Skip to main content
Clinical and Developmental Immunology logoLink to Clinical and Developmental Immunology
. 2011 Dec 29;2011:160250. doi: 10.1155/2011/160250

More Than Just Tumor Destruction: Immunomodulation by Thermal Ablation of Cancer

Sebastian P Haen 1, 2,2,*, Philippe L Pereira 3, Helmut R Salih 1, Hans-Georg Rammensee 2, Cécile Gouttefangeas 2
PMCID: PMC3254009  PMID: 22242035

Abstract

Over the past decades, thermoablative techniques for the therapy of localized tumors have gained importance in the treatment of patients not eligible for surgical resection. Anecdotal reports have described spontaneous distant tumor regression after thermal ablation, indicating a possible involvement of the immune system, hence an induction of antitumor immunity after thermoinduced therapy. In recent years, a growing body of evidence for modulation of both adaptive and innate immunity, as well as for the induction of danger signals through thermoablation, has emerged. Induced immune responses, however, are mostly weak and not sufficient for the complete eradication of established tumors or durable prevention of disease progression, and combination therapies with immunomodulating drugs are being evaluated with promising results. This article aims to summarize published findings on immune modulation through radiofrequency ablation, cryoablation, microwave ablation therapy, high-intensity focused ultrasound, and laser-induced thermotherapy.

1. Introduction

The local application of high or low temperatures is frequently used to induce protein denaturation, tissue necrosis, and tumor destruction in order to curatively or palliatively treat localized primary or secondary tumors [1]. Thermal ablative procedures in clinical practice comprise radiofrequency (RF) ablation, microwave ablation therapy (MWA), high-intensity focused ultrasound (HIFU), and laser-induced thermotherapy (LITT) with the use of high temperatures, as well as cryoablation with induction of low temperatures. Primarily all these techniques were applied for the palliative treatment of patients not eligible for surgical resection or frail patients with a reduced functional reserve capacity and many comorbidities [2, 3]. Local thermal ablative methods present several advantages as compared with surgery which include less damage to surrounding healthy tissue, greater patient comfort, for example, less pain and limitation in exercise due to wound healing, improved cosmetic results, and—in times of critical financial situations in the medical facilities—reduced cost and shorter periods of hospitalization [2, 4]. For selected patients, local thermoablative techniques have similar clinical outcomes as compared with historical controls of surgical resection [58]. However, except for early hepatocellular carcinoma, no large randomized clinical trial has been performed to directly compare thermoablation and surgical resection so far [9]. In clinical routine, thermal ablation techniques have gained further importance in the treatment of small tumors as an alternative to surgical resection. Their application is limited by the size of the tumor lesions since large tumors (>4 cm) require more expanded treatment with an increased rate of complications and local recurrence [10, 11].

The choice of the most suitable thermal ablation modality depends on different premises. Tumors located in tissues with a high impedance like lung or bone can be better treated with cryoablation or MWA [1214]. Other factors for the assignment to an ablation modality depend on patient characteristics and comorbidities, on the physician's choice and availability of a certain method in a respective hospital, as well as on tumor location and relative position to other anatomic structures [1]. The clinical indications and characteristic features of each technique are summarized in Table 1.

Table 1.

Thermal ablative methods in clinical use for the treatment of cancer and described effects on the immune system.

Immune Modulation
Treatment Indication Characteristics/principle Component Effect Ref. Species
Radiofrequency Clinical indication: primary and Mechanism: application of Cytokines + [5154] Human
(RF) ablation secondary malignancies in liver, alternating RF current through tip Danger + [5561] Animal
kidney, lung, and bone applicator placed around and in signals + [62, 63] Human
[2, 8, 17, 49] tumor tissue resulting in heat and Granulocytes + [64, 65] Animal
coagulative necrosis [54, 66] Human
Experimental application: tumors NK cells + [61] Animal
of the breast [50] Approach: percutaneous, open, + [67] Human
and intraoperative Monocytes/ + [66] Animal
Macrophages
Image guidance: US, CT, and MRI DC + [68, 69] Animal
[51] Human
T cells* + [49, 57, 65, 6874] Animal
+ [70, 7578] Human
Treg [54] Human
B cells + [75] Human
Antibodies* + [78] Human

Cryoablation Clinical indication: primary and Mechanism: application of cold Cytokines + [39, 83] Animal
secondary malignancies in liver, through gaseous evaporation at + [36, 84, 85] Human
kidney, and prostate, as well as the tip of a cryoprobe. Repetitive Danger ?
dermatologic and ophthalmologic freezing and thawing cycles lead to signals
tumors [4, 33, 79, 80]. direct cellular damage through ice Granulocytes + [86] Animal
Experimental Application: tumors crystals, vascular and endothelial NK cells + [83] Animal
of the breast [81]. injury, and eventually thrombosis + [87] Human
and ischemia [79, 82] resulting in Monocytes/ + [86, 88] Animal
coagulative necrosis and apoptosis Macrophages
at the ablation margin DC + [69] Animal
Approach: percutaneous, open, T cells + [38, 69, 79, 83, 8998] Animal
intraoperative + [85, 87, 99102] Human
Treg + [103, 104] Animal
[105] Human
Image guidance: US, CT, and MRI B cells + [91] Animal
Antibodies* + [86, 106113] Animal
+ [41, 87, 114116] Human

Microwave Clinical indication: mainly used for Mechanism: application of Cytokines ? [117] Animal
ablation therapy treatment of HCC, but also other microwaves through tip applicator Danger +
(MWA) primary and leading to coagulative necrosis signals
secondary [21] Granulocytes ?
malignancies of the liver [21, 22] NK cells + [118] Animal
Approach: percutaneous, open, + [119] Human
and intraoperative Monocytes/ + [119] Human
Macrophages
DC ?
Image guidance: US, CT, and MRI T cells + [118] Animal
+ [22, 119, 120] Human
Treg ?
B cells + [22] Human
Antibodies ?

High-intensity Experimental application: primary Mechanism: application of focused Cytokines ?
focused and secondary malignancies in ultrasound beams of Danger + [122] Human
Ultrasound breast, liver, pancreas, kidney, high-intensity resulting in signals
(HIFU) bone, prostate, and coagulative necrosis Granulocytes ?
soft-tissue-tumors [121] NK cells + [123] Human
Monocytes/ ?
Approach: noninvasive Macrophages
DC ?
Image guidance: noninvasive T cells + [121, 124] Animal
real-time US + [27, 125, 126] Human
Treg ?
B cells + [27] Human
Antibodies ?

Laser induced Clinical indication: broadly applied Mechanism: placement of multiple Cytokines + [130] Human
thermotherapy for photocoagulation in retinal simultaneous fired laser fibers into Danger signals + [131] Animal
(LITT) disease [127], primary, and a tumor resulting in coagulative necrosis [129] Granulocytes
secondary malignancies of the NK cells ?
liver [128]
Experimental application: primary Approach: percutaneous Monocytes/ ?
and secondary malignancies of the Macrophages
breast, brain, bone, and prostate DC ?
[129] Image guidance: MRI, CT, and US T cells + [128, 132] Animal
Treg ?
B cells ?
Antibodies ?

Asterisks indicate allocation of T-cell or antibody responses to defined antigens.

Ref., reference number.

The concept of thermal treatment for cancer is not new. The first patients with cerebral tumors were already treated with RF ablation in the early 20th century, but it took until the 1990s for RF ablation to become an accepted, commonly used treatment option for primarily unresectable tumor lesions in liver, kidney, bones, and lung [15]. During RF treatment, one or more RF applicators are placed in the target tissue and high-frequency alternating current is generated, leading to frictional heating above 60°C up to 100°C inducing coagulative necrosis [2, 16]. Higher temperatures would result in desiccation and subsequent increase in tissue impedance which limits further conduction of electricity into the tissue [12]. Recent studies have shown that the clinical outcome after RF ablation is comparable or even better in comparison to that of surgical resection. Consequently, RF ablation is currently being discussed as a possible new standard for elimination of metastatic liver lesions and oligofocal hepatocellular carcinoma (HCC) [5, 8, 17] and further as a curative treatment option in HCC and metastatic stages of colorectal carcinoma (CRC) when combined with surgery [18, 19]. Early-stage non-small cell lung cancer (NSCLC) can also be successfully treated with RF ablation. However, retrospective comparative analyses of survival have shown a strong tendency to increased survival benefits for NSCLC patients treated with surgery compared to RF ablation (46 versus 33 months, P = 0.054) [20], limiting the application of RF ablation to patients with contraindications against surgery.

MWA represents a relatively new technique using electromagnetic waves to induce high temperatures of up to more than 100°C. Here also, an active microwave antenna is placed into the tumor. Since MWA does not require the conduct of electric current, temperatures above 100°C do not result in a decline of therapeutic efficacy [12]. This method could therefore be effectively applied in tissues with higher impedance like lung and bone [12]. In humans, MWA is currently mainly applied for the treatment of HCC [21, 22].

During HIFU, ultrasound beams of high energy are applied to focus acoustic energy on a well-defined region inducing tissue vibration. Although single ultrasound beams can penetrate tissue without causing significant heat, focussing beams from multiple directions into a selected region results in a temperature rise to over 60°C and subsequently in coagulative necrosis [23, 24]. HIFU also induces acoustic cavitation which represents an additional mechanical mechanism of tissue destruction. Acoustic cavitation (the expansion and contraction of gaseous nuclei in cells through acoustic pressure) leads to collapse of mitochondria, endoplasmic reticulum, as well as nuclear and cell membranes [25]. This procedure is the only noninvasive thermal technique and allows real-time imaging of the treatment progress by ultrasound (US) [25]. However, the clinical application of HIFU is limited since the size of the multidirectional ultrasound focus is confined by technical boundaries and a treatment time as short as possible is required for an accurate ablation [24]. HIFU has been applied for the treatment of breast, liver, pancreas, kidney, bone, prostate, and soft-tissue tumors [2527].

Laser fibers placed into a tumor lesion are used for laser ablation where photon energy conduction induced heating can reach temperatures of over 50°C. The tissue penetration depth of laser light is only 0.4 mm which implies that multiple laser fibers have to be positioned into a tumor to ensure optimal tissue destruction [28]. However, this limited penetration can facilitate the monitoring and accuracy of the ablation. This technique is experimentally used for the treatment of breast, brain, liver, bone, and prostate tumors [29, 30]. More extensively used is the clinical exertion of laser photocoagulation in retinal diseases, here also leading to retinal scarring [31].

In contrast to all the techniques mentioned above, cryoablation utilizes not high, but extremely low temperatures that sink to −160°C [32]. Cryoablation involves the evaporation of liquid gases and is a purely thermal process which does not require application of electrical current, leading to a broader applicability in high impedance tissues like lung or bone. The extent of tissue destruction can be easily monitored by direct monitoring of ice-ball formation with all conventional imaging modalities [1]. Cryoablation is used in broad clinical application, even for the treatment of retinoblastoma in children [33]. In 1–6% of cases cryoablation causes a systemic inflammatory response syndrome (SIR), termed as the cryoshock phenomenon which represents a potentially life-threatening complication [3438] and limits its clinical application, especially for liver tumors [39].

The observation that spontaneous regression of untreated tumors can occur after thermoablation of distant tumor masses may indicate an involvement of immune activation upon thermoablation [3942]. The initiation, maintenance, and termination of an effective antitumor immune response requires a complex interplay between cellular (immune cells including effector and regulatory subsets) and humoral components (cytokines, chemokines, antibodies). Various constitutive or inducible danger signals released by injured cells are known to play a determinant role in alarming the immune system against self-damage. In this danger model, cells dying by physiological processes such as apoptosis will be rapidly eliminated and ignored by the immune system whereas necrotic cells releasing their content in the extracellular space will trigger an immune response [43, 44]. In particular, heat shock proteins (HSP) constitute a group of molecular chaperones which stimulate the maturation of dendritic cells (DC) and carry antigenic peptides from their cells of origin inducing subsequent priming of antigen-specific T cells [4548]. Local ablative treatment induces necrosis which may naturally modulate all of these parameters by inducing inflammatory processes finally leading to the development of an antitumor specific immune response. A growing series of reports describing inflammatory responses, antigen release and uptake by professional APC and antitumor adaptive immunity shows that this can indeed be the case. This review aims to summarize findings on the modulation of the immune system through high- or low- temperature-induced thermal tissue ablation of cancer in animal tumor models and cancer patients. The respective techniques are presented in the order of common clinical use.

2. Radiofrequency Ablation

RF ablation has the broadest application in cancer treatment. It is therefore not surprising that most recent data relating to the activation of the immune system through thermoablation have been obtained using this method (Table 2).

Table 2.

Studies reporting immune modulation in cancer patients and animal models treated with RF ablation.

Species Tumor Model Immunologic effect References
Human HCC (n = 1) HSP-70, HSP-90 (cytoplasm, membrane)↑ [62]
HCC (n = 8) Activation of myeloid dendritic cells (blood) [51]
IL-1β, TNF (serum)↑
HCC (n = 20) CD4+ and CD8+ cells (blood)↑ [70]
CD3CD56+, CD56+CD16+ cells (blood)↑
Activity of tumor-specific T cells↑
HCC (n = 37) CD3CD56dim⁡ cells (blood)↑ [67]
Activity of CD3CD56dim⁡ cells↑
HCC (n = 20) Tumor-antigen specific T cells (blood)↑ [77]
RCC (n = 6) CD3+HLA-DR+, CD4+ and CD8+ cells (blood)↑ [71]
CD56+CD16+cells (blood)↓
Liver metastases (n = 8) Neutrophils (blood)↑ [66]
HCC (n = 4)
Liver metastases (n = 6) CD4+ and CD8+ cells (blood)↑ [76]
HCC (n = 6)
Liver metastases of CRC IL-6 (serum)↑ [52]
(n = 10)
Liver metastases (n = 9) IL-6 (serum)↑ [53]
HCC (n = 2)
Liver metastases (n = 13) CD4+ cells (blood)↓ [75]
HCC (n = 4) MUC-1 specific T cells (blood)↑
B cells (blood)↑ (only in metastatic cancer patients)
Trafficking of CD62L+ T cells into tissues
Lung metastases (n = 4) IL-8, MIP-1α, MIP-1β (serum)↑ [54]
NSCLC (n = 10) IL-10 (serum)↑
CD4+CD25+Foxp3+ cells (blood)↓
Neutrophils (blood)↑
Liver metastases (n = 13) IL-6 (serum)↑ [133]
HCC (n = 4) IL-10 (serum)↑
Metastases (n = 29) ± chemotherapy CD4+ and CD8+ responses against tumor-specific [78]
Primary tumors (n = 26) antigens (blood)↑
Metastases (n = 16) Tumor-specific antibodies (serum)↑
HCC (n = 4) HSP-70 (serum)↑ [63]
RCC (n = 2)

Mouse CRC CT26 hEpCam ± huKS-IL2 Antitumor activity (splenocytes)↑ [74]
(BALB/c) Tumor growth (distant tumor)↓
Tumor growth (rechallenge)↓
C26 CD4+ cells (perinecrotic)↑ [65]
Neutrophils (perinecrotic)↑
Neutrophils and lymphocytes (distant metastases)↑
HCC BNL ± CCL3 CD11c+ cells (blood)↑ [73]
CD11c+ cells (tumor)↑
CD4+ and CD8+ cells (tumor)↑
Tumor-specific cells (tumor)↑
Tumor growth (distant tumor)↓
Tumor growth (rechallenge)↓

Mouse Melanoma B16-OVA ± CTLA4-mAb CD8+ tumor-antigen specific T cells (blood)↑ [72]
(C57BL/6) ± Treg depletion Tumor growth (rechallenge)↓
B16-OVA ± CTLA4-mAb Antigen loaded DC, DC maturation (draining [69]
lymphnodes)↑
Tumor growth (rechallenge)↓
B16-OVA ± DC HSP-70, gp96 (tumor)↑ [57]
HMGB1 (tumor)↑
CD8+ tumor-specific T cells (spleen, draining
lymph nodes)↑
Local recurrence↓
Tumor growth (rechallenge)↓
Urothelial carcinoma MB49 ± DC CD4+, CD8+ antitumor responses (splenocytes)↑ [49]
CD11c+cells (tumor)↑
Tumor growth (rechallenge)↓

Mouse CRC HT29 HSP-70 mRNA (cytoplasm)↑ [56]
(NIH (S)-nu)

Rabbit Hepatoma VX2 Lymphocytes, plasma cells, and neutrophils [68]
(tumor)↑
Tumor-specific T cells (blood)↑

Rat (Fisher) Mammary MatBIII CD161+ cells (tumor-surrounding tissue)↑ [61]
HSP-70 (tumor-surrounding tissue)↑
R3230 ± liposomal HSP-70 (around central coagulation zone)↑ [59]
doxorubicin

Rat (rNU) Hepatoma SK-HEP-1 HSP-70, HSP-90 (cytoplasm, membrane)↑ [55]

2.1. Cytokines and Stress Response

Several groups have evaluated the systemic release of cytokines, chemokines, and various stress factors after RF ablation. Serum levels of proinflammatory cytokines like interleukins IL-1 β, -6, and -8, as well as TNF were found to be either increased [5154] or unchanged [133, 134]. In general, changes were modest and transient (several hours to days after ablation) [38, 52, 53, 134]. Moreover, IL-10 could be elevated in the serum postinterventionally [54, 133]. Over all, no case of severe SIR with multiorgan failure and coagulopathy, but significant increases in body temperature, mean arterial blood pressure, and concomitantly increased serum levels of adrenaline, nor adrenaline or C-reactive protein (CRP) have been reported shortly after RF ablation [53, 54, 133, 134].

In murine models, RF ablation induced strong upregulation of mRNA and/or protein levels of HSP-70, HSP-90, and glycoprotein 96 (gp96) as well as translocation of nuclear high-mobility group protein B1 (HMGB1) into the cytoplasm of tumor cells and into the intercellular space [5557]. More specifically, increased HSP-70 expression was shown to occur at the margin of the ablation zone, the so-called transition zone, both in animals [55, 58, 59] and in human liver cells in vivo [62]. The time frame of maximal HSP-70 expression is described to be no more than 24 hours after ablation [56, 60, 62], the protein remaining upregulated in the necrosis surrounding tissue three days after RF ablation [61]. Several factors may influence local expression of heat shock proteins after RF: in rats receiving thermal ablation in different zones of the liver, the degree of HSP-70 expression was observed to be dependent on the relative spatial position of the ablated area to larger liver vessels since the blood stream of these vessels can nourish adjacent cells preserving cellular metabolism and, hence, expression of HSP in these cells [60]. Further experiments in nude rats transplanted with human HCC also suggested a correlation between applied energy and level of expression of HSP-70 and -90 [55]. As we recently described, a significant systemic release of HSP-70 into the serum can also be detected one day after treatment in RF-treated cancer patients, but serum levels did not correlate with ablation volumes, histological tumor type, and other clinical or laboratory parameters [63].

2.2. Cellular Immunity

2.2.1. Changes in Peripheral and Intratumoral Immune Cell Subsets

Postinterventional changes in peripheral leukocyte subsets have been observed by several groups and taken as evidence for the immune modulatory effect of RF ablation. Of note, antibody tools for cell subset identification, timepoints of observation, and patient cohorts differed between published studies. A decrease of circulating CD4+CD25+Foxp3+ regulatory T cells (Treg) was observed in patients 1 month after RF ablation of lung nodules [54]. In another study including 20 HCC patients, no significant changes in T-cell subsets were detected 1 month after RF (naïve or memory CD4+, CD8+) while increased percentages of activated T cells and circulating NK cells were noted in randomly selected patients from the study cohort [70]. The same group later described a marked expansion of CD3CD56dim⁡ effector NK cells 1 week and 4 weeks after treatment [67]. Matuszewski and colleagues evaluated lymphocyte subpopulations after RF ablation of renal cell carcinoma (RCC) in 6 patients and found a globally increased proportion of activated T cells in the majority of patients (CD3+HLA-DR+) whereas effects on CD4+, CD8+, and NK (CD56+CD16+) cells varied among individuals and at different timepoints [71]. In patients with colorectal liver metastases, but not with HCC, a transient decrease in CD3+CD4+ T cells was noted shortly (day 2) after treatment [75]. Although these and further observations are heterogenous, they collectively suggest an impact of RF ablation on various peripheral cell subsets, including T and NK cells [61], but also neutrophils, monocytes, B lymphocytes, and even DC [51, 54, 66, 75].

The assessment of tumor-infiltrating cells before and following RF ablation is intrinsically difficult in patients and available data have been obtained in various animal models. Most reports describe infiltration of immune cells in the transition zone hours to days after treatment. Granulocytes, macrophages, plasma cells, DC, CD3+, and CD4+ cells were found [49, 64, 68]. Interestingly, neutrophils and lymphocytes could also infiltrate distant, untreated metastases [65].

2.2.2. Antitumor Specific Responses

Few data addressing the adaptive immune response to tumors after RF ablation are available.

In a transplant-tumor model of VX2-hepatoma, rabbits were randomly assigned to treatment with RF ablation or to observation. Two weeks after RF ablation, the activation of tumor-lysate specific T cells was detected and persisted over a postinterventional observation period of 6 weeks [68]. Animals in the RF-treated group had a significant survival increase [68].

Antigen-specificity of RF-induced antitumor T-cell responses was investigated in several reports. Dromi and coworkers used a murine urothelial carcinoma expressing the male minor histocompatibility antigen HY which was inoculated to female mice. T-cell responses against MHC-class I and class II HY-derived epitopes were significantly increased in the group of mice having received tumor RF ablation as compared to control animals. This was accompanied by an enhanced control of tumor growth, including upon rechallenge [49].

In a mouse model of OVA-expressing melanoma, adoptive transfer of splenocytes from RF-treated to naïve mice led to a growth retardation of OVA+-, but not OVA-tumors after rechallenge, and to complete tumor elimination in 20% of the mice. The treatment could also induce long-lasting immunity since RF-treated mice surviving the first tumor inoculation were completely protected after a second challenge 70 days later [72]. Moreover, intratumor injection of tagged-OVA led to antigen uptake and maturation of CD11c+ cells in the tumor-draining lymphnode, albeit to a lesser extent than after cryoablation which was directly compared to RF ablation in this model [69].

In patients, HCC-reactive T cells were detected with IFNγ ELISPOT in 4/20 patients before RF ablation upon stimulation of PBMC with lysate of autologous tumor cells obtained either before or after treatment. One month after RF treatment, cellular reactivity was observed in 9/20 patients, strongly suggesting an in vivo immunization effect after RF-intervention [70]. Similar results were reported in two further cohorts of HCC and CRC patients [76].

Three recent publications have addressed the antigen-specificity of the RF-induced T-cell responses in patients. Napoletano and colleagues detected an increased IFNγ production upon stimulation with MUC-1-derived glycopeptides in 2 patients treated for liver metastases and also an increase in circulating CD3CD19+ B cells. However, the specificity of antibodies was not studied [75]. Hiroishi and coworkers investigated CD8+ T-cell responses against MAGE-1, NY-ESO-1, and GPC3 antigens in patients with HCC and found that antigen-specific T cells were already detectable in samples obtained before RF ablation, and increased in approximately half of the patients [77]. Recently, we evaluated the occurrence of tumor-antigen specific T cells or antibodies after RF ablation in 55 cancer patients and found an increase in antigen-specific antibodies, and CD4+ or CD8+ T cells in several individuals receiving RF ablation alone or shortly after chemotherapy [78].

2.2.3. Combination Therapies

All the results presented above show that RF ablation is able to induce tumor-directed immunity; however, the observed therapeutic effects are limited. Combination therapies have therefore been already tested in preclinical models, with the aim to enhance antitumor responses and protection. For OVA-expressing melanoma, CTLA-4 blockade or Treg depletion (with anti-CD25 mAb) showed improvement in tumor control and enhanced induction of OVA-specific CD8+ T cells whereas CTLA-4 mAb application without RF ablation did not mediate the same effects [72].

The coadministration of the monocyte attracting chemokine ligand 3 and inflammatory protein-1α (CCL3/MIP-1α) [73], antibody-conjugated IL-2 [74] or even chemotherapy (liposomal doxorubicin) [59] also enhanced the effects of RF ablation. Finally, whereas intratumor injection of unloaded DC did not synergize with RF-treatment, application of tumor-lysate loaded DC was reported to abrogate tumor relapse in most animals. Interestingly, while vaccination with DC alone was ineffective with regard to survival benefits, the combination with RF ablation significantly improved the survival of tumor bearing mice [49, 57].

All these reports provide a strong rationale for testing the combination of RF therapy with immune-modulating agents in cancer patients. It has to be noted that—besides RF ablation—many patients currently receive additional therapies like chemotherapy which may also influence the development of tumor-specific immune responses as recently recognized [137].

2.3. Immune Response and Clinical Course

The relationship between occurrence of antitumor immunity after RF ablation and clinical outcome still remains elusive. In HCC patients with induced tumor-specific T-cell reactivity after RF ablation, the local- and distant-site recurrence was similar [70]. In contrast, Hiroishi and colleagues observed a correlation between the frequency of tumor-antigen specific T cells and a favorable tumor-free survival in HCC patients [77]. Here, it has to be noted that patients could additionally receive transarterial chemoembolization (TACE). We have recently observed a tendency to a better survival for patients who presented with at least a twofold increase of HSP-70 in the serum one day after treatment [63]. Since patient cohorts were small in all three studies, results need further confirmation.

3. Cryoablation

3.1. Special Premises of Cryoablation

While thermal techniques utilizing lethal high temperatures have been so far mostly described to stimulate immune responses, cryoablation has been described to exert both stimulatory and suppressive effects on the immune system. These particular features could be due to the specific physiological mechanisms of cold injury including (i) direct cellular damage through formation of ice crystals, and (ii) vascular and endothelial injury with potential ischemia [82]. Whereas most other thermoablative techniques are believed to induce essentially coagulative necrosis, apoptotic cells might be also present at the outer rim of the ablation zone after cryoablation. According to the danger model, apoptotic cells do not release their cellular content (antigens, HSP, and HMGB1) and induce immunological tolerance [43, 44]. It has been proposed that larger numbers of apoptotic cells might cause tissue protection and lead to immunosuppression while larger numbers of necrotic cells could serve as immunostimulators [4]. More recently, they showed that the cryoablation modality itself, that is, rate of freeze, influences both tumor growth and T cell recruitment [79]. Moreover, technique and rate of freezing cycles could play a role in the precise mechanisms of the watershed between immunosuppression and immunostimulation after cryoablation [79]. However, this model is questioned by more recent reports showing that apoptotic cells can also exhibit significant immunostimulatory capacity [138, 139]. One other influential factor for these contradictory observations could be the timepoint of immunomonitoring: early assessment might miss immune activation and antitumor activity. Interestingly, clinical improvement could be recorded rather late after cryotreatment (up to 10 weeks) [140, 141] which is in line with the new concept that assessment of tumor response upon immunotherapy should be performed later than after conventional cytostatic therapy [142].

Table 3 only presents recent immunological observations of the past decade. Many observations reporting immunosuppression by cryoablation were made earlier and are discussed below, but are not presented in the table.

Table 3.

Recent Studies reporting immune modulation in cancer patients and animal models treated with cryoablation.

Species Tumor Model Immunologic effect References
Human CRC (n = 110) Gangliosides (GM2, GD1a, GT1b; serum)↑ [116]
Antiganglioside antibodies (serum)↑
HCC (n = 111) CD4+CD25+Foxp3+ cells (blood, ablation [105]
zone surrounding tissue)↓
Prostate (n = 20) IFNγ↑, TNF (serum)↑ [85]
Tumor-specific T-cell responses (blood)↑
Prostate (n = 12) ± GM-CSF Tumor-specific T-cell responses (blood)↑ [85]
RCC (n = 6) + GM-CSF Tumor-specific T-cell responses (blood)↑ [87]
Tumor-specific antibodies (serum)↑
Liver metastases IL-6, TNF (serum)↑ [84]
(n = 12) Th1/Th2 ratio (blood)↑
CCC (n = 3)

Mouse (BALB/c) CRC Colon-26 ± krestin CD8+ antitumor T-cell reactivity (spleen) [94]
(↑)
Number of metastases↓
Colon-26 ± Treg Tumor-specific CD8+ T cells (spleen)↑ [96]
depletion ± DC + BCG Tumor growth (distant tumors)↓
Colon-26 ± Tumor-specific T cells (spleen, draining [97]
cyclophosphamide lymph nodes)↑
Tumor growth (rechallenge)↓
Mammary MT-901 IFNγ, IL-12 (serum)↑ [83]
Tumor-specific T cells (draining
lymph nodes but not in spleen)↑
NK cell activity (spleen)↑
Tumor growth (rechallenge)↓
T cells (draining lymph nodes)↑ [95]
Tumor-specific T cells (draining lymph
nodes)↑
Pulmonary metastases↓
Tumor-specific T cells (draining lymph [79]
nodes)↑
Pulmonary metastases (high-intensity
freezing)↓
Pulmonary metastases (low-intensity
freezing)↑
Melanoma B16-OVA ± imiquimod Tumor-specific T cell proliferation↑ [98]
Tumor growth (rechallenge)↓
B16-MO5 ± DC Tumor growth (rechallenge)↓ [92]

Mouse (C57BL/6) Melanoma B16-OVA ± DC maturation and antigen uptake [69]
CTLA4-mAb (TDLN)↑
Tumor growth (rechallenge)↓
B16-OVA ± CpG DC (TDLN)↑ [135]
CD4+, CD8+ T cells (TDLN)↑
OVA-specific T cells (TDLN)↑
Tumor growth (rechallenge after
peritumoral CpG administration)↓

Mouse (NIH Melanoma IIB-MEL-J (human) Neutrophils (RB6-5CG+) [86]
(S)-nu) ± GM-CSF (tumor-surrounding tissue)↑
macrophages (F4/80+; tumor-surrounding tissue)↑
DC (DEC205+; tumor-surrounding
tissue)↑

Mouse (OT-I T cell Lung Lewis lung tumor Tumor-specific CD8+ T-cell [92]
receptor (Vα2/Vβ5) D122 ± DC proliferation↑
transgenic) Th1 responses↑
Tumor growth (lung metastases)↓

Rat (Wistar) Glioma C6 CD3+ and CD4+ T-cell percentages (blood)↑ [89]
CD4+/CD8+ ratio (blood)↑

(↑) Weak induction.

3.2. Cytokines and Stress Response

Unlike the other thermoablative methods, cryoablation induces a cytokine release syndrome (SIR—1–6.4% of all cases, with a mortality rate of 0.2–4%) [34, 35], assimilated to the cryoshock phenomenon which is clinically manifested by thrombocytopenia, disseminated intravascular coagulation (DIC), and pulmonary failure [3538]. Cryoshock is mainly limited to ablation of hepatocytes [39]. In sheep and rats, the frequency of SIR correlated positively with the extent of cryoablated liver tissue, animals with more than 35% of ablated tissue presenting an elevated risk of SIR [37]. Moreover, cryoablation leads to significant increases of serum IFNγ TNF, IL-6, and IL-12, but not IL-10 within several hours after intervention [36, 83, 84]. In a rat model, cytokine release after cryoablation, RF ablation, and LITT was compared. Between 1 and 6 hours after cryoablation, significantly elevated serum levels of IL-6 were observed. IL-10 serum levels were slightly, but not significantly elevated [38]. In patients, TNF and IFNγ could remain elevated for up to four weeks [85]. In a model of transgenic mice overexpressing HSP-70 only a slight increase of HSP-70 expression could be observed which proved to be tissue-protective against cryonecrosis in skeletal muscle cells. Here, is has to be noted that no complete cryoablation, but only cryolesioning of skin and skeletal muscle was performed [88]. To our knowledge, no data on HSP expression after necrosis induction through cryoablation are available.

3.3. Antibodies

The earliest reports on immune modification after cryoablation described autoantibody production against ablated normal and tumor tissues in rabbits and monkeys, as well as in patients [41, 106112, 114]. These antibodies were essentially IgG and IgM in the serum [41, 86, 114] and at the vicinity of the ablated lesion mainly IgG and IgA [115] appearing within two weeks after intervention [41, 86, 114]. In contrast, Müller and colleagues treated osteosarcoma in mice with cryoablation and found a decrease of tumor-binding antibodies [113].

Another effect of cryoablation was detected by Ravindranath and colleagues who observed a release of gangliosides into the circulation of CRC patients after cryoablation but not after RF ablation or surgery. At the same time, the group also described increasing titers of anti-ganglioside IgM antibodies [116]. Since anti-ganglioside antibodies have inhibitory effects on primary tumors, such as the induction of complement mediated killing [143] or apoptosis [144], production of antitumor antibodies might be one of the mechanisms underlying the immune-mediated tumor rejection following cryoablation [116].

3.4. Cellular Immunity

3.4.1. Changes in Peripheral and Intratumoral Immune Cell Subsets

In rats, significantly elevated peripheral leukocyte counts—especially CD3+ and CD4+ T cells—were detectable between 1 and 14 days after intervention [38, 89]. In humans, cryoablation led to an increase of circulating T cells in few patients [99, 100]. In a randomized trial, cryoablation—compared to conventional surgery—led to increased numbers of helper T cells and activated T cells [101]. In a cohort of patients with liver metastases, an increase of the Th1/Th2 ratio was observed in the peripheral blood [84] whereas Zhou and colleagues reported a decrease of circulating CD4+CD25+Foxp3+Treg after cryolesioning of HCC [105].

In tumor draining lymph nodes (TDLN), increased cellularity was observed both in T-cell (paracortical) and B-cell (germinal center) areas one week after treatment. Immunologic activity could remain increased over a time span of up to 10 weeks [90, 91]. Using a xenograft model of human melanoma in nude mice, Gazzaniga and coworkers further described a massive intravascular and peritumoral recruitment of leukocytes, essentially neutrophils and macrophages after cryoablation [86]. In a mouse mammary cancer model, the number of CD4+ T cells in TDLN was augmented. Interestingly, CD4+CD25+Treg were more numerous after low rate freeze [79].

3.4.2. Antitumor Specific Responses

Assessment of the immune modulation by cryoablation has yielded contradictory results. Older works have pointed out immunosuppressive effects: an increase of circulating immune effector cells was not of functional relevance for tumor rejection, rather, tumor outgrowth and increased metastasis was promoted. This indicated that cryoablation might mediate deleterious effects possibly by induction of suppressor T cells, today referred to as regulatory T cells, as well as delayed development of antitumor immunity [103, 104, 145]. In line with these findings, Machlenkin and colleagues did not observe cellular activation through cryotherapy as a monotherapy [92].

In contrast, other groups demonstrated immunologic activation in cryotreated animals (Table 3). Kimura and colleagues found an increased cytotoxic activity of peripheral lymphocytes and splenocytes against a murine leukemia virus-induced lymphoma [146]. Regression of distant metastases and resistance to tumor rechallenge was described by Bagley and colleagues who found that splenic lymphocytes isolated from sarcoma-bearing mice treated with cryoablation exhibited significantly increased cytotoxic activity against sarcoma cells as compared to those obtained from mice undergoing limb amputation [93]. Increased immunological activity could be delayed up to ten weeks after intervention [140].

Urano and colleagues observed an increased activity of tumor-specific cytotoxic T lymphocytes (CTL) seven days after cryoablation in a mouse colon-carcinoma model. These effects were only observed after ablation of a single nodule while ablation of several lesions abrogated immune-related tumor regression. Here, a threshold of ablated tissue volume that governed immune stimulation or suppression was proposed [94]. Interestingly, tumor-specific effector cells isolated from TDLN but not from the spleen or peripheral blood secreted a higher amount of IFNγ (between days 3 and 7 after treatment) as compared to cells obtained following surgical resection.

In an OVA-expressing melanoma model, den Brok and colleagues observed an increase of antigen-loaded DC in draining lymph nodes both after cryoablation and RF-ablation. Of note, almost double as high cell numbers were observed compared to the induction through treatment with RF ablation [69]. Moreover, the numbers of infiltrating lymphocytes in TDLN were increased. These lymphocytes produced approximately 10-fold greater amounts of IFNγ upon stimulation with irradiated mammary adenocarcinoma cells after cryoablation than after surgical resection, delayed tumor growth and reduced the number of pulmonary metastases after adoptive transfer of TDLN cells of the cryoablated tumor [95]. The protection against a tumor rechallenge with B16-OVA cells was enhanced after cryoablation (50% surviving mice after 70 days) compared with the protective effect observed after RF ablation (20% surviving after 70 days) [79, 83, 95].

3.4.3. Combination Therapies

While Machlenkin and colleagues observed no clinical benefit with cryoablation alone, the combination with an intratumoral injection of immature DC induced robust activation of CD4+ and CD8+ CTL [92]. This synergistic effect was further improved after pretreatment with anti-CD4 or anti-CD25 mAb for Treg depletion [96].

In an OVA-expressing melanoma model, CTLA-4 blockade and depletion of regulatory T cells could further enhance cryoinduced tumor-specific T-cell responses [69]. Alternatively, concomitant injection of CpG 1668, a TLR 9 ligand, had a similar effect on T-cell recruitment. The route of adjuvant injection was crucial for immune induction, peritumoral CpG application showing to be superior to distant site. Although tumor growth was delayed after combination therapy, survival benefit was not superior to treatment with cryoablation alone [135].

Conditioning with cyclophosphamide injected one day before cryoablation led to increased IFNγ  production of tumor-antigen specific CD4+ T cells in a mouse colon cancer model as detected in intracellular cytokine staining, enhanced survival and even some complete remission. Three out of four animals cured with the combination therapy also survived a tumor rechallenge with no macroscopically visible tumor upon autopsy [97]. Moreover, adoptive transfer of spleen and lymph node cells from surviving mice led to an improved survival in tumor-bearing mice. Depletion experiments showed that CD8+ effectors were responsible for tumor elimination indicating that immunological memory had developed [97].

In the same OVA-expressing melanoma model used by den Brok and colleagues, Redondo and coworkers observed a clear survival advantage for mice treated with cryoablation combined with repeated topical application of imiquimod as an adjuvant indicating that TLR-7 activation can enhance tumor-specific immune responses induced by thermal treatment [98].

To sum up, all these reports suggest that combination of cryoablation with check point blockade or immunoadjuvants is a promising approach in the treatment of cancer patients.

3.5. Immune Response and Clinical Course

In patients with hormone refractory prostate carcinoma, a combination of cryoablation with injection of GM-CSF as adjuvant was evaluated. T-cell reactivity against autologous tumor tissue lysates as determined in IFNγ  ELISPOT was found to be weakly increased after therapy, and no correlation could be established between the breadth of the immune response and the clinical course as measured by analysis of PSA serum levels [85, 102]. Also in a small cohort of patients with RCC, increased cytotoxic T-cell activity and increased antitumor serum antibodies in selected patients were observed which only weakly correlated with a favorable clinical response. Here also, GM-CSF was applied as an adjuvant [87].

4. Microwave Ablation

As with RF ablation, microwave ablation therapy (MWA) induces hyperthermia leading to coagulative necrosis. The clinical application of MWA is, however, more limited than the thermoablative methods discussed above and only few groups have evaluated the immunomodulatory effects of MWA (Table 4).

Table 4.

Studies reporting immune modulation in cancer patients and animal models treated with MWA.

Species Tumor Model Immunologic effect References
Human HCC (n = 82) CD3+ cells, CD56+ cells (treated and distant tumors)↑ [119]
CD68+ cells (treated and distant tumors)↑
HCC (n = 10) ± DC Phase I study: [120]
CD4+CD25high cells (blood)↓
CD8+CD28 cells (blood)↑

Mouse HCC Hepa 1–6 Activity of tumor-specific CD4+, CD8+ cells (spleen)↑ [118]
(C57BL/6) ±GM-CSF NK1.1+ cells (spleen)↑
± CTLA4-mAb Tumor growth (rechallenge)↓

4.1. Cytokines and Stress Response

MWA was described to induce HSP-70 expression in normal kidney tissue lysates obtained from treated rats, as detected with specific ELISA. However, HSP-70 expression was significantly lower upon MWA as compared to animals treated with RF ablation and cryoablation [117].

4.2. Cellular Immunity

In a mouse tumor model of HCC, only 2/10 animals experienced tumor rejection upon rechallenge after MWA, suggesting an existing but suboptimal protective antitumor immunity [118]. However, the protective effect could be improved by intratumoral coadministration of GM-CSF loaded microspheres, and even more by intraperitoneal CTLA-4 blockade. The triple combination not only led to rejection of newly inoculated tumors, but was also effective in the rejection of established distant tumors. Splenocytes isolated from the treated mice killed hepatoma cells in vitro, but not an unrelated tumor cell line. In vitro depletion experiments using mAb could further show that cytotoxicity was mediated by T cells (both CD4+ and CD8+) and NK cells, confirming that antitumor immunity was induced upon combination therapy [118].

One month after MWA, 10 patients with hypersplenism that had developed as a result of portal hypertension exhibited a transient peripheral increase of T helper cells (CD3+CD4+) and B cells, but not of cytotoxic (CD3+CD8+) T cells [22]. In a larger cohort of patients suffering from HCC, immune cell infiltration was studied by immunohistochemistry analyses of biopsy tumor samples taken either before or at different timepoints (3–30 days) after MWA application. A markedly increased infiltration of lymphocytes (predominantly CD3+ T cells, CD56+ NK cells, and macrophages, but not of B cells) was detected after MWA inside the ablated lesions, in the adjacent normal tissue and in distant untreated lesions [119].

4.3. Immune Response and Clinical Course

The density of infiltrates of lymphocytes, macrophages, and CD56+ cells into MWA-treated liver tissue correlated inversely with the risk of local recurrence [119].

Zhou and colleagues performed a phase I clinical study in ten HCC patients with chronic hepatitis B by combining local microwave tumor ablation with immunotherapy, which was applied at 3 timepoints, that is, on the day of the MWA and then on days 11 and 100. Immature and mature monocyte-derived DC loaded with autologous tumor lysate were injected into the rim between the ablation zone and normal liver parenchyma and into the groin lymph nodes, respectively. Additionally, in vitro activated lymphocytes were applied intravenously. A modest and transient effect on peripheral T-cell subsets (decrease of CD4+CD25high—possibly Treg—and increase of CD8+CD28—differentiated CD8+ T cells—was reported one month after treatment concomitant with a reduction in hepatitis B virus load observed in some patients, but analyses of the antitumor specific responses were not performed in this study. Of note, this clinical setting does not allow determining whether the observed effects were due to the MWA itself, to the immunotherapy regimen or to the combination of both treatments [120].

5. High-Intensity Focused Ultrasound (HIFU)

In addition to mere hyperthermia, HIFU also exerts nonthermal mechanistic constraints (acoustic cavitation) on treated tissues that might contribute to and modulate its effects on the immune system [122] (Table 5).

Table 5.

Studies reporting immune modulation in cancer patients and animal models treated with HIFU.

Species Tumor Model Immunologic effect References
Human Breast carcinoma (n = 23) HSP-70 (membrane)↑ [122]
Breast carcinoma (n = 48) CD3+, CD4+, CD8+ cells (tumor)↑ [27]
CD20+ cells (tumor)↑
CD57+ cells (tumor)↑
Pancreatic carcinoma (n = 15) NK cells (blood)↑* [123]
Uveal melanoma (n = 5) CD4+ cells (blood)↑ [125]
Osteosarcoma (n = 6) CD4+ cells (blood)↑ [126]
HCC (n = 5)
RCC (n = 5)

Mouse (Ajax) Neuroblastoma C1300 ± adriamycin Tumor growth (rechallenge)↓ [136]

Mouse (C57BL/6J) HCC H22 ± DC Activation of CD8+ cells (spleen)↑ [124]
Tumor growth (rechallenge)↓
H22 ± tumor lysate vaccine Cytolytic activity (spleen)↑ [121]
Tumor growth (rechallenge)↓

*NK cell phenotype was not specified.

5.1. Cytokines and Stress Response

In breast cancer patients, increased HSP-70 expression was detected on the cell membrane of treated cancer cells. HSP expression was mainly found in the central necrosis zone while only a few positively stained cells were observed in the periphery [122].

5.2. Cellular Immunity

5.2.1. Changes in Peripheral and Intratumoral Immune Cell Subsets

In patients with posterior uveal melanoma [125], pancreatic carcinoma [123], osteosarcoma, HCC, and RCC [126] that were treated with HIFU, increased percentages of CD4+ T cells and a higher CD4+/CD8+ ratio were observed [125, 126]. Another study observed only statistically significant higher NK cell percentages in the peripheral blood, while other leukocyte subsets remained stable [123].

In human breast cancer specimens collected 1-2 weeks after HIFU treatment, immunohistochemistry analyses showed a significant increase of T and B cells at the margin of the ablated region as compared to HIFU-untreated tumor samples. Interestingly, a subset of these cells were activated (CD57+) and expressed perforin and granzyme B, indicating the presence of activated cytotoxic effectors [27].

5.2.2. Antitumor Specific Responses

In a model of experimental neuroblastoma, reduced secondary tumor growth after HIFU treatment was observed, although involvement of immune cells was not evaluated further [136].

Zhang and coworkers immunized mice with a vaccine consisting of a lysate of the H22 hepatoma cell line either untreated or pretreated in vivo with HIFU. Ten days after vaccination, animals received a subcutaneous tumor challenge. Tumor growth was significantly delayed in mice vaccinated with previously HIFU-treated tumor cells. However, survival was not different between the vaccination groups [121]. In another model, the same group utilized a DC vaccine loaded with cell debris from HIFU-treated or -untreated tumor cells. While tumor growth was again reduced, no survival advantage could be observed. However, increased activity of CD8+ splenocytes could be detected in IFNγ ELISPOT [124]. These were supported by in vitro experiments showing activation of bone-marrow derived DC upon incubation with tumor lysates and increased tumor killing by splenocytes harvested from HIFU-treated animals [121].

In contrast, several studies describe increased numbers of peripheral T cells following HIFU, which did not exert enhanced antitumor immunity [123, 125, 126].

Several groups observed a loss in tumor antigen expression in ablated prostate [23] or breast carcinoma [147, 148] lesions after HIFU. Such downregulation would be expected to lead to a reduced recognition of tumor tissue through antigen-specific T cells. Further investigations will be needed to determine whether the HIFU method is generally appropriate for efficient induction of antitumor T-cell immunity in patients.

6. Laser Ablation

Laser-induced thermotherapy (LITT) is applied widely for photocoagulation in retinal diseases, where the release of proinflammatory cytokines [127] or the activation of retina-specific T cells after panretinal photocoagulation (PRP) [149] have been described. In cancer patients, however, laser ablation is still experimental, and only scarce publications have addressed the modulation of cellular immunity through LITT (Table 6).

Table 6.

Studies reporting immune modulation in cancer patients and animal models treated with LITT.

Species Tumor Model Immunologic Effect References
Human CRC (n = 4) IL-6, TNF-R1 (serum)↑ [130]
HCC (n = 3)
Other (n = 6)

Mouse (CBA) Liver metastases of CRC MoCR HSP-70 (cytoplasm, nuclear)↑ [131]
Subcutaneous CRC tumors MoCR CD3+ cells (tumor-host interface)↑ [132]
Spontaneous IFNγ production (spleen, lymph nodes, tumor,
and distant tumors)↑

Rat (WAG) CRC CC531 CD8, CD86, MHC-II, CD11a, and ICAM1 expression (invasion [128]
front of distant tumors)↑

6.1. Cytokines and Stress Response

In patients, laser ablation led to increased levels of IL-6 and TNF-receptor 1 in the serum of patients suffering from primary and secondary malignant lesions of the liver 72 hours after treatment. Changes in the level of other proinflammatory cytokines such as TNF and IL-1β were not observed in this study [130].

In a murine model of colorectal liver metastases, LITT was also shown to enhance expression of HSP-70 at the margin of the coagulated tissue, with cytoplasmic and nuclear expression in sublethally damaged mouse hepatocytes and extraparenchymal cells. In tumor cells, this upregulation was detected between 12 hours and 7 days after intervention, with a peak at 24 hours [131].

6.2. Cellular Immunity

In WAG rats, Isbert and colleagues induced two independent tumors in the left and right liver lobes. One of the two tumors was either ablated with LITT or surgically removed, and immune cell infiltration into the untreated remaining tumor was compared to that observed in an untreated control group. Expression of CD8, CD86, MHC-class II, and adhesion molecules was found to be increased between 1 and 10 days after LITT at the tumor invasion front as compared to resection or no treatment, indicating an influx of immune cells [128]. Moreover, the growth of the untreated tumors was found to be considerably reduced in LITT-treated animals.

Using a murine CRC model, Lin and coworkers observed an increased infiltration of CD3+ T cells into the tumor-host interface and into the tumor, as well as into the liver parenchyma and—to a certain extent—also into distant tumor lesions. Moreover, increased activation of splenocytes and tumor infiltrating lymphocytes was reported in ex vivo IFNγ ELISPOT without antigen restimulation [132]. Of note, these results were all obtained in animal models, and immune modulation after LITT has not been reported for cancer patients yet.

Taken altogether, the available results strongly suggest that laser therapy, as shown for other thermoablation methods, can stimulate antitumor immune effector cells in vivo [128, 132].

7. Conclusion and Perspectives: Implications for Anticancer Therapy

During the past two decades, numerous publications in animal models and patients have shown that local thermoablative techniques can induce or enhance tumor-specific immune responses that contribute to tumor control. Although the sequential mechanisms involved are not yet fully elucidated, several pieces of the puzzle have been identified: thermal treatment induces necrosis and can (i) lead to local inflammation, release of danger signals—for example, heat shock proteins—which may even been detected systemically; (ii) stimulate the recruitment and activation of immune effector cells, including DC, at the vicinity and most probably inside the damaged tumoral tissue. Both processes occur rapidly, that is, within a few hours to days following intervention; (iii) activate antitumor adaptive immunity, including CD4+, CD8+ T cells, and antibody production which can contribute to local tumor elimination, control distant tumors including micrometastases, and establish long lasting antitumor immunological memory [69, 72, 83, 93, 97, 121]. The source of tumor-associated antigens for inducing specific T cells may be either necrotic dying cells [138, 150, 151] or sublethally damaged cells [62, 63]. Besides these direct mechanisms, the removal of tumor tissue leads to depletion of Treg and more generally may overcome local immunosuppression shifting favorably the balance towards effective antitumor immunity [61, 96, 105].

Hence, thermoablation can trigger physiological cascades necessary and sufficient for a protective immune response. Obviously, several methods can be applied successfully, suggesting that the key element is the induction of local necrosis, which can be achieved by using different settings and temperatures. High temperatures (RF ablation, MWA, HIFU, and LITT) seem rather to sustain antitumor activity whereas both immunomodulatory and immunosuppressive effects have been reported upon cryoablation. Of note, the lesions induced by high-temperature thermoablation are probably not solely of necrotic nature but may also exhibit apoptotic cells [16, 152, 153]. Whether opposite immunological outcomes are hence related to a different balance between apoptosis, necrosis, and secondary necrosis, with apoptotic cells acting more in a tolerizing or immunosuppressive fashion and necrotic cells more immunogenic, is at the moment unclear [138, 139]. Because necrosis induction can be easily visualized during thermoablative intervention, controlled necrosis might be an ideal tool for inducing enhanced immunogenic cell death [154]. Interestingly, local hyperthermia between 40°C and 44°C has been also been described to modulate immunity, as reviewed elsewhere [155].

However, it should be noted that the reported effects of thermoablation alone on the immune system are generally modest, suggesting that such treatment as a monotherapy is in general not capable of inducing sufficient immune responses for full tumor protection [97, 118]. Thermotherapy should be therefore most effective in case of a limited tumor burden, ideally without detectable tumor postinterventionally, and not in advanced cancer where it is applied in most cases so far. Notably, combined therapies in order to enhance tumor-specific immune responses showed extremely promising results. Several strategies, such as check point blockade (anti-CTLA-4 mAb, Treg depletion) [69] or application of adjuvants (interleukins or chemokines, GM-CSF, TLR agonists) have been evaluated in preclinical models but very little in clinical application yet. Randomized trials have still to be conducted.

In summary, thermal ablation represents a promising component for cancer immunotherapy in the treatment of small or subclinical tumor lesions which can be attacked by the patient's immune system. By controlled induction of physiological stress, it offers the possibility of letting the “natural” immune response develop in its whole by breaking self-tolerance. So, thermal ablation of cancer provides a therapeutic implementation of the danger model. However, the induced antitumor immunity is weak and probably not sufficient alone to eradicate established tumors, but it can synergize with some chemotherapies and immunomodulating strategies. Selecting the appropriate thermoablative method and finding optimal combinations for individual patients will be an exciting challenge for the upcoming years.

Conflict of Interests

None of the authors has any commercial interests or conflicts of interests to declare.

Acknowledgments

The authors wish to thank Lynne Yakes for editorial assistance. Work of the authors was supported by the Deutsche Forschungsgemeinschaft (DFG, Grant no. DFG RA 369/7-1), the Else-Übelmesser Stiftung, the Studienstiftungdes deutschen Volkes, the Deutsche Jose Carreras Leukemia Foundation, and Hölle & Hüttner AG, as well as the fortüne Program of the Eberhard Karls University of Tuebingen (Grant no. 1530-0-0). Sebastian P. Haen is supported by the Deutsche José Carreras Leukemia Foundation.

Abbreviations

CD

Cluster of differentiation

CRC

Colorectal carcinoma

CT

Computed tomography

CTLA

Cytotoxic T-lymphocyte antigen

DC

Dendritic cell

HCC

Hepatocellular carcinoma

HIFU

High-intensity focused ultrasound

HMGB1

High-mobility group protein B1

HSP

Heat shock protein

ICAM

Intercellular adhesion molecule

IFN

Interferon

IL

Interleukin

LITT

Laser-induced thermo therapy

mAb

Monoclonal antibody

MIP

Macrophage inflammatory protein

MRI

Magnetic resonance imaging

MWA

Microwave ablation therapy

n.a.

Not applicable

NSCLC

Nonsmall cell lung cancer

RCC

Renal Cell Carcinoma

RF

Radiofrequency

TDLN

Tumor draining lymph node

TLR

Toll-like receptor

TNF

Tumor necrosis factor

US

Ultrasound.

References

  • 1.Webb H, Lubner MG, Hinshaw JL. Thermal ablation. Seminars in Roentgenology. 2011;46(2):133–141. doi: 10.1053/j.ro.2010.08.002. [DOI] [PubMed] [Google Scholar]
  • 2.Pereira PL. Actual role of radiofrequency ablation of liver metastases. European Radiology. 2007;17(8):2062–2070. doi: 10.1007/s00330-007-0587-0. [DOI] [PubMed] [Google Scholar]
  • 3.Katsanos K, Ahmad F, Dourado R, Sabharwal T, Adam A. Interventional radiology in the elderly. Clinical Interventions in Aging. 2009;4(1):1–15. [PMC free article] [PubMed] [Google Scholar]
  • 4.Sabel MS. Cryo-immunology: a review of the literature and proposed mechanisms for stimulatory versus suppressive immune responses. Cryobiology. 2009;58(1):1–11. doi: 10.1016/j.cryobiol.2008.10.126. [DOI] [PubMed] [Google Scholar]
  • 5.Lencioni R, Cioni D, Crocetti L, et al. Early-stage hepatocellular carcinoma in patients with cirrhosis: long-term results of percutaneous image-guided radiofrequency ablation. Radiology. 2005;234(3):961–967. doi: 10.1148/radiol.2343040350. [DOI] [PubMed] [Google Scholar]
  • 6.Gillams AR, Lees WR. Five-year survival in 309 patients with colorectal liver metastases treated with radiofrequency ablation. European Radiology. 2009;19(5):1206–1213. doi: 10.1007/s00330-008-1258-5. [DOI] [PubMed] [Google Scholar]
  • 7.Thumar AB, Trabulsi EJ, Lallas CD, Brown DB. Thermal ablation of renal cell carcinoma: triage, treatment, and follow-up. Journal of Vascular and Interventional Radiology. 2010;21(8):S233–S241. doi: 10.1016/j.jvir.2010.01.050. [DOI] [PubMed] [Google Scholar]
  • 8.Zemlyak A, Moore WH, Bilfinger TV. Comparison of survival after sublobar resections and ablative therapies for stage I non-small cell lung cancer. Journal of the American College of Surgeons. 2010;211(1):68–72. doi: 10.1016/j.jamcollsurg.2010.03.020. [DOI] [PubMed] [Google Scholar]
  • 9.Lü MD, Kuang M, Liang LJ, et al. Surgical resection versus percutaneous thermal ablation for early-stage hepatocellular carcinoma: a randomized clinical trial. National Medical Journal of China. 2006;86(12):801–805. [PubMed] [Google Scholar]
  • 10.Paulet E, Aubé C, Pessaux P, et al. Factors limiting complete tumor ablation by radiofrequency ablation. CardioVascular and Interventional Radiology. 2008;31(1):107–115. doi: 10.1007/s00270-007-9208-1. [DOI] [PubMed] [Google Scholar]
  • 11.Allen BC, Remer EM. Percutaneous cryoablation of renal tumors: patient selection, technique, and postprocedural imaging. Radiographics. 2010;30(4):887–900. doi: 10.1148/rg.304095134. [DOI] [PubMed] [Google Scholar]
  • 12.Wright AS, Sampson LA, Warner TF, Mahvi DM, Lee FT. Radiofrequency versus microwave ablation in a hepatic porcine model. Radiology. 2005;236(1):132–139. doi: 10.1148/radiol.2361031249. [DOI] [PubMed] [Google Scholar]
  • 13.McTaggart RA, Dupuy DE. Thermal ablation of lung tumors. Techniques in Vascular and Interventional Radiology. 2007;10(2):102–113. doi: 10.1053/j.tvir.2007.09.004. [DOI] [PubMed] [Google Scholar]
  • 14.Eradat J, Abtin F, Gutierrez A, Suh R. Evaluation of treatment response after nonoperative therapy for early-stage non-small cell lung carcinoma. Cancer Journal. 2011;17(1):38–48. doi: 10.1097/PPO.0b013e31820a0948. [DOI] [PubMed] [Google Scholar]
  • 15.Siperstein AE, Gitomirski A. History and technological aspects of radiofrequency thermoablation. Cancer Journal. 2000;6(4):5293–5303. [Google Scholar]
  • 16.Clasen S, Krober SM, Kosan B, et al. Pathomorphologic evaluation of pulmonary radiofrequency ablation: proof of cell death is characterized by DNA fragmentation and apoptotic bodies. Cancer. 2008;113(11):3121–3129. doi: 10.1002/cncr.23882. [DOI] [PubMed] [Google Scholar]
  • 17.Guenette JP, Dupuy DE. Radiofrequency ablation of colorectal hepatic metastases. Journal of Surgical Oncology. 2010;102(8):978–987. doi: 10.1002/jso.21658. [DOI] [PubMed] [Google Scholar]
  • 18.Leung EYL, Roxburgh CSD, Leen E, Horgan PG. Combined resection and radiofrequency ablation for bilobar colorectal cancer liver metastases. Hepato-Gastroenterology. 2010;57(97):41–46. [PubMed] [Google Scholar]
  • 19.Cheung TT, Ng KK, Chok KS, et al. Combined resection and radiofrequency ablation for multifocal hepatocellular carcinoma: prognosis and outcomes. World Journal of Gastroenterology. 2010;16(24):3056–3062. doi: 10.3748/wjg.v16.i24.3056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kim SR, Han HJ, Park SJ, et al. Comparison between surgery and radiofrequency ablation for stage I non-small cell lung cancer. doi: 10.1016/j.ejrad.2010.12.091. European Journal of Radiology. In press. [DOI] [PubMed] [Google Scholar]
  • 21.Seki T, Wakabayashi M, Nakagawa T, et al. Ultrasonically guided percutaneous microwave coagulation therapy for small hepatocellular carcinoma. Cancer. 1994;74(3):817–825. doi: 10.1002/1097-0142(19940801)74:3<817::aid-cncr2820740306>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 22.Duan YQ, Gao YY, Ni XX, Wang Y, Feng L, Liang P. Changes in peripheral lymphocyte subsets in patients after partial microwave ablation of the spleen for secondary splenomegaly and hypersplenism: a preliminary study. International Journal of Hyperthermia. 2007;23(5):467–472. doi: 10.1080/02656730701474533. [DOI] [PubMed] [Google Scholar]
  • 23.Van Leenders GJLH, Beerlage HP, Ruijter ET, De La Rosette JJMCH, Van De Kaa CA. Histopathological changes associated with high intensity focused ultrasound (HIFU) treatment for localised adenocarcinoma of the prostate. Journal of Clinical Pathology. 2000;53(5):391–394. doi: 10.1136/jcp.53.5.391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fischer K, Gedroyc W, Jolesz FA. Focused ultrasound as a local therapy for liver cancer. Cancer Journal. 2010;16(2):118–124. doi: 10.1097/PPO.0b013e3181db7c32. [DOI] [PubMed] [Google Scholar]
  • 25.Kennedy JE. High-intensity focused ultrasound in the treatment of solid tumours. Nature Reviews Cancer. 2005;5(4):321–327. doi: 10.1038/nrc1591. [DOI] [PubMed] [Google Scholar]
  • 26.Kramer G, Steiner GE, Gröbl M, et al. Response to sublethal heat treatment of prostatic tumor cells and of prostatic tumor infiltrating T-cells. Prostate. 2004;58(2):109–120. doi: 10.1002/pros.10314. [DOI] [PubMed] [Google Scholar]
  • 27.Lu P, Zhu XQ, Xu ZL, Zhou Q, Zhang J, Wu F. Increased infiltration of activated tumor-infiltrating lymphocytes after high intensity focused ultrasound ablation of human breast cancer. Surgery. 2009;145(3):286–293. doi: 10.1016/j.surg.2008.10.010. [DOI] [PubMed] [Google Scholar]
  • 28.Tan AHH, Gilling PJ. Free-beam and contact laser soft-tissue ablation in urology. Journal of Endourology. 2003;17(8):587–593. doi: 10.1089/089277903322518581. [DOI] [PubMed] [Google Scholar]
  • 29.Lindner U, Trachtenberg J, Lawrentschuk N. Focal therapy in prostate cancer: modalities, findings and future considerations. Nature Reviews Urology. 2010;7(10):562–571. doi: 10.1038/nrurol.2010.142. [DOI] [PubMed] [Google Scholar]
  • 30.Zhao Z, Wu F. Minimally-invasive thermal ablation of early-stage breast cancer: a systemic review. European Journal of Surgical Oncology. 2010;30(12):1149–1155. doi: 10.1016/j.ejso.2010.09.012. [DOI] [PubMed] [Google Scholar]
  • 31.Kriechbaum K, Bolz M, Deak GG, Prager S, Scholda C, Schmidt-Erfurth U. High-Resolution Imaging of the Human Retina In Vivo after Scatter Photocoagulation Treatment Using a Semiautomated Laser System. Ophthalmology. 2010;117(3):545–551. doi: 10.1016/j.ophtha.2009.07.031. [DOI] [PubMed] [Google Scholar]
  • 32.Chiu D, Niu L, Mu F, et al. The experimental study for efficacy and safety of pancreatic cryosurgery. Cryobiology. 2010;60(3):281–286. doi: 10.1016/j.cryobiol.2010.01.006. [DOI] [PubMed] [Google Scholar]
  • 33.Kim JW, Abramson DH, Dunkel IJ. Current management strategies for intraocular retinoblastoma. Drugs. 2007;67(15):2173–2185. doi: 10.2165/00003495-200767150-00005. [DOI] [PubMed] [Google Scholar]
  • 34.Weaver ML, Atkinson D, Zemel R. Hepatic cryosurgery in treating colorectal metastases. Cancer. 1995;76(2):210–214. doi: 10.1002/1097-0142(19950715)76:2<210::aid-cncr2820760208>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
  • 35.Seifert JK, Morris DL. World survey on the complications of hepatic and prostate cryotherapy. World Journal of Surgery. 1999;23(2):109–114. doi: 10.1007/pl00013173. [DOI] [PubMed] [Google Scholar]
  • 36.Seifert JK, France MP, Zhao J, et al. Large volume hepatic freezing: association with significant release of the cytokines interleukin-6 and tumor necrosis factor α in a rat model. World Journal of Surgery. 2002;26(11):1333–1341. doi: 10.1007/s00268-002-6139-5. [DOI] [PubMed] [Google Scholar]
  • 37.Chapman WC, Debelak JP, Blackwell TS, et al. Hepatic cryoablation-induced acute lung injury: pulmonary hemodynamic and permeability effects in a sheep model. Archives of Surgery. 2000;135(6):667–672. doi: 10.1001/archsurg.135.6.667. [DOI] [PubMed] [Google Scholar]
  • 38.Jansen MC, van Hillegersberg R, Schoots IG, et al. Cryoablation induces greater inflammatory and coagulative responses than radiofrequency ablation or laser induced thermotherapy in a rat liver model. Surgery. 2010;147(5):686–695. doi: 10.1016/j.surg.2009.10.053. [DOI] [PubMed] [Google Scholar]
  • 39.Gravante G, Sconocchia G, Ong SL, Dennison AR, Lloyd DM. Immunoregulatory effects of liver ablation therapies for the treatment of primary and metastatic liver malignancies. Liver International. 2009;29(1):18–24. doi: 10.1111/j.1478-3231.2008.01915.x. [DOI] [PubMed] [Google Scholar]
  • 40.Soanes WA, Ablin RJ, Gonder MJ. Remission of metastatic lesions following cryosurgery in prostatic cancer: immunologic considerations. Journal of Urology. 1970;104(1):154–159. doi: 10.1016/s0022-5347(17)61690-2. [DOI] [PubMed] [Google Scholar]
  • 41.Ablin RJ, Soanes WA, Gonder MJ. Elution of in vivo bound antiprostatic epithelial antibodies following multiple cryotherapy of carcinoma of prostate. Urology. 1973;2(3):276–279. doi: 10.1016/0090-4295(73)90463-9. [DOI] [PubMed] [Google Scholar]
  • 42.Sanchez-Ortiz RF, Tannir N, Ahrar K, Wood CG. Spontaneous regression of pulmonary metastases from renal cell carcinoma after radio frequency ablation of the primary tumor: an in situ tumor vaccine? Journal of Urology. 2003;170(1):178–179. doi: 10.1097/01.ju.0000070823.38336.7b. [DOI] [PubMed] [Google Scholar]
  • 43.Matzinger P. Tolerance, danger, and the extended family. Annual Review of Immunology. 1994;12:991–1045. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
  • 44.Matzinger P. The danger model: a renewed sense of self. Science. 2002;296(5566):301–305. doi: 10.1126/science.1071059. [DOI] [PubMed] [Google Scholar]
  • 45.Blachere NE, Udono H, Janetzki S, Li Z, Heike M, Srivastava PK. Heat shock protein vaccines against cancer. Journal of Immunotherapy with Emphasis on Tumor Immunology. 1993;14(4):352–356. doi: 10.1097/00002371-199311000-00016. [DOI] [PubMed] [Google Scholar]
  • 46.Srivastava PK. Heat shock proteins in immune response to cancer: the fourth paradigm. Experientia. 1994;50(11-12):1054–1060. doi: 10.1007/BF01923461. [DOI] [PubMed] [Google Scholar]
  • 47.Blachere NE, Li Z, Chandawarkar RY, et al. Heat shock protein-peptide complexes, reconstituted in vitro, elicit peptide-specific cytotoxic T lymphocyte response and tumor immunity. Journal of Experimental Medicine. 1997;186(8):1315–1322. doi: 10.1084/jem.186.8.1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Calderwood SK, Mambula SS, Gray PJ., Jr. Extracellular heat shock proteins in cell signaling and immunity. Annals of the New York Academy of Sciences. 2007;1113:28–39. doi: 10.1196/annals.1391.019. [DOI] [PubMed] [Google Scholar]
  • 49.Dromi SA, Walsh MP, Herby S, et al. Radiofrequency ablation induces antigen-presenting cell infiltration and amplification of weak tumor-induced immunity. Radiology. 2009;251(1):58–66. doi: 10.1148/radiol.2511072175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kinoshita T, Iwamoto E, Tsuda H, Seki K. Radiofrequency ablation as local therapy for early breast carcinomas. Breast Cancer. 2010;18(1):10–17. doi: 10.1007/s12282-009-0186-9. [DOI] [PubMed] [Google Scholar]
  • 51.Ali MY, Grimm CF, Ritter M, et al. Activation of dendritic cells by local ablation of hepatocellular carcinoma. Journal of Hepatology. 2005;43(5):817–822. doi: 10.1016/j.jhep.2005.04.016. [DOI] [PubMed] [Google Scholar]
  • 52.Evrard S, Menetrier-Caux C, Biota C, et al. Cytokines pattern after surgical radiofrequency ablation of liver colorectal metastases. Gastroenterologie Clinique et Biologique. 2007;31(2):141–145. doi: 10.1016/s0399-8320(07)89344-4. [DOI] [PubMed] [Google Scholar]
  • 53.Jansen MC, van Wanrooy S, van Hillegersberg R, et al. Assessment of systemic inflammatory response (SIR) in patients undergoing radiofrequency ablation or partial liver resection for liver tumors. European Journal of Surgical Oncology. 2008;34(6):662–667. doi: 10.1016/j.ejso.2007.06.009. [DOI] [PubMed] [Google Scholar]
  • 54.Fietta AM, Morosini M, Passadore I, et al. Systemic inflammatory response and downmodulation of peripheral CD25+Foxp3+ T-regulatory cells in patients undergoing radiofrequency thermal ablation for lung cancer. Human Immunology. 2009;70(7):477–486. doi: 10.1016/j.humimm.2009.03.012. [DOI] [PubMed] [Google Scholar]
  • 55.Schueller G, Kettenbach J, Sedivy R, et al. Expression of heat shock proteins in human hepatocellular carcinoma after radiofrequency ablation in an animal model. Oncology Reports. 2004;12(3):495–499. [PubMed] [Google Scholar]
  • 56.Yang WL, Nair DG, Makizumi R, et al. Heat shock protein 70 is induced in mouse human colon tumor xenografts after sublethal radiofrequency ablation. Annals of Surgical Oncology. 2004;11(4):399–406. doi: 10.1245/ASO.2004.08.013. [DOI] [PubMed] [Google Scholar]
  • 57.Liu Q, Zhai B, Yang W, et al. Abrogation of local cancer recurrence after radiofrequency ablation by dendritic cell-based hyperthermic tumor vaccine. Molecular Therapy. 2009;17(12):2049–2057. doi: 10.1038/mt.2009.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rai R, Richardson C, Flecknell P, Robertson H, Burt A, Manas DM. Study of apoptosis and heat shock protein (HSP) expression in hepatocytes following radiofrequency ablation (RFA) Journal of Surgical Research. 2005;129(1):147–151. doi: 10.1016/j.jss.2005.03.020. [DOI] [PubMed] [Google Scholar]
  • 59.Solazzo SA, Ahmed M, Schor-Bardach R, et al. Liposomal doxorubicin increases radiofrequency ablation-induced tumor destruction by increasing cellular oxidative and nitrative stress and accelerating apoptotic pathways. Radiology. 2010;255(1):62–74. doi: 10.1148/radiol.09091196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bhardwaj N, Dormer J, Ahmad F, et al. Heat shock protein 70 expression following hepatic radiofrequency ablation is affected by adjacent vasculature. doi: 10.1016/j.jss.2010.09.040. Journal of Surgical Research. In press. [DOI] [PubMed] [Google Scholar]
  • 61.Todorova VK, Klimberg VS, Hennings L, Kieber-Emmons T, Pashov A. Immunomodulatory effects of radiofrequency ablation in a breast cancer model. Immunological Investigations. 2010;39(1):74–92. doi: 10.3109/08820130903428291. [DOI] [PubMed] [Google Scholar]
  • 62.Schueller G, Kettenbach J, Sedivy R, et al. Heat shock protein expression induced by percutaneous radiofrequency ablation of hepatocellular carcinoma in vivo. International Journal of Oncology. 2004;24(3):609–613. [PubMed] [Google Scholar]
  • 63.Haen SP, Gouttefangeas C, Schmidt D, et al. Elevated serum levels of heat shock protein 70 can be detected after radiofrequency ablation. Cell Stress and Chaperones. 2011;16(5):495–504. doi: 10.1007/s12192-011-0261-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hänsler J, Neureiter D, Strobel D, et al. Cellular and vascular reactions in the liver to radio-frequency thermo-ablation with wet needle applicators: study on juvenile domestic pigs. European Surgical Research. 2002;34(5):357–363. doi: 10.1159/000064000. [DOI] [PubMed] [Google Scholar]
  • 65.Nijkamp MW, Borren A, Govaert KM, et al. Radiofrequency ablation of colorectal liver metastases induces an inflammatory response in distant hepatic metastases but not in local accelerated outgrowth. Journal of Surgical Oncology. 2010;101(7):551–556. doi: 10.1002/jso.21570. [DOI] [PubMed] [Google Scholar]
  • 66.Rughetti A, Rahimi H, Rossi P, et al. Modulation of blood circulating immune cells by radiofrequency tumor ablation. Journal of Experimental and Clinical Cancer Research. 2003;22(4):247–250. [PubMed] [Google Scholar]
  • 67.Zerbini A, Pilli M, Laccabue D, et al. Radiofrequency thermal ablation for hepatocellular carcinoma stimulates autologous NK-cell response. Gastroenterology. 2010;138(5):1931–1942. doi: 10.1053/j.gastro.2009.12.051. [DOI] [PubMed] [Google Scholar]
  • 68.Wissniowski TT, Hünsler J, Neureiter D, et al. Activation of tumor-specific T lymphocytes by radio-frequency ablation of the VX2 hepatoma in rabbits. Cancer Research. 2003;63(19):6496–6500. [PubMed] [Google Scholar]
  • 69.den Brok MHMGM, Sutmuller RPM, Nierkens S, et al. Efficient loading of dendritic cells following cryo and radiofrequency ablation in combination with immune modulation induces anti-tumour immunity. British Journal of Cancer. 2006;95(7):896–905. doi: 10.1038/sj.bjc.6603341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zerbini A, Pilli M, Penna A, et al. Radiofrequency thermal ablation of hepatocellular carcinoma liver nodules can activate and enhance tumor-specific T-cell responses. Cancer Research. 2006;66(2):1139–1146. doi: 10.1158/0008-5472.CAN-05-2244. [DOI] [PubMed] [Google Scholar]
  • 71.Matuszewski M, Michajłowski J, Michajłowski I, et al. Impact of radiofrequency ablation on PBMC subpopulation in patients with renal cell carcinoma. doi: 10.1016/j.urolonc.2009.11.021. Urologic Oncology. In press. [DOI] [PubMed] [Google Scholar]
  • 72.den Brok MHMGM, Sutmuller RPM, van der Voort R, et al. In situ tumor ablation creates an antigen source for the generation of antitumor immunity. Cancer Research. 2004;64(11):4024–4029. doi: 10.1158/0008-5472.CAN-03-3949. [DOI] [PubMed] [Google Scholar]
  • 73.Iida N, Nakamoto Y, Baba T, et al. Antitumor effect after radiofrequency ablation of murine hepatoma is augmented by an active variant of CC chemokine ligand 3/macrophage inflammatory protein-1á. Cancer Research. 2010;70(16):6556–6565. doi: 10.1158/0008-5472.CAN-10-0096. [DOI] [PubMed] [Google Scholar]
  • 74.Johnson EE, Yamane BH, Buhtoiarov IN, et al. Radiofrequency ablation combined with KS-IL2 immunocytokine (EMD 273066) results in an enhanced antitumor effect against murine colon adenocarcinoma. Clinical Cancer Research. 2009;15(15):4875–4884. doi: 10.1158/1078-0432.CCR-09-0110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Napoletano C, Taurino F, Biffoni M, et al. RFA strongly modulates the immune system and anti-tumor immune responses in metastatic liver patients. International Journal of Oncology. 2008;32(2):481–490. [PubMed] [Google Scholar]
  • 76.Hänsler J, Wissniowski TT, Schuppan D, et al. Activation and dramatically increased cytolytic activity of tumor specific T lymphocytes after radio-frequency ablation in patients with hepatocellular carcinoma and colorectal liver metastases. World Journal of Gastroenterology. 2006;12(23):3716–3721. doi: 10.3748/wjg.v12.i23.3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hiroishi K, Eguchi J, Baba T, et al. Strong CD8+ T-cell responses against tumor-associated antigens prolong the recurrence-free interval after tumor treatment in patients with hepatocellular carcinoma. Journal of Gastroenterology. 2010;45(4):451–458. doi: 10.1007/s00535-009-0155-2. [DOI] [PubMed] [Google Scholar]
  • 78.Widenmeyer M, Shebzukhov Y, Haen SP, et al. Analysis of tumor antigen-specific T cells and antibodies in cancer patients treated with radiofrequency ablation. International Journal of Cancer. 2011;128(11):2653–2662. doi: 10.1002/ijc.25601. [DOI] [PubMed] [Google Scholar]
  • 79.Sabel MS, Su G, Griffith KA, Chang AE. Rate of freeze alters the immunologic response after cryoablation of breast cancer. Annals of Surgical Oncology. 2010;17(4):1187–1193. doi: 10.1245/s10434-009-0846-1. [DOI] [PubMed] [Google Scholar]
  • 80.Ahmed M, Brace CL, Lee FT, Jr., Goldberg SN. Principles of and advances in percutaneous ablation. Radiology. 2011;258(2):351–369. doi: 10.1148/radiol.10081634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sabel MS. Cryoablation for breast cancer: no need to turn a cold shoulder. Journal of Surgical Oncology. 2008;97(6):485–486. doi: 10.1002/jso.20887. [DOI] [PubMed] [Google Scholar]
  • 82.Hoffmann NE, Bischof JC. The cryobiology of cryosurgical injury. Urology. 2002;60(2):40–49. doi: 10.1016/s0090-4295(02)01683-7. [DOI] [PubMed] [Google Scholar]
  • 83.Sabel MS, Nehs MA, Su G, Lowler KP, Ferrara JLM, Chang AE. Immunologic response to cryoablation of breast cancer. Breast Cancer Research and Treatment. 2005;90(1):97–104. doi: 10.1007/s10549-004-3289-1. [DOI] [PubMed] [Google Scholar]
  • 84.Osada S, Imai H, Tomita H, et al. Serum cytokine levels in response to hepatic cryoablation. Journal of Surgical Oncology. 2007;95(6):491–498. doi: 10.1002/jso.20712. [DOI] [PubMed] [Google Scholar]
  • 85.Si T, Guo Z, Hao X. Immunologic response to primary cryoablation of high-risk prostate cancer. Cryobiology. 2008;57(1):66–71. doi: 10.1016/j.cryobiol.2008.06.003. [DOI] [PubMed] [Google Scholar]
  • 86.Gazzaniga S, Bravo A, Goldszmid SR, et al. Inflammatory changes after cryosurgery-induced necrosis in human melanoma xenografted in nude mice. Journal of Investigative Dermatology. 2001;116(5):664–671. doi: 10.1046/j.0022-202x.2001.01313.x. [DOI] [PubMed] [Google Scholar]
  • 87.Thakur A, Littrup P, Paul EN, Adam B, Heilbrun LK, Lum LG. Induction of specific cellular and humoral responses against renal cell carcinoma after combination therapy with cryoablation and granulocyte-macrophage colony stimulating factor: a pilot study. Journal of Immunotherapy. 2011;34(5):457–467. doi: 10.1097/CJI.0b013e31821dcba5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Miyabara EH, Martin JL, Griffin TM, Moriscot AS, Mestril R. Overexpression of inducible 70-kDa heat shock protein in mouse attenuates skeletal muscle damage induced by cryolesioning. American Journal of Physiology. 2006;290(4):C1128–C1138. doi: 10.1152/ajpcell.00399.2005. [DOI] [PubMed] [Google Scholar]
  • 89.Li M, Zhang S, Zhou Y, Guo Y, Jiang X, Miao L. Argon-helium cryosurgery for treatment of C6 gliomas in rats and its effect on cellular immunity. Technology in Cancer Research and Treatment. 2010;9(1):87–93. doi: 10.1177/153303461000900110. [DOI] [PubMed] [Google Scholar]
  • 90.Jacob G, Li AKC, Hobbs KEF. A comparison of cryodestruction with excision or infarction of an implanted tumor in rat liver. Cryobiology. 1984;21(2):148–156. doi: 10.1016/0011-2240(84)90206-2. [DOI] [PubMed] [Google Scholar]
  • 91.Miya K, Saji S, Morita T, et al. Immunological response of regional lymph nodes after tumor cryosurgery: experimental study in rats. Cryobiology. 1986;23(4):290–295. doi: 10.1016/0011-2240(86)90034-9. [DOI] [PubMed] [Google Scholar]
  • 92.Machlenkin A, Goldberger O, Tirosh B, et al. Combined dendritic cell cryotherapy of tumor induces systemic antimetastatic immunity. Clinical Cancer Research. 2005;11(13):4955–4961. doi: 10.1158/1078-0432.CCR-04-2422. [DOI] [PubMed] [Google Scholar]
  • 93.Bagley DH, Faraci RP, Marrone JC, Beazley RM. Lymphocyte mediated cytotoxicity after cryosurgery of a murine sarcoma. Journal of Surgical Research. 1974;17(6):404–406. doi: 10.1016/0022-4804(74)90151-6. [DOI] [PubMed] [Google Scholar]
  • 94.Urano M, Tanaka C, Sugiyama Y, Miya K, Saji S. Antitumor effects of residual tumor after cryoablation: the combined effect of residual tumor and a protein-bound polysaccharide on multiple liver metastases in a murine model. Cryobiology. 2003;46(3):238–245. doi: 10.1016/s0011-2240(03)00039-7. [DOI] [PubMed] [Google Scholar]
  • 95.Sabel MS, Arora A, Su G, Chang AE. Adoptive immunotherapy of breast cancer with lymph node cells primed by cryoablation of the primary tumor. Cryobiology. 2006;53(3):360–366. doi: 10.1016/j.cryobiol.2006.07.004. [DOI] [PubMed] [Google Scholar]
  • 96.Udagawa M, Kudo-Saito C, Hasegawa G, et al. Enhancement of immunologic tumor regression by intratumoral administration of dendritic cells in combination with cryoablative tumor pretreatment and Bacillus Calmette-Guerin cell wall skeleton stimulation. Clinical Cancer Research. 2006;12(24):7465–7475. doi: 10.1158/1078-0432.CCR-06-1840. [DOI] [PubMed] [Google Scholar]
  • 97.Levy MY, Sidana A, Chowdhury WH, et al. Cyclophosphamide unmasks an antimetastatic effect of local tumor cryoablation. Journal of Pharmacology and Experimental Therapeutics. 2009;330(2):596–601. doi: 10.1124/jpet.109.152603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Redondo P, del Olmo J, López-Diaz De Cerio A, et al. Imiquimod enhances the systemic immunity attained by local cryosurgery destruction of melanoma lesions. Journal of Investigative Dermatology. 2007;127(7):1673–1680. doi: 10.1038/sj.jid.5700777. [DOI] [PubMed] [Google Scholar]
  • 99.Renziehausen K, Schroder M, Seeber C, Krafft W, Kleinschmidt R. Immunologic studies in relation to cryotherapy of gynecologic diseases. Zentralblatt fuer Gynaekologie. 1975;97(24):1492–1501. [PubMed] [Google Scholar]
  • 100.Eastham RJ, III, Mason JM, Jennings BR. T cell rosette test in squamous cell carcinoma of the head and neck. Archives of Otolaryngology. 1976;102(3):171–175. doi: 10.1001/archotol.1976.00780080093013. [DOI] [PubMed] [Google Scholar]
  • 101.Weyer U, Petersen I, Ehrke C, et al. Immunomodulation induced by cryosurgery in malignant melanoma. Onkologie. 1989;12(6):291–296. doi: 10.1159/000216665. [DOI] [PubMed] [Google Scholar]
  • 102.Si T, Guo Z, Hao X. Combined cryoablation and GM-CSF treatment for metastatic hormone refractory prostate cancer. Journal of Immunotherapy. 2009;32(1):86–91. doi: 10.1097/CJI.0b013e31818df785. [DOI] [PubMed] [Google Scholar]
  • 103.Yamashita T, Hayakawa K, Hosokawa M. Enhanced tumor metastases in rats following cryosurgery of primary tumor. Gann. 1982;73(2):222–228. [PubMed] [Google Scholar]
  • 104.Wing MG, Rogers K, Jacob G, Rees RC. Characterisation of suppressor cells generated following cryosurgery of an HSV-2-induced fibrosarcoma. Cancer Immunology Immunotherapy. 1988;26(2):169–175. doi: 10.1007/BF00205611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhou L, Fu JL, Lu YY, et al. Regulatory T cells are associated with post-cryoablation prognosis in patients with hepatitis B virus-related hepatocellular carcinoma. Journal of Gastroenterology. 2010;45(9):968–978. doi: 10.1007/s00535-010-0243-3. [DOI] [PubMed] [Google Scholar]
  • 106.Yantorno C, Soanes WA, Gonder MJ, Shulman S. Studies in cryo-immunology. I. The production of antibodies to urogenital tissue in consequence of freezing treatment. Immunology. 1967;12(4):395–410. [PMC free article] [PubMed] [Google Scholar]
  • 107.Riera CM, Brandt EJ, Shulman S. Studies in cryo-immunology. IV. Antibody development in rabbits after iso-immunization followed by freezing. Immunology. 1968;15(6):779–787. [PMC free article] [PubMed] [Google Scholar]
  • 108.Shulman S, Brandt EJ, Yantorno C. Studies in cryo-immunology. II. Tissue and species specificity of the autoantibody response and comparison with iso-immunization. Immunology. 1968;14(2):149–158. [PMC free article] [PubMed] [Google Scholar]
  • 109.Shulman S, Bronson P, Riera C, Brandt EJ, Yantorno C. Studies in cryo-immumology. 3. The immunoglobulin nature of the antibody response. Immunology. 1968;14(4):541–551. [PMC free article] [PubMed] [Google Scholar]
  • 110.Blackwood CE, Cooper IS. Response of experimental tumor systems to cryosurgery. Cryobiology. 1972;9(6):508–515. doi: 10.1016/0011-2240(72)90172-1. [DOI] [PubMed] [Google Scholar]
  • 111.Ablin RJ. Cryosurgery of the rabbit prostate. Comparison of the immune response of immature amd mature bucks. Cryobiology. 1974;11(5):416–422. doi: 10.1016/0011-2240(74)90108-4. [DOI] [PubMed] [Google Scholar]
  • 112.Ablin RJ, Jagodzinski RV, Prox C. Cryosurgery of the monkey (macaque) prostate. I. Humoral immunologic responsiveness following cryostimulation. Cryobiology. 1976;13(1):47–53. doi: 10.1016/0011-2240(76)90158-9. [DOI] [PubMed] [Google Scholar]
  • 113.Muller LC, Micksche M, Yamagata S, Kerschbaumer F. Therapeutic effect of cryosurgery of murine osteosarcoma—influence on disease outcome and immune function. Cryobiology. 1985;22(1):77–85. doi: 10.1016/0011-2240(85)90010-0. [DOI] [PubMed] [Google Scholar]
  • 114.Ablin RJ. Serum proteins in prostatic cancer. VI. Reduction of the suppressive (“blocking”?) properties of serum on in vitro parameters of cell mediated immunologic responsiveness following cryosurgery. Urologia Internationalis. 1977;32(1):65–73. doi: 10.1159/000280112. [DOI] [PubMed] [Google Scholar]
  • 115.Kogel H, Grundmann R, Fohlmeister I, Pichlmaier H. Cryotherapy of rectal carcinoma. Immunological results. Zentralblatt fur Chirurgie. 1985;110(2-3):147–154. [PubMed] [Google Scholar]
  • 116.Ravindranath MH, Wood TF, Soh D, et al. Cryosurgical ablation of liver tumors in colon cancer patients increases the serum total ganglioside level and then selectively augments antiganglioside IgM. Cryobiology. 2002;45(1):10–21. doi: 10.1016/s0011-2240(02)00102-5. [DOI] [PubMed] [Google Scholar]
  • 117.Ahmad F, Gravante G, Bhardwaj N, et al. Renal effects of microwave ablation compared with radiofrequency, cryotherapy and surgical resection at different volumes of the liver treated. Liver International. 2010;30(9):1305–1314. doi: 10.1111/j.1478-3231.2010.02290.x. [DOI] [PubMed] [Google Scholar]
  • 118.Chen Z, Shen S, Peng B, Tao J. Intratumoural GM-CSF microspheres and CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a subcutaneous murine hepatoma model. International Journal of Hyperthermia. 2009;25(5):374–382. doi: 10.1080/02656730902976807. [DOI] [PubMed] [Google Scholar]
  • 119.Dong BW, Zhang J, Liang P, et al. Sequential pathological and immunologic analysis of percutaneous microwave coagulation therapy of hepatocellular carcinoma. International Journal of Hyperthermia. 2003;19(2):119–133. doi: 10.1080/0265673021000017154. [DOI] [PubMed] [Google Scholar]
  • 120.Zhou P, Liang P, Dong B, Yu X, Han Z, Xu Y. Phase I clinical study of combination therapy with microwave ablation and cellular immunotherapy in hepatocellular carcinoma. Cancer Biology and Therapy. 2011;11(5):450–456. doi: 10.4161/cbt.11.5.14669. [DOI] [PubMed] [Google Scholar]
  • 121.Zhang Y, Deng J, Feng J, Wu F. Enhancement of antitumor vaccine in ablated hepatocellular carcinoma by high-intensity focused ultrasound. World Journal of Gastroenterology. 2010;16(28):3584–3591. doi: 10.3748/wjg.v16.i28.3584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wu F, Wang ZB, Cao YD, et al. Expression of tumor antigens and heat-shock protein 70 in breast cancer cells after high-intensity focused ultrasound ablation. Annals of Surgical Oncology. 2007;14(3):1237–1242. doi: 10.1245/s10434-006-9275-6. [DOI] [PubMed] [Google Scholar]
  • 123.Wang X, Sun J. High-intensity focused ultrasound in patients with late-stage pancreatic carcinoma. Chinese Medical Journal. 2002;115(9):1332–1335. [PubMed] [Google Scholar]
  • 124.Deng J, Zhang Y, Feng J, Wu F. Dendritic cells loaded with ultrasound-ablated tumour induce in vivo specific antitumour immune responses. Ultrasound in Medicine and Biology. 2010;36(3):441–448. doi: 10.1016/j.ultrasmedbio.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 125.Rosberger DF, Coleman DJ, Silverman R, Woods S, Rondeau M, Cunningham-Rundles S. Immunomodulation in choroidal melanoma: reversal of inverted CD4/CD8 ratios following treatment with ultrasonic hyperthermia. Biotechnology Therapeutics. 1994;5(1-2):59–68. [PubMed] [Google Scholar]
  • 126.Wu F, Wang ZB, Lu P, et al. Activated anti-tumor immunity in cancer patients after high intensity focused ultrasound ablation. Ultrasound in Medicine and Biology. 2004;30(9):1217–1222. doi: 10.1016/j.ultrasmedbio.2004.08.003. [DOI] [PubMed] [Google Scholar]
  • 127.Shimura M, Yasuda K, Nakazawa T, et al. Panretinal photocoagulation induces pro-inflammatory cytokines and macular thickening in high-risk proliferative diabetic retinopathy. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2009;247(12):1617–1624. doi: 10.1007/s00417-009-1147-x. [DOI] [PubMed] [Google Scholar]
  • 128.Isbert C, Ritz JP, Roggan A, et al. Enhancement of the immune response to residual intrahepatic tumor tissue by Laser-Induced Thermotherapy (LITT) compared to hepatic resection. Lasers in Surgery and Medicine. 2004;35(4):284–292. doi: 10.1002/lsm.20097. [DOI] [PubMed] [Google Scholar]
  • 129.Stafford RJ, Fuentes D, Elliott AA, Weinberg JS, Ahrar K. Laser-induced thermal therapy for tumor ablation. Critical Reviews in Biomedical Engineering. 2010;38(1):79–100. doi: 10.1615/critrevbiomedeng.v38.i1.70. [DOI] [PubMed] [Google Scholar]
  • 130.Kallio R, Sequeiros R, Surcel HM, Ohtonen P, Kiviniemi H, Syrjälä H. Early cytokine responses after percutaneous magnetic resonance imaging guided laser thermoablation of malignant liver tumors. Cytokine. 2006;34(5-6):278–283. doi: 10.1016/j.cyto.2006.06.003. [DOI] [PubMed] [Google Scholar]
  • 131.Nikfarjam M, Muralidharan V, Su K, Malcontenti-Wilson C, Christophi C. Patterns of heat shock protein (HSP70) expression and Kupffer cell activity following thermal ablation of liver and colorectal liver metastases. International Journal of Hyperthermia. 2005;21(4):319–332. doi: 10.1080/02656730500133736. [DOI] [PubMed] [Google Scholar]
  • 132.Lin WX, Fifis T, Malcontenti-Wilson C, et al. Induction of Th1Immune responses following laser ablation in a murine model of colorectal liver metastases. Journal of Translational Medicine. 2011;9(1, article 83) doi: 10.1186/1479-5876-9-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Schälte G, Henzler D, Waning C, Tacke J, Rossaint R, Mahnken AH. Case study of hepatic radiofrequency ablation causing a systemic inflammatory response under total intravenous anesthesia. Korean Journal of Radiology. 2010;11(6):640–647. doi: 10.3348/kjr.2010.11.6.640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Schell SR, Wessels FJ, Abouhamze A, Moldawer LL, Copeland EM. Pro- and antiinflammatory cytokine production after radiofrequency ablation of unresectable hepatic tumors. Journal of the American College of Surgeons. 2002;195(6):774–781. doi: 10.1016/s1072-7515(02)01333-9. [DOI] [PubMed] [Google Scholar]
  • 135.Nierkens S, den Brok MH, Roelofsen T, et al. Route of administration of the TLR9 agonist CpG critically determines the efficacy of cancer immunotherapy in mice. PloS One. 2009;4(12):p. e8368. doi: 10.1371/journal.pone.0008368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yang R, Reilly CR, Rescorla FJ, et al. Effects of high-intensity focused ultrasound in the treatment of experimental neuroblastoma. Journal of Pediatric Surgery. 1992;27(2):246–251. doi: 10.1016/0022-3468(92)90321-w. [DOI] [PubMed] [Google Scholar]
  • 137.Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nature Reviews Immunology. 2008;8(1):59–73. doi: 10.1038/nri2216. [DOI] [PubMed] [Google Scholar]
  • 138.Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nature Medicine. 1999;5(11):1249–1255. doi: 10.1038/15200. [DOI] [PubMed] [Google Scholar]
  • 139.Zitvogel L, Kepp O, Senovilla L, Menger L, Chaput N, Kroemer G. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clinical Cancer Research. 2010;16(12):3100–3104. doi: 10.1158/1078-0432.CCR-09-2891. [DOI] [PubMed] [Google Scholar]
  • 140.Misao A, Sakata K, Saji S, Kunieda T. Late appearance of resistance to tumor rechallenge following cryosurgery. A study in an experimental mammary tumor of the rat. Cryobiology. 1981;18(4):386–389. doi: 10.1016/0011-2240(81)90111-5. [DOI] [PubMed] [Google Scholar]
  • 141.Matsumura K, Sakata K, Saji S, Misao A, Kunieda T. Antitumor immunologic reactivity in the relatively early period after cryosurgery: experimental studies in the rat. Cryobiology. 1982;19(3):263–272. doi: 10.1016/0011-2240(82)90152-3. [DOI] [PubMed] [Google Scholar]
  • 142.Wolchok JD, Hoos A, O’Day S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clinical Cancer Research. 2009;15(23):7412–7420. doi: 10.1158/1078-0432.CCR-09-1624. [DOI] [PubMed] [Google Scholar]
  • 143.Nishinaka Y, Ravindranath MH, Irie RF. Development of a human monoclonal antibody to ganglioside G(M2) with potential for cancer treatment. Cancer Research. 1996;56(24):5666–5671. [PubMed] [Google Scholar]
  • 144.Nakamura K, Hanibuchi M, Yano S, et al. Apoptosis induction of human lung cancer cell line in multicellular heterospheroids with humanized antiganglioside GM2 monoclonal antibody. Cancer Research. 1999;59(20):5323–5330. [PubMed] [Google Scholar]
  • 145.Hayakawa K, Yamashita T, Suzuki K, et al. Comparative immunological studies in rats following cryosurgery and surgical excision of 3-methylcholanthrene-induced primary autochthonous tumors. Gann, The Japanese Journal of Cancer Research. 1982;73(3):462–469. [PubMed] [Google Scholar]
  • 146.Kimura H. Comparative immunological studies on cryosurgery and surgical operation using Moloney murine sarcoma virus-induced primary tumors in BALB/c mice. Gann. 1978;69(4):507–515. [PubMed] [Google Scholar]
  • 147.Wu F, Wang ZB, Cao YD, et al. Changes in biologic characteristics of breast cancer treated with high-intensity focused ultrasound. Ultrasound in Medicine and Biology. 2003;29(10):1487–1492. doi: 10.1016/s0301-5629(03)01034-2. [DOI] [PubMed] [Google Scholar]
  • 148.Wu F, Wang ZB, Cao YD, et al. Heat fixation of cancer cells ablated with high-intensity-focused ultrasound in patients with breast cancer. American Journal of Surgery. 2006;192(2):179–184. doi: 10.1016/j.amjsurg.2006.03.014. [DOI] [PubMed] [Google Scholar]
  • 149.Vrabec TR, Reber RN, Magargal LE, Donoso LA. S-antigen. Identification of human T-cell lymphocyte proliferation sites. Archives of Ophthalmology. 1990;108(10):1470–1473. doi: 10.1001/archopht.1990.01070120118040. [DOI] [PubMed] [Google Scholar]
  • 150.Pulendran B. Immune activation: death, danger and dendritic cells. Current Biology. 2004;14(1):R30–R32. doi: 10.1016/j.cub.2003.12.020. [DOI] [PubMed] [Google Scholar]
  • 151.Zeh HJ, Lotze MT. Addicted to death: invasive cancer and the immune response to unscheduled cell death. Journal of Immunotherapy. 2005;28(1):1–9. doi: 10.1097/00002371-200501000-00001. [DOI] [PubMed] [Google Scholar]
  • 152.Nikfarjam M, Muralidharan V, Christophi C. Mechanisms of focal heat destruction of liver tumors. Journal of Surgical Research. 2005;127(2):208–223. doi: 10.1016/j.jss.2005.02.009. [DOI] [PubMed] [Google Scholar]
  • 153.Chida S, Okada K, Suzuki N, Komori C, Shimada Y. Infiltration by macrophages and lymphocytes in transplantable mouse sarcoma after irradiation with high-intensity focused ultrasound. Anticancer Research. 2009;29(10):3877–3882. [PubMed] [Google Scholar]
  • 154.Srivastava PK. Hypothesis: controlled necrosis as a tool for immunotherapy of human cancer. Cancer Immun. 2003;3:p. 4. [PubMed] [Google Scholar]
  • 155.Schildkopf P, Ott OJ, Frey B, et al. Biological rationales and clinical applications of temperature controlled hyperthermia—implications for multimodal cancer treatments. Current Medicinal Chemistry. 2010;17(27):3045–3057. doi: 10.2174/092986710791959774. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Developmental Immunology are provided here courtesy of Wiley

RESOURCES