work in several ways to
radioresponse of normal tissues, these agents are enhance the effectiveness of radiation. In some
able to broaden the therapeutic window, resulting in instances the agents directly enhance individual
Altering the response to radiation 851

tumor cell radiosensitivity. Most radiation sensitizers in concept of the radiation dose modification factor
this class target the cell cycle, DNA repair, or pathways (DMF), defined by the ratio of radiation doses without
known to be involved in survival after irradiation. and with the modifier at an iso-survival level (typically
Alternatively, agents may take advantage of the unique at 1 log of cell kill). Hence, the DMF provides a
aspects of tumor physiology to enhance the sensitivity measure of the treatment response over a range of
of resistant tumor clones, for example by targeting doses as opposed to a modification factor for a single
vasculature or selectively sensitizing hypoxic cells. radiation dose. For the hypothetical sensitizer Drug X
Other agents may have minimal radiation enhancing in Figure 2A, the DMF is 1.5, meaning that the addition
effects but may provide an enhancement of local of Drug X results in the radiation dose being equivalent
control via a direct cytotoxic effect, which in turn to 1.5-fold the effect obtained with radiation alone. The
reduces the number of surviving tumor clones that DMF for different modifiers can be compared for
require sterilization by radiation. These agents, different cell lines and conditions. Agents with larger
although considered additive and not synergistic, may DMFs are anticipated to deliver a greater degree of
have therapeutic efficacy. sensitization; however, this depends on the agent
The distinction between additivity and synergy is an being studied as some agents might target vasculature
important concept. Agents that kill tumor cells without or other physiologic processes that are inadequately
any alteration of the radiation response provide a modeled in vitro. Determining the DMF with regard to
therapeutic benefit by reducing the number of surviv- timing of the agent and radiation exposure can greatly
ing tumor cells, but any efficacy is additive. These facilitate translation to in vivo models as well as
agents may be useful clinically if the toxicity of therapy identifying if possible the mechanism of action and
is tolerable and if the additivity can lead to enhanced biomarkers that might inform whether the drug is
tumor control. In contrast, agents that enhance the actually achieving its potential in vivo.
efficacy of radiotherapy through synergy directly func- Agents that have efficacy in clonogenic studies are
tion to enhance the cytotoxicity of radiotherapy, thus typically further evaluated in xenograft studies. The two
providing a level of tumor cell killing superior to that most common xenograft models used to evaluate tumor
which would be obtained with either agent given alone cure (TCD50, tumor control dose in 50% of the mice) or
or sequentially. tumor regrowth delay. Both single radiation doses or
fractionated radiation delivery are used; however, most
researchers favor fractionated radiation delivery as it is
EVALUATING RADIATION SENSITIZERS
more relevant to the radiation clinic. Because of the
The preclinical and clinical evaluation of radiation high cost and long evaluation times, TCD50 studies are
sensitizers is tailored to the target of the drug, the used less than regrowth delay studies.
chemical and pharmacokinetic and dynamic proper- To test a candidate radiation modifier, xenograft
ties of the drug, and the anticipated clinical outlet. studies typically include at least 4 conditions: con-
Preclinical assessment of an agent typically involves trol, radiation with vehicle, radiation with the can-
several steps including enhancement of radiation- didate drug, and the candidate drug alone. These
induced cell killing, timing of drug administration in studies compare the various groups by determining
relation to radiation exposure, mechanistic studies, the length of time (days) it takes a tumor to grow to
and finally in vivo assessment (xenografts). Preclin- a pre-specified volume (typically at least a tripling in
ical evaluation of potential radiation sensitizers size of the tumor volume from the initiation of
should include assessment of cell survival by use of treatment) (Figure 2B). From these data, a DMF of
clonogenic assays. The clonogenic assay is the gold the agent in vivo can be calculated. Comparing
standard for in vitro assessment of radiation modifi- tumor volume at a single point is not sufficient to
cation, and directly assesses the ability of single cells determine the degree of tumor growth delay and can
to form macroscopic colonies.15,16 While automated inadvertently over or underestimate the degree of
cell assays such as the dye exclusion or MTT (3-(4,5- effect. For these studies, it is also critical that tumors
dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro- are a similar size at the time of treatment as
mide) assays may be useful in large-scale screening sensitivity and radiation can vary with tumor size
of agents, they should not be used as the primary due to hypoxia or other physiologic processes.18
means of assessing an agent for radiation modifica-
tion.17 As shown in Figure 2A, radiation survival
curves obtained through at least 2 logs of survival
MECHANISMS OF RADIATION
allow for adequate assessment of potential radiation
SENSITIZATION
modifiers. Note that it is imperative to correct for the
cytotoxicity of the drug alone. Omission of this The vast majority of agents used clinically as radia-
correction can result in the appearance of synergy tion sensitizers are cytotoxic chemotherapies that
when none exists. Figure 2A also illustrates the independently induce DNA damage or inhibit DNA
852 D.E. Citrin and J.B. Mitchell

Surviving Fraction
0.1

0.01
vehicle
Drug X (not corrected for cytotoxicity)
Drug X (cytotoxicity corrected)

0 2 4 6 8
Dose (Gy)
Dose (Gy) for survival of 0.1 with vehicle 6
DMF drug X = = = 1.5
Dose (Gy) for survival of 0.1 with drug X 4

2000
Tumor size (mm3)

1500

1000
Control
3 Gy
500 50mg
50mg + 3Gy

0 10 20 30 40 50 60 70

Days after randomization

Figure 2. Evaluation of radiation dose modification in vitro and in vivo. (A) Cells are plated at varying densities after
exposure to Drug X or vehicle followed by exposure to radiation. Following incubation for 10–14 days, colonies of 50 or
more cells are counted. The number of colonies that are counted is compared to the number of cells plated and a
surviving fraction is generated. Plotting the surviving fraction over a range of radiation doses allows for the generation of a
survival curve. The survival of the cell line when treated with vehicle and radiation is compared to that obtained when cells
are treated with Drug X and radiation. The surviving fraction of the drug and radiation combination must include a
correction for the cytotoxicity of the drug alone (open squares). If this correction is not made, a dose modifying factor
cannot be calculated and the ability to sensitize or protect cannot be accurately determined. In this example, lack of
correction for cytotoxicity (grey squares) appears to make the drug much more effective as a sensitizer than it actually is.
(B) A dose modifying factor may also be calculated for tumor regrowth. Mice are implanted with a tumor xenograft. Once
tumors reach a measurable and consistent size, mice are randomized to treatment with vehicle, vehicle with radiation (in
this example 3 Gy), drug (in this example, 50 mg of AZD6244), or radiation with drug. The dose modifying factor is
calculated by determining the number of days it takes tumors in each group triple in volume. Comparing volumes at only
one time point (dotted line at 20 days) does not allow a calculation of DMF and may over or underestimate the efficacy of
the investigational agent. Figure 2B reproduced with permission.31

repair. DNA damage, particularly unrepaired DNA may sensitize tumor cells to irradiation by several
double strand breaks, are a potentially lethal event different mechanisms,21 impaired DNA double strand
following exposure to ionizing radiation.19 Agents that break repair is a major contributing factor.22 A number
increase the extent of DNA damage or that inhibit DNA of agents that target DNA repair, such as poly ADP
double strand break repair often sensitize tumor cells ribose polymerase (PARP) inhibitors and ATM/ATR
to irradiation.20 An excellent example of a clinically (ataxia telangiectasia mutated/ataxia telangiectasia
useful modifier in this category is 5-FU. Although 5-FU and Rad3-related protein) inhibitors have been tested
Altering the response to radiation 853

0 0
10 10

Surviving Fraction

Surviving Fraction
10-1

-1
10

10-2

10-3 10-2
0 2 4 6 8 10 0 2 4 6 8 10
Dose (Gy) Dose (Gy)

Control Control
p < 0.0077
AZD7762 AZD7762
10 10
2 Gy 2 Gy
p < 0.0087
2 Gy + AZD7762 2 Gy + AZD7762
Average MC per Cell (%)

8 Average MC per Cell (%)


8
p < 0.01
p < 0.0038

6 6

4 4

2 2

0 0
24 hr 48 hr 72 hr 24 hr 48 hr 72 hr
Time (hr) Time (hr)

Figure 3. The effects of radiation with and without the Chk1/2 inhibitor AZD7762 (100 nmol/L, added 1 hour prior to
and 24 hours post-radiation) on radiation-induced cell killing and mitotic catastrophe. In this study the influence of p53
on Chk1/2 inhibition/radiation response was evaluated using a H460 lung cancer cell line (wild-type p53) as opposed to
H460 cells containing a dominant negative p53 construct (H460 DN p53). Wild-type p53 cells (A and C) were not
sensitized by the combination of AZD7762 and radiation, whereas the radiosensitivity H460 DN p53 cells was significantly
enhanced by AZD7762 (B), as well as DNA damage as measured by mitotic catastrophe (D). Adapted with permission.26

preclinically and are in varying stages of clinical tumor versus normal tissues or no improvement in
development.20,23,24 therapeutic ratio will be realized with their use.
Aside from DNA damage, other aspects of tumor A number of other signaling pathways are known
cell biology can alter radiation sensitivity. For example, to be activated after exposure to ionizing radiation.
the location of a tumor cell in the cell cycle can alter In some cases, basal activation of these pathways
the sensitivity to radiation.25 Cells in G1 and late S are though mutation or amplification can result in a
relatively resistant to radiation compared to cells in G2 reduction in radioresposiveness. An example of such
and M. This is likely partially due to the ability of cells a pathway is the Ras-MAPK pathway, which is
in G1 and S to activate cell cycle checkpoints that rapidly activated following ionizing radiation.27,28
allow for DNA repair. Thus, agents that redistribute Tumor cells expressing mutant conditionally active
tumor cells into a more sensitive phase of the cell cycle Ras are more resistant to radiation-induced cell
or that prevent checkpoint activation are capable of death.29,30 Targeting of these pathways, such as with
sensitizing cells to irradiation. An example of such an MEK inhibition, is capable of sensitizing tumor cells
agent is AZD7762, an inhibitor of Chk1. This agent in vitro and in vivo to radiation.31,32 An ever-
selectively sensitizes p53 mutant cells to radiation increasing number of modifiers that target such
through abrogation of the G2 checkpoint after irradi- pro-survival pathways, such as epidermal growth
ation (Figure 3).26 It is important to reiterate that factor receptor (EGFR), Akt, and mammalian target
agents targeting this and other pathways must have of rapamycin (mTOR), has been described.24,33
selective uptake, retention, or activity in regards to Indeed, EGFR inhibition has been shown to sensitize
854 D.E. Citrin and J.B. Mitchell

to irradiation both in vitro and in vivo34,35 and is nutrient status and defects in metabolism can result
the only molecularly targeted radiation sensitizing in compromised production of ATP, needed to fuel
strategy to be translated successfully to the clinic DNA damage repair systems.42 Lastly, genetic muta-
with US Food and Drug Administration (FDA) tion status of specific driver genes has been shown to
approval.36 differ markedly when multiple biopsies are taken
The majority of preclinical in vitro research on from the same tumor,43 potentially complicating the
radiation modifiers and for that matter molecularly application of effective molecularly targeted therapy.
targeted agents use tumor cells in log phase growth Thus, the inherent heterogeneity of solid tumors
maintained under ambient oxygen conditions (21%). across microenvironmental and genetic landscapes
While cycling tumor cells are an important compo- presents a formidable challenge to the identification
nent of solid tumors, many other subpopulations can of radiation modifiers. This reality speaks to the
be present in solid tumors whose response to importance of including xenograft models in preclin-
radiation and radiation modifiers may differ from ical regrowth delay studies when evaluating radiation
aerobic log phase cells. Abnormal vasculature in solid modifiers/sensitizers.
tumors can result in a complex local microenviron-
ment consisting of reduced levels of oxygenation
RADIATION PROTECTORS
(hypoxia), areas of low nutrient status, and low pH.37
Cells under hypoxic conditions (o0.5 % oxygen) Sensitizing tumor cells to irradiation is one possi-
have long been known to be resistant to radiation by ble method to enhance the therapeutic ratio. An
a factor of 3.38,39 Hypoxia also has been shown to alternative is to prevent radiation injury in normal
decrease the efficacy of cytotoxic chemotherapy tissues. Improvements in radiation delivery and
agents.40 The combination of reduced oxygen levels imaging technology have reduced the amount of
and low nutrient status can result in non-cycling cell normal tissues exposed to high-dose radiation; how-
populations (plateau phase, G0 arrested), whose ever, a margin of clinically uninvolved normal tissue
response to cell-cycle specific agents combined with is often treated to account for set-up error
radiation can be compromised.41 Further, low and microscopic tumor extension that cannot be

Figure 4. Sequence of events following radiation exposure. The chart is divided into two parts by dashed lines suggesting
events and reactions that might be modified by radiation protectors (top), radiation mitigators, and treatment (bottom).
Reproduced with permission.14
Altering the response to radiation 855

visualized with modern imaging technology. Thus, Amifostine is a drug that is metabolized into a free
technological advancements can improve the ther- radical scavenging thiol compound and is an exam-
apeutic ratio only to a point, with residual normal ple of a chemical radioprotector. Amifostine is
tissue toxicity dominated by the effects of irradiation thought to selectively protect normal tissues from
on the tissues included in this obligatory margin. irradiation due to transiently increased uptake in
Radiation injury in normal tissues is highly specific to normal tissues coupled with the higher pH of normal
the location of exposure and can occur as either an tissues and differential activity of alkaline phos-
acute injury and/or a late injury. Acute radiation injury phates in tumor versus normal tissues.50 Levels of
occurs during or shortly after treatment, and typically amifostine in experimental tumors eventually
involves rapidly dividing cells such as skin and mucosa. reached comparable levels to normal tissue, making
Examples of acute injury are dermatitis and mucositis, the timing of the radiation treatment critical to
but these are temporary and not permanent. Late achieve maximal radioprotection (Figure 5).48 Ami-
radiation injury occurs months to years after treatment. fostine is the only FDA-approved radiation protector;
Examples of late radiation injury are myelopathy, however, controversy remains about the potential
fibrosis, fracture, and ulceration. These injuries can be for tumor protection.51
permanent, progressive and even fatal. These toxicities
are typically manifestations of cell loss, tissue/organ
atrophy, and sublethal injury to surviving cells. EVALUATING RADIATION PROTECTORS
The DNA damage and free radical generation that Many of the techniques used to evaluate candi-
result from irradiation occur rapidly after exposure; date radiation sensitizers also can be used in the
however, the deleterious effects of this injury may evaluation of radiation protectors. Clonogenic assays
not be manifest until months to years after treatment
is completed (Figure 4). Agents that prevent the
initial injury from occurring or reduce the extent of
the injury are termed radioprotectors. Radioprotec-
tors are expected to reduce both acute and late
toxicities as they reduce the initial extent of normal
tissue damage. Radiation mitigators are agents that
function after the initial insult but prior to the
manifestation of toxicity. These agents target the
late toxicities of radiation exposure, such as fibro-
sis.44 An example is delivery of bone morrow
stromal cells six weeks after exposure to a fibrosing
dose of radiation. These stromal cells are capable of
reducing subsequent fibrosis.45 Finally, agents that
are aimed at reducing or reversing the injury after it
is manifest are called treatment. Examples of such an
agent are pentoxifylline and vitamin E, which have
been used in combination to effectively treat cuta-
neous radiation fibrosis.46
As described above, radiation protectors are
agents that prevent the initial injury caused by
radiation exposure. The vast majority of these agents
prevent DNA damage by scavenging free radicals,
the molecules that are responsible for the majority of Figure 5. Serum, tissue, and tumor levels of WR-2721
DNA damage after ionizing radiation. These chemical (amifostine) in Fischer 344 rats as a function of time after
radioprotectors are often anti-oxidants; however, not an intraperitoneal injection. Notice that normal tissue
all anti-oxidants are radioprotectors.47 To be an levels rapidly achieve millimolar concentrations of WR-
effective radioprotector of this class, and agent must 2721, whereas tumor levels are quite low initially. With
time tumor levels gradually increase, while normal tissue
be able to enter the cell nucleus and reside in close
levels begin to decrease. This study demonstrates that
proximity to DNA as free radicals have an extremely
chemical radioprotector tissue concentrations in the
short half-life and range. By scavenging the free millimolar range can be achieved quickly. Further the
radicals that are generated in this process, the agents study underscores the importance of the timing of
prevent DNA double-strand breaks and the resulting radiation exposure with within 15-30 min after injection
negative consequences. Two examples of chemical to take advantage of the differential concentration of
radioprotectors are the aminothiol amifostine48 and WR-2721 in normal as opposed to tumor tissue. Adapted
the nitroxide tempol.49 with permission.64
856 D.E. Citrin and J.B. Mitchell

can provide evidence of cellular radioprotection.


Typically, these include the evaluation of non-
malignant cell lines, such as fibroblasts, and tumor
cells lines to evaluate if radioprotectors have selec-
tivity for normal cells versus tumor. Data from these
assays can be used to determine a DMF in a similar
fashion to that used for radiation sensitizers.
For in vivo testing of radiation protectors, a
number of models exist. Total body irradiation is
usually used to test the ability of agents to protect
bone marrow52,53 and intestines from irradiation.54
Lung fibrosis and pneumonitis can be assessed after
pulmonary irradiation using objective endpoints
such as survival, inflammation, and collagen con-
tent.55–57 Models for mucositis, dermatitis, and skin
fibrosis also have been developed to test radiation
protectors.58–60 In addition to assessment of survival,
measures of fibrosis, and scores for acute toxicity,
functional assays such as breathing rate for lung
injury models, can be used as endpoints.61
Lastly, but perhaps most importantly, pre-clinical
studies identifying radioprotectors should also deter-
mine if the agent will protect against tumor
regrowth delay. Agents that protect both tumor
and normal tissue to the same degree offer no
benefit in regards to the therapeutic ratio.
Figure 6. The effects of selumetinib (a MEK inhibitor)
and 5-FU on tumor cell and xenograft radiosensitivity.
(A) The HCT116 cell line was exposed to 15 μmol/L
CONSIDERATIONS IN THE DEVELOPMENT OF of 5-FU (or vehicle) for 18 hours and selumetinib (or
vehicle) for 2 hours and irradiated with graded doses
NOVEL RADIATION MODIFIERS
of x-rays. Colony forming efficiency was determined
It is clear that current in vitro and in vivo models 10–12 days later and survival curves generated after
used to identify radiation modifiers have limitations. normalizing for cell killing by selumetinib, 5-FU or
There is always concern as to whether in vitro selumetinib þ 5-FU in the absence of irradiation.
models are relevant to the in vivo condition. There (B) Mice harboring HCT116 xenografts were randomized
into four groups: vehicle, selumetinib, radiation þ 5-FU,
is a huge disparity between the growth rates of
or selumetinib þ radiation þ 5-FU. Each drug was given
tumor xenografts and tumors in humans. The xeno-
as a single dose prior to a single dose of radiation. The
graft is further complicated in that the tumor is study demonstrates that radiation modifiers can be
supported by a rodent vascular system and infiltrated evaluated in the context of chemoradiotherapy and
by rodent host cells. The question also arises as to highlights the need to account for the toxicity of both
the location of the tumor on the mouse (leg, flank, or the investigational agent and chemotherapy in calcula-
orthotopic). Gene expression profiles of some glio- tions of modifying capability. Adapted with permission.32
blastoma cell lines have been shown to vary consid-
erably when grown in vitro, in the flank of mice, or
orthotopically in the brain of mice.62,63 Whether this include an evaluation of any interaction of the new
variation in gene expression governs the response to agent with the established regimen. For example,
radiation with or without modifiers is not known. the addition of a MEK inhibitor to a standard regimen
The relevance of radiation-induced toxicities in of radiation and 5-FU for eventual clinical translation
rodent normal tissues to human injury also can be for rectal cancer required the evaluation of the
questioned. Despite these limitations, preclinical effects of MEK inhibition (selumetinib) on 5-FU
studies are able to identify agents that can sensitize efficacy in tumor, an evaluation of the effects of
or protect at the cellular and tissue level that may MEK inhibition on radiation sensitivity of tumor cells
hold clinical promise. when given with radiation or the combination of 5-
An additional complication of developing novel FU and radiation (Figure 6).32 The evaluation of the
radiation modifiers is the frequent use of sensitizing chemotherapy agent and the modifier is critical to
chemotherapy in modern treatment regimens. As a rule out any decrease in sensitivity to chemotherapy
result, the use of a radiosensitizer or protector must with the combination. Many candidate radiation
Altering the response to radiation 857

sensitizers that target signal transduction also result


in cell cycle arrest in G1. As cycling is often
important for sensitivity to cytotoxic chemotherapy,
this interaction must be probed. The sequencing of
the agents for both preclinical testing and clinical
translation becomes of critical importance to under-
standing possible positive and negative interactions.
Likewise, the addition of a protector to these
standard chemoradiation regimens presents similar
concerns, namely, that the radiation protector may
interfere with the efficacy of the chemotherapy as a
single agent or with the radiation modification
offered by the chemotherapy. An example of this
scenario is the addition of tempol, a radioprotector,
to the regimen of radiation and cisplatin in antici-
pation of use in the treatment of head and neck
cancer.58 As shown in Figure 7A, tempol protected 2000
Control
against radiation/cisplatin mucositis, but a crucial FX
question in such studies is whether tempol would 1750 CDDP
CDDP-FX
lessen the radiation/cisplatin tumor response.
Tumor Volume (mm3)
1500 TPL-CDDP-FX
Figure 7B shows that tempol did not interfere with
the chemoradiation enhancement in the tumor. 1250
Preclinical studies that include an evaluation of 1000
radiation, chemotherapy, and an investigational radi-
ation modifier are by nature complex, costly, and 750
time consuming. Nonetheless as the standard of care
500
in many cancers has moved toward the use of
concurrent chemotherapy with radiation, radiopro- 250
tector studies must be evaluated in preclinical
0
models that include these agents. 0 5 10 15 20 25
An additional complication to the clinical trans- Time (days)
lation of radiation modifiers is determining and
delivering the optimal schedule for sensitization. Figure 7. The effect of the radioprotector tempol on
Many drugs that are developed that may have radio- oral mucositis resulting from fractionated irradiation and
sensitizing properties are tested with dosing sched- cisplatin (CDDP). To measure ulceration tongues were
ules geared towards chemotherapy. As radiotherapy removed from the various groups and stained with
is given more continuously than many chemother- Toluidine blue. Ulcerations were clearly present in ton-
gues from mice treated with the combination of CDDP
apy regimens, these schedules must be tested pre-
and radiation (panel A, second row, marked by arrows).
clinically to provide safety data for clinical trans-
Tempol either delivered systemically or topically afforded
lation. near complete protection (A, third and fourth rows).
Finally, incorporation of biomarkers and surrogate Using the same tempol/CDDP/radiation protocol as used
markers into clinical trials is of increasing impor- in A, SCCVII tumor regrowth for the various groups is
tance. These markers may be used to screen for shown in B. Tempol did not interfere with CDDP
presence of the target, effective modulation of the mediated enhancement of radiation. Adapted with
target with the investigational drug, and upregula- permission.58
tion of the target with radiotherapy. Ideally, a non-
invasive or minimally invasive marker, such as a
circulating marker or imaging test, could be used for and interactions with standard therapy. Thoughtful
this purpose. clinical translation requires the consideration of
available preclinical and clinical data to rationally
design the appropriate method and sequence of
FUTURE DIRECTIONS AND CONCLUSIONS
delivery. Incomplete evaluation of agents, such as
A number of promising targets for radiation lack of assessment of effects on both normal and
sensitization and protection have been described, tumor tissue, incomplete studies on sequencing, and
and agents targeting these pathways are in develop- incomplete understanding of mechanism of effect
ment. Successful clinical translation requires rigor- may result in failure at the time of clinical
ous preclinical testing, careful study of sequencing translation.
858 D.E. Citrin and J.B. Mitchell

REFERENCES 16. Franken NA, Rodermond HM, Stap J, Haveman J, van


1. Gilbert M. Radiotherapy in Hodgkin’s disease (malig- Bree C. Clonogenic assay of cells in vitro. Nat Protoc.
nant granulomatosis); anatomic and clinical founda- 2006;1:2315–9.
tions; governing principles, results. Am J Roentgenol. 17. Harrington KJ, Billingham LJ, Brunner TB, et al. Guide-
1939;41:198–241. lines for preclinical and early phase clinical assessment of
2. Peters M. A study in survivals in Hodgkin’s disease treated novel radiosensitisers. Br J Cancer. 2011;105:628–39.
radiologically. Am J Roentgenol. 1950;63:299–311. 18. Suit H, Schlachter L, Andrews JR. “Oxygen effect” and
3. Coutard H. Roentgen therapy of epitheliomas of the tumor size as related to response of C3H/Ba adeno-
tonsillar region, hypopharynx, and larynx from. 1920 carcinoma to local X irradiation. J Natl Cancer Inst.
to. 1926. AJR Am J Roentgenol. 1932;28:313. 1960;24:1271–81.
4. Minchinton AI, Tannock IF. Drug penetration in solid 19. Cornforth MN, Bedford JS. A quantitative comparison
tumours. Nat Rev Cancer. 2006;6:583–92. of potentially lethal damage repair and the rejoining of
5. Kuban DA, Tucker SL, Dong L, et al. Long-term results interphase chromosome breaks in low passage normal
of the M. D. Anderson randomized dose-escalation trial human fibroblasts. Radiat Res. 1987;111:385–405.
20. Thoms J, Bristow RG. DNA repair targeting and radio-
for prostate cancer. Int J Radiat Oncol Biol Phys.
therapy: a focus on the therapeutic ratio. Semin Radiat
2008;70:67–74.
Oncol. 2010;20:217–22.
6. Bartelink H, Horiot JC, Poortmans PM, et al. Impact of a
21. Lawrence TS, Tepper JE, Blackstock AW.
higher radiation dose on local control and survival in
Fluoropyrimidine-radiation interactions in cells and
breast-conserving therapy of early breast cancer: 10-year
tumors. Semin Radiat Oncol. 1997;7:260–6.
results of the randomized boost versus no boost EORTC
22. Lawrence TS, Davis MA, Tang HY, Maybaum J.
22881-10882 trial. J Clin Oncol. 2007;25:3259–65.
Fluorodeoxyuridine-mediated cytotoxicity and radio-
7. Andrews DW, Scott CB, Sperduto PW, et al. Whole
sensitization require S phase progression. Int J Radiat
brain radiation therapy with or without stereotactic
Biol. 1996;70:273–80.
radiosurgery boost for patients with one to three brain
23. Chalmers AJ, Lakshman M, Chan N, Bristow RG. Poly
metastases: phase III results of the RTOG 9508
(ADP-ribose) polymerase inhibition as a model for
randomised trial. Lancet. 2004;363:1665–72.
synthetic lethality in developing radiation oncology
8. Darby S, McGale P, Correa C, et al. Effect of radio-
targets. Semin Radiat Oncol. 2010;20:274–81.
therapy after breast-conserving surgery on 10-year
24. Choudhury A, Cuddihy A, Bristow RG. Radiation and
recurrence and 15-year breast cancer death: meta-
new molecular agents part I: targeting ATM-ATR
analysis of individual patient data for 10,801 women checkpoints, DNA repair, and the proteasome. Semin
in 17 randomised trials. Lancet. 2011;378:1707–16. Radiat Oncol. 2006;16:51–8.
9. Overgaard M, Jensen MB, Overgaard J, et al. Post- 25. Terasima T, Tolmach LJ. Changes in x-ray sensitivity
operative radiotherapy in high-risk postmenopausal of HeLa cells during the division cycle. Nature. 1961;
breast-cancer patients given adjuvant tamoxifen: Dan- 190:1210–1.
ish Breast Cancer Cooperative Group DBCG 82c 26. Mitchell JB, Choudhuri R, Fabre K, et al. In vitro and
randomised trial. Lancet. 1999;353:1641–8. in vivo radiation sensitization of human tumor cells by
10. Warde P, Mason M, Ding K, et al. Combined androgen a novel checkpoint kinase inhibitor, AZD7762.
deprivation therapy and radiation therapy for locally Clin Cancer Res. 2010;16:2076–84.
advanced prostate cancer: a randomised, phase 3 trial. 27. Kasid U, Suy S, Dent P, Ray S, Whiteside TL, Sturgill
Lancet. 2011;378:2104–11. TW. Activation of Raf by ionizing radiation. Nature.
11. Fisher B, Jeong JH, Anderson S, Bryant J, Fisher ER, 1996;382:813–6.
Wolmark N. Twenty-five-year follow-up of a random- 28. Kharbanda S, Saleem A, Shafman T, Emoto Y, Weich-
ized trial comparing radical mastectomy, total mastec- selbaum R, Kufe D. Activation of the pp90rsk and
tomy, and total mastectomy followed by irradiation. mitogen-activated serine/threonine protein kinases
N Engl J Med. 2002;347:567–75. by ionizing radiation. Proc Natl Acad Sci U S A. 1994;
12. Recht A, Gray R, Davidson NE, et al. Locoregional 91:5416–20.
failure 10 years after mastectomy and adjuvant chemo- 29. Sklar MD. The ras oncogenes increase the intrinsic
therapy with or without tamoxifen without irradia- resistance of NIH 3T3 cells to ionizing radiation.
tion: experience of the Eastern Cooperative Oncology Science. 1988;239:645–7.
Group. J Clin Oncol. 1999;17:1689–700. 30. Bernhard EJ, Stanbridge EJ, Gupta S, et al. Direct
13. Russo A, Mitchell J, Kinsella T, Morstyn G, Glatstein E. evidence for the contribution of activated N-ras and
Determinants of radiosensitivity. Semin Oncol. 1985; K-ras oncogenes to increased intrinsic radiation resist-
12:332–49. ance in human tumor cell lines. Cancer Res. 2000;
14. Citrin D, Cotrim AP, Hyodo F, Baum BJ, Krishna MC, 60:6597–600.
Mitchell JB. Radioprotectors and mitigators of 31. Chung EJ, Brown AP, Asano H, et al. In vitro and
radiation-induced normal tissue injury. Oncologist. in vivo radiosensitization with AZD6244 (ARRY-
2010;15:360–71. 142886), an inhibitor of mitogen-activated protein
15. Puck TT, Marcus PI, Cieciura SJ. Clonal growth of kinase/extracellular signal-regulated kinase 1/2 kinase.
mammalian cells in vitro; growth characteristics of Clin Cancer Res. 2009;15:3050–7.
colonies from single HeLa cells with and without a 32. Urick ME, Chung EJ, Shield WP, 3rd, et al. Enhance-
feeder layer. J Exp Med. 1956;103:273–83. ment of 5-fluorouracil-induced in vitro and in vivo
Altering the response to radiation 859

radiosensitization with MEK inhibition. Clin Cancer 48. Yuhas JM, Storer JB. Differential chemoprotection of
Res. 2011;17:5038–47. normal and malignant tissues. J Natl Cancer Inst.
33. Chinnaiyan P, Allen GW, Harari PM. Radiation and 1969;42:331–5.
new molecular agents, part II: targeting HDAC, HSP90, 49. Soule BP, Hyodo F, Matsumoto K, et al. The chemistry
IGF-1R, PI3K, and Ras. Semin Radiat Oncol. and biology of nitroxide compounds. Free Radic Biol
2006;16:59–64. Med. 2007;42:1632–50.
34. Huang SM, Harari PM. Modulation of radiation 50. Kouvaris JR, Kouloulias VE, Vlahos LJ. Amifostine: the
response after epidermal growth factor receptor first selective-target and broad-spectrum radioprotec-
blockade in squamous cell carcinomas: inhibition of tor. Oncologist. 2007;12:738–47.
damage repair, cell cycle kinetics, and tumor angio- 51. Brizel DM, Overgaard J. Does amifostine have a role in
genesis. Clin Cancer Res. 2000;6:2166–74. chemoradiation treatment? Lancet Oncol. 2003;4:
35. Harari PM, Huang S. Radiation combined with EGFR 378–81.
signal inhibitors: head and neck cancer focus. Semin 52. Hahn SM, Tochner Z, Krishna CM, et al. Tempol, a
Radiat Oncol. 2006;16:38–44. stable free radical, is a novel murine radiation protec-
36. Bonner JA, Harari PM, Giralt J, et al. Cetuximab tor. Cancer Res. 1992;52:1750–3.
prolongs survival in patients with locoregionally 53. Davis RM, Sowers AL, DeGraff W, et al. A novel
advanced squamous cell carcinoma of head and neck: nitroxide is an effective brain redox imaging contrast
a phase III study of high dose radiation therapy with or agent and in vivo radioprotector. Free Radic Biol Med.
without cetuximab. J Clin Oncol (Meeting Abstracts). 2011;51:780–90.
2004;22:5507. 54. Travis EL, Thames HD, Jr., Tucker SL, Watkins TL, Kiss
37. Brown JM. Tumor microenvironment and the I. Protection of mouse jejunal crypt cells by WR-2721
response to anticancer therapy. Cancer Biol Ther. after small doses of radiation. Int J Radiat Oncol Biol
2002;1:453–8. Phys. 1986;12:807–14.
38. Thomlinson RH, Gray LH. The histological structure of 55. Travis EL, Harley RA, Fenn JO, Klobukowski CJ,
some human lung cancers and the possible implica- Hargrove HB. Pathologic changes in the lung follow-
tions for radiotherapy. Br J Cancer. 1955;9:539–49. ing single and multi-fraction irradiation. Int J Radiat
39. Bristow RG, Hill RP. Hypoxia and metabolism. Hypo- Oncol Biol Phys. 1977;2:475–90.
xia, DNA repair and genetic instability. Nat Rev 56. Citrin DE, Shankavaram U, Horton JA, et al. Role of
Cancer. 2008;8:180–92. type II pneumocyte senescence in radiation-induced
40. Teicher BA, Holden SA, al-Achi A, Herman TS. Classi- lung fibrosis. J Natl Cancer Inst. 2013;105:1474–84.
fication of antineoplastic treatments by their differ- 57. Chiang CS, Liu WC, Jung SM, et al. Compartmental
ential toxicity toward putative oxygenated and responses after thoracic irradiation of mice: strain
hypoxic tumor subpopulations in vivo in the FSaIIC differences. Int J Radiat Oncol Biol Phys. 2005;62:
murine fibrosarcoma. Cancer Res. 1990;50:3339–44. 862–71.
41. Liebmann J, Cook JA, Fisher J, Teague D, Mitchell JB. 58. Cotrim AP, Yoshikawa M, Sunshine AN, et al. Pharma-
In vitro studies of Taxol as a radiation sensitizer in cological protection from radiation þ/- cisplatin-
human tumor cells. J Natl Cancer Inst. 1994;86: induced oral mucositis. Int J Radiat Oncol Biol Phys.
441–6. 2012;83:1284–90.
42. Bailey KM, Wojtkowiak JW, Hashim AI, Gillies RJ. 59. Fleischer G, Dorr W. Amelioration of early radiation
Targeting the metabolic microenvironment of tumors. effects in oral mucosa (mouse) by intravenous or
Adv Pharmacol. 2012;65:63–107. subcutaneous administration of amifostine. Strah-
43. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor lenther Onkol. 2006;182:567–75.
heterogeneity and branched evolution revealed by multi- 60. Stone HB. Leg contracture in mice: an assay of normal
region sequencing. N Engl J Med. 2012;366:883–92. tissue response. Int J Radiat Oncol Biol Phys.
44. Williams JP, Jackson IL, Shah JR, Czarniecki CW, 1984;10:1053–61.
Maidment BW, DiCarlo AL. Animal models and medi- 61. Travis EL, Down JD, Holmes SJ, Hobson B. Radiation
cal countermeasures development for radiation- pneumonitis and fibrosis in mouse lung assayed by
induced lung damage: report from an NIAID Work- respiratory frequency and histology. Radiat Res.
shop. Radiat Res. 2012;177:e0025-e0039. 1980;84:133–43.
45. Horton JA, Hudak KE, Chung EJ, et al. Mesenchymal 62. Camphausen K, Purow B, Sproull M, et al. Influence of
stem cells inhibit cutaneous radiation-induced fibrosis in vivo growth on human glioma cell line gene
by suppressing chronic inflammation. Stem Cells. expression: convergent profiles under orthotopic con-
2013;31:2231-2241. ditions. Proc Natl Acad Sci U S A. 2005;102:8287–92.
46. Delanian S, Porcher R, Rudant J, Lefaix JL. Kinetics of 63. Camphausen K, Purow B, Sproull M, et al. Orthotopic
response to long-term treatment combining pentoxifyl- growth of human glioma cells quantitatively and
line and tocopherol in patients with superficial radiation- qualitatively influences radiation-induced changes in
induced fibrosis. J Clin Oncol. 2005;23:8570–9. gene expression. Cancer Res. 2005;65:10389–93.
47. Camphausen K, Citrin D, Krishna MC, Mitchell JB. 64. Yuhas JM. Active versus passive absorption kinetics as
Implications for tumor control during protection of the basis for selective protection of normal tissues by
normal tissues with antioxidants. J Clin Oncol. S-2-(3-aminopropylamino)-ethylphosphorothioic acid.
2005;23:5455–7. Cancer Res. 1980;40:1519–24.

You might also like