Вы находитесь на странице: 1из 18

Heat-Based Tumor Ablation: Role

of the Immune Response 8


Feng Wu

Abstract
The ideal cancer therapy not only induces the death of all localized tumor
cells with less damage to surrounding normal tissue, but also activates a
systemic antitumor immunity. Heat-based tumor ablation has the potential
to be such a treatment as it can minimal-invasively ablate a targeted tumor
below the skin surface, and may subsequently augment host antitumor
immunity. This chapter primarily introduces increasing pre-clinical and
clinical evidence linking antitumor immune response to thermal tumor
ablation, and then discusses the potential mechanisms involved in ablation-
enhanced host antitumor immunity. The seminal studies performed so far
indicate that although it is not possible to make definite conclusions on the
connection between thermal ablation and antitumor immune response, it is
nonetheless important to conduct extensive studies on the subject in order
to elucidate the processes involved.

Keywords
Heat • Ablation • Neoplasm • Thermal ablation • Immunity • High inten-
sity focused ultrasound • Radiofrequency • Microwave • Laser •
Cryoablation • Antigen presenting cell • Cytotoxic T lymphocyte • Tumor
infiltrating lymphocyte • Heat shock protein • Tumor vaccine

8.1 Introduction
F. Wu
Nuffield Department of Surgical Sciences, As an alternative approach to surgical interven-
HIFU Unit, Churchill Hospital, University of Oxford, tion, heat-based tumor ablation with minimally
Oxford OX3 9 DU, UK
invasive techniques has received increasingly
Institute of Ultrasonic Engineering in Medicine, widespread interest in the local management of
College of Biomedical Engineering, Chongqing
solid malignancy. It employs various kinds of
Medical University, Chongqing 400016, China
e-mail: mfengwu@yahoo.com; physical energy for in-situ destruction of a tar-
feng.wu@nds.ox.ac.uk geted tumor, instead of local tumor removal. This

© Springer International Publishing Switzerland 2016 131


J.-M. Escoffre, A. Bouakaz (eds.), Therapeutic Ultrasound, Advances in Experimental
Medicine and Biology, Vol. 880, DOI 10.1007/978-3-319-22536-4_8
132 F. Wu

is achieved by either raising the temperature 8.2 Methods of Thermal


between 56 and 100 °C, or by using extremely Ablation Technique
cold temperatures. The main advantage of this
alternative is a decrease in less invasiveness ver- Local tumor destruction occurs when physical
sus surgical procedures, resulting in an associated energy is transmitted into a tumor lesion and all
reduction in mortality, morbidity, hospital stay, targeted cancer cells can be completely destroyed.
cost and improved quality of life for cancer Minimally invasive thermal techniques rely on
patients (Cabibbo et al. 2009; Timmerman et al. heat as the major mode of tumor ablation. They
2009; Liapi and Geschwind 2007; Hong et al. vary based on the processes involved in heat gen-
2006; Hafron and Kaouk 2007, Dacadt and eration and their delivery. Due to differences in
Siriwardena 2004). Due to various energy sources, energy sources, these thermal techniques can be
the thermal ablation techniques include high- classified into five categories as follows: High-
intensity focused ultrasound (HIFU), radiofre- intensity focused ultrasound (HIFU) ablation,
quency, laser, microwave and cryoablation. All of radiofrequency ablation (RFA), laser ablation
them can selectively destroy a targeted tumor via (LA), microwave ablation (MWA) and cryoabla-
either percutaneous or extracorporeal approaches. tion. Each method has unique characteristics for
As curative and palliative intentions, they have tumor ablation with regards to the method of
partially replaced some open surgery procedures energy delivery through the skin, conduction of
in the clinical treatment of patients with solid energy and length of time required, real-time
tumors, including those of the liver, prostate, kid- imaging for targeting/monitoring and a variety of
ney, lung, breast, pancreas, brain, bone and soft other specific issues. A summary comparing the
tissues. varied methods is shown in Table 8.1.
It has been observed for a long time that large
amounts of tumor debris remain in-situ after ther-
mal ablation. As a normal process of the healing 8.2.1 High-Intensity Focused
response, tumor debris is gradually reabsorbed Ultrasound Ablation
and then replaced by scar tissue in the patient. It
usually takes a period from months to years, Concerning all minimally invasive therapies, HIFU
which depends on the size of the ablated tumor. ablation is the only non-invasive approach proposed
However, it is still unclear what kind of biologi- to date (Kennedy 2005). It employs extracorporeal
cal significance may exist during the absorption ultrasound energy to ablate a targeted tumor at
period of the ablated tumor. Some studies have depth without any needle insertion. Thus, there is no
recently shown that an active immune response to damage to the skin and overlying tissues. Ultrasound
the treated tumor could be developed after ther- is a high frequency pressure wave. It can be brought
mal ablation, and the host immune system could to a tight focus at a distance from its source while
become more sensitive to the tumor cells (Wu propagating through tissues. If the concentrated
et al. 2007a, b; Gravante et al. 2009; Fagnoni energy is sufficient, energy absorption by the living
et al. 2008; Sabel 2009). This may lead to a tissue causes measurable temperature rises (56–
potential procedure that reduces or perhaps elim- 100 °C), resulting in coagulation necrosis of the tis-
inates metastases, thus preventing local recur- sue solely within the focal volume (Chaussy et al.
rence in cancer patients who have had original 2005). In addition, non-thermal effects, such as
dysfunction of antitumor immunity before treat- cavitation, can induce local tissue destruction due to
ment. In this chapter we will introduce the stud- cavitation-induced high pressures and temperatures
ies that focused on the host immune responses (Wu 2006). A single exposure ablation zone (1–3 s)
after thermal tumor ablation, and provide experi- is small, ellipsoidal sized, approximately
mental and clinical data available to assess 1.5 × 15 mm under normal exposure parameters at
whether they could be potential for understand- 1.0 MHz. By placing numerous individual ablation
ing of this complex phenomenon. zones side-by-side, conformal confluent ablation
8 Heat-Based Tumor Ablation: Role of the Immune Response 133

Table 8.1 Comparison of thermal ablation methods for tumors


Energy Imaging Ablation
Methods conduction Ablation volume Energy delivery guidance time (min) Ablation expense
HIFU Heat Conformal Transcutaneous Ultrasound 30–120 Expensive device
cavitation ablation No probe MRI No probe charge
No tumor size insertion
limitation
RFA Heat No conformal Percutaneous Ultrasound 10–30 Cheap device
ablation Electrode probe CT Expensive probe
Tumor size
limitation
Laser Heat Not Conformal Percutaneous Ultrasound 25–30 Expensive device
ablation Optical fiber MRI Probe charge
Tumor size
limitation
Microwave Heat Not Conformal Percutaneous Ultrasound 20–60 Cheap device
ablation Electrode CT/MRI Probe charge
Tumor size Antenna
limitation
Cryoablation Cold Not conformal Percutaneous Ultrasound 15–30 Cheap device
ablation Applicator CT Expensive
Tumor size applicator
limitation

volumes of clinically relevant size can be achieved 8.2.3 Laser Ablation


(ter Haar 2007). On the other hand, while HIFU
ablation only takes 1–3 s per exposure; the total The term LA is also referred to as laser photo-
time can be substantial, longer than other minimally coagulation or laser interstitial thermal ther-
invasive therapies. apy (Goldberg et al. 2005). LA employs
infrared light energy to produce heat and
ablate a targeted cancer. The light energy is
8.2.2 Radiofrequency Ablation transmitted through an optical fiber with a
bare tip, and thus induces coagulation necrosis
RFA uses an electromagnetic energy source with of the targeted tumor while it diffuses through
frequencies less than 900 kHz to generate heat the target (Gough-Palmer and Gedroyc 2008).
(Decadt and Siriwardena 2004). An electrode The Nd-YAG (neodymium:yttrium aluminum
probe is percutaneously placed into a targeted garnet) laser with a wavelength of 1064 nm,
tumor. Through the probe, there is transmission and diode laser with shorter wavelengths
of low voltage alternating current that creates (800–980 nm), are the most widely used
ionic agitation and heating (Lau and Lai 2009). devices for laser ablation of solid tumors.
Ablation temperatures reach 50–100 °C, result- They can both induce tissue photocoagulation
ing in the coagulation necrosis of the targeted at low power, or vaporization and cavitation at
tumor (Curley 2001). While the tissue surround- a higher output. The extent of tissue necrosis
ing the tip of the probe reaches in excess of is typically limited, dependent on the amount
100 °C, it will vaporize and char. This decreases of deposited energy. Thus, multiple fiber
the absorption of the energy, and reduces the applicators are necessary in clinical applica-
ablative size of the surrounding tissue (Goldberg tion for ablation of larger lesions (Sabharwal
et al. 1996). et al. 2009).
134 F. Wu

8.2.4 Microwave Ablation temperatures are increased to 42–45 °C for a


period of 30–60 min, cells become more subject
MWA employs electromagnetic energy to ablate a to damage by other agents such as radiotherapy
targeted tumor via an electrode-antenna placed and chemotherapy (Hill and ter Haar 1995).
within the lesion (Carrafiello et al. 2008). While Increasing the temperature can obviously shorten
electromagnetic microwaves (900–3,000 kHz) the exposure time for therapeutic effects. If the
travel through the tissues, they evoke agitation temperature is increased a few degrees more to
and vibration of ionic molecules, such as water 50–52 °C and maintained for 4–6 min, irrevers-
molecules, within cells. The rapid motion of these ible cellular damage is induced (Thomsen 1991).
ionic molecules causes frictional heating, raising Between 60 and 100 °C, instantaneous induction
the local temperature range from 60–100 °C in the of protein coagulation occurs, resulting in the
cellular environment, resulting in tissue coagula- permanent destruction of key mitochondrial
tion necrosis (Simon et al. 2005). Compared to enzymes and nucleic acid-histone complexes
RFA, microwave ablation can actively heat a (Goldberg et al. 2000). Temperatures greater than
much larger area, with less effect on the heat sink, 105 °C can cause tissue vaporization and carbon-
but there is no tissue boiling and charring during ization (Goldberg et al. 1996).
ablation procedure (Beland et al. 2007). Thermal ablation technique is a different ther-
apy to hyperthermia, which has been applied by
physical heating technology to elevate targeted
8.2.5 Cryoablation regions to temperatures in the 42–45 °C range.
This “conventional” hyperthermia usually main-
Cryoablation is an alternative technique that uses tains uniform temperature distributions in a nar-
extreme cold to freeze a targeted tumor in the row therapeutic range for a period of 30–60 min,
form of an “ice-ball.” It is one of the oldest abla- and is applied once or twice a week (Diederich
tion methods, with less peri- and post-procedural and Hynynen 1999). However, the temperature
pain (Rybak 2009). Cryoablation has recently distributions induced in-vivo are usually non-
gained an increased interest due to the use of an uniform because of tissue cooling by blood flow,
argon-gas cryotherapy technique, which induces and it is extremely difficult to avoid local cold
controlled tissue freezing by inserting a percuta- spots that do not reach the required therapeutic
neous applicator into a targeted lesion (Dumot temperature level (Lubbe and Bergemann 1994).
and Greenwald 2008). A typical cryoablation ses- The efficiency of hyperthermia is highly depen-
sion involves a freeze-thaw-freeze cycle. The dent on the ability to localize and control the
argon and helium gases are alternately delivered successful temperature distributions, which are
to achieve extra- and intra-cellular ice crystal for- often influenced by tissue heterogeneities and
mation and tissue osmosis. This process causes blood flow. As a result, hyperthermia cannot be
protein denaturation, cell membrane rupture and used alone in its clinical application, but can be
cellular death (Babaian et al. 2008). only implemented as an adjuvant method to
combine with either radiation therapy or chemo-
therapy in the treatment of malignant tumors
8.3 Mechanisms of Thermal (Dewey 1994). Two types of mechanism are
Ablation and Immune commonly involved to explain the rational for
Response this combined therapy. Heat is a radio-sensitizer
that increases radiation damage and prevents
The absorption of physical energy delivered by subsequent repair. Hyperthermia can also pro-
thermal ablation technique can result in a mea- duce biological effects on targeted tumors,
surable temperature elevation in living tissue. including direct cellular toxicity, hypoxia, low
The thermal effects on tissue are directly depen- pH and indirect blood perfusion deprivation in
dent on how heat interacts with the tissue. When the tumor (Overgaard 1989).
8 Heat-Based Tumor Ablation: Role of the Immune Response 135

Thermal ablation can cause direct and indirect Histological changes are evident in tumor tis-
damages to a targeted tumor. Direct heat injury sue after thermal ablation (Clement 2004). In
occurs during the period of heat deposition, and it addition to HIFU ablation, four cellular change
is predominately determined by the total energy zones are described in the liver after thermal
delivered to the targeted tumor (Nikfarjam et al. ablation as follows: Application, central, transi-
2005a, b, c). Indirect heat injury usually occurs tion and reference tissue zones (Ozaki et al.
after thermal ablation, which produces a progres- 2003a, b; Germer et al. 1998a, b; Ohno et al.
sion in tissue damage. It may involve a balance of 2001). The application zone is where the heat
several factors, including microvascular damage, source contacts the tissue. The central zone
cellular apoptosis, Kupffer cell activation and immediately surrounds the application zone and
altered cytokine release (Nikfarjam et al. 2005a, consists of damaged tissue. The transition zone
b, c). Direct injury is generally better defined contains apparently undamaged tissue, but exhib-
than the secondary indirect effects. its signs of subacute hemorrhage. The reference
zone refers to normal tissue surrounding the tran-
sition zone.
8.3.1 Direct Thermal and Non-
thermal Effects on Tumor
8.3.2 Direct Thermal Effects
The effects of thermal ablation on a targeted tumor on Tumor Blood Vessels
are determined by increased temperatures, thermal
energy deposition, rate of heat removal and the Structural and functional changes are directly
specific thermal sensitivity of the tissue. As the tis- observed in tumor blood vessels after thermal
sue temperature rises, the time required to achieve ablation. These changes are not as well described
irreversible cellular damage decreases exponen- as the thermal effects on tissues, but they do rely
tially. At temperatures between 50 and 55 °C, cel- on varying temperatures. At temperatures
lular death occurs instantaneously in cell culture between 40 and 42 °C, there is no significant
(Wheatley et al. 1989). Protein denaturation, change in tumor blood flow after 30–60 min
membrane rupture, cell shrinkage, pyknosis and exposure (Ozaki et al. 2003a, b). Beyond
hyperchromasia occur ex-vitro between 60 and 42–44 °C, there is an irreversible decrease in
100 °C, leading to immediate coagulation necrosis tumor blood flow, with vascular stasis and throm-
(Wheatley et al. 1989). Additional to this necrosis, bosis resulting in heat trapping and progressive
tissue vaporization and boiling occur at tempera- tissue damage (Emami and Song 1984). While
tures greater than 105 °C. Carbonization, charring temperatures exceed 60 °C, immediate destruc-
and smoke generation occur when the temperature tion of tumor microvasculature occurs (Tranberg
is over 300 °C (Heisterkamp et al. 1997). 2004). It cuts the blood supply to the tumor
In addition, acoustic cavitation, one of mechan- directly through the cauterization of the tumor
ical effects induced by HIFU ablation, is the most feeder vessels, leading to nutrient and oxygen
important non-thermal mechanism for tissue dis- deprivation. Thus, tissue destruction can be
ruption in the ultrasound field (Germer et al. enhanced by the damage caused by thermal abla-
1998a, b). The presence of small gaseous nuclei tion to tumor blood vessels.
within subcellular organelles and tissue fluids are
the source of cavitation. These bubbles can
expand and contract under influence of the acous- 8.3.3 Indirect Effects After Thermal
tic pressure. During the collapse of bubbles, the Ablation
acoustic pressure is more than several thousand
Pascals, and the temperatures reach several thou- Indirect injury is a secondary damage to tissue
sand degrees Celsius, resulting in the local that progresses after the cessation of thermal
destruction of the tissue (Maris and Balibar 2000). ablation stimulus (Muralidharan et al. 2004). It is
136 F. Wu

based on histological evaluation of tissue damage et al. 2005). These cytokines may have direct
at various time points after thermal ablation cytotoxic effects, such as inducing tumor endo-
(Matsumoto et al. 1992). The full extent of the thelial injury and rendering tumor cells more sen-
secondary tissue damage becomes evident 1–7 sitive to heat-induced damage (Watanabe et al.
days after thermal ablation, depending on the 1988; Isbert et al. 2004). However, contrasting
model and energy source used (Wiersinga et al. results are obtained for TNF-α levels in two stud-
2003; Benndorf and Bielka 1997). The exact ies (Evrard et al. 2007a, b; Schell et al. 2002) and
mechanism of this process is still unknown. IL-1 level in one study (Schell et al. 2002), where
However, it may represent a balance of several levels remain unchanged after thermal ablation.
promoting and inhibiting mechanisms, including Cryoablation may cause pathophysiological
induction of apoptosis, Kupffer cell activation changes, which are similar to those observed
and cytokine release. after endotoxin administration (Chapman et al.
Cellular apoptosis may contribute to progres- 2000a, b, c; Wudel et al. 2003). These changes
sive tissue injury after thermal ablation. It is well cause significant increases in capillary permea-
established that apoptosis increases in a bility in the lung, leading to secondary injury
temperature-dependent manner, and tempera- (Washington et al. 2001). It is generally believed
tures between 40 and 45 °C cause inactivation of that all alterations may be associated with post-
vital enzymes, thus initiating tumor cell apopto- cryosurgery activation in the lungs of the nuclear
sis (Barry et al. 1990; Hori et al. 1989). Thermal factor-κB factor and derived cytokines, including
ablation creates a temperature gradient that pro- TNF-α and macrophage inflammatory protein-2,
gressively decreases away from the site of probe along with an increase in serum thromboxane
insertion. The induction of apoptosis at a distance levels (Seifert et al. 2002; Sadikot et al. 2002).
from the heat source may potentially contribute
to the progression of injury. An increased apopto-
sis rate is observed in the liver 24 h after micro- 8.4 Antitumor Immune
wave ablation (Ohno et al. 2001). The stimulation Response After Thermal
of apoptosis may be directly induced by tempera- Ablation
ture elevations, alterations in tissue microenvi-
ronment and the release of various cytokines 8.4.1 HIFU Ablation
after thermal ablation. Kupffer cell activity may
be one of the major factors involved in progres- As shown in Table 8.2, there is increasing evi-
sive injury after thermal ablation (Heisterkamp dence from animal studies that indicate that
et al. 1997). Heat induces Kupffer cell IL-1 HIFU may modulate host antitumor immunity
(Decker et al. 1989) and tumor necrosis factor-α after tumor ablation. Yang and colleagues (Yang
(TNF-α) (Adams and Hamilton 1984) secretion, et al. 1992) used HIFU to treat C1300 neuroblas-
which are known to have in-vivo antitumor activ- toma implanted in mouse flanks, followed by the
ity and increase cancer cell apoptosis (Hori et al. re-challenge of the same tumor cells. A signifi-
1989). Kupffer cells also induce the production cantly slower growth of re-implanted tumors was
of interferon that augments liver-associated natu- observed in these mice compared with the con-
ral killer cell activity (Kirn et al. 1982). trols. After HIFU treatment, the cytotoxicity of
Thermal ablation may induce both regional cytotoxic T lymphocytes (CTLs) and the number
and systemic production of cytokines through of activated tumor-specific CTLs was signifi-
activation of inflammatory cells. Compared with cantly increased in the H22 tumor bearing mice
controls, the circulating level of IFN-γ and vascu- treated with HIFU. Adoptive transfer of the acti-
lar endothelial growth factor levels markedly vated lymphocytes could provide better long-
increase after RFA (Napoletano et al. 2008; term survival and lower metastatic rates in the
Evrard et al. 2007a, b). The increased level of mice re-challenged by the same tumor cells when
IL-1 and TNF-α is also observed after RFA (Ali compared with sham-HIFU and control groups.
8
Table 8.2 Antitumor Immune Response to HIFU Alone in Animal Studies
References Tumor cell line/model HIFU parameters Endpoint Results Additional observations
Yang et al. (1992) C1300 Neuroblatoma 4 MHz Resistance to rechallenge Significant inhibition of tumor Single & repeated
Ajax (A/J) mice 550 W/cm2 growth in mice treated with HIFU could prolong
curative HIFU compared to the survival rates in the
untreated controls tumor bearing mice
Xia et al. (2012) H22 Hepatocarcinoma 9.5 MHz Measurement of CTL Significant increase in CTL Significantly increased
C57BL/6 J mice 5W cytotoxicity & resistance to cytotoxicity number of activated
180–240 s rechallenge after adoptive Superior protection after adoptive tumor-specific CTLs
transfer of the activated immunotherapy
lymphocytes
Xing et al. (2008) B16F10-LucG5 melanoma 3.3 MHz Measurement of CTL Significant increase in CTL HIFU couldn’t increase
C57Bl/6 mice cytotoxicity cytotoxicity in mice treated with the risk of distant
curative HIFU compared to the metastasis
untreated controls
Hu et al. (2007) MC-38 colon adenocarcinoma 3.3 MHz Resistance to rechallenge & Superior protection & tumor- HIFU could enhance
C57Bl/6 mice cytotoxicity assays of splenic specific lymphocyte mediated dendritic cell (DC)
lymphocytes cytotoxicity after both thermal & infiltration in the
mechanical HIFU treatments treated tumor &
Antitumor immunity induced by subsequent migration
Heat-Based Tumor Ablation: Role of the Immune Response

cavitation-based HIFU was to draining lymph


stronger compared to thermal nodes
HIFU
Zhang et al. H22 hepatocarcinoma 9.5 MHz Resistance to rechallenge Significant increase in CTL HIFU treatment alone
(2010) C57BL/6 J 5W Measurement of DC cytotoxicity & DC activation could enhance CTL
180–240 s activation & CTL Superior protection in mice cytotoxicity &
cytotoxicity after immunized with HIFU-generated resistance to
immunization with HIFU- tumor vaccine when compared rechallenge
generated tumor vaccine with the controls
Deng et al. (2010) H22 Hepatocarcinoma 9.5 MHz Measurement of DC Significant increase in DC
C57BL/6 J mice 5W activation & CTL activation & CTL cytotoxicity
180–240 s cytotoxicity with inhibition of tumor growth
Resistance to rechallenge in mice immunized by DCs
after immunization of DCs loaded with HIFU-treated tumor
loaded with HIFU-treated when compared with the controls
tumor
(continued)
137
Table 8.2 (continued)
138

References Tumor cell line/model HIFU parameters Endpoint Results Additional observations
Hu et al. (2005) MC-38 mouse colon 1.1 MHz Measurement of endogenous Release of ATP and HSP-60
adenocarcinoma P+ 12/P− 6.7 MPa danger signals released from from HIFU-treated tumor cells
In-vitro P+ 31.7/P− 10.7 MPa HIFU-treated cells Activation of DCs &
DC 30 or 3 % Activation of APCs macrophages
5 s or 30s
Kruse et al. (2008) Transgenic reporter mouse for 1.5 MHz Skin HSP-70 expression after Upregulated HSP-70 expression
HSP70-Luc2AeGFP 53–353 W/cm2 1 s HIFU treatment after HIFU treatment
Hundt et al. Transfected HSP-70-Luc M21 1 MHz HSP-70 expression in tumor Increased HSP-70 expression
(2007) Melanoma 28–179 W/cm2 cells after either thermal after both thermal stress & HIFU
NIH-3 T3 mouse fibroma stress or HIFU treatment treatment
SCCVII mouse squamous cell Higher expression observed at
carcinoma cells in-vitro HIFU-induced lower
temperatures than thermal stress
alone
Liu et al. (2010) B16 melanoma 3.3 MHz Measurement of DC Significant increase in local DC Sparse-scan HIFU was
C57BL/6 mice P+19.5/P−7.2 MPa infiltration & maturation in infiltration & maturation after more effective than
4s HIFU-treated tumor HIFU treatment compared to the dense-scan HIFU in
controls enhancing DC
infiltration &
maturation in-situ
Zhou et al. (2007) H22 hepatocarcinoma 9.5 MHz Resistance to rechallenge Significant protection in mice A significant increase
Chinese Kun Ming mice 5W after immunization with immunized with HIFU-treated in CD4+ levels &
180–240 s HIFU-treated tumor vaccine tumor compared to heat-treated CD4+/CD8+ ratio in
tumor group both HIFU & thermal
Activation of DCs and groups
Macrophages
F. Wu
8 Heat-Based Tumor Ablation: Role of the Immune Response 139

This is indicative that HIFU ablation could acti- HSP70 (Kruse et al. 2008; Hundt et al. 2007).
vate tumor-specific T lymphocytes, thus inducing This is an intracellular molecular chaperone that
antitumor cellular immunity in the mice (Xia can enhance tumor cell immunogenicity, result-
et al. 2012). Similar results were confirmed in the ing in potent cellular immune responses.
mice implanted with MC-38 colon adenocarci- The potency of DC infiltration and activation
noma and melanoma after HIFU ablation. following mechanical lysis and sparse-scan
HIFU treatment could also induce an enhanced HIFU was much stronger than that from thermal
CTL activity in-vivo, thus providing protection necrosis and dense-scan HIFU exposure, sug-
against subsequent tumor re-challenge (Xing gesting that optimization of a HIFU ablation
et al. 2008). In addition, HIFU could enhance strategy may help in enhancing immune responses
infiltration of dendritic cells (DCs) in the treated after treatment (Liu et al. 2010). Heat and acous-
tumor and subsequent migration to the draining tic cavitation are two major mechanisms involved
lymph nodes. Compared to thermal HIFU treat- in HIFU-induced tissue damage, while cavitation
ment, antitumor immunity induced by mechani- is a HIFU-unique effect when compared with
cal HIFU treatment (being a pulsed HIFU other thermal ablation techniques. It causes
exposure with no significantly elevated tempera- membranous organelles to collapse, including
ture increase in tumor tissue and thermal necro- mitochondria and endoplasmic reticulum, as well
sis) was significantly stronger in terms of DC and as cell and nuclear membranes. This breaks
CTL activation, and a superior protection against tumor cells up into small pieces, by which the
tumor re-challenge was reported (Hu et al. 2007). tumor antigens can remain intact, or it may lead
After HIFU ablation, large amounts of tumor to the exposure of an immunogenic moiety that is
debris remain in-situ, and the host gradually normally hidden in tumor antigens. Zhou and
reabsorbs the debris as the normal process of the colleagues (Zhou et al. 2007) used either heat-
healing response. Using a murine hepatocellular exposed or HIFU-treated H22 tumor vaccines to
carcinoma model, Zhang and colleagues (Zhang inoculate naïve mice. The vaccination times were
et al. 2010) demonstrated that the remaining four sessions once a week for four consecutive
tumor debris induced by HIFU could be immu- weeks, and each mouse was challenged with H22
nogenic, thus an effective vaccine to elicit tumor- tumor cells 1 week after the last vaccination.
specific immune responses. In this study, these They found that the HIFU-treated tumor vaccine
included induction of CTL cytotoxic activity, could significantly inhibit tumor growth and
enhanced activation of DCs and protection increase survival rates in the vaccinated mice,
against lethal tumor challenge in naïve mice. suggesting that acoustic cavitation could play an
When the tumor debris was loaded with imma- important role in stimulating the host antitumor
ture DCs, it could significantly induce DC matu- immune system.
ration, increase cytotoxicity and CTL TNF-α and Emerging clinical results revealed that sys-
IFN-γ secretion, thus initiating a host-specific temic cellular immune response was observed in
immune response after H22 challenge in the vac- cancer patients after HIFU treatment, as shown in
cinated mice (Deng et al. 2010). Immediately Table 8.3. Rosberger and colleagues (Rosberger
after HIFU exposure to MC-38 colon adenocarci- et al. 1994) reported five consecutive cases of
noma cells in-vitro, the release of endogenous posterior choroidal melanoma treated with
danger signals, including HSP60, was observed HIFU. Three patients had abnormal, and two
from the damaged cells. These signals could sub- patients normal CD4+/CD8+ ratios before treat-
sequently activate antigen-presenting cells ment. One week after treatment, the ratio in two
(APCs), leading to an increased expression of co- patients reverted to normal, while another was
stimulatory molecules and enhanced secretion of noted to have a 37 % increase in CD4+ T cells
IL-12 by DCs and TNF-α by macrophages (Hu relative to CD8+ cells. Wang and Sun (2002) used
et al. 2005). In addition, HIFU could upregulate multiple-session HIFU to treat 15 patients with
in-vitro and ex-vitro molecular expression of late stage pancreatic cancer. Although there was
Table 8.3 Antitumor immune response to HIFU alone in clinical studies
140

Reference Tumor/nb. patients HIFU parameters Endpoint Results Additional observations


Rosberger et al. (1994) Choroidal melanoma /5 4.6 MHz Measurement of T cells & CD4+/CD8+ ratio reverted to 37 % increase in CD4+
2 W/cm2 subsets in peripheral blood normal after HIFU in 2 of 3 relative to CD8+ in the
patients with previously remaining one patient
abnormal CD4+/CD8+ ratio
Wang and Sun (2002) Pancreatic cancer/15 0.5–1.6 kW Measurement of T cells, A significant increase in NK An increase in CD3+, CD4+
30–80 s subsets, NK cell activity in cell activity after HIFU & CD4+/ CD8+ ratios in 10
peripheral blood treatment patients after HIFU
treatment, but not
statistically significant
Wu et al. (2004) Osteosarcoma/6 0.8 MHz Measurement of circulating NK, A significant increase in The abnormal levels of
Hepatocarcinoma/5 5–20 kW/cm2 T cells & subsets CD4+ & CD4+/ CD8+ ratios CD3+ returned to normal in
Renal cell carcinoma/5 after HIFU treatment 2 patients, CD4+/CD8+
ratios in 3 patients, CD19+
in 1 patient & NK cells in
1 patient
Zhou et al. (2008) Liver cancer/13 0.8 MHz Measurement of serum A significant decrease in
Sarcoma/3 5–20 kW/cm2 immunosuppressive cytokines serum VEGF, TGF-β1 & β-2
in peripheral blood levels after HIFU treatment
Madersbacher et al. Prostate cancer/5 4 MHz Measurement of HSP-27 A significant increase of
(1998) Bladder cancer/4 1.26–2.2 kW/cm2 expression in HIFU-treated HSP-27 expression after
tumor and prostate tissue HIFU treatment compared to
the controls
Kramer et al. (2004) Prostate cancer/6 4 MHz Measurement of HSP A significant upregulated A significant decrease in
1.26–2.2 kW/cm2 expression & Th1- & Th2- expression of HSP-72, TIL-released Th2-
cytokine release from TILs in HSP-73, glucose GRP-75 & cytokines (IL-4, -5, -10)
HIFU-treated tumor GRP-78 after HIFU treatment
Significant increase in
TIL-released IL-2, IFN-γ &
TNF-α after HIFU treatment
F. Wu
8

Wu et al. (2007a, b) Breast cancer/23 4 MHz Measurement of expression of 100 % positive rate of Varied expressions of ER,
1.26–2.2 kW/cm2 13 proteins on tumor cells HSP-70 in HIFU-treated PR, CA15-3, VEGF,
including HSPs cancer cells compared to the TGF-β1, TGFβ2, IL-6,
control IL-10 and EMA in HIFU
treated tumor, with no
expression of PCNA,
MMP-9 and CD44v6
Xu et al. (2009) Breast cancer/23 4 MHz Measurement of APC A significant increase in local
1.26–2.2 kW/cm2 infiltration & activation in infiltration & activation of
HIFU-treated tumor DCs & macrophages
compared to the control
Liu et al. (2010) Breast cancer/23 4 MHz Measurement of TIL infiltration A significant decrease in
1.26–2.2 kW/cm2 & activation in HIFU-treated tumor-infiltrating CD3+,
tumor CD4+, CD8+, CD4+/CD8+, B
lymphocytes, NK cells,
FasL+, Granzyme+ and
perforin+ TILs when
compared with the control
Heat-Based Tumor Ablation: Role of the Immune Response
141
142 F. Wu

an increase in average numbers of NK cells, T and CD86 molecules. Activated APCs may take
lymphocytes and subsets in ten patients after up the HSP-tumor peptide complex, which
HIFU treatment, a significant statistical differ- remains in the tumor debris and present the chap-
ence was only observed in NK cell activity before eroned peptides directly to tumor-specific T lym-
and after HIFU treatment (p < 0.05). Wu and col- phocytes with high efficiency, resulting in potent
leagues (Wu et al. 2004) observed changes in cir- cellular immune responses against tumor cells
culating NK, T lymphocyte and subsets in 16 after HIFU treatment.
patients with solid malignancy before and after Furthermore, HIFU could induce significant
HIFU treatment. The results showed a significant infiltration of TILs in human breast cancer,
increase in the CD4+ T lymphocyte population including CD3+, CD4+, CD8+, B lymphocytes
(p < 0.01) and the ratio of CD4+/CD8+ cells and NK cells. The number of the activated CTLs
(p < 0.05) after HIFU treatment. The abnormal expressing FasL+, granzyme+ and perforin+ sig-
levels of CD3+ lymphocytes returned to normal nificantly increased in the HIFU-treated tumor,
in two patients, CD4+/CD8+ ratio in three patients, suggesting that specific cellular antitumor immu-
CD19+ lymphocytes in one patient and NK cells nity could be locally triggered after HIFU treat-
in one patient, in comparison to the values in the ment (Lu et al. 2010).
control group. In addition, serum levels of immu-
nosuppressive cytokines, including VEGF, TGF-
β1 and TGF-β2, were significantly decreased in 8.4.2 Radiofrequency Ablation
peripheral blood of cancer patients after HIFU
treatment, indicating that HIFU may decrease With regards to minimally invasive therapies,
tumor-induced immunosuppression and renew RFA is only one technique that has been widely
host antitumor immunity (Zhou et al. 2008). used in the clinical management of solid tumors,
Clinical evidence suggests that HIFU treat- particularly in hepatocellular carcinoma (HCC).
ment may also enhance local antitumor immunity As coagulative necrosis is immediately induced
in cancer patients. Kramer and colleagues in a targeted tumor after thermal ablation,
(Madersbacher et al. 1998; Kramer et al. 2004) necrotic cell death can be recognized by the
found that HIFU treatment could alter the presen- immune system as a result of dangerous events,
tation of tumor antigens in prostate cancer according to the “danger” model of immunity by
patients, which was most likely to be stimulatory. Matzinger (Gallucci et al. 1999; Matzinger
Histological examination showed significantly 2002). It is also accompanied by the release of
upregulated expression of HSP72, HSP73 and “danger signals” from the heat-stressed cells,
glucose regulated protein (GRP) 75 and 78 at the such as acute phase proteins, pro-inflammatory
border zone of HIFU treatment in prostate can- cytokines and heat shock proteins (HSPs), thus
cer. Heated prostate cancer cells exhibited developing a temporary inflammatory stress.
increased Th1-cytokine (IL-2, IFN-γ, TNF-α) This stress may be associated with positive pro-
release, but decreased Th2-cytokine (IL-4, -5, cesses similar to the healing of injured tissues,
-10) release from tumor infiltrating lymphocytes but could also lead to the stimulation of tumor
(TIL). The upregulated expression of HSP70 was growth (Gravante et al. 2009). After RFA treat-
confirmed in the tumor debris of breast cancer ment, a moderate and temporary systemic inflam-
after HIFU ablation (Wu et al. 2007a, b), indicat- matory response has been observed in cancer
ing that HIFU may modify tumor antigenicity to patients, as demonstrated by the increase in
produce a host immune response. plasma levels of pro-inflammatory cytokines and
Xu and colleagues (2009) found the number acute phase reactants (Evrard et al. 2007a, b;
of tumor-infiltrating APCs, including DCs and Schell et al. 2002; Meredith et al. 2007; Schueller
macrophages, increased significantly along the et al. 2003; Fietta et al. 2009).
margin of HIFU-treated human breast cancer, HSPs are families of highly conserved pro-
with an increased expression of HLA-DR, CD80 teins involved in mechanisms of cell repair. They
8 Heat-Based Tumor Ablation: Role of the Immune Response 143

are intracellular molecular chaperones that physi- with increased infiltration of DCs after subtotal
ologically bind tumor peptide antigens and RF ablation (Dromi et al. 2009). These results
enhance tumor cell immunogenicity (Pockley suggest that the generation of heat-altered tumor
2003). APCs take up HSP-tumor peptide com- antigens, in combination with the “dangerous
plex and present the chaperoned peptides directly signals”, may help to overcome immune toler-
to tumor-specific T lymphocytes with high effi- ance or allergy towards the remaining tumor.
ciency, resulting in potent cellular immune The effects of RFA on antitumor T cell
responses against tumor cells (Todryk et al. responses have been studied in animal models. A
2003). Around the necrotic ablated area, RFA local influx of immune cells was observed after
produced sub-lethal injury in the zone of RFA in tumor-free domestic pigs and in the livers
transition that showed apoptosis, and increased of rabbits implanted with epithelial tumors
HSP70 expression in the liver of normal swine (Hänsler et al. 2002). The latter was located in the
(Schueller et al. 2004). Schueller and colleagues periphery of the coagulated area, and consisted of
found that there was an increased synthesis and lymphocytic and plasma cell infiltrates.
cell surface expression of HSPs (HSP70, 90) Concomitantly, a specific T cell proliferative
after RFA in nude rats bearing human hepatocel- response to the tumor cells was also detected in
lular carcinoma (Rai et al. 2005). In addition, the peripheral blood of RFA-treated animals
large amounts of tumor debris could induce local (Wissniowski et al. 2003). Den Brok and col-
infiltration of activated DCs, the most potent leagues (2004) found that a weak, but detectable,
APC for induction of adaptive immunity against immune response was present after RFA in mice
cancer (Melief 2008). Activating signals, includ- bearing ovalbumin-transfected melanoma. This
ing necrotic tumor cells and HSPs, could induce antitumor immunity was mediated by antigen-
the progression of infiltrating DCs from an imma- specific CD8+ T cells, and adoptive transfer of
ture to a mature stage, resulting in the presenta- splenocytes could induce partial protection
tion of tumor antigens by mature DCs to naïve T against tumor challenge in syngenic mice.
lymphocytes in a MHC-restricted fashion (Lutz Compared to surgical resection and control
and Schuler 2002). Ali and colleagues demon- groups, RFA could efficiently stimulate activa-
strated that a transient function of myeloid DCs tion and proliferation of splenocytes in mice
could be activated in HCC patients 7–14 days bearing H22 tumor, and the cytotoxicity of sple-
after RFA, with an increased ability to stimulate nocytes to tumor cells was significantly enhanced
CD4+ T cells (Ali et al. 2005). Up to 7 % of DCs in RFA-treated animals, with an increased secre-
present in the draining lymph nodes contained tion of IL-2 and IFN-gamma (Zhang et al. 2006).
tumor antigens in the ablated tumor after After in-situ RFA of liver tumor, resistance to
RFA. Compared to untreated HCC and normal local and systemic tumor rechallenge was
liver tissue, expression of costimulatory mole- increased in mice bearing CC531 colon carci-
cules, such as CD80 and CD86, was significantly noma (van Duijnhoven et al. 2005). However, no
enhanced by incubation with RFA-treated HCC inhibitory effect on tumor growth was observed
(den Brok et al. 2006). Similar results were also in the nearby untreated liver tumors.
demonstrated by Zerbini and colleagues (2008) Similar results have been also demonstrated in
in HCC patients; indicative that local tumor abla- cancer patients treated with RFA. Zerbini and
tion could lead to efficient antigen loading, colleagues (2006) showed convincing evidence
migration and maturation of APCs, including that RFA could activate a systemic antitumor T
DCs and monocytes. Direct evidence has recently cell response in 20 HCC patients. Using an
been found that RFA could induce weak tumor- ELISPOT assay, the reactivity of circulating T
induced immunity in a murine tumor model har- cells to autologous HCC lysate was assessed
boring APC infiltration and amplification, and before and after RFA treatment. They found that
that enhanced systemic antitumor T cell immune the specific T cell response was increased in three
responses and tumor regression was associated patients immediately after RFA, compared to no
144 F. Wu

patients before treatment. Importantly, this boost- responses against tumors, inhibited tumor devel-
ing effect still persisted at 4 weeks after RFA, and opment and lung metastasis, and reduced myeloid-
the number of patients showing the same T cell derived suppressor cells (Habibi et al. 2009).
response increased up to nine. These data were
confirmed by another study where both HCC and
colorectal liver metastasis were treated with RFA 8.4.3 Laser Ablation
(Hänsler et al. 2006). After RFA treatment, both
HCC and colorectal cancer cells could signifi- In addition to local destruction with thermal
cantly stimulate a specific immune response, energy, LA can induce an immunogenic effect
resulting in an increase of circulating CD4+ and on cancer in both animal tumor models and
CD8+ T cells and cytotoxic activity. In contrast, cancer patients. Compared to surgical resec-
one study observed a decrease in circulating tion, LA could reduce metastatic liver tumor
CD3+ and CD4+ T cells after RFA treatment in spread in rats bearing a liver adenocarcinoma
metastatic cancer patients, together with no (Möller et al. 1998). Furthermore, with HSP70
change in HCC patients. However, RFA induced shifts from the cytoplasm to the nucleus in
trafficking of naïve and memory CD62L+ T cells LA-treated liver cancer cells, an increase in
from the circulation to tissues, and enhanced the HSP70 immunoreactivity in tumors was
function of T cells, including in-vitro responses observed, leading to increased numbers of
to phytohaemagglutinin (PHA) and tumor associ- tumor-infiltrating macrophages and an
ated MUC1 antigen (Napoletano et al. 2008). increased presence of HSP70 in the membrane
In order to improve the RFA-induced weak and cytoplasm of these macrophages (Ivarsson
immune response, the combination of RFA with et al. 2003). LA could also induce a significant
immunotherapy has been investigated in labora- increase in HSP70 expression in the colorectal
tory settings. RFA could be efficiently combined liver metastasis mouse model (Nikfarjam et al.
with immune modulation by anti-CTLA-4 anti- 2005a, b, c) and prostate cancer (Paulus et al.
bodies or regulatory T cell depletion. These com- 1993; Rylander et al. 2006). While two inde-
bination treatments protected mice from the pendent adenocarcinomas were implanted into
outgrowth of tumor challenges and led to in-vivo both lobes of the liver in rats (one as a control
enhancement of tumor-specific T cell numbers, in the right and one treated with LA in the left
which produced more IFN-γ upon activation (den lobe), the control tumor volumes were signifi-
Brok et al. 2006). Saji and colleagues (2006) cantly smaller in the LA group than those in the
demonstrated that RFA, plus intratumoral injec- hepatic resection group. The expression of CD8
tion of naïve DCs, could induce DC migration to and B7-2 (CD86) was significantly higher in
regional lymph nodes and induce adoptive antitu- the control tumor after LA (Isbert et al. 2004).
mor immunity in a mouse tumor model. The com- Moreover, compared to surgical extirpation,
bination of RFA with IFN injection could complete eradication of reimplanted tumor, as
significantly increase antitumor effects in an well as increased local infiltration of ED1 mac-
orthotopic murine model with squamous cell car- rophages and CD8 lymphocytes, were observed
cinoma, upon comparison with single therapy and in the LA group 48 days after tumor challenge
control groups (Saito et al. 2005). In this study, (Ivarsson et al. 2005). Overall, this suggests
the RFA treatment stimulated tumor specific T that LA could enhance antitumor immune
cells to move to tumor sites, whereas IFN acti- response to eradicate a challenging tumor,
vated DCs and enhanced antigen presentation. All which might be associated with increased num-
of the mice survived for 50 days in the combined bers of tumor-infiltrating macrophages and
therapy group. Using both neu-overexpressing CD8+ lymphocytes.
mouse mammary carcinoma in FVBN202 trans- Immunological assays followed by LA proce-
genic mice and 4 T1 tumors in Balb/c mice, RFA dure for cancer patients are still limited in the
treatment was followed by the administration of clinical setting. An early systemic inflammatory
intratumor IL-7 and IL-15. This induced immune reaction was observed after LA in patients with
8 Heat-Based Tumor Ablation: Role of the Immune Response 145

malignant liver tumors (Kallio et al. 2006). secondary tumor growth after rechallenge in cryo-
Serum level of IL-6, TNFRI and CRP increased treated rats (Hoffmann et al. 2001). Cryoablation
significantly up to 72 h after LA procedure, while alone couldn’t directly cause a tumor-specific
the TNF-α, IL-1β and IL-10 levels remained CTL response and a protective anti-metastatic
unchanged. Using an IFN-γ secretion assay and impact when compared to cryotherapy combined
flow cytometry, Vogl and colleagues (2009) stud- with subsequent in-situ injection of immature
ied peripheral T lymphocyte (CD3+, CD4+, CD8+) DCs (Udagawa et al. 2006; Machlenkin et al.
activation against autologous tumor tissue, and T 2005). Moreover, immunosuppressive effects
cell cytotoxicity against allogenic colorectal can- induced by cryoablation on host antitumor immu-
cer cells (CaCo). This was carried out before and nity were also observed in tumor-bearing animals,
after LA in patients with liver metastases of resulting in a decreased resistance to a secondary
colorectal cancer. They found that tumor-specific tumor challenge and an increase in pulmonary
cytotoxic T cell stimulation was detected after metastases after cryoablation (Shibata et al. 1998;
LA treatment, with a significant increase in cyto- Hanawa 1993; Miya et al. 1987). This has led to
lytic activity against CaCo cells, indicative that controversy whether a cryo-immunologic
LA could trigger T lymphocyte-mediated antitu- response would exist after cryoablation of malig-
mor immune response against autologous tumor nant tissue.
tissue in patients. Recently, due to a better understanding of the
relationships between the innate and adaptive
arms of the immune response, more detailed
8.4.4 Cryoablation studies of the mechanism behind cryo-
immunology have offered insight into why cryo-
In the early introduction of cryoablation to clinical ablation may alternate between immune
practice, there were occasional reports of patients enhancement and immune suppression. It is evi-
with spontaneous regression of tumor metastases dent that several changes induced by cryoabla-
after ablation of a primary tumor, suggesting a tion (cytokine profile, availability of tumor
potential systemic benefit to a local therapy (Sabel antigens processed by APCs, mechanism of cell
2009). However, the mechanisms behind the exis- death (apoptosis or necrosis)), and the subsets of
tence of a cryo-immunologic response were phagocytic cells (DCs or macrophages) respon-
unclear because immunologic assays were limited sible for ablated cell clearance, may either posi-
at the time of many of these observations. tively or negatively impact the immune response
Subsequently, an immune response induced by (Sabel 2009). For instance, although apoptosis
cryoablation was investigated using a variety of and necrosis are the primary mechanisms of
animal tumor models. The results revealed that tumor cell death, they have a significantly differ-
tumor-specific immunity, as measured by resis- ent impact on the immune response (Viorritto
tance to rechallenge in tumor-bearing animals et al. 2007). Apoptosis results in the uptake of
undergoing cryoablation of primary tumor, was cellular debris without causing inflammation or
significantly greater in the cryoablation-treated releasing the intracellular contents. APCs that
animals when compared with surgical excision or take up the apoptotic cells do not only not gener-
naïve animals (Redondo et al. 2007; den Brok ate an immune response, but also can lead to
et al. 2006; Sabel et al. 2005). In addition, cryoab- clonal deletion and anergy (Viorritto et al. 2007;
lation could significantly inhibit the growth of Peng et al. 2007; Savill et al. 2002; Liu et al.
contralateral tumors (Joosten et al. 2001; Shibata 2002). In contrast, necrotic cell death is charac-
et al. 1998), and reduce metastatic deposits in the terized by cellular breakdown and release of
lung and liver in tumor-bearing animals (Müller intracellular contents, many of which are danger
et al. 1985; Urano et al. 2003). signals. These signals promote cross-presentation,
On the contrary, some studies found that cryo- maturation of DCs and ultimately the activation
ablation failed to induce antitumor immune of antigen-specific T cells (Gallucci et al. 1999;
responses. There was no significant inhibition on Sauter et al. 2000; Skoberne et al. 2004). As both
146 F. Wu

necrosis and apoptosis play a role in tumor cell indicating that cryoablation could improve tumor-
death after cryoablation, the relative contribution specific cytolytic activity of CTLs in prostate can-
of necrosis and apoptosis in the death of the cer patients. This immune response was only
tumor cells may shift the immune response from sufficiently maintained for a period of 4 weeks.
stimulatory to suppressive. Cryoablative tech- However, when cryoablation was combined with
niques that result in large areas of apoptotic cell granulocyte macrophage colony-stimulating factor
death, as opposed to necrosis, may result in (GM-CSF) administration to treat metastatic hor-
immunosuppression. However, some studies mone refractory prostate cancer, the response
have suggested that apoptotic tumor cells may be could last for at least 8 weeks (Si et al. 2009).
superior to necrotic cells in stimulating an antitu- For the case of freezing large tumors, cryoabla-
mor immune response (Rock et al. 2006; Scheffer tion may cause a serious complication known as
et al. 2003; Schnurr et al. 2002). “cryoshock”, a syndrome of coagulopathy, dis-
In addition to animal models, some clinical seminated intravascular coagulation and multior-
studies have recently attempted to reveal how gan failure (Seifert and Junginger 2004). As it is
cryoablation has profound effects on the immune similar to those observed after endotoxin adminis-
system in cancer patients. Osada and colleagues tration and other systemic inflammatory stimuli,
(2007) measured serum levels of IL- 2, IL-4, IL-6, cryoshock is believed to be caused by the systemic
IL-10, TNF-α and IFN-γ in 13 patients with unre- release of inflammatory cytokines after cryoabla-
sectable hepatic tumors before and after cryoabla- tion, including IL-1, IL-6 and TNF-α (Chapman
tion. Decreased levels of serum tumor markers and et al. 2000a, b, c; Sadikot et al. 2002; Seifert et al.
local tumor necrosis detected on CT scan were 2002). This is different from the RFA-treated liver
observed in all patients, including five cases who tissue, where there is a coagulative destruction of
presented evidence of necrosis in metastatic the hepatocyte organelles within an intact plasma
tumors away from the treated lesions. Serum IL-6 membrane (Chapman et al. 2000a, b, c). Cryoshock
level was increased in all patients after cryoabla- remains rare in the cryoablation of renal and pros-
tion, but no change in the IL-2 level was observed. tate tumors, but a more common side effect of
There was a significant increase in serum TNF-α hepatic cryoablation.
level and Th1/Th2 ratios in the patients showing
necrosis of secondary tumors. The effects of cryo-
ablation on humoral immune compartments were 8.4.5 Microwave Ablation
also analyzed by Ravindranath and colleagues
(2002) in 35 patients with liver metastases origi- The effect of microwaves on immune cells was
nated from colon cancer. They found an increase initially investigated in murine B16 melanoma
in the production of IgM antibodies against tumor- models. Microwave hyperthermia, in combination
released gangliosides. Interestingly, these antibod- with ethanol injection, could significantly prolong
ies were not significantly increased in patients the survival of the tumor bearing mice with an
undergoing RFA or routine surgery. Si and col- increased infiltration of T lymphocytes and NK
leagues (2008) observed a specific cytotoxic T cell cells in the ablated melanoma (Nakayama et al.
response induced by cryoablation in 20 patients 1997a, b). Whole body microwave hyperthermia
with high-risk prostate cancer. Four weeks after could cause a significant enhancement in TNF-α
cryoablation, there was a significant increase in secretion in murine peritoneal macrophages and
serum TNF-α and IFN-γ levels, as well as Th1/ splenic T lymphocytes (Fesenko et al. 1999). Yao
Th2 ratios, when compared with the values before and Yang (2007) found that a murine CT-26 tumor
cryoablation. However, no changes were observed treated with microwave ablation could sensitize
in the serum levels of IL-4 or IL-10. Tumor- immature DCs, which subsequently induced in-
specific T cell responses were significantly vitro proliferation of T cells and activated CTL
increased 4 weeks after cryoablation, while cytotoxicity. In addition, the sensitized DCs could
peripheral blood mononuclear cells were co-incu- significantly inhibit in-vivo tumor growth and
bated with human prostate cancer cells (LNCaP), prolong the survival of the mice.
8 Heat-Based Tumor Ablation: Role of the Immune Response 147

Clinical studies related to the immune response ture, could enhance the cellular immune response
were initially conducted on prostate cancer treated once compared with the control group, improv-
by microwave energy. A transient, yet significant ing survival time and reducing local recurrence in
increase in the CD4+/CD8+ ratio, PHA and Con-A HCC patients (Lin et al. 2005; Han et al. 2009).
transformation indices was observed after micro-
wave hyperthermia in 15 prostate cancer patients, Conclusion
and the peak effect of this immune response was As a minimally invasive therapy, thermal abla-
noted at 2 months, followed by a subsequent tion has been increasingly used in clinical
decrease (Szmigielski et al. 1991). Fan and col- practice for the local treatment of solid malig-
leagues (1996) treated 58 patients with malignant nancy. Beyond optimization of technical and
bone tumors by surgical procedure in combina- physiological parameters, it is clear that ther-
tion with microwave hyperthermia and adjuvant mal ablation should be undertaken when there
immunotherapy. The immune response, including is precise knowledge not only of the number
T cell subsets, IL-2 and sIL-2, was monitored and location of the lesions, but also of the bio-
3–38 months (mean 19 months) after the com- logical characteristics and natural history of
bined therapies. The immune function was sig- the tumor. The goal of tumor therapy is that all
nificantly improved in the majority of the patients, cancer cells should be completely killed in the
though oncologic outcome was similar to that patient’s body. A similar multidisciplinary
obtained by limb-saving procedure. approach including other modalities is impor-
The MWA-induced immune response was tant in the treatment of solid malignancies. For
studied in majority by Dr Dong and colleagues in patients with cancer, the therapeutic strategy
78 patients with hepatocellular carcinoma. for the disease should be a multiple treatment
Ultrasound-guided core needle biopsy was per- plan, which includes local treatments, such as
formed after treatment for determining the local surgery and radiotherapy, and systemic ther-
infiltration of immunocytes within the treated apy, such as chemotherapy and immunother-
lesion. The results demonstrated a significantly apy. Thus, success achieved in the application
increased infiltration of T lymphocytes, memory of thermal ablation is mainly dependent not
T lymphocytes, NK cells and monocytes in the only on the ablation technique, but also on a
ablated tumor, with no change in B lymphocytes, better understanding of the natural character-
suggesting that MWA could only enhance cellu- istics of tumors.
lar immune response in HCC patients (Zhang A review of the literature strongly supports
et al. 2002; Dong et al. 2002, 2003). This response academic evidence that thermal ablation may
was maximal on the third day after thermal abla- elicit a systemic antitumor immune response.
tion, but persisted to day 30. The extent of infil- It may lead to a post-ablative procedure that
tration was negatively related to serum reduces, or perhaps eliminates distant disease,
α-fetoprotein and tumor size (Dong et al. 2002). and prevents local recurrence through the
However, interestingly, patients with a high immune system in cancer patients who have
degree of immune cell infiltration in the treated had original dysfunction of antitumor immu-
tumor had lower recurrence rates than those with nity after ablation. Evidence ranges from
low levels of infiltration, and there is a statistically anecdotal observations in a clinical setting, a
significant correlation between survival outcome variety of animal models and correlative
and the extent of immunocyte infiltration (Dong immune studies in patients undergoing ther-
et al. 2003). In addition, IL-6 serum levels, IL-1ra mal ablation. It is not surprising that there is
and C-reactive protein were significantly elevated great concern about a close relationship
1 day after laparoscopic MWA, and returned to between thermal ablation and antitumor
the preoperative levels at day 7 postoperatively immune response, as thermal ablation may
(Sadamori et al. 2003). Furthermore, MWA com- have the potential to be both local and sys-
bined with either local injection of staphylococin, temic therapies. However, the generation of an
or oral uptake of Shenqi (a Chinese herb) mix- antitumor immune response is complex, and
148 F. Wu

several factors can not only preclude the Babaian RJ, Donnelly B, Bahn D, Baust JG, Dineen M,
development of a positive response, but tilt Ellis D, Katz A, Pisters L, Rukstalis D, Shinohara K,
Thrasher JB (2008) Best practice statement on cryo-
that response towards immunosuppression surgery for the treatment of localized prostate cancer.
that is observed in cryoablation. As the clini- J Urol 180:1993–2004
cal use of thermal ablation expands, it becomes Barry MA, Behnke CA, Eastman A (1990) Activation of
increasingly crucial that we better understand programmed cell death (apoptosis) by cisplatin, other
anticancer drugs, toxins and hyperthermia. Biochem
the means by which thermal ablation modu- Pharmacol 40:2353–2362
lates the immune system in cancer patients. Beland M, Mueller PR, Gervais DA (2007) Thermal abla-
Although the mechanism for ablation- tion in interventional oncology. Semin Roentgenol
42:175–190
induced immunologic changes is still unclear,
Benndorf R, Bielka H (1997) Cellular stress response:
several possibilities have been hypothesized stress proteins – physiology and implications for can-
based on previous study results. Firstly, host cer. Recent Results Cancer Res 143:129–144
immune suppression, induced by tumor cells, Cabibbo G, Latteri F, Antonucci M, Craxì A (2009)
Multimodal approaches to the treatment of hepatocel-
may be decreased or relieved after thermal abla-
lular carcinoma. Nat Clin Pract Gastroenterol Hepatol
tion as the tumor is completely ablated, leading 6:159–169
to renewed host antitumor immunity. Secondly, Carrafiello G, Laganà D, Mangini M, Fontana F, Dionigi
thermal ablation may modify tumor antigenicity G, Boni L, Rovera F, Cuffari S, Fugazzola C (2008)
Microwave tumors ablation: principles, clinical appli-
and upregulate expression of HSPs, which act
cations and review of preliminary experiences. Int J
as tumor vaccines to produce potent cellular Surg 6:S65–S69
immune responses. Thirdly, cytokines are Chapman WC, Debelak JP, Blackwell TS, Gainer KA,
secreted by immune cells at the inflammatory Christman JW, Pinson CW, Brigham KL, Parker RE
(2000a) Hepatic cryoablation, but not radiofrequency
margin of the ablation-treated region, present-
ablation, results in lung inflammation. Arch Surg
ing a milieu for the development of mature 135:667–672
CTLs. Finally, large amounts of cellular debris Chapman WC, Debelak JP, Blackwell TS, Gainer KA,
are gradually phagocytized by macrophages Christman JW, Pinson CW, Brigham KL, Parker RE
(2000b) Hepatic cryoablation-induced acute lung
and other cells that can function as APCs.
injury: pulmonary hemodynamic and permeability
It is increasingly apparent that thermal effects in a sheep model. Arch Surg 135:667–672
ablation alone may not be sufficient to gener- Chapman WC, Debelak JP, Wright Pinson C, Washington
ate a clinically relevant immune response and MK, Atkinson JB, Venkatakrishnan A, Blackwell TS,
Christman JW (2000c) Hepatic cryoablation, but not
to consistently stimulate the host immune sys-
radiofrequency ablation, results in lung inflammation.
tem. A strategy to combine thermal ablation Ann Surg 231:752–761
with active immunological stimulation, such Chaussy C, Thuroff S, Rebillard X, Gelet A (2005)
as immunoadjuvants, may augment the effi- Technology insight: high-intensity focused ultrasound
for urologic cancers. Nat Clin Pract Urol 2:191–198
cacy of ablation-induced antitumor immunity
Clement GT (2004) Perspectives in clinical uses of
specifically against the targeted tumors, if the high-intensity focused ultrasound. Ultrasonics 42:
tumor destruction releases tumor antigens or 1087–1093
improves tumor immunogenicity. This com- Curley SA (2001) Radiofrequency ablation of malignant
liver tumors. Oncologist 6:14–23
bined approach may become an important part
Decadt B, Siriwardena AK (2004) Radiofrequency abla-
in the thermal ablation of solid malignancy. tion of liver tumours: systematic review. Lancet Oncol
5:550–560
Decker T, Lohmann-Matthes ML, Karck U, Peters T,
Decker K (1989) Comparative study of cytotoxicity,
References tumor necrosis factor, and prostaglandin release after
stimulation of rat Kupffer cells, murine Kupffer cells,
Adams DO, Hamilton TA (1984) The cell biology of mac- and murine inflammatory liver macrophages. J Leukoc
rophage activation. Annu Rev Immunol 2:283–318 Biol 45:139–146
Ali MY, Grimm CF, Ritter M, Mohr L, Allgaier HP, Weth den Brok MH, Sutmuller RP, van der Voort R, Bennink EJ,
R, Bocher WO, Endrulat K, Blum HE, Geissler M Figdor CG, Ruers TJ, Adema GJ (2004) In situ tumor
(2005) Activation of dendritic cells by local ablation ablation creates an antigen source for the generation of
of hepatocellular carcinoma. J Hepatol 43:817–822 antitumor immunity. Cancer Res 64:4024–4029

Вам также может понравиться