Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 12.
Published in final edited form as: J Vasc Interv Radiol. 2014 Oct 23;25(11):1691–705.e4. doi: 10.1016/j.jvir.2014.08.027

Image-Guided Tumor Ablation: Standardization of Terminology and Reporting Criteria—A 10-Year Update

Muneeb Ahmed 1, Luigi Solbiati 2, Christopher L Brace 3, David J Breen 4, Matthew R Callstrom 5, J William Charboneau 6, Min-Hua Chen 7, Byung Ihn Choi 8, Thierry de Baère 9, Gerald D Dodd III 10, Damian E Dupuy 11, Debra A Gervais 12, David Gianfelice 13, Alice R Gillams 14, Fred T Lee Jr 15, Edward Leen 16, Riccardo Lencioni 17, Peter J Littrup 18, Tito Livraghi 19, David S Lu 20, John P McGahan 21, Maria Franca Meloni 22, Boris Nikolic 23, Philippe L Pereira 24, Ping Liang 25, Hyunchul Rhim 26, Steven C Rose 27, Riad Salem 28, Constantinos T Sofocleous 29, Stephen B Solomon 30, Michael C Soulen 31, Masatoshi Tanaka 32, Thomas J Vogl 33, Bradford J Wood 34, S Nahum Goldberg 35, International Working Group on Image-Guided Tumor Ablation, Interventional Oncology Sans Frontières Expert Panel, Technology Assessment Committee of the Society of Interventional Radiology, and the Standard of Practice Committee of the Cardiovascular and Interventional Radiological Society of Europe
PMCID: PMC7660986  NIHMSID: NIHMS894127  PMID: 25442132

Abstract

Image-guided tumor ablation has become a well-established hallmark of local cancer therapy. The breadth of options available in this growing field increases the need for standardization of terminology and reporting criteria to facilitate effective communication of ideas and appropriate comparison among treatments that use different technologies, such as chemical (eg, ethanol or acetic acid) ablation, thermal therapies (eg, radiofrequency, laser, microwave, focused ultrasound, and cryoablation) and newer ablative modalities such as irreversible electroporation. This updated consensus document provides a framework that will facilitate the clearest communication among investigators regarding ablative technologies. An appropriate vehicle is proposed for reporting the various aspects of image-guided ablation therapy including classification of therapies, procedure terms, descriptors of imaging guidance, and terminology for imaging and pathologic findings. Methods are addressed for standardizing reporting of technique, follow-up, complications, and clinical results. As noted in the original document from 2003, adherence to the recommendations will improve the precision of communications in this field, leading to more accurate comparison of technologies and results, and ultimately to improved patient outcomes.


In 2003, the International Working Group on Image-Guided Tumor Ablation published a document titled “Image-Guided Tumor Ablation: Proposal for Standardization of Terms and Reporting Criteria” (1). At the time, image-guided tumor ablation, and indeed, the subspecialty of interventional oncology, was in its infancy. Nevertheless, it was acknowledged by the members of the Working Group that the new field of image-guided tumor ablation required standardization of terminology and reporting criteria to facilitate effective communication of ideas and appropriate comparison among different technologies. The main objective of the document was “improved precision and communication in this field that leads to more accurate comparison of technologies and results and ultimately to improved patient outcomes” (1). Originally published in 2003 in Radiology, the document was subsequently reviewed at regular intervals in conjunction with the Society of Interventional Radiology (SIR) Technology Assessment Committee and republished in near original form in 2005 and 2009. As a testament to its intended utility, this document has been cited by over 600 studies on tumor ablation.

Ten years later, the field of tumor ablation continues to evolve. Tumor ablation modalities that were still being developed at the time of original preparation, such as microwave and focused ultrasound, now have multiple commercially available clinical platforms in routine clinical use (2, 3). Newer ablation modalities, such as irreversible electroporation (IRE), have been introduced and clinical niches are being defined (4). Preliminary clinical studies have matured into larger longer-term series with 5- and 10-year follow-up data on par with the surgical and medical oncology literature (57). Several randomized, controlled studies have been published or are under way (8, 9). Over the interim, our initial document has also given rise to several additional position statements within the field of interventional oncology and been the source for more focused societal statements on tumor ablation of liver, kidney, and musculoskeletal tumors.

Given the number of changes that have taken place in the field of tumor ablation in the past 10 years, the members of the original Working Group and additional interventional oncology experts have taken advantage of the opportunity to meet at the Interventional Oncology Sans Frontiers meeting in Lake Como, Italy, in May 2013 and to incorporate recent advances in this updated document. It is our intention to ensure that this highly utilized standardization continues to remain relevant as it unites all investigators and clinicians practicing interventional oncology by providing a common language to describe therapies and outcomes, develop studies, and communicate with other medical specialties. As was done previously, this document has again been vetted and approved by the Technology Assessment Committee of SIR. In an attempt to attain greater worldwide adoption, this version has also received official approval of the Cardiovascular and Interventional Radiological Society of Europe, CIRSE, and additionally includes more prominent authors from Asia than the initial document.

SCOPE

The main objective of this document is to improve precision in communication in the field of image-guided tumor ablation, leading to more accurate comparison of technologies, results, and ultimately to improve patient outcomes. Here, we outline a standardized set of terminology to be used and requisite clinical and technical information that should be provided when reporting on tumor ablation. Since our original document, clinical uses and imaging evaluation of tumor ablation have expanded significantly to the point that it is challenging to fully encompass all aspects of tumor ablation in one document. Accordingly, standardization of imaging techniques, imaging findings, and tumor-specific follow-up recommendations will now be reported separately in a companion document. Similarly, despite the authors’ commitment to improving all aspects of consensus in the field of interventional oncology, detailed reviews of any specific ablation modality (such as radiofrequency [RF] or microwave ablation) or clinical indication (such as liver or kidney ablation) are beyond the scope of this document.

CLASSIFICATION OF THERAPIES

Image-Guided Tumor Ablation

The term tumor ablation is defined as the direct application of chemical (ie, nonenergy) or energy-based (ie, thermal and nonthermal) therapies to eradicate or substantially destroy focal tumors (1, 1012). The term “direct” aims to distinguish these often applicator-based therapies from others that are applied orally or via an intravascular or peripheral venous route. The concept of image guidance and planning is emphasized in the title given our radiology perspective and to highlight that imaging (throughout the treatment cycle) is critical to the optimal success of ablative therapies (11, 12). Given that most ablative therapies can be performed using a host of imaging modalities (ie, ultrasonography [US], computed tomography [CT], magnetic resonance [MR] imaging, positron emission tomography [PET], and fluoroscopy), the more general term of image guidance is preferred, unless a particular imaging modality is mandated as part of the technique. However, virtually all available ablation techniques can theoretically be used with more than one image-guidance modality.

While some have previously referred to these procedures as “minimally invasive” or “percutaneous” therapies, these terms should only be used where appropriate. Minimally invasive therapies refer to all therapeutic procedures that are less invasive than open, conventional surgery. All percutaneous procedures are therefore minimally invasive; however, not all minimally invasive therapies are performed or applied percutaneously. Indeed, the term “minimally invasive” is often used by surgeons to refer to procedures performed with mini-laparotomy or with laparoscopy. Although less invasive than open surgery, these are clearly more invasive than percutaneous image-guided tumor ablation procedures. Including the term “percutaneous” as a prefix to “image-guided tumor ablations” is often too limiting, as it does not reflect the fact that tumor ablation procedures can also be performed laparoscopically, endoscopically, or surgically (13, 14).

Individual procedures and therapies have often received multiple different names by various investigators, which can potentially lead to confusion. Hence, we propose and recommend a unified approach to the terminology regarding these therapies. The primary aim of this classification is to provide simplicity and clarity, most notably by eliminating extraneous detail and many acronyms. We acknowledge that some acronyms (such as RF and RFA for radiofrequency ablation and HIFU for high-intensity focused ultrasound) have gained widespread international acceptance. Nevertheless, the creation of additional niche acronyms for individual techniques should be avoided.

When discrimination between the ablation of malignant versus nonmalignant tissue is needed, the descriptive term “ablation” should still be used, with the type of ablated tissue stated afterwards (eg, acetic acid ablation of hepatocellular carcinoma, or radiofrequency ablation of angiomyolipoma, etc) In other words, the term “thermal (or laser, microwave, etc) ablation” should be used regardless of what is being ablated.

Our original document divided the different methods of tumor ablation in use at the time into two large classifications (chemical and thermal) to establish a basis for comparing modalities that differed in their specific mechanism of action but were broadly similar in application methodology or mechanism of tissue injury (1). For example, both ethanol and acetic acid instillation were considered “chemical ablation” and radiofrequency and microwave-based ablation were considered “thermal ablation.” However, we now recognize that, while the utility of our classification system remains, tumor ablation has expanded to include modalities that are not completely suited to the original classification. A key case in point is the interval development of IRE as an ablative modality, as IRE is energy based with a mechanism of cellular injury that is largely nonthermal, but clearly not chemical (15). Accordingly, it is now most appropriate to divide ablative modalities into: (a) chemical ablation (ie, nonenergy ablation) or (b) energy-based ablation (ie, thermal and nonthermal). We recognize that there will be some potential crossover, as several modalities may have more than one type of mechanism of tissue injury (16). Thus, when necessary, ablation modalities should be assigned a category/classification based on the dominant mode of injury. For example, several studies have used direct injection of two or more chemicals to achieve a localized high-temperature thermal reaction to induce tissue injury—this would be considered a “thermal ablation” based on the mechanism of tissue injury (17).

Other interventional oncologic therapeutic approaches including the percutaneous delivery of genetic material, drug delivery, radiation sensitization, low-temperature hyperthermia protocols, radioactive seeds or beads, radiation segmentectomy, and the transcatheter delivery of chemoembolization may ultimately require better definition but are beyond the scope of this current position article. Nevertheless, many of the issues discussed concerning reporting criteria may likely be equally appropriate for clinical trials with those therapies as well.

Chemical Ablation

These therapies are to be classified based on the universally accepted chemical nomenclature of the agent(s) such as ethanol, acetic acid, et cetera, that induce coagulation necrosis and cause tumor ablation (18, 19). For example, the term ethanol ablation should replace “PEI” (percutaneous ethanol instillation or injection), “PAI” (percutaneous alcohol instillation), and others (18, 19). The Materials and Methods section of the manuscript should specify the route (intravenous, intraarterial, or interstitial), method of substance preparation when not commercially available or when combining agents, substances and amounts injected, delivery vehicle (size and type of needle or catheter), and rate of delivery (rapid injection or a defined rate of infusion). The intended effect should be reported, if different from complete tissue destruction (such as using ablation to enhance drug delivery, radiation sensitization, in combination with other ablation modalities). The term “instillation” for the direct delivery of pharmacologic agents is preferred given that many pharmaceuticals can be injected (a process that implies rapid percutaneous delivery) or delivered intravascularly with a catheter. This category also includes newer chemical-based therapies that have variable mechanisms of actions (such as inducing thermal injury through the concomitant injection of acid and base solutions) (17).

Energy-Based Ablation

This category includes modalities that destroy a tumor either through thermal (heat or cold) or nonthermal mechanisms. For thermal therapies, energy is “applied.” The term “irradiation of energy,” particularly in regard to microwave ablation, is a misnomer and should therefore be avoided. The following energy-based modalities have been described.

RF ablation.

This term (6, 7, 20) applies to coagulation induction from all electromagnetic energy sources within the RF spectrum (3 KHz to 300 GHz), including available “radiofrequency” and “microwave” devices (10, 21). However, currently available devices traditionally designated for “radiofrequency ablation” function in the 375–500-KHz range. The term radiofrequency should be written as a single nonhyphenated word. Most devices currently used are monopolar in that there is a single “active” or “interstitial” electrode, with current dissipated at one or more return grounding pads. Bipolar devices have two “active” electrode applicators, usually placed in close proximity to achieve contiguous coagulation between the two electrodes (either needlelike or multitined), or on a single electrode (22). Since less common, in clinical practice today, bipolar RF ablation should be specified as such.

Microwave Ablation.

By convention, the term “microwave ablation” (3, 23) has been used for electromagnetic methods for inducing tumor destruction using devices with frequencies from 300 MHz to 300 GHz (21). Therefore, technically, microwave ablation devices also function within the RF spectrum and are therefore a subset of RF ablation. However, due to a different mechanism of heating and practical device and applicator differences compared with RF ablation (and described in more detail below), this category should be reported separately. Currently available microwave ablation devices function at the 915-MHz or 2.45-GHz frequencies designated for industrial, scientific, and medical (ISM) use. The term “microwave ablation” should replace the less succinct terminology of “percutaneous microwave coagulation therapy” or “microwave coagulation therapy.”

Ultrasound Ablation.

There are currently two methods (2, 24) for the application of ultrasound energy—extracorporeal (or transcutaneous) (25) and direct (or interstitial) for percutaneous application with a needlelike applicator and for intracavitary (and intracardiac) devices (26). Hence, additional nomenclature is required to distinguish between these two groups. For transcutaneous ultrasound ablation (which does not require placement of an applicator within the target tissue), high intensity focused ultrasound is the preferred term, as this denotes that more than one ultrasound beam is “focused” to create an ablation. Additionally, extracorporeal focused ablation can also be used. Both of these terms are separate from the direct application of ultrasound energy through an applicator placed within the target tissue, which should be referred to as interstitial ultrasound ablation. We feel that this revised nomenclature provides a more concise and clear description of different methodologies being studied and more closely aligns several classifications being used in the literature (25, 26).

Laser Ablation.

The term laser ablation (27, 28) should replace terminology such as “laser interstitial tumor therapy” (or LITT), “laser coagulation therapy,” and “laser interstitial photocoagulation.” This term should be used for all types of ablation using light energy. Given multiple laser technologies and application methods, including superficial therapy (contact/noncontact mode) or transcutaneous ablation, the term “interstitial” or “direct” can be reported to clarify that laser energy is applied with fibers directly inserted into the tissue.

Cryoablation.

This term (2730) should be exclusively used for all methods of destroying tissue by the application of freezing temperatures, or alternating freezing and thawing or slight heating (31). The phrase “cryo” as a freestanding term is to be avoided, as “cryo” is a prefix and not a word. The more antiquated terms “cryotherapy” or “cryosurgery” are also to be avoided as imprecise given the introduction of newer applicators that can be introduced percutaneously, endocavitarily, or endovascularly in a minimally invasive fashion.

Rapid tissue freezing and thawing produce the greatest cytotoxic effects by disrupting cellular membranes and inducing cell death (31). In the past, liquid nitrogen was placed directly on tissue, but with a few exceptions, this method is no longer used. In the neck, chest, abdomen/pelvis, and extremities, cryoablation is generally performed using one or more closed cryoprobe(s) that are placed in close proximity to or inside of the target tumor. The most common clinically available cryoablation systems utilize the Joule-Thomson effect, which relies on the expansion of a cryogen (argon gas or liquid nitrogen) at the cryoprobe tip to cause internal temperature fluctuation. Other cooling mechanisms have also been described, but all rely on a heat sink inside of the cryoprobe and thermal conduction through the probe wall from the tissue. For publication purposes, the type of cryoablation system, the gases used, probe dimensions including tip length and total length and number of freeze-thaw cycles (active or passive thawing) should also be specified.

Irreversible Electroporation.

This term (IRE or IRE ablation) (4, 16) should be used for those technologies and devices that cause cell death through the repeated application of short-duration high-voltage electrical pulses that create “irreversible” injuries to cellular membranes (15). While there may be some hyperthermic ablative changes with higher-power applications, the mechanism of cell death with IRE is thought to be predominantly nonthermal (16). When describing IRE applications, relevant energy parameters that have been shown to affect outcome (including the number and length of pulses, their spacing in time, current applied, and voltage) must be adequately described (32).

ABLATION PARAMETERS

In the original version of this document, ablation parameters, such as the number of applicators and the algorithms for energy application had largely been described and developed for RF-based devices, and were described as such in a single general category. Now, there are a wide range of applicator types, device modifications, and application techniques for several modalities, and these parameters should be clearly delineated in any reporting, so as to ensure the reproducibility of any ablation technique. We now discuss reporting terminology for applicators, application parameters, and tissue characteristics separately, and highlight modality-specific topics as needed.

Applicators

General Applicator Descriptions.

Although the devices are often referred to as “needles” or other nonspecific terms, they do not always conform to these precise classifications. Hence, the term applicator should be used when generally describing energy-based devices. Similarly, while generic needles are often used to inject agents for chemical ablation, if a device designed specifically for injection of chemical ablation agents is used, this should also be referred to as an “applicator” (33). For precision, RF and IRE applicators are electrodes, microwave applicators are antennas (rather than “antennae”), and laser applicators are fibers. By convention and consensus, cryoprobes are used to freeze tissue during cryoablation. For reporting completeness, a reference describing the appropriate applicator(s) should be cited if available; otherwise, an appropriate figure and/or schematic should be provided. A description of the applicator should also include length, a description of the active component (eg, for a needlelike RF applicator, this might include a “2-cm active tip”), and gauge size (eg, 17 gauge) (34). Gauge is preferred, as this is the common nomenclature for needle equipment used in percutaneous procedures.

Modality-Specific Applicator Descriptions.

A description of pertinent applicator characteristics relevant to a specific ablation modality is required. For RF ablation, the geometry of the electrode (eg, active tip length) should be provided (34). For microwave ablation, the energy frequency and a basic antenna design description (eg, dipole, slot, etc) is necessary to understand energy deposition around the antenna (21). For laser ablation, in addition to the laser source (Nd:YAG, erbium, holmium, etc) and precise wavelength, additional device characteristics must be specified, including the following: (a) type of laser fiber (flexible/glass dome); (b) modifications to the tip (ie, flexible diffusor tip, or scattering dome) with dimensions and materials specified; and (c) length of applicator and diameter of the optic fiber (35). For IRE, active tip length, number of electrodes in the array, and interelectrode spacing should be specified (32). For cryoablation, probe caliber, gases used, applicator length, and number of probes used should be specified.

Multitined Expandable Applicators, Cluster Electrodes, and Multielement Antennas.

This standard terminology refers to a family of applicators that are currently available from several manufacturers for RF platforms (36) but have also been developed or are in development for chemical ablation (33) and microwave platforms (37), respectively. For RF ablation, the usual embodiment of this type of device is that of an array of multiple electrode tines that expand from a single centrally positioned larger needle cannula (36). These have been previously referred to as umbrella electrodes, multitined electrodes, Christmas tree electrodes, multiple hooked electrodes, or arrays, but this has led to confusion. Given the number of electrode types that have become available and the fact that several multitined devices are now available with variable deployment lengths, the exact electrode model and diameter of electrode array used must be specified. Also, if a stepped deployment with incremental extension of the tines was performed with a multitined device, this too needs to be explained in detail regarding the length and time of deployment. One RF ablation device uses an applicator with three parallel electrodes closely spaced together that are separately introduced into the body but have a common hub (38). This should be referred to as a “cluster electrode” [not “clustered”] and is most appropriate to describe internally cooled electrode devices in which three or more closely spaced (< 1 cm) electrodes are used simultaneously to approximate a larger-diameter electrode (38). Many refer to these electrodes as “an array,” which may not adequately reflect the true underlying mechanism for enhanced energy deposition and ablation.

Internally Cooled Applicators and Perfusion Electrodes.

Some devices use a cooling agent (such as saline, water, or gas) that flows within internal lumina and does not come in direct contact with patient tissues (3840). These should be referred to as “internally cooled applicators,” and should not be confused with perfusion electrodes. When internal cooling is used, specific parameters (cooling agent used, approximate temperature of the agent, perfusate volume, and rate of infusion) should be provided where applicable. Cooled applicators should also describe whether the perfusion was performed in a closed system (with no communication with the tissue) or an open system (with free infusion into tissue) (40). Perfusion electrodes have been described for RF ablation and have small apertures at the active tip or along the distal shaft allowing fluids (ie, normal or hypertonic saline) to be infused or injected into the tissue before, during, or after the ablation procedure should be referred to as perfusion electrodes. The term replaces descriptions such as “cool-wet,” “wet,” or “saline-enhanced” electrodes, which should be avoided.

Multipolar Ablations.

Most RF ablation devices are “monopolar,” applying energy through one active tip with the current dissipated on a return grounding pad. Several ablation technologies (such as multipolar RF ablation or IRE) use energy application between two or more applicators to create a zone of ablation between applicators (10, 22). For multipolar applications, the number of applicators, length of active tip, spacing between applicators, and application algorithms (such as the order of energy application between different applicators) should be described (22, 32).

Device and Application Parameters

Energy Application Parameters and Algorithm of Energy Deposition.

For all energy-based ablation systems, energy application parameters should be provided, including power (in appropriate terminology for the specific energy source) and duration of application. As the methods used for applying energy have undergone continuous modification and improvement, this has led to substantial confusion and difficulty comparing the results of studies performed by different groups of investigators. When reporting results, pulsing techniques and other methods for amplifying energy deposition should be succinctly elaborated on in the Materials and Methods (41). Whenever possible, a reference for the precise algorithm used (eg, ramped energy deposition or impedance regulated) and the model number of the generator should be cited. Additionally, other parameters including the use of monopolar or bipolar systems, the amount of energy applied (current and/or watts), and the total or incremental duration of ablation should be provided.

For microwave ablation, sufficient parameters must be given to at least estimate the total energy delivered. The cables that transfer power from the generator to the antenna, and within the antenna apparatus itself, are lossy and can absorb a substantial fraction of the generated power (over 50% in some systems) (21). Therefore, an estimate of the actual power delivered to the tissue should be included when describing microwave ablation results.

Multiple Applicator Insertions of a Single Applicator.

When multiple overlapping ablations are performed to achieve a single large ablation zone or an ablation zone of specific configuration, the number of ablations, mean ablation times, and the end point used (ie, imaging end point, or predetermined number of ablations) should be reported (42). If a complex composite ablation is performed, details regarding spacing and degree of overlap should be described in a manner that allows reproducibility (with many advocating for a schematic as well) (43, 44).

Multiple Separate Applicators Inserted Simultaneously.

If several applicators are inserted simultaneously for simultaneous application of power (45), multipolar ablations (described above), or simultaneous ablations using “switching” technology (in which energy is applied to a single applicator at any given time, but energy application rapidly alternates between two or more applicators) (46), then specific application algorithms, ablation times, and applicator spacing should also be reported. Similarly, for multiple microwave antenna arrays, the approximate phase between electromagnetic waves applied to each antenna (if known and controlled, or acknowledged if not controlled), total power and time applied to each antenna, and pulsing parameters (if used), should also be described (47). Similar descriptions should be provided for equivalent platforms for cryoablation, and for newer ablative modalities such as ultrasound ablation or IRE (32).

Tissue Properties

Tissue-specific properties have been shown to affect the success of ablative technologies in achieving adequate tumor destruction. Characteristics of the primary organ (ie, lung, bone, liver, etc) and the tumor type (ie, hypervascular hepatocellular carcinoma vs hypovascular liver metastasis) both influence the extent of tissue injury (48). Additionally, variability in tissue characteristics in the same organ may occur based on ablation location (ie, RF ablation may be limited near the main portal vein compared with a small peripheral branch). Finally, specific ablation modalities will be affected more by one tissue characteristic than another (48, 49). In general and for publication, tissue type and the effect of tissue properties on ablation (eg, proximity to adjacent blood vessels when this might impact study end point) should be acknowledged and discussed whenever relevant. Terminology specific to certain tissue characteristics has been previously described and is addressed herein.

Blood Flow and Airflow.

Blood flow can negatively counteract the intended modulation of tissue temperatures during thermal ablation by cooling heated tissues or warming cooled tissues (50). Similar effects have been observed from airflow in ventilated lung during pulmonary ablation (51). The term heat sink effect refers to the buffering effect of patent blood vessels or ventilated bronchi adjacent to the ablation zone (50, 51). The shape of the thermal zone of ablation is altered away from the vessel, and the overall ablation size is diminished (50). Although this phenomenon serves to protect blood vessels and prevent bleeding from large vessels, it is also a major source of incomplete tumor ablation in many studies involving thermal ablation (52). Perfusion mediated tissue cooling (or heating) is a more encompassing term that refers to both the effects of the larger heat sinking vessels, as well as the substantial effects of capillary level microperfusion (53). Several strategies have been developed to overcome this problem, ranging from pharmacologically decreasing blood flow, to temporary vascular balloon occlusion of a specific vessel during ablation (ie, hepatic artery, hepatic vein, and/or portal vein during intrahepatic ablation), to intraarterial embolization and chemoembolization, to performing a Pringle maneuver (ie, temporary hepatic arterial and portal venous occlusion by direct compression of the vessels) while performing RF or cryoablation at laparotomy (10). Finally, it is further acknowledged that other fluids can be used to alter or retard uniform heating and thereby protect critical structures (such as chilled perfusate in the ureter) (54). The fluid instillation method should be adequately reported whenever employed.

Other Properties.

Other tissue properties that influence tissue and tumor heating during thermal ablation include thermal conductivity, electrical conductivity (for RF ablation and IRE), tissue elasticity or fibrosis, and tissue water content and permittivity (microwave ablation). These should be acknowledged and discussed on an ablative modality and an organ/tumor-specific basis. Authors should also set out to describe the tissue homogeneity of the target tumors or stratify/quantify those with substantial cystic components, calcification, metallic structures, graft material (eg, diaphragm or thoracoabdominal mesh), suture lines, stents (eg, ureteral or biliary) or appreciable (> 1 mm) tumoral vessels.

ABLATION PROCEDURE

Procedure Terms

As was outlined in our original standards, we continue to recommend using the term procedure rather than “operation,” as the latter implies open surgery. We consider the term session to be synonymous with procedure. A procedure refers to a single intervention event that consists of one or more ablations performed on one or more tumors. We acknowledge that multiple ablations may be performed, either in the same procedure or as separate serial events, but as part of an overall treatment plan. The term course of treatment (akin to terminology currently used in radiation therapy) is now recommended to be used to describe this series of ablations. Whenever possible, this “course of treatment” should be intention based, within a well-defined time frame, and with a clearly defined end point described. The number of planned sessions and key deviations from the original course of treatment should also be explained. We acknowledge that a course of treatment may include planned treatments other than tumor ablation (eg, performing embolization prior to ablation). Thus, specific details regarding additional nonablative treatments should also be provided.

Indications

Clinical indications for tumor ablation are divided into ablations performed for curative intent (ie, achieving the goal of complete eradication of all known tumor cells within the index tumor[s], and without any other known tumor foci in the body) or palliative intent (ie, complete ablation of the index tumor[s] [6,7,19] with other known nontarget tumor foci within the body or complete or partial ablation to treat sufficient portions of the index tumor to achieve symptom relief) (29). As one cannot “palliate” asymptomatic tumors, the term debulking should be used when describing a procedure performed with the sole intent of reducing tumor burden or controlling disease progression.

Additionally, the specified well-defined rationale for palliative therapy and an appropriate method for assessing outcomes must be provided (ie, the intended partial ablation of given tumor). For example, when tumor ablation is used as a vehicle for pain reduction (such as pain from osseous metastases), pre- and postprocedure pain scales and medication use (using commonly used scales such as morphine equivalent dose) should be obtained (55, 56). If ablation is employed to reduce symptoms of a syndrome (such as carcinoid or other hormonally active or paraneoplastic tumors), appropriate documentation of laboratory results from blood or urine before and after therapy must be provided, and other symptomatic end points and grading systems must be specified and employed. Standardized questionnaires should also be used for quality of life assessment when appropriate (55).

Complete ablation of symptomatic benign tumors (such as osteoid osteomas, tender breast fibroadenomas, or hormonally active benign adrenal aldosteronomas) to complete symptomatic relief can also be considered curative (5759).

Adjuvant Therapies

In the original standards document, “adjuvant therapies” referred to those therapies administered concomitantly with or during ablation to potentiate local effects of ablation. For example, the percutaneous instillation of sodium chloride solutions was used to alter electrical and thermal conductivity during RF ablation. Increasingly though, tumor ablation is now being combined with a multitude of agents, ranging from those given to potentiate the local antitumor effects of ablation, to the concurrent or staged administration of systemic chemotherapy while simultaneously performing local ablation (7, 60). As such, the original general description of “adjuvant therapies” is felt to be sufficiently nonspecific and archaic. The more precise following descriptions should replace this term.

Concomitant Agents.

This includes those agents that are being used to potentiate the local effects of tumor ablation (without having a specific independent antitumoral effect). For example, sodium chloride fluid or iron oxide particles injected into the target tumor prior to RF ablation have been described. Hence, specific details of the agent used (ie, agent/substance/liquid concentration, route and rate of administration, timing in relation to the ablation) must be provided. Whenever possible a reference for the precise algorithm and the rationale for the selected concomitant agent should be provided. An additional term, “sensitizers,” is used to describe certain treatment-enhancing agents in radiation therapy, and may be appropriate here as well (61).

Combination Therapies.

This includes cytotoxic or chemotherapeutic agents that, while having known independent antitumor effects, are administered in conjunction with (and temporally close to) ablation with the specific intent of inducing a synergistic effect (eg, RF ablation combined with transarterial chemoembolization [or TACE], antiangiogenic agents such as sorafenib, liposomal doxorubicin, or ethanol) (8, 60). Specific details of the agents used should be provided, along with a rationale for their use (whenever possible).

Concurrent Therapies.

This includes agents that have known antitumor effects that are administered at the time (or around) of ablation, but either have not been shown to interact with ablation, or are without clear mechanisms of synergy, or are administered without intent to potentiate effects of one or the other therapy (eg, systemic chemotherapy or radiation therapy, or cementoplasty after bone ablation) (7, 62). Specific administration timing related to ablation (and any predetermined periods of cessation around ablation) should still be described, as these therapies may ultimately be proven to effect end-point outcomes.

We also acknowledge that with greater understanding of potential systemic effects of local ablation, agents may be combined with tumor ablation to modulate secondary systemic effects (without intended effect on local tumor ablation efficacy). Examples of this include modulating antitumor immunity after tumor ablation using vaccines or immunomodulatory agents (63). Yet, this area of research is too premature to provide a well-defined classification system. Regardless, in all circumstances, specific details of administration, rationale, and use should be provided. For example, in clinical studies in patients treated with ablation, details regarding prior or concurrent systemic chemotherapy treatment (first- and second-line regimens) should be provided.

Image Guidance

While all procedures referred to in this communication refer to tumor ablations guided by imaging, it is important to understand what is meant by the term “image guidance.” First, guidance refers to procedures in which imaging techniques (eg, fluoroscopy, US, CT, PET, and MR imaging) are used during the procedure. Imaging is used in five separate and distinct ways: planning, targeting, monitoring, intraprocedural modification, and assessing treatment response (64). Different imaging techniques can be used, alone or in combination, to successfully perform each of the procedural steps described. While CT and MR imaging use have been traditionally described, contrast material–enhanced US is also now well established and commonly used in performing image guidance for all parts of an ablation procedure, and in many different organs (65). Treatments are planned before the procedure, and the assessment of treatment response occurs after the procedure is completed. Targeting, monitoring, and intraprocedural modification are all performed during the procedure. The meaning of these terms is described further as follows.

Planning.

Imaging techniques, including US, CT, MR imaging, and more recently PET/CT, are used to help determine whether patients are suitable candidates for these procedures. Imaging aspects that are particularly important include tumor size and shape, number, and location within the organ relative to blood vessels, as well as critical structures that might be at risk for injury during an ablative procedure (66). Additionally, disease-specific cancer staging (which may include additional imaging of nontarget areas) should also be provided. Adopting similar terms to radiation therapy is acceptable, such as “planned treatment volume” (or PTV).

Targeting.

This term is used to describe the step during an ablation procedure that involves placement of an applicator (eg, an RF electrode or cryoprobe) into the tumor. While much of the current image-guided tumor ablation literature describes the use of techniques such as contrast-enhanced US and CT to target tumors for purposes of ablating them, targeting is only one aspect of intraprocedural image guidance. Ideal qualities of a targeting technique include clear delineation of the tumor(s) and the surrounding anatomy, coupled with real-time imaging, and multiplanar and interactive capabilities. For example, US (66) and some MR imaging systems (67) have all of these qualities.

Image-fusion and navigation systems that combine multiple modalities (such as US and MR imaging with CT) have also been developed and are used with ever increasing frequency for tumor targeting (68, 69). These devices should be appropriately described, including the type of source/reference images and real-time images incorporated into the fusion and projections displayed. Methods of registration should be described (ie, rigid vs elastic, fiducial-based vs landmark selection, software source, and level of automation clarified where appropriate). Errors should be described in terms of overall accuracy (system error), registration error (root mean square error where applicable), and target to registration error (or TRE) (68, 69).

Monitoring.

Monitoring is the term that is used to describe the process by which therapy effects are viewed during a procedure. Changes in imaging that occur during a procedure can and should be used to determine treatment effects. For example, the zone of cryoablation can be effectively monitored with US, CT, and MR imaging by virtue of appreciable changes in tissue reflectivity, density, and phase as tissues solidify with freezing, respectively. Important aspects of monitoring include how well the tumor/target is being covered (ie, included and/or encompassed) by the ablation zone, and whether any adjacent normal structures are being affected at the same time. Not all image-guidance techniques provide the same degree and types of monitoring. For example, MR imaging is currently the only modality with well-validated techniques for near real-time temperature monitoring. For thermal monitoring, temperature measurements within the applicator and/or the ablation zone, when reported, should include specification as to where the temperature was measured (ie, where the temperature sensor is located in the applicator, or if a separate thermocouple was used), and when during the ablation temperature measurements were acquired. For noninvasive thermal monitoring (ie, with MR imaging), additional descriptions of how this was performed (eg, number of sections and imaging plane), and specific imaging sequences used, should be provided. If other forms of monitoring are used, such as measuring evoked potentials during ablation near nerves or of intramuscular tumors, then detailed descriptions should be provided. The term “monitoring” should not be used to describe response to treatment; for this, “treatment assessment” or “follow-up” is used.

Intraprocedural Modification.

This term was previously referred to as “controlling” and is used to describe the intraprocedural tools and techniques that are used to perform “real-time” modification of the ablation treatment. In order to control an image-guided ablation procedure, the treatment should be monitorable, such that the operator can utilize the image-based information obtained during monitoring to modify the ablation treatment as needed to control it. This may simply be repositioning of a therapy applicator based on physician experience, imaging findings, and thermal feedback, or it could be as sophisticated as an automated system that automatically terminates the ablation at a critical point in the procedure. This also includes intraprocedural imaging with cone-beam CT or CT, PET, MR imaging, or US, when used for assessment of effect or repositioning.

Assessment of Immediate Treatment Response.

Imaging used to immediately assess an image-guided tumor ablation procedure occurs after the procedure is completed (1012). Immediate assessment after ablation procedure should demonstrate that the target end point has been reached. When ablation is performed with curative intent, assessment should demonstrate that the ablation zone encompasses the target tumor including a circumferential ablative margin (at least 5 mm, and ideally 10 mm all around the tumor) (70). Use of a contrast agent during procedures should be well described, including agent volume and timing of imaging.

Ancillary Procedures.

As one of the main considerations in thermal ablative strategies has been nontarget injury to nearby structures, several techniques have been described to separate critical nontarget structures from the target ablation zone (54). One key technique involves injection of fluid using a separately introduced hollow-bore needle to create separation, and was initially termed “hydrodissection.” This concept has now expanded to include the injection of air, creation of artificial ascites or pneumothorax, and mechanical displacement using balloon catheters. Additionally, mixing injected fluid with an iodinated contrast agent to improve visibility has also been described. When these techniques are used, a description of the injected agent (such as saline or sterile water, with or without contrast agent), the technique used to introduce the agent (such as needle caliber and length), and the end point (such as specific distance between structures or a set volume of the agent), should also be included. Likewise, denoting the agent used with the prefix hydro- or pneumo- combined with dissection is also recommended. Displacement is the appropriate term to describe separation of the target from the nontarget structure.

The use of saline and/or externally applied warming or cooling bags for overlying skin protection are additional examples of ancillary procedures. Use of thermal balloons to control/protect surrounding tissue temperatures should also be noted, such as for ablations near ureters, the urethra and/or esophagus. Similarly, intraluminal perfusion to protect nontarget structures, such as for renal pelvicalyceal, ureteral, and bile duct protection, should also be specified, when used.

PATHOLOGIC AND IMAGING FINDINGS

The difference between pathologic findings and imaging findings must be stressed by the appropriate selection of terminology. Although in many cases there is a good correlation or overlap between radiologic and pathologic findings, this is not invariably the case, as over- and underreporting of the true extent of disease has occurred (12, 71). The classic example of this is assuming that imaging findings (ie, the zone of abnormality on the image) are equivalent to the pathologic findings (ie, the true zone of tumor destruction/treatment effect), which may not be the case. Hence, careful differentiation between imaging findings and pathologic findings must be made. This distinction is critical given that our accuracy at assessing the extent of tumor destruction by using imaging is limited by the resolution of imaging and uncertainty about the viability of cells within the radiographic margins of the zone of ablation (72).

Zone of Cell Death at Pathologic Examination

As newer technologies such as IRE induce tissue injury through nonthermal mechanisms, thus the term treatment effect should be used globally to describe the gross pathologic changes from ablation. For thermal ablation, the gross pathologic appearance of treated tissue should continue to be referred to as coagulation (which is associated with those pathologic findings associated with high-temperature thermal injury). Given that many tumors undergo central necrosis without ablation therapy, the term “coagulation” is preferred over the use of “necrosis,” as it denotes that the ablation intervention is actively leading to tumor destruction. The more generalized term “coagulation” is preferred over the term “coagulative necrosis,” as the latter term has a well-defined meaning within the pathology literature including absence of visible nuclei within the dead cells. In actuality, the zone of coagulation, while predominantly comprised of coagulative necrosis, often lacks the classic, well-defined histologic appearance of coagulative necrosis in the acute postablation period or even within some zones of adequately ablated tissue for many months following ablation (73). Additionally, for thermal ablation, short-duration high-temperature exposure results in a well-known “thermal fixation” effect, which preserves cellular architecture despite cell death, making interpretation of pathologic findings based on traditional features of “coagulation necrosis” difficult (74). When histopathologic evaluation of the ablation zone is performed, tumor cells identified in morphologic stains (hematoxylin-eosin) should undergo additional evaluation with specialized immunohistochemical stains to determine viability or irreversible cell death (72, 75). Both histopathologic and immunohistochemical evaluation of the ablation zone are recommended for articles reporting on pathologic findings or performing radiologic-pathologic correlation after tumor ablation (73). The term “coagulation” should also be used to describe pathologic findings caused by newer ablation technologies, such as microwave ablation and IRE, as well.

Another important issue is defining the zone of ablation at gross pathologic examination. Most thermal therapies induce a central “white zone” of coagulation, a pathologic finding that is generally accepted to represent coagulated tissue, surrounded by a variable “red zone” of hyperemia, which is most often absent in ex vivo specimens (76). However, there has been controversy in measuring and hence comparing the “true” size of induced zones of ablation based on the fact that some have reported that this more peripheral “red” zone also represents ablated tissue and include it in their measurements. To avoid confusion, both measurements (the zone of complete ablation alone and the extent of the inflammatory zone) should be provided. Furthermore, these descriptions apply closely to thermal ablation, but may not be as applicable to other modalities such as IRE or chemical ablation (77). Therefore, terminology such as “central ablation” and “peripheral inflammation” can also be used. This should be differentiated from the thickness of the ablation transition zone, which describes how much spatial zone resides between devascularized and dead tissue and normal/unaffected tissue. This has been called the “hyperemic rim” or “benign periablational enhancement” (at imaging), but could be described simply as the “transition zone.” At a minimum, the zones included in gross pathologic measurement should be specified. Where appropriate, for newer technologies, histopathologic results with viability staining should be correlated to gross pathologic changes.

Zone of Ablation at Postprocedural Imaging

Appropriate terminology must reflect the fact that although we rely on imaging to define the gross extent of induced coagulation, our accuracy is limited by both spatial and contrast resolution to approximately 2–3 mm depending on the imaging modality employed (73). Hence, postprocedural imaging findings are only a rough guide to the success of ablation therapy because microscopic foci of residual disease cannot be expected to be identified with standard imaging. The term “ablation zone” can be used to describe the radiologic region or zone of induced treatment effect (ie, the area of gross tumor destruction visualized by using imaging). The term “lesion” is to be avoided given potential confusion as to the intended meaning, as the term “lesion” has been used to refer to both the “ablation zone,” as well as the underlying tumor to be ablated itself. Reporting of the ablation zone should be made in relation to the target tumor. In order for the ablation to be considered successful, the target tumor should be completely covered by the ablation zone that includes at least a 5–10-mm margin all around the expected tumor margin (70).

There are two types of imaging findings that are identified following an ablation procedure, those related to zones of decreased perfusion (73) and those in which the signal intensity (at MR imaging), echogenicity (at US), attenuation (at CT), or tracer uptake (at PET) are altered (78). Hence, the imaging strategy employed and the criteria used to define ablation must be specified. Timing of early or “immediate” imaging should be described when performed. For contrast-enhanced studies, it is important to recognize that in some organ sites, and in particular the kidney, minimal contrast enhancement (ie, for CT, < 20 HU) early after ablation can be identified in areas that are subsequently proven at pathologic examination to be uniformly dead tissue (79). This finding is not well understood but may be due to pseudoenhancement, as has recently been described for renal cysts, or alternatively to represent true minimal enhancement from leaky capillaries at the treatment margin.

Finally, we acknowledge that imaging findings after tumor ablation differ based on ablation modality, imaging modality, tumor type, and organ site of ablation. Our original document included specific imaging features of thermal ablation of the liver, where terminology at the time was unclear or poorly defined. The field of image-guided tumor ablation has expanded sufficiently that standardization of descriptive terminology for postablation imaging findings that are modality and organ/tumor specific now falls beyond the scope of this document. Key terminology for imaging will be reviewed and reported in a separate consensus document.

Ablative Margin

For many disease processes and particularly for tumors in the liver, the ablation of appropriate margins beyond the borders of the tumor is necessary to achieve complete tumor destruction. The term “ablative margin” is used to describe the region that should ideally be ablated in these cases (1, 44, 70). This term is preferable to “surgical margin,” as there is no surgery. Although most investigators place this at 5–10 mm for many processes, particularly those in the liver, lung, and kidney, data are currently lacking to support definitive recommendations regarding the ideal margin size at this time (70, 80). Accordingly, the extent of desired or intended ablative margin should be specifically mentioned. It is important to stress that an extensive ablative margin, while desirable in curative ablation, is not always necessary or desired when sparing of uninvolved organ parenchyma is required. For example, when attempting to destroy focal tumors in the kidney in patients having a tendency toward the development of multiple tumors such as those with von Hippel-Lindau syndrome, nephron sparing and more limited ablation are desired to preserve renal function and avoid dialysis (81).

For normally vascular organs such as the kidney and liver, creation of an ablative margin results in zones of low attenuation and absent perfusion extending into the parenchyma (78, 82). Increased attenuation occurs in low-density tissues such as perinephric fat (for exophytic renal or adrenal tumors) and in the lungs where the term “ground glass opacity” is used to describe the imaging findings of the treatment zone surrounding and including the ablated lung tumor.

Involution of the Ablation Zone

The term “involution” should describe the process by which the body eliminates the zone of induced coagulation over weeks to months. The term “shrinkage” should be avoided as being imprecise. The term “regression” is likewise to be avoided given that it is commonly used in the medical oncology literature to describe involution of just the tumor itself, rather than the induced coagulation that often involves both tumor and the surrounding tissues (ie, the ablative margin). It is important to note that the lack of or minimal involution does not imply treatment failure. This is a finding that has been described for multiple ablation modalities (eg, RF ablation, and more recently, IRE) (78). Cicatrization may accompany involution, where nearby tissue is retracted toward the treatment zone.

Reporting of Tumor and Ablation Sizes

Appropriate uniform guidelines and standards are needed for the reporting of the extent of induced coagulation. In the past, comparison between technologies has been made somewhat difficult based on the fact that some authors report the largest diameter of induced coagulation, others report the average diameter, while some report the short-axis diameter. Additionally, coagulation has occasionally been reported as a volume of ablated tissue without any definition of dimensional measurements. Finally, zones of coagulation often demonstrate nonspherical shapes, and variations in cross-sectional axis can introduce variability in ablation size measurements. Hence, uniform standards of comparison are essential and must be adopted. It is also important to acknowledge that volumetric assessment for staging is also not yet uniform or standardized in the oncology community, but will likely be increasingly important for ablation, as noted below.

A three-dimensional, or whenever possible volumetric evaluation, should be performed to measure the ablation zone (80). While software to perform volumetric quantification of the ablation zone is being developed and not in widespread clinical use, we recognize that this technology may ultimately provide a means for detailed evaluation (83). At a minimum, characterization with multiplanar imaging (which is now widely available in clinical practice) should be performed. Additionally, it is important to acknowledge variability in postablation size measurements, which can be more or less significant depending on the ablation modality. For example, microwave ablation and, to a lesser degree, RF ablation can lead to significant tissue contraction after ablation, resulting in a smaller apparent ablation zone at postprocedure imaging (84). The visible “ice ball” during cryoablation likely overestimates the size of the ablation zone, as the cytotoxic isotherm is several millimeters inside the ice-ball margin (85). Finally, a successful ablation zone will be significantly larger than the target tumor and therefore traditional Response Evaluation Criteria in Solid Tumors, or RECIST, do not address successful ablation (86). Therefore, the first postablation imaging (eg, contrast-enhanced CT or MR imaging) is the new baseline imaging for further assessment of the ablation zone and detection of subsequent local tumor progression.

Ablation Index Tumor.

Ablation index tumor is the preferred term for the initially identified tumor prior to ablation. This tumor should not be referred to as a “lesion,” as this term could be confused with the zone of induced coagulation or the region of ablation at imaging. This should be distinguished from other “index tumors” defined by response criteria for prior courses of systemic chemotherapy or radiation therapy.

Size Classification of Tumors.

Actual tumor sizes (mean ± standard deviation, and range if applicable) should be reported. Given that the appropriate ablation of adequate margins often represents the rate-limiting step for treatment efficacy, the maximum diameter of the original tumor must be specified (based on Response Evaluation Criteria in Solid Tumors 1.1). However, many investigators perform analyses of their results based on stratification of tumor sizes. In this regard, there is often too much ambiguity and variability in the categorization of tumors by size. Different investigators have reported an upper limit of 2, 2.5, 3, and 5 cm as “small tumors” and 5 or 10 cm as large. This has made the direct comparison of results using different technologies challenging. We therefore continue to recommend that if such categorization is performed that the tumor size classification should be standardized according to the following scale: small tumors as 3 cm or smaller in diameter, 3–5-cm tumors as intermediate, and tumors larger than 5 cm as large. This classification was determined as most practical because it parallels the current technical capabilities and efficacy for most image-guided ablation therapies and has proven to be reproducible in clinical practice (7, 8).

Comparing Zones of Coagulation among Different Ablation Techniques

Often the extent of induced coagulation is reported in experimental studies as a vehicle for comparing different ablation technologies and parameter modifications (87). The extent of induced coagulation should include reporting of the short-axis diameter, given that this parameter influences the overall extent of necrosis that can be achieved from a single application of energy, and is likely to be an important factor influencing technical success in clinical practice. Hence, while additional parameters can certainly be provided and may be potentially useful, at a minimum, this should be the standard that is reported to enable honest comparison between techniques. Of course, given that the ablation of a tumor is performed in three dimensions, ideally, all three-dimensional measurements of the ablation zone and tumor, and less ideally both measurements of the cross-sectional area should be provided. If volume is to be used as the only reported parameter, then a rationale must be specified. Average diameters should only be accepted if the tumor or zone of ablation is truly spherical, varying not more than 2–3 mm in cross-sectional diameter. It is further well known that many devices produce irregularly shaped zones of coagulation. Hence, the degree of uniformity or irregularity in the shape of the ablation zone should be specified. Finally, some ablation technologies, most notably microwave ablation and to a lesser extent RF ablation, can cause relatively immediate local tissue contraction secondary to collagen and other protein remodeling, profound water evaporation, and tissue dehydration in the ablation zone (84). As a result, postablation measurements of the ablation zone at imaging or gross inspection likely underestimate the preablation tissue dimensions. Because the amount of contraction varies with ablation time, temperatures, and energy type, postablation measurements alone may not be suitable for directly comparing all technologies.

It is important to stress that reliance on minimum and maximum sizes for the zone of ablation may not be useful for predicting clinical technical efficacy, as other technical factors are likely to be equally important. For instance, depending on the orientation of the energy applicator, a 1 × 2-cm tumor may be adequately treated by using a 2 × 3-cm zone of ablation, but not by using a 3 × 2-cm zone of ablation. Ablation diameter or volume may also not tell the entire story. Although a 3-cm zone of coagulation may completely cover a 2-cm tumor when correctly positioned, if off the mark, it will fail to destroy the entire tumor.

STANDARDIZATION OF FOLLOW-UP

Currently, defining appropriate length of follow-up and the time points for defining technical success are not well established. One investigator’s long-term follow-up is often another’s short-term follow-up. Hence, specific guidelines need to be adhered to depending on the type of disease treated, and the intended goal of the study. Particularly, if existing standards for overall length of follow-up exist for a specific type of tumor, then those practice guidelines should be followed when treating those cancers with ablative therapies. Treatment study goals are generally related to one or more of the following four categories, which usually need to be distinguished from one another: (a) technical success, or was the tumor treated according to protocol?, (b) technique efficacy, or was the tumor effectively ablated? (c) morbidity, or were critical structures and complications avoided? and (d) outcomes, or was there some improvement in tumor control, patient survival, quality of life, or palliation?

Technical Success

This term simply addresses whether the tumor was treated according to protocol and was covered completely by the ablation zone. Tumor coverage can be assessed either during or immediately following the procedure, most often with contrast-enhanced CT or contrast-enhanced US. A tumor that is treated according to protocol and covered completely (ie, ablation zone completely overlaps or encompasses target tumor plus an ablative margin), as determined at the time of the procedure, is “technically successful.” The importance of this term is to help investigators separate out those patients in whom the protocol could not be executed completely, either for technical reasons or for reasons related to comorbid disease, from those who were treated according to protocol. As outlined above, a predefined course of treatment may include several ablation procedures spaced out over time. Primary technical success should be determined at the first follow-up imaging study after completion of the predetermined course of treatment.

Technique Efficacy

Distinction between “technical success” and “technique efficacy” must be made for each treated tumor. Efficacy can only be demonstrated with appropriate clinical follow-up. “Technique efficacy” should therefore refer to a prospectively defined time point (ie, immediately following the last course of a defined ablation protocol, 1 week, or 1 month after treatment) at which point “complete ablation” of macroscopic tumor, as evidenced by imaging follow-up (or another specified end point), was achieved. The number of sessions (ie, the number of interventional procedures) to achieve the specified end point should likewise be defined. Authors are encouraged to report whether or not this complete ablation included an ablative margin and how this was determined (ie, what imaging modality).

Comparison of technical success and efficacy between various ablation protocols has been challenging, as many authors have adopted different terminology or guidelines. This problem is further compounded by our ability, and often the clinical need, to ablate a tumor over many sessions and the possibility of ablating growing foci of local tumor progression months after the initial course of therapy. A window of initial therapy for each ablation technique during which it is reasonably expected for the tumor to be completely ablated should be defined. For percutaneous thermal ablation, ideally this should not exceed an upper limit of either one to four procedures or a specified time frame (up to 1–3 months), depending on the size, type, and location of the tumor, as well as the rationale for therapy. We have purposefully left definition of this end point as a broad range, given evolving consensus on defining more specific parameters, as each disease process may vary. If complete ablation cannot be achieved within these specified parameters, the tumor should be classified as “unsuccessfully treated.”

Primary and Secondary Technique Efficacy Rates.

Given that multiple treatments of image-guided tumor ablation therapy are often given over the course of the disease, primary and secondary technique efficacy rates should be reported. The primary efficacy rate is defined as the percentage of target tumors successfully eradicated following the initial procedure or a defined course of treatment. The secondary or assisted efficacy rate is defined as including tumors that have undergone successful repeat ablation following identification of local tumor progression. The term retreatment should be reserved for describing ablation of locally progressive tumor, in cases where complete ablation was initially thought to have been achieved based on imaging demonstrating “adequate” ablation of the tumor.

The technical success and technique efficacy rates are very important as we define the limitations of our technologies, ideally in a manner similar to other disciplines (ie, surgical resection articles typically report a positive margin rate). Nevertheless, for some protocols, the concepts of local technical success and local tumor progression (ie, technique efficacy) may have limited impact on the most important outcome parameter—patient survival. For example, using three to four procedures or 1 month as the window may be of secondary importance if the patient lives for 5 years because of the treatment, or if the tumor is completely eradicated over multiple courses of ablation therapy over many years.

Disease Progression.

Disease progression may be considered in three ways.

  1. Residual unablated tumor versus local tumor progression. When initial follow-up imaging demonstrates residual tumor at the ablative margin, this is referred to as residual unablated tumor. Local tumor progression describes the appearance of tumor foci at the edge of the ablation zone, after at least one contrast-enhanced follow-up study has documented adequate ablation and an absence of viable tissue in the target tumor and surrounding ablation margin by using imaging criteria. This term applies regardless of when tumor foci were discovered either early or late in the course of imaging follow-up.

    The term local tumor recurrence implies the appearance of new tumor foci at the ablative margin after local eradication of all tumor cells with ablation. However, pathologic determination of a “clear margin” cannot be made after most cases of image-guided ablation (6, 7). Accordingly, the appearance of tumor at the ablative margin at imaging likely represents residual untreated microscopic tumor, and therefore, this term should be avoided.

  2. Causes of disease progression. The distinction between local incomplete therapy (local tumor progression), new foci of disease within the target organ (especially the liver), and distant malignancy should be distinguished whenever possible and reported on. Discrimination between “local tumor progression” and new tumor is important for determining the potential utility (ie, local treatment success rate) of a given method, in the setting of many potentially confounding causes for the demise of a given patient. Additionally, for patients with cirrhosis, the causes of mortality should be differentiated between hepatic disease and others.

  3. Complete ablation versus partial ablation. When complete ablation is not achieved, classification of the degree of partial ablation should be avoided. For example, either reporting a percentage of the tumor ablated, or using descriptions of “near complete ablation” (to refer to ablation zones that encompass 90%–95% of the tumor) should be avoided. This kind of classification of partial ablation is not warranted given that adequate data are lacking to support a difference in outcome between different levels of partial ablation. Furthermore, such percentages are often estimates and may be inaccurate. Hence, for cases with curative intent, partial ablations should either be considered technical failures or simply noted as incomplete ablations as appropriate.

Complications

Classification.

The unified standardized SIR grading system should be used as outlined (88). Complications should be reported using the most recent version of the SIR Classification standard table so that they can be categorized consistently according to severity. The definition of death is self-explanatory and should be reported on a per-patient basis. Any patient death within 30 days of image-guided tumor ablation should be addressed (SIR classification F). The specific cause of death should be reported, with the potential and degree of causality to the ablation procedure clearly specified. Major and minor complications and side effects should be reported based on the number of ablation sessions on a per-session basis. However, ideally, the number of ablations performed should be included, as multiple ablations increase the likelihood of complications.

The definition of major complication is an event that leads to substantial morbidity and disability (eg, results in the unexpected loss of an organ) that increases the level of care, or results in hospital admission, or substantially lengthens the hospital stay (SIR classifications C–E). This includes any case in which a blood transfusion or interventional drainage procedure is required. All other complications are considered minor. It is important to stress that several complications such as pneumothorax or tumor seeding can be either a major or minor complication depending on severity. For tumor seeding this would depend on whether or not the ectopic tumor focus can be successfully ablated or otherwise treated.

Differentiation among immediate complications (up to 6–24 hours following the procedure), periprocedural complications (within 30 days), and delayed complications (greater than 30 days after ablation) is advised. This stratification will give the reader an idea when specific complications/side effects are most likely to occur and assist in defining when and how to take adequate precautions. Ablation-related complications should include problems encountered within the periprocedural (30–day) time period that can be related in any way to the procedure, as well as additional complications that were identified at delayed follow-up imaging that were judged to be highly likely due to the ablation therapy (biliary ductal stricture, tumor seeding along the needle track, etc). Additionally, it should be specified which complications are being reported on a patient-by-patient basis (such as death) and for which the denominator represents the number of sessions, or by the number of tumors.

Alternative classifications exist, and can be used if a compelling reason is provided. For example, the Common Terminology Criteria for Adverse Events (CTCAE) v4.0 of the National Cancer Institute and the Clavien-Dindo classification system are commonly used systems in oncologic and surgical practice (89, 90).

Side Effects.

Side effects are expected, undesired consequences of the procedure that although occurring commonly, rarely, if ever, result in substantial morbidity. These include pain, the postablation syndrome, and asymptomatic pleural effusions and minimal asymptomatic perihepatic (or renal) fluid or blood collections seen at imaging (78). Another such side effect would include asymptomatic imaging evidence of minimal thermal damage to adjacent structures without other evidence for negative sequelae (ie, “collateral damage”). An example of this would include when the zone of ablation extends beyond the liver capsule to include small portions of the diaphragm or kidney. These are not true complications, as they do not lead to an unexpected increased level of care.

Pain.

Even with appropriate conscious sedation techniques, patients may experience pain during ablation procedures. Additionally, depending on the organ site, many patients may experience grade 1–2 pain for several days, occasionally lasting 1–2 weeks following an ablation procedure. Last, thermal ablation, particularly RF and cryoablation, are being used with increased frequency as a method for treating refractory metastatic and primary bone tumor pain. We therefore propose adopting the Common Terminology Criteria for Adverse Events (CTCAE) v4.0 of the National Cancer Institute for the reporting of pain (90).

Postablation Syndrome.

This syndrome is a transient, self-limiting symptom/sign complex of low-grade fever, nausea, vomiting, and general malaise. The duration depends on the volume of necrosis produced and the overall condition of the patient. If small areas are treated, the patient is unlikely to experience postablation syndrome at all. If very large areas of liver tumors are ablated, the syndrome may persist for 2–3 weeks. The majority of patients who get this syndrome will experience some malaise for 2–7 days, depending on the volume of tumor and surrounding tissue ablated and the integrity of the patient’s immune system (ie, patients receiving steroids or with small tumors may have no postablation syndrome).

Follow-up and Outcomes

Outcomes of interest may include the following: local response (by imaging assessment), systemic response (pain, cancer syndromes, etc), quality of life, time to progression (or progression-free survival) or overall survival. For those studies that deal with quality of life, some form of objective measurement must be used both before and after treatment (91). Ideally, previously validated scales or metrics should be used and appropriately referenced.

Imaging Follow-up.

Currently, despite a reliance on imaging findings to determine the extent of “unablated residual tumor,” there is a lack of consensus on a standard follow-up interval regimen for imaging. The most common approach taken by members of the Working Group include contrast-enhanced imaging (US, CT, MR imaging, or PET) within 6 weeks of the initial ablation to determine whether or not additional ablation therapy is required (many centers perform this on the day of the initial procedure), and thereafter every 3–4 months, to determine technique efficacy. Imaging intervals may also vary depending on the type of underlying tumor and the goals of treatment. At a minimum, the intervals at which imaging follow-up were performed should be clearly specified. A more comprehensive, separate document describing approaches and standardization of imaging follow-up is forthcoming.

Although standard imaging criteria for response assessments have been defined for evaluation of other cancer therapies, these criteria focus almost exclusively on tumor size. Yet, exclusive reliance on tumor size does not provide a complete imaging assessment of tumor response, and may even lead to erroneous conclusions as to the efficacy of the therapy (86). Therefore, in addition to reporting index tumor diameter and the diameter of the zone of ablation, assessment of tumor enhancement or lack thereof should also be included in the imaging response assessment following ablation therapy. This approach is consistent with the incorporation of tumor enhancement as a measure of treatment response in newer imaging criteria (92).

Length of Follow-up.

Compared with the original document, now much of the data from clinical studies has matured, and 5- and 10-year follow-up data are becoming available (68, 92). In this context, some standardization of reporting clinical follow-up is required. Here, we define these end points as (a) technical success and early safety data should have 6-month follow-up, (b) preliminary clinical outcome results should have a minimum of 1-year follow-up, (c) intermediate-term data should have 3-year follow-up, and long-term data should have at least 5-year (and ideally longer) data, clearly specifying whether this is mean or median follow-up. Adopting this approach ensures that clinical data for ablation meet benchmarks used by other specialties. When assessing survival and disease-free survival, an appropriate length of follow-up should be selected based on tumor biology and accepted criteria for other therapies for a given tumor type. For example, surgical literature has required long-term follow-up of greater than 5 years for determining the impact of various therapies on survival for colorectal metastases to the liver or hepatocellular carcinoma (93). For other tumors, the appropriate length of follow-up may vary, and indeed for more rapidly growing tumors such as in the lung, the length of follow-up may be shorter. For slow-growing tumors, such as low-grade primary renal cell carcinoma, the length of follow-up may need to be longer (6).

Clinical Outcomes.

For all studies reporting intermediate or long-term ablation outcomes, metrics of overall survival (OS) should be reported (including percentage survival at specified time points, and mean and median survival times). OS should be calculated from the start of ablation treatment rather than treatment completion. Additionally, OS should also be reported from the date of cancer diagnosis. The time interval between treatment initiation and disease progression, time to tumor progression (TTP) (and its associated metric, progression-free survival [PFS] ) is also increasingly used as a measure of how effective tumor ablation is in achieving local tumor control, particularly in patients receiving more than one treatment where interpreting the effect of ablation on OS can be difficult. “Local TTP or PFS” (reflecting the incidence of progression in the ablated index tumor[s]) should be differentiated from “organ-specific TTP or PFS” (representing tumor progression or lack thereof in the diseased organ, such as liver), and both reported. For “tumor-related” death, determination of local TTP or PFS (eg, differentiating death due to local tumor progression or diffuse metastatic burden) will often be useful, as it can potentially shed further light on the efficacy of local therapy. As for OS, PFS should be calculated from the time of treatment initiation. Definitions of “progression” should also be provided (eg, percentage increase in tumor size), and any imaging response assessment criteria used should be specified. Where tumor ablation is performed for symptom relief, symptom-free survival may be a more appropriate descriptor. For some oncologic populations (such as early-stage renal cell cancers or small hepatocellular cancers), substantial non–cancer-related patient mortality (unrelated to or even masking ablation efficacy) may be anticipated, particularly in clinical studies with long-term follow-up. In this case, the cause of death should be specified as related or unrelated to the patient’s underlying malignancy (cancer-specific survival). Finally, risk adjustors should be reported as appropriate for the organ/disease involved (eg, performance status using Eastern Cooperative Oncology Group or Karnofsky scores).

OTHER IMPORTANT ASPECTS REQUIRING ATTENTION WHEN REPORTING CLINICAL RESULTS

Technique Parameters to Be Provided for Publication

It is our belief that many published series do not provide enough technical detail to permit duplication of the investigators’ efforts. This problem is compounded by the fact that there are many different types of ablation equipment on the market and in development, and these often change. Hence, the specification of the parameters such as duration of application energy applied, manufacturer, et cetera, must be provided. Particularly, in clinical trials where several different devices and techniques are used, a clear description of device and applicator selection, and determination of ablation end point should be provided. A clear table that details how often specific devices and techniques used should also be provided. Also, the number of treatment sessions for each tumor should be specified. The procedure approach (ie, whether the procedure was performed percutaneously, laparoscopically, or endoscopically) should also be clearly specified. Additional parameters to be provided for publication should include the following: (a) whether the procedure is performed under general anesthesia or conscious sedation (the specifics of anesthesia and medications administered during the procedure and in the recovery phase should always be reported, including agent, dose, route, etc), (b) the types of imaging guidance (CT, CT fluoroscopy, US, PET, and/or MR imaging), (c) whether or not the patient was hospitalized, (d) the number of sessions required to initially achieve technical success, and (e) the subsequent rates of other tumors requiring additional ablation therapy. Furthermore, any repositioning of the applicator during the ablation and the procedure for applicator removal (ie, use of tract ablation, fiber enclosure, or other closure devices) should be noted. Last, the frequency of use within a reported series of all ancillary procedures should be provided to establish the procedural complexity that is required to achieve a specific outcome. This will also formally differentiate more complex procedures (requiring more time, equipment, resources, and ultimately, reimbursement) from simpler procedures requiring less time, associated equipment costs, and risk.

Other Study Population Data to Be Reported

The study population should be rigorously described, including inclusion/exclusion criteria, tumor type and size, or other patient selection criteria. The degree of proof of disease required for entry into the study (ie, biopsy, imaging, or serologic criteria) should be clearly specified. Pretreatment evaluation also needs to be reported. In addition to an appropriate focus on anatomy (ie, the organ, tumor size, location, and number), the pretreatment evaluation should also include tumor stage (ie, spread elsewhere), patient comorbidities, age, gender, and overall clinical debility as outcomes such as mortality will depend on these factors. Obviously, a debilitated, cachectic patient with widespread metastases will have a worse outcome following liver ablation than an otherwise well patient.

Studies have also suggested the potential complementary effects of chemotherapy and radiation therapy on ablation efficacy. Hence, the administration of either of these therapies to patients enrolled in clinical trials of ablation should be specified. This should be further classified as having received the conventional oncologic therapies previously, around the time of ablation (within 1 month), or during the follow-up period. The specific therapy protocol, duration of therapy, and time interval in relation to ablation therapy should also be provided.

Accurate and Complete Delineation of Ablation Procedures

Substantial confusion and difficulty in comparing results has arisen regarding the success and complication rates due to the fact that patients may have had one or more tumors treated over multiple procedure sessions. Ideally, all four parameters (number of patients, tumors, treatment sessions, and ablations) should be reported whenever possible. Additionally, results are often reported for heterogeneous populations of patients for which varied rationales for the procedure (ie, cure vs palliation) or outcomes (ie, hepatic metastases vs hepatocellular carcinoma) have been reported. Stratification of patients into appropriate categories is therefore advised to avoid confusion and best facilitate extraction of clinically meaningful conclusions.

Minimizing Technical Jargon

Although substantial technical jargon and marketing terminology appear within our literature, these should not be used. For example, colloquial phrasing such as “lesioning” and “burning” are to be avoided when describing the application of thermal energy. The term “roll off” that describes the impedance control algorithm of a particular manufacturer’s RF device should not be used.

Comparison with Other Treatments

Given that many reports of image-guided therapy, particularly with newer technologies, are relatively small case series, a major benefit of uniform reporting standards is the ability to perform meta-analyses of outcomes to compare therapies. Clinical research studies should be reported in such a manner that the results can be directly compared with various cancer therapies including other forms of image-guided ablation, surgery, radiation, and chemotherapy. The gold/reference standard in oncology is survival, disease-free survival, and quality of life stratified by disease stage and patient functional status. While studies addressing these outcomes after ablation are becoming increasingly available, nevertheless, there continue to be limited data addressing these issues for many diseases treated with image-guided ablation (94). Thus, we wish to stress the need for studies on an organ-by-organ and disease-by-disease basis. Randomized, controlled and blinded studies are considered the gold standard for pivotal studies and should be performed when possible (95). By the same token, we acknowledge both the very real obstacles to performing such studies (including patient recruitment, long periods of data collection, expense, multicenter organization, etc), as well as the benefit of reporting less robust forms of data including retrospective studies, case series, and case reports (96). Finally, in our current worldwide financial climate, we strongly encourage and advocate for the performance of both cost-effectiveness analysis and comparative effectiveness studies, recognizing that these are essential to optimally positioning tumor ablation in comparison with other available treatments (97, 98).

Statistical Evaluation

Regardless of the study type, rigorous statistical evaluation appropriate for the data collected should be presented. The primary and secondary study end points should be clearly stated. Bearing in mind that the data from individual studies may need to be treated differently, in general survival outcomes should be reported using life-table (Kaplan-Meier) analysis. Patients should be randomized, if possible, and results reported based on intention to treat, treated as randomized, as treated per protocol (ie, excluding protocol violations). Outcomes may further need to be stratified according to multiple factors (tumor type, grade and stage, functional status, comorbidities, etc) Appropriate methodology for assessment of quality of life should be likewise selected (99).

More Relevant Studies

Since the original document, many additional relevant studies have been published. Appendix E1 (available online at www.jvir.org) includes more relevant studies that could not be included because of article length limitations.

CONCLUSIONS

The original intent of this standardization of terminology was to provide an appropriate vehicle for reporting the various aspects of image-guided ablation therapy. Our intent continues to be to provide such a framework in order to facilitate the clearest communication between investigators, and the greatest flexibility in comparison among the many newer, exciting, and emerging technologies. Clearly, 10 years later, this is an ongoing process that will require that we adapt to our greater understanding of improving existing technologies and emerging novel treatments. As the original version of this document has been successful in providing a framework for the evaluation of ablation therapies worldwide, we encourage all of our colleagues to adopt the terminology and reporting strategies outlined in this updated proposal to facilitate worldwide communication of scientific advances.

ABBREVIATIONS

IRE

irreversible electroporation

OS

overall survival

PFS

progression-free survival

RF

radiofrequency

SIR

Society of Interventional Radiology

TTP

time to tumor progression

APPENDIX E1

Given the expansion of tumor ablation–related studies since the original document, many additional relevant studies should be cited. Due to limitations in manuscript length for print publication, we have included a list of additional references on a section-by-section basis as Appendix E1.

ADDITIONAL CLINICAL STUDIES: LONG-TERM STUDIES AND RANDOMIZED, CONTROLLED STUDIES

  1. Ambrogi MC, Fanucchi O, Cioni R, et al. Long-term results of radiofrequency ablation treatment of stage I non-small cell lung cancer: a prospective intention-to-treat study. J Thorac Oncol 2011; 6:2044–2051. [DOI] [PubMed] [Google Scholar]
  2. Francica G, Saviano A, De Sio I, et al. Long-term effectiveness of radiofrequency ablation for solitary small hepatocellular carcinoma: a retrospective analysis of 363 patients. Dig Liver Dis 2013; 45:336–341. [DOI] [PubMed] [Google Scholar]
  3. Gillams AR, Lees WR. Five-year survival following radiofrequency ablation of small, solitary, hepatic colorectal metastases. J Vasc Interv Radiol 2008; 19:712–717. [DOI] [PubMed] [Google Scholar]
  4. Ma Y, Bedir S, Cadeddu JA, Gahan JC. Long-term outcomes in healthy adults following radiofrequency ablation of T1a renal tumours. BJU Int 2014; 113:51–55. [DOI] [PubMed] [Google Scholar]
  5. Simon CJ, Dupuy DE, DiPetrillo TA, et al. Pulmonary radiofrequency ablation: long-term safety and efficacy in 153 patients. Radiology 2007; 243:268–275. [DOI] [PubMed] [Google Scholar]
  6. Tohme S, Geller DA, Cardinal JS, et al. Radiofrequency ablation compared to resection in early-stage hepatocellular carcinoma. HPB (Oxford) 2013; 15:210–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Shiina S, Tateishi R, Arano T, et al. Radiofrequency ablation for hepatocellular carcinoma: 10-year outcome and prognostic factors. Am J Gastroenterol 2012; 107:569–577; quiz 578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Feng K, Yan J, Li X, et al. A randomized controlled trial of radiofrequency ablation and surgical resection in the treatment of small hepatocellular carcinoma. J Hepatol 2012; 57:794–802. [DOI] [PubMed] [Google Scholar]
  9. Guan W, Bai J, Liu J, et al. Microwave ablation versus partial nephrectomy for small renal tumors: intermediate-term results. J Surg Oncol 2012; 106:316–321. [DOI] [PubMed] [Google Scholar]
  10. Petre EN, Jia X, Thornton RH, et al. Treatment of pulmonary colorectal metastases by radiofrequency ablation. Clin Colorectal Cancer 2013; 12:37–44. [DOI] [PubMed] [Google Scholar]

ADDITIONAL SOCIETY POSITION STATEMENTS ON TUMOR ABLATION

  1. Brown DB, Gould JE, Gervais DA, et al. Transcatheter therapy for hepatic malignancy: standardization of terminology and reporting criteria. J Vasc Interv Radiol 2009; 20:S425–S434. [DOI] [PubMed] [Google Scholar]
  2. Callstrom MR, York JD, Gaba RC, et al. Research reporting standards for image-guided ablation of bone and soft tissue tumors. J Vasc Interv Radiol 2009; 20:1527–1540. [DOI] [PubMed] [Google Scholar]
  3. Clark TW, Millward SF, Gervais DA, et al. Reporting standards for percutaneous thermal ablation of renal cell carcinoma. J Vasc Interv Radiol 2009; 20:S409–S416. [DOI] [PubMed] [Google Scholar]
  4. Rose SC, Dupuy DE, Gervais DA, et al. Research reporting standards for percutaneous thermal ablation of lung neoplasms. J Vasc Interv Radiol 2009; 20:S474–S485. [DOI] [PubMed] [Google Scholar]

DIFFERENT METHODS OF TUMOR ABLATION

  1. Ward RC, Healey TT, Dupuy DE. Microwave ablation devices for interventional oncology. Expert Rev Med Devices 2013; 10:225–238. [DOI] [PubMed] [Google Scholar]
  2. Cheung W, Kavnoudias H, Roberts S, Szkandera B, Kemp W, Thomson KR. Irreversible electroporation for unresectable hepatocellular carcinoma: initial experience and review of safety and outcomes. Technol Cancer Res Treat 2013; 12:233–241. [DOI] [PubMed] [Google Scholar]
  3. Cressman EN, Shenoi MM, Edelman TL, et al. In vivo comparison of simultaneous versus sequential injection technique for thermochemical ablation in a porcine model. Int J Hyperthermia 2012; 28:105–112. [DOI] [PubMed] [Google Scholar]
  4. Shiina S, Tagawa K, Niwa Y, et al. Percutaneous ethanol injection therapy for hepatocellular carcinoma: results in 146 patients. AJR Am J Roentgenol 1993; 160:1023–1028. [DOI] [PubMed] [Google Scholar]
  5. Lin SM, Lin CJ, Lin CC, Hsu CW, Chen YC. Randomised controlled trial comparing percutaneous radiofrequency thermal ablation, percutaneous ethanol injection, and percutaneous acetic acid injection to treat hepatocellular carcinoma of 3 cm or less. Gut 2005; 54:1151–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Ahmed M, Weinstein J, Liu Z, et al. Image-guided percutaneous chemical and radiofrequency tumor ablation in an animal model. J Vasc Interv Radiol 2003; 14:1045–1052. [DOI] [PubMed] [Google Scholar]
  7. Aguilar-Garib JA. Medical applications: the ISM band [letter]. J Microw Power Electromagn Energy 2010; 44:131. [DOI] [PubMed] [Google Scholar]
  8. Cheung TT, Fan ST, Chan SC, et al. High-intensity focused ultrasound ablation: an effective bridging therapy for hepatocellular carcinoma patients. World J Gastroenterol 2013; 19:3083–3089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Kariniemi J, Ojala R, Hellström P, Sequeiros RB. MRI-guided percutaneous laser ablation of small renal cell carcinoma: initial clinical experience. Acta Radiol 2010; 51:467–472. [DOI] [PubMed] [Google Scholar]
  10. Silverman SG, Tuncali K, Adams DF, et al. MR imaging-guided percutaneous cryotherapy of liver tumors: initial experience. Radiology 2000; 217:657–664. [DOI] [PubMed] [Google Scholar]
  11. Cheung TT, Fan ST, Chan SC, et al. High-intensity focused ultrasound ablation: an effective bridging therapy for hepatocellular carcinoma patients. World J Gastroenterol 2013; 19:3083–3089. [DOI] [PMC free article] [PubMed] [Google Scholar]

REPORTING APPLICATOR, DEVICE, AND ENERGY APPLICATION/ALGORITHIM PARAMETERS

  1. Meijerink MR, van den Tol P, van Tilborg AA, van Waesberghe JH, Meijer S, van Kuijk C. Radiofrequency ablation of large size liver tumours using novel plan-parallel expandable bipolar electrodes: initial clinical experience. Eur J Radiol 2011; 77:167–171. [DOI] [PubMed] [Google Scholar]
  2. Aguilar-Garib JA. Medical applications: the ISM band. J Microw Power Electromagn Energy 2010; 44:131. [DOI] [PubMed] [Google Scholar]
  3. Brace CL. Dual-slot antennas for microwave tissue heating: parametric design analysis and experimental validation. Med Phys 2011; 38:4232–4240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Koda M, Tokunaga S, Matono T, Sugihara T, Nagahara T, Murawaki Y. Comparison between different thickness umbrella-shaped expandable radiofrequency electrodes (SuperSlim and CoAccess): experimental and clinical study. Exp Ther Med 2011; 2:1215–1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Miao Y, Ni Y, Yu J, Zhang H, Baert A, Marchal G. An ex vivo study on radiofrequency tissue ablation: increased lesion size by using an “expandable-wet” electrode. Eur Radiol 2001; 11:1841–1847. [DOI] [PubMed] [Google Scholar]
  6. Hirakawa M, Ikeda K, Kobayashi M, et al. Randomized controlled trial of a new procedure of radiofrequency ablation using an expandable needle for hepatocellular carcinoma. Hepatol Res 2013; 43:846–852. [DOI] [PubMed] [Google Scholar]
  7. Rempp H, Voigtländer M, Clasen S, et al. Increased ablation zones using a cryo-based internally cooled bipolar RF applicator in ex vivo bovine liver. Invest Radiol 2009; 44:763–768. [DOI] [PubMed] [Google Scholar]
  8. Burdío F, Tobajas P, Quesada-Diez R, et al. Distant infusion of saline may enlarge coagulation volume during radiofrequency ablation of liver tissue using cool-tip electrodes without impairing predictability. AJR Am J Roentgenol 2011; 196:W837–W843. [DOI] [PubMed] [Google Scholar]
  9. Abitabile P, Maurer CA. Radiofrequency ablation of liver tumors: a novel needle perfusion technique enhances efficiency. J Surg Res 2010; 159:532–537. [DOI] [PubMed] [Google Scholar]
  10. Clasen S, Schmidt D, Boss A, et al. Multipolar radiofrequency ablation with internally cooled electrodes: experimental study in ex vivo bovine liver with mathematic modeling. Radiology 2006; 238:881–890. [DOI] [PubMed] [Google Scholar]
  11. Goldberg SN, Stein MC, Gazelle GS, Sheiman RG, Kruskal JB, Clouse ME. Percutaneous radiofrequency tissue ablation: optimization of pulsed-radiofrequency technique to increase coagulation necrosis. J Vasc Interv Radiol 1999; 10:907–916. [DOI] [PubMed] [Google Scholar]
  12. Wong SN, Santi GE, Nurjadin H, Aguilar R, Gosalvez-Pe S. Temperature-dependent electrode repositioning for multiple overlapping radiofrequency ablation in ex vivo porcine livers. J Vasc Interv Radiol 2010; 21:1733–1738. [DOI] [PubMed] [Google Scholar]
  13. Woo S, Lee JM, Yoon JH, et al. Small- and medium-sized hepatocellular carcinomas: monopolar radiofrequency ablation with a multiple-electrode switching system—mid-term results. Radiology 2013; 268:589–600. [DOI] [PubMed] [Google Scholar]
  14. Laeseke PF, Lee FT Jr, van der Weide DW, Brace CL. Multiple-antenna microwave ablation: spatially distributing power improves thermal profiles and reduces invasiveness. J Interv Oncol 2009; 2:65–72. [PMC free article] [PubMed] [Google Scholar]

TISSUE PROPERTIES AFFECTING ABLATION

  1. Ahmed M, Liu Z, Humphries S, Goldberg SN. Computer modeling of the combined effects of perfusion, electrical conductivity, and thermal conductivity on tissue heating patterns in radiofrequency tumor ablation. Int J Hyperthermia 2008; 24:577–588. [DOI] [PubMed] [Google Scholar]
  2. Ahmed M, Liu Z, Afzal KS, et al. Radiofrequency ablation: effect of surrounding tissue composition on coagulation necrosis in a canine tumor model. Radiology 2004; 230:761–767. [DOI] [PubMed] [Google Scholar]
  3. Yu NC, Raman SS, Kim YJ, Lassman C, Chang X, Lu DS. Microwave liver ablation: influence of hepatic vein size on heat-sink effect in a porcine model. J Vasc Interv Radiol 2008; 19:1087–1092. [DOI] [PubMed] [Google Scholar]
  4. Chang I, Mikityansky I, Wray-Cahen D, Pritchard WF, Karanian JW, Wood BJ. Effects of perfusion on radiofrequency ablation in swine kidneys. Radiology 2004; 231:500–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Kim C, O’Rourke AP, Will JA, Mahvi DM, Webster JG. Finite-element analysis of hepatic cryoablation around a large blood vessel. IEEE Trans Biomed Eng 2008; 55:2087–2093. [DOI] [PubMed] [Google Scholar]
  6. Patterson EJ, Scudamore CH, Owen DA, Nagy AG, Buczkowski AK. Radiofrequency ablation of porcine liver in vivo: effects of blood flow and treatment time on lesion size. Ann Surg 1998; 227:559–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Solbiati L, Livraghi T, Goldberg SN, et al. Percutaneous radiofrequency ablation of hepatic metastases from colorectal cancer: long-term results in 117 patients. Radiology 2001; 221:159–166. [DOI] [PubMed] [Google Scholar]
  8. Yang W, Chen MH, Wang MQ, et al. Combination therapy of radiofrequency ablation and transarterial chemoembolization in recurrent hepatocellular carcinoma after hepatectomy compared with single treatment. Hepatol Res 2009; 39:231–240. [DOI] [PubMed] [Google Scholar]
  9. Curley SA, Izzo F, Ellis LM, Nicolas Vauthey J, Vallone P. Radiofrequency ablation of hepatocellular cancer in 110 patients with cirrhosis. Ann Surg 2000; 232:381–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Seki T, Tamai T, Nakagawa T, et al. Combination therapy with transcatheter arterial chemoembolization and percutaneous microwave coagulation therapy for hepatocellular carcinoma. Cancer 2000; 89:1245–1251. [PubMed] [Google Scholar]
  11. Yang W, Chen MH, Wang MQ, et al. Combination therapy of radiofrequency ablation and transarterial chemoembolization in recurrent hepatocellular carcinoma after hepatectomy compared with single treatment. Hepatol Res 2009; 39:231–240. [DOI] [PubMed] [Google Scholar]
  12. Liu Z, Ahmed M, Weinstein Y, Yi M, Mahajan RL, Goldberg SN. Characterization of the RF ablation-induced ‘oven effect’: the importance of background tissue thermal conductivity on tissue heating. Int J Hyperthermia 2006; 22:327–342. [DOI] [PubMed] [Google Scholar]
  13. Cantwell CP, Wah TM, Gervais DA, et al. Protecting the ureter during radiofrequency ablation of renal cell cancer: a pilot study of retrograde pyeloperfusion with cooled dextrose 5% in water. J Vasc Interv Radiol 2008; 19:1034–1040. [DOI] [PubMed] [Google Scholar]
  14. Wah TM, Koenig P, Irving HC, Gervais DA, Mueller PR. Radiofrequency ablation of a central renal tumor: protection of the collecting system with a retrograde cold dextrose pyeloperfusion technique. J Vasc Interv Radiol 2005; 16:1551–1555. [DOI] [PubMed] [Google Scholar]
  15. Goldberg SN, Ahmed M, Gazelle GS, et al. Radio-frequency thermal ablation with NaCl solution injection: effect of electrical conductivity on tissue heating and coagulation—phantom and porcine liver study. Radiology 2001; 219:157–165. [DOI] [PubMed] [Google Scholar]
  16. Ben-David E, Ahmed M, Faroja M, et al. Irreversible electroporation: treatment effect is susceptible to local environment and tissue properties. Radiology 2013; 269:738–747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. O’Rourke AP, Lazebnik M, Bertram JM, et al. Dielectric properties of human normal, malignant and cirrhotic liver tissue: in vivo and ex vivo measurements from 0.5 to 20 GHz using a precision open-ended coaxial probe. Phys Med Biol 2007; 52:4707–4719. [DOI] [PubMed] [Google Scholar]
  18. Brace CL. Temperature-dependent dielectric properties of liver tissue measured during thermal ablation: toward an improved numerical model. Conf Proc IEEE Eng Med Biol Soc 2008; 2008:230–233. [DOI] [PubMed] [Google Scholar]

CONCOMITANT, COMBINATION, AND CONCURRENT THERAPIES

  1. Berber E, Flesher N, Siperstein AE. Laparoscopic radiofrequency ablation of neuroendocrine liver metastases. World J Surg 2002; 26:985–990. [DOI] [PubMed] [Google Scholar]
  2. Merkle EM, Goldberg SN, Boll DT, et al. Effects of superparamagnetic iron oxide on radio-frequency-induced temperature distribution: in vitro measurements in polyacrylamide phantoms and in vivo results in a rabbit liver model. Radiology 1999; 212:459–466. [DOI] [PubMed] [Google Scholar]
  3. Hakimé A, Hines-Peralta A, Peddi H, et al. Combination of radiofrequency ablation with antiangiogenic therapy for tumor ablation efficacy: study in mice. Radiology 2007; 244:464–470. [DOI] [PubMed] [Google Scholar]
  4. Ahmed M, Liu Z, Lukyanov AN, et al. Combination radiofrequency ablation with intratumoral liposomal doxorubicin: effect on drug accumulation and coagulation in multiple tissues and tumor types in animals. Radiology 2005; 235:469–477. [DOI] [PubMed] [Google Scholar]
  5. Wood BJ, Poon RT, Locklin JK, et al. Phase I study of heat-deployed liposomal doxorubicin during radiofrequency ablation for hepatic malignancies. J Vasc Interv Radiol 2012; 23:248–255, e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Lane MD, Le HB, Lee S, et al. Combination radiofrequency ablation and cementoplasty for palliative treatment of painful neoplastic bone metastasis: experience with 53 treated lesions in 36 patients. Skeletal Radiol 2011; 40:25–32. [DOI] [PubMed] [Google Scholar]
  7. Gameiro SR, Higgins JP, Dreher MR, et al. Combination therapy with local radiofrequency ablation and systemic vaccine enhances antitumor immunity and mediates local and distal tumor regression. PLoS ONE 2013; 8:e70417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Kroeze SG, Daenen LG, Nijkamp MW, et al. Radio frequency ablation combined with interleukin-2 induces an antitumor immune response to renal cell carcinoma in a murine model. J Urol 2012; 188:607–614. [DOI] [PubMed] [Google Scholar]

IMAGE GUIDANCE IN ABLATION

  1. Jolesz FA, Silverman SG. Interventional magnetic resonance therapy. Semin Intervent Radiol 1995; 12:20–27. [Google Scholar]
  2. Kurup AN, Morris JM, Schmit GD, et al. Neuroanatomic considerations in percutaneous tumor ablation. RadioGraphics 2013; 33:1195–1215. [DOI] [PubMed] [Google Scholar]
  3. Ryan ER, Sofocleous CT, Schöder H, et al. Split-dose technique for FDG PET/CT-guided percutaneous ablation: a method to facilitate lesion targeting and to provide immediate assessment of treatment effectiveness. Radiology 2013; 268:288–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Gazelle GS, Haaga JR. Guided percutaneous biopsy of intraabdominal lesions. AJR Am J Roentgenol 1989; 153:929–935. [DOI] [PubMed] [Google Scholar]
  5. Lewin JS, Connell CF, Duerk JL, et al. Interactive MRI-guided radiofrequency interstitial thermal ablation of abdominal tumors: clinical trial for evaluation of safety and feasibility. J Magn Reson Imaging 1998; 8:40–47. [DOI] [PubMed] [Google Scholar]
  6. Jung EM, Friedrich C, Hoffstetter P, et al. Volume navigation with contrast enhanced ultrasound and image fusion for percutaneous interventions: first results. PLoS ONE 2012; 7:e33956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Onik G, Kane R, Steele G, et al. Society of Gastrointestinal Radiologists Roscoe E. Miller Award. Monitoring hepatic cryosurgery with sonography. AJR Am J Roentgenol 1986; 147:665–669. [DOI] [PubMed] [Google Scholar]
  8. Wu B, Xiao YY, Zhang X, Zhang AL, Li HJ, Gao DF. Magnetic resonance imaging-guided percutaneous cryoablation of hepatocellular carcinoma in special regions. Hepatobiliary Pancreat Dis Int 2010; 9:384–392. [PubMed] [Google Scholar]
  9. Rempp H, Unterberg J, Hoffmann R, et al. Therapy monitoring of magnetic resonance-guided radiofrequency ablation using T1- and T2-weighted sequences at 1.5 T: reliability of estimated ablation zones. Invest Radiol 2013; 48:429–436. [DOI] [PubMed] [Google Scholar]
  10. Holbrook AB, Santos JM, Kaye E, Rieke V, Pauly KB. Real-time MR thermometry for monitoring HIFU ablations of the liver. Magn Reson Med 2010; 63:365–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Tsoumakidou G, Buy X, Garnon J, Enescu J, Gangi A. Percutaneous thermal ablation: how to protect the surrounding organs. Tech Vasc Interv Radiol 2011; 14:170–176. [DOI] [PubMed] [Google Scholar]
  12. Rempp H, Clasen S, Pereira PL. Image-based monitoring of magnetic resonance-guided thermoablative therapies for liver tumors. Cardiovasc Intervent Radiol 2012; 35:1281–1294. [DOI] [PubMed] [Google Scholar]
  13. Zientara GP, Saiviroonporn P, Morrison PR, et al. MRI monitoring of laser ablation using optical flow. J Magn Reson Imaging 1998; 8:1306–1318. [DOI] [PubMed] [Google Scholar]
  14. Park MJ, Kim TH, Lee KM, Cheong JY, Kim JK. Radiofrequency ablation of metastatic liver masses: recurrence patterns and prognostic factors based on radiologic features. Hepatogastroenterology 2013; 60:563–567. [DOI] [PubMed] [Google Scholar]
  15. Makino Y, Imai Y, Igura T, et al. Utility of computed tomography fusion imaging for the evaluation of the ablative margin of radiofrequency ablation for hepatocellular carcinoma and the correlation to local tumor progression. Hepatol Res 2013; 43:950–958. [DOI] [PubMed] [Google Scholar]
  16. Anderson EM, Lees WR, Gillams AR. Early indicators of treatment success after percutaneous radiofrequency of pulmonary tumors. Cardiovasc Intervent Radiol 2009; 32:478–483. [DOI] [PubMed] [Google Scholar]

ANCILLARY PROCEDURES

  1. Chen MH, Yang W, Yan K, et al. Radiofrequency ablation of problematically located hepatocellular carcinoma: tailored approach. Abdom Imaging 2008; 33:428–436. [DOI] [PubMed] [Google Scholar]
  2. Gervais DA, Arellano RS, McGovern FJ, McDougal WS, Mueller PR. Radiofrequency ablation of renal cell carcinoma. II. Lessons learned with ablation of 100 tumors. AJR Am J Roentgenol 2005; 185:72–80. [DOI] [PubMed] [Google Scholar]
  3. Lee SJ, Choyke LT, Locklin JK, Wood BJ. Use of hydrodissection to prevent nerve and muscular damage during radiofrequency ablation of kidney tumors. J Vasc Interv Radiol 2006; 17:1967–1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Kam AW, Littrup PJ, Walther MM, Hvizda J, Wood BJ. Thermal protection during percutaneous thermal ablation of renal cell carcinoma. J Vasc Interv Radiol 2004; 15:753–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Uehara T, Hirooka M, Ishida K, et al. Percutaneous ultrasound-guided radiofrequency ablation of hepatocellular carcinoma with artificially induced pleural effusion and ascites. J Gastroenterol 2007; 42:306–311. [DOI] [PubMed] [Google Scholar]
  6. Song I, Rhim H, Lim HK, Kim YS, Choi D. Percutaneous radiofrequency ablation of hepatocellular carcinoma abutting the diaphragm and gastrointestinal tracts with the use of artificial ascites: safety and technical efficacy in 143 patients. Eur Radiol 2009; 19:2630–2640. [DOI] [PubMed] [Google Scholar]
  7. DeBenedectis CM, Beland MD, Dupuy DE, Mayo-Smith WW. Utility of iodinated contrast medium in hydrodissection fluid when performing renal tumor ablation. J Vasc Interv Radiol 2010; 21:745–747. [DOI] [PubMed] [Google Scholar]
  8. Campbell C, Lubner MG, Hinshaw JL, Muñoz del Rio A, Brace CL. Contrast media-doped hydrodissection during thermal ablation: optimizing contrast media concentration for improved visibility on CT images. AJR Am J Roentgenol 2012; 199:677–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Sudheendra D, Dromi S, Wood BJ. Subdermal fluid for skin protection during superficial palliative thermal ablation. J Vasc Interv Radiol 2006; 17:1545–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Raman SS, Aziz D, Chang X, et al. Minimizing central bile duct injury during radiofrequency ablation: use of intraductal chilled saline perfusion—initial observations from a study in pigs. Radiology 2004; 232:154–159. [DOI] [PubMed] [Google Scholar]

PATHOLOGY AND IMAGING

  1. Sofocleous CT, Garg S, Petrovic LM, et al. Ki-67 is a prognostic biomarker of survival after radiofrequency ablation of liver malignancies. Ann Surg Oncol 2012; 19:4262–4269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Snoeren N, Huiskens J, Rijken AM, et al. Viable tumor tissue adherent to needle applicators after local ablation: a risk factor for local tumor progression. Ann Surg Oncol 2011; 18:3702–3710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Fujisawa S, Romin Y, Barlas A, et al. Evaluation of YO-PRO-1 as an early marker of apoptosis following radiofrequency ablation of colon cancer liver metastases. Cytotechnology 2014; 66:259–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Zervas NT, Kuwayama A. Pathological characteristics of experimental thermal lesions. Comparison of induction heating and radiofrequency electrocoagulation. J Neurosurg 1972; 37:418–422. [DOI] [PubMed] [Google Scholar]
  5. Wu F, Wang ZB, Cao YD, et al. Heat fixation of cancer cells ablated with high-intensity-focused ultrasound in patients with breast cancer. Am J Surg 2006; 192:179–184. [DOI] [PubMed] [Google Scholar]
  6. Yamashiki N, Kato T, Bejarano PA, et al. Histopathological changes after microwave coagulation therapy for patients with hepatocellular carcinoma: review of 15 explanted livers. Am J Gastroenterol 2003; 98:2052–2059. [DOI] [PubMed] [Google Scholar]
  7. Sofocleous CT, Garg SK, Cohen P, et al. Ki 67 is an independent predictive biomarker of cancer specific and local recurrence-free survival after lung tumor ablation. Ann Surg Oncol 2013; 20:S676–S683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Schneider T, Reuss D, Warth A, et al. The efficacy of bipolar and multipolar radiofrequency ablation of lung neoplasms: results of an ablate and resect study. Eur J Cardiothorac Surg 2011; 39:968–973. [DOI] [PubMed] [Google Scholar]
  9. 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; 113:3121–3129. [DOI] [PubMed] [Google Scholar]
  10. Appelbaum L, Ben-David E, Sosna J, Nissenbaum Y, Goldberg SN. US findings after irreversible electroporation ablation: radiologic-pathologic correlation. Radiology 2012; 262:117–125. [DOI] [PubMed] [Google Scholar]
  11. Dupuy DE, Aswad B, Ng T. Irreversible electroporation in a swine lung model. Cardiovasc Intervent Radiol 2011; 34:391–395. [DOI] [PubMed] [Google Scholar]
  12. Schraml C, Schwenzer NF, Clasen S, et al. Navigator respiratory-triggered diffusion-weighted imaging in the follow-up after hepatic radiofrequency ablation: initial results. J Magn Reson Imaging 2009; 29:1308–1316. [DOI] [PubMed] [Google Scholar]
  13. Georgiades C, Rodriguez R, Azene E, et al. Determination of the nonlethal margin inside the visible “ice-ball” during percutaneous cryoablation of renal tissue. Cardiovasc Intervent Radiol 2013; 36:783–790. [DOI] [PubMed] [Google Scholar]
  14. Abtin FG, Eradat J, Gutierrez AJ, Lee C, Fishbein MC, Suh RD. Radiofrequency ablation of lung tumors: imaging features of the postablation zone. RadioGraphics 2012; 32:947–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Singnurkar A, Solomon SB, Gönen M, Larson SM, Schöder H. 18F-FDG PET/CT for the prediction and detection of local recurrence after radiofrequency ablation of malignant lung lesions. J Nucl Med 2010; 51:1833–1840. [DOI] [PubMed] [Google Scholar]
  16. Pavlovich CP, Walther M, Choyke PL, et al. Percutaneous radio frequency ablation of small renal tumors: initial results. J Urol 2002; 167:10–15. [PMC free article] [PubMed] [Google Scholar]
  17. Liu CH, Arellano RS, Uppot RN, Samir AE, Gervais DA, Mueller PR. Radiofrequency ablation of hepatic tumours: effect of postablation margin on local tumour progression. Eur Radiol 2010; 20:877–885. [DOI] [PubMed] [Google Scholar]
  18. Brook OR, Mendiratta-Lala M, Brennan D, Siewert B, Faintuch S, Goldberg SN. Imaging findings after radiofrequency ablation of adrenal tumors. AJR Am J Roentgenol 2011; 196:382–388. [DOI] [PubMed] [Google Scholar]
  19. Ganguli S, Brennan DD, Faintuch S, Rayan ME, Goldberg SN. Immediate renal tumor involution after radiofrequency thermal ablation. J Vasc Interv Radiol 2008; 19:412–418. [DOI] [PubMed] [Google Scholar]
  20. Arima K, Yamakado K, Suzuki R, et al. Image-guided radiofrequency ablation for adrenocortical adenoma with Cushing syndrome: outcomes after mean follow-up of 33 months. Urology 2007; 70:407–411. [DOI] [PubMed] [Google Scholar]
  21. Yasui K, Kanazawa S, Sano Y, et al. Thoracic tumors treated with CT-guided radiofrequency ablation: initial experience. Radiology 2004; 231:850–857. [DOI] [PubMed] [Google Scholar]
  22. Frich L, Hagen G, Brabrand K, et al. Local tumor progression after radiofrequency ablation of colorectal liver metastases: evaluation of ablative margin and three-dimensional volumetric analysis. J Vasc Interv Radiol 2007; 18:1134–1140. [DOI] [PubMed] [Google Scholar]
  23. Keil S, Bruners P, Schiffl K, et al. Radiofrequency ablation of liver metastases: software-assisted evaluation of the ablation zone in MDCT—tumor-free follow-up versus local recurrent disease. Cardiovasc Intervent Radiol 2010; 33:297–306. [DOI] [PubMed] [Google Scholar]
  24. Fujioka C, Horiguchi J, Ishifuro M, et al. A feasibility study: evaluation of radiofrequency ablation therapy to hepatocellular carcinoma using image registration of preoperative and postoperative CT. Acad Radiol 2006; 13:986–994. [DOI] [PubMed] [Google Scholar]
  25. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009; 45:228–247. [DOI] [PubMed] [Google Scholar]
  26. Schmit GD, Atwell TD, Callstrom MR, et al. Percutaneous cryoablation of renal masses >or=3 cm: efficacy and safety in treatment of 108 patients. J Endourol 2010; 24:1255–1262. [DOI] [PubMed] [Google Scholar]
  27. Andreano A, Huang Y, Meloni MF, Lee FT Jr, Brace C. Microwaves create larger ablations than radiofrequency when controlled for power in ex vivo tissue. Med Phys 2010; 37:2967–2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. de Baere T, Denys A, Wood BJ, et al. Radiofrequency liver ablation: experimental comparative study of water-cooled versus expandable systems. AJR Am J Roentgenol 2001; 176:187–192. [DOI] [PubMed] [Google Scholar]
  29. Hoffmann R, Rempp H, Erhard L, et al. Comparison of four microwave ablation devices: an experimental study in ex vivo bovine liver. Radiology 2013; 268:89–97. [DOI] [PubMed] [Google Scholar]

REPORTING COMPLICATIONS AND SIDE EFFECTS

  1. Livraghi T, Solbiati L, Meloni MF, Gazelle GS, Halpern EF, Goldberg SN. Treatment of focal liver tumors with percutaneous radiofrequency ablation: complications encountered in a multicenter study. Radiology 2003; 226:441–451. [DOI] [PubMed] [Google Scholar]
  2. Rhim H, Yoon KH, Lee JM, et al. Major complications after radiofrequency thermal ablation of hepatic tumors: spectrum of imaging findings. RadioGraphics 2003; 23:123–134; discussion 134–136. [DOI] [PubMed] [Google Scholar]
  3. Vogl TJ, Straub R, Eichler K, Woitaschek D, Mack MG. Malignant liver tumors treated with MR imaging–guided laser-induced thermotherapy: experience with complications in 899 patients (2,520 lesions). Radiology 2002; 225:367–377. [DOI] [PubMed] [Google Scholar]
  4. Dodd GD 3rd, Napier D, Schoolfield JD, Hubbard L. Percutaneous radiofrequency ablation of hepatic tumors: postablation syndrome. AJR Am J Roentgenol 2005; 185:51–57. [DOI] [PubMed] [Google Scholar]
  5. Narayanan G, Froud T, Lo K, Barbery KJ, Perez-Rojas E, Yrizarry J. Pain analysis in patients with hepatocellular carcinoma: irreversible electroporation versus radiofrequency ablation-initial observations. Cardiovasc Intervent Radiol 2013; 36:176–182. [DOI] [PubMed] [Google Scholar]
  6. Carrafiello G, Laganà D, Ianniello A, et al. Post-radiofrequency ablation syndrome after percutaneous radiofrequency of abdominal tumours: one centre experience and review of published works. Australas Radiol 2007; 51:550–554. [DOI] [PubMed] [Google Scholar]

FOLLOW-UP AND OUTCOMES

  1. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92:205–216. [DOI] [PubMed] [Google Scholar]
  2. Li H, Guo Z, Si T, Wang H. EASL and mRECIST responses are independent predictors of survival in hepatocellular carcinoma patients treated with cryoablation. Eur J Gastroenterol Hepatol 2013; 25:620–627. [DOI] [PubMed] [Google Scholar]
  3. Shim JH, Lee HC, Kim SO, et al. Which response criteria best help predict survival of patients with hepatocellular carcinoma following chemoembolization? a validation study of old and new models. Radiology 2012; 262:708–718. [DOI] [PubMed] [Google Scholar]
  4. Fong Y, Blumgart LH, Cohen AM. Surgical treatment of colorectal metastases to the liver. CA Cancer J Clin 1995; 45:50–62. [DOI] [PubMed] [Google Scholar]
  5. Molmenti EP, Marsh JW, Dvorchik I, Oliver JH 3rd, Madariaga J, Iwatsuki S. Hepatobiliary malignancies: primary hepatic malignant neoplasms. Surg Clin North Am 1999; 79:43–57, viii. [DOI] [PubMed] [Google Scholar]
  6. Tracy CR, Raman JD, Donnally C, Trimmer CK, Cadeddu JA. Durable oncologic outcomes after radiofrequency ablation: experience from treating 243 small renal masses over 7.5 years. Cancer 2010; 116:3135–3142. [DOI] [PubMed] [Google Scholar]
  7. Hwang JE, Kim SH, Jin J, et al. Combination of percutaneous radiofrequency ablation and systemic chemotherapy are effective treatment modalities for metachronous liver metastases from gastric cancer. Clin Exp Metastasis 2014; 31:25–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Stangl DK, Berry DA. Meta-analysis in medicine and health policy. New York, NY: Dekker, 2000. [Google Scholar]
  9. Fisher B Clinical trials for the evaluation of cancer therapy. Cancer 1984; 54:2609–2617. [DOI] [PubMed] [Google Scholar]
  10. Peto R, Pike MC, Armitage P, et al. Design and analysis of randomized clinical trials requiring prolonged observation of each patient. I. Introduction and design. Br J Cancer 1976; 34:585–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Begg CB, McNeil BJ. Assessment of radiologic tests: control of bias and other design considerations. Radiology 1988; 167:565–569. [DOI] [PubMed] [Google Scholar]
  12. Armitage P, Berry G. Statistical methods in medical research. Oxford, England: Blackwell Scientific Publications, 1987. [Google Scholar]
  13. Rothmann M, Li N, Chen G, Chi GY, Temple R, Tsou HH. Design and analysis of non-inferiority mortality trials in oncology. Stat Med 2003; 22:239–264. [DOI] [PubMed] [Google Scholar]
  14. Link RE, Permpongkosol S, Gupta A, Jarrett TW, Solomon SB, Kavoussi LR. Cost analysis of open, laparoscopic, and percutaneous treatment options for nephron-sparing surgery. J Endourol 2006; 20:782–789. [DOI] [PubMed] [Google Scholar]

Footnotes

M.A. No relevant conflicts of interest to disclose. L.S. No relevant conflicts of interest to disclose. C.L.B. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: received consultancy fees from NeuWave Medical, author and institution received payment for patents (planned, pending, or issued) from University of Wisconsin and NeuWave Medical, received payment for stock/stock options from NeuWave Medical. Other relationships: none to disclose. D.J.B. Financial activities related to the present article: paid for occasional proctoring by Galil Medical. Financial activities not related to the present article: none to disclose. Other relationships: none to disclose. M.R.C. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: received grants from Galil Medical, Siemens Medical, GE Medical, Thermedical. Other relationships: none to disclose. J.W.C. No relevant conflicts of interest to disclose. M.H.C. No relevant conflicts of interest to disclose. B.I.C. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: institution received a research grant from Samsung Electronics. Other relationships: none to disclose. T.d.B. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: received personal fees from Terumo, a grant from HS-Medical, and payment for travel to a meeting from Boston-Scientific. Other relationships: none to disclose. G.D.D. No relevant conflicts of interest to disclose. D.E.D. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: paid for board membership by BSD Medical, received consultancy fees from BSD Medical and Covidien, received grants or has grants pending from NeuWave Medical, received payment for lectures including service on speakers bureaus from BSD Medical, was paid royalties by Springer Verlag and UpToDate, received stock or stock options from BSD Medical. Other relationships: none to disclose. D.A.G. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: received a research grant from Covidien/Valleylab. Other relationships: none to disclose. D.G. No relevant conflicts of interest to disclose. A.R.G. Financial activities related to the present article: received consulting fee or honorarium from Covidien, received fees for participatient in review activities such as data monitoring boards, statistical analysis, end point committees, and the like from Covidien. Financial activities not related to the present article: was paid for board membership, consultancy, and lectures including service on speakers bureaus by Covidien. Other relationships: none to disclose. F.T.L. Financial activities related to the present article: received payment from NeuWave Medical. Financial activities not related to the present article: received royalties from Covidien, is a stockholder and on the Board of Directors for NeuWave Medical, has a patent on the Switching Controller with royalties paid from Covidien, has multiple patents on heat-based thermal therapies for microwave ablation systems licensed to NeuWave Medical. Other relationships: none to disclose. E.L. No relevant conflicts of interest to disclose. R.L. No relevant conflicts of interest to disclose. P.J.L. No relevant conflicts of interest to disclose. T.L. No relevant conflicts of interest to disclose. D.S.L. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: institution received grants or has grants pending from Endocare and Ethicon Endo-Surgery, received payment for lectures including service on speakers bureaus from Philips. Other relationships: none to disclose. J.P.M. No relevant conflicts of interest to disclose. M.F.M. No relevant conflicts of interest to disclose. B.N. No relevant conflicts of interest to disclose. P.L.P. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: received a grant from Celon Olympus; fees for conferences from Siemens Medical Solutions; consultant’s honoraries from Terumo Europe, Terumo Germany, and Microsulis; materials from AngioDynamics. Other relationships: none to disclose. P.L. No relevant conflicts of interest to disclose. H.R. No relevant conflicts of interest to disclose. S.C.R. No relevant conflicts of interest to disclose. R.S. No relevant conflicts of interest to disclose. C.T.S. No relevant conflicts of interest to disclose. S.B.S. Financial activities related to the present article: institution received a grant from GE Healthcare; author received consulting fee or honorarium from AngioDyamics, Covidien, and GE Healthcare. Financial activities not related to the present article: none to disclose. Other relationships: none to disclose. M.C.S. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: author received consultancy fees from Guerbet LLC, BTG, Merit Medical; institution received grants or has grants pending from Guerbet LLC and BTG; author received payment for lectures including service on speakers bureaus from Sirtex; author received royalties from Cambridge University Press. Other relationships: none to disclose. M.T. No relevant conflicts of interest to disclose. T.J.V. No relevant conflicts of interest to disclose. B.J.W. Financial activities related to the present article: none to disclose. Financial activities not related to the present article: institution received grants or has grants pending from Cooperative Research and Development Agreement (CRADA) Philips Healthcare, CRADA Biocompatibles BTG, CRADA Perfint, CRADA Celsion, CRADA Invivo; the National Institutes of Health has intellectual property in the field. Other relationships: none to disclose. S.N.G. Financial activities related to the present article: institution received a grant for sponsored research from AngioDynamics; author received consulting fee or honorarium from AngioDynamics. Financial activities not related to the present article: author received consultancy fees from Cosman Medical and institution received grants or has grants pending from Cosman Medical. Other relationships: none to disclose.

Contributor Information

Muneeb Ahmed, Department of Radiology, Beth Israel Deaconess Medical Center 1 Deaconess Rd, WCC-308B, Boston, MA 02215.

Luigi Solbiati, Department of Radiology, Ospedale Generale, Busto Arsizio, Italy.

Christopher L. Brace, Departments of Radiology, Biomedical Engineering, and Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.

David J. Breen, Department of Radiology, Southampton University Hospitals, Southampton, England.

Matthew R. Callstrom, Department of Radiology, Mayo Clinic, Rochester, Minnesota.

J. William Charboneau, Department of Radiology, Mayo Clinic, Rochester, Minnesota.

Min-Hua Chen, Department of Ultrasound, School of Oncology, Peking University, Beijing, China.

Byung Ihn Choi, Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea.

Thierry de Baère, Department of Imaging, Institut de Cancérologie Gustave Roussy, Villejuif, France.

Gerald D. Dodd, III, Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.

Damian E. Dupuy, Department of Diagnostic Radiology, Rhode Island Hospital, Providence, Rhode Island.

Debra A. Gervais, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.

David Gianfelice, Medical Imaging, University Health Network, Laval, Quebec, Canada.

Alice R. Gillams, Imaging Department, the London Clinic, London, England.

Fred T. Lee, Jr., Department of Radiology, University of Wisconsin Hospital and Clinics, Madison, Wisconsin.

Edward Leen, Department of Radiology, Royal Infirmary, Glasgow, Scotland.

Riccardo Lencioni, Department of Diagnostic Imaging and Intervention, Cisanello Hospital, Pisa University Hospital and School of Medicine, University of Pisa, Pisa, Italy.

Peter J. Littrup, Department of Radiology, Karmonos Cancer Institute, Wayne State University, Detroit, Michigan.

Tito Livraghi, Busto Arsizio, Italy.

David S. Lu, Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, California.

John P. McGahan, Department of Radiology, Ambulatory Care Center, UC Davis Medical Center, Sacramento, California.

Maria Franca Meloni, Department of Radiology, Ospedale Valduce, Como, Italy.

Boris Nikolic, Department of Radiology, Albert Einstein Medical Center, Philadelphia, Pennsylvania.

Philippe L. Pereira, Clinic of Radiology, Minimally-Invasive Therapies and Nuclear Medicine, Academic Hospital Ruprecht-Karls-University Heidelberg, Heilbronn, Germany.

Ping Liang, Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China.

Hyunchul Rhim, Department of Diagnostic Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.

Steven C. Rose, Department of Radiology, University of California, San Diego, San Diego, California.

Riad Salem, Department of Radiology, Northwestern University, Chicago, Illinois.

Constantinos T. Sofocleous, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.

Stephen B. Solomon, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.

Michael C. Soulen, Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.

Masatoshi Tanaka, Department of Hepatology, Yokokura Hospital, Miyama, Japan.

Thomas J. Vogl, Institute for Diagnostic and Interventional Radiology, University Hospital Frankfurt, Johann Wolfgang Goethe-University, Frankfurt, Germany.

Bradford J. Wood, Radiology and Imaging Science, National Institutes of Health, Bethesda, Maryland.

S. Nahum Goldberg, Department of Radiology, Image-Guided Therapy and Interventional Oncology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel.

REFERENCES

  • 1.Goldberg SN, Charboneau JW, Dodd GD III, et al. Image-guided tumor ablation: proposal for standardization of terms and reporting criteria. Radiology 2003; 228:335–345. [DOI] [PubMed] [Google Scholar]
  • 2.Froeling V, Meckelburg K, Schreiter NF, et al. Outcome of uterine artery embolization versus MR-guided high-intensity focused ultrasound treatment for uterine fibroids: long-term results. Eur J Radiol 2013; 82:2265–2269. [DOI] [PubMed] [Google Scholar]
  • 3.Groeschl RT, Pilgrim CH, Hanna EM, et al. Microwave ablation for hepatic malignancies: a multiinstitutional analysis. Ann Surg 2014; 259:1195–1200. [DOI] [PubMed] [Google Scholar]
  • 4.Narayanan G, Hosein PJ, Arora G, et al. Percutaneous irreversible electroporation for downstaging and control of unresectable pancreatic adenocarcinoma. J Vasc Interv Radiol 2012; 23:1613–1621. [DOI] [PubMed] [Google Scholar]
  • 5.Kim YS, Lim HK, Rhim H, et al. Ten-year outcomes of percutaneous radiofrequency ablation as first-line therapy of early hepatocellular carcinoma: analysis of prognostic factors. J Hepatol 2013; 58:89–97. [DOI] [PubMed] [Google Scholar]
  • 6.Psutka SP, Feldman AS, McDougal WS, McGovern FJ, Mueller P, Gervais DA. Long-term oncologic outcomes after radiofrequency ablation for T1 renal cell carcinoma. Eur Urol 2013; 63:486–492. [DOI] [PubMed] [Google Scholar]
  • 7.Solbiati L, Ahmed M, Cova L, Ierace T, Brioschi M, Goldberg SN. Small liver colorectal metastases treated with percutaneous radiofrequency ablation: local response rate and long-term survival with up to 10-year follow-up. Radiology 2012; 265:958–968. [DOI] [PubMed] [Google Scholar]
  • 8.Peng ZW, Zhang YJ, Chen MS, et al. Radiofrequency ablation with or without transcatheter arterial chemoembolization in the treatment of hepatocellular carcinoma: a prospective randomized trial. J Clin Oncol 2013; 31:426–432. [DOI] [PubMed] [Google Scholar]
  • 9.Ruers T, Punt C, Van Coevorden F, et al. Radiofrequency ablation combined with systemic treatment versus systemic treatment alone in patients with non-resectable colorectal liver metastases: a randomized EORTC Intergroup phase II study (EORTC 40004). Ann Oncol 2012; 23:2619–2626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ahmed M, Brace CL, Lee FT Jr, Goldberg SN. Principles of and advances in percutaneous ablation. Radiology 2011; 258:351–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dodd GD III, Soulen MC, Kane RA, et al. Minimally invasive treatment of malignant hepatic tumors: at the threshold of a major breakthrough. RadioGraphics 2000; 20:9–27. [DOI] [PubMed] [Google Scholar]
  • 12.Goldberg SN, Gazelle GS, Mueller PR. Thermal ablation therapy for focal malignancy: a unified approach to underlying principles, techniques, and diagnostic imaging guidance. AJR Am J Roentgenol 2000; 174:323–331. [DOI] [PubMed] [Google Scholar]
  • 13.Herbold T, Wahba R, Bangard C, Demir M, Drebber U, Stippel DL. The laparoscopic approach for radiofrequency ablation of hepatocellular carcinoma: indication, technique and results. Langenbecks Arch Surg 2013; 398:47–53. [DOI] [PubMed] [Google Scholar]
  • 14.Vavra P, Dostalik J, Zacharoulis D, Khorsandi SE, Khan SA, Habib NA. Endoscopic radiofrequency ablation in colorectal cancer: initial clinical results of a new bipolar radiofrequency ablation device. Dis Colon Rectum 2009; 52:355–358. [DOI] [PubMed] [Google Scholar]
  • 15.Golberg A, Yarmush ML. Nonthermal irreversible electroporation: fundamentals, applications, and challenges. IEEE Trans Biomed Eng 2013; 60:707–714. [DOI] [PubMed] [Google Scholar]
  • 16.Faroja M, Ahmed M, Appelbaum L, et al. Irreversible electroporation ablation: is all the damage nonthermal? Radiology 2013; 266:462–470. [DOI] [PubMed] [Google Scholar]
  • 17.Cressman EN, Jahangir DA. Dual mode single agent thermochemical ablation by simultaneous release of heat energy and acid: hydrolysis of electrophiles. Int J Hyperthermia 2013; 29:71–78. [DOI] [PubMed] [Google Scholar]
  • 18.Fartoux L, Arrive L, Andreani T, et al. Treatment of small hepatocellular carcinoma with acetic acid percutaneous injection. Gastroenterol Clin Biol 2005; 29:1213–1219. [DOI] [PubMed] [Google Scholar]
  • 19.Livraghi T, Giorgio A, Marin G, et al. Hepatocellular carcinoma and cirrhosis in 746 patients: long-term results of percutaneous ethanol injection. Radiology 1995; 197:101–108. [DOI] [PubMed] [Google Scholar]
  • 20.Yan K, Chen MH, Yang W, et al. Radiofrequency ablation of hepatocellular carcinoma: long-term outcome and prognostic factors. Eur J Radiol 2008; 67:336–347. [DOI] [PubMed] [Google Scholar]
  • 21.Brace CL. Microwave tissue ablation: biophysics, technology, and applications. Crit Rev Biomed Eng 2010; 38:65–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sommer CM, Lemm G, Hohenstein E, et al. Bipolar versus multipolar radiofrequency (RF) ablation for the treatment of renal cell carcinoma: differences in technical and clinical parameters. Int J Hyperthermia 2013; 29:21–29. [DOI] [PubMed] [Google Scholar]
  • 23.Vogl TJ, Naguib NN, Gruber-Rouh T, Koitka K, Lehnert T, Nour-Eldin NE. Microwave ablation therapy: clinical utility in treatment of pulmonary metastases. Radiology 2011; 261:643–651. [DOI] [PubMed] [Google Scholar]
  • 24.Merckel LG, Bartels LW, Köhler MO, et al. MR-guided high-intensity focused ultrasound ablation of breast cancer with a dedicated breast platform. Cardiovasc Intervent Radiol 2013; 36:292–301. [DOI] [PubMed] [Google Scholar]
  • 25.Haar GT, Coussios C. High intensity focused ultrasound: physical principles and devices. Int J Hyperthermia 2007; 23:89–104. [DOI] [PubMed] [Google Scholar]
  • 26.Lafon C, Melodelima D, Salomir R, Chapelon JY. Interstitial devices for minimally invasive thermal ablation by high-intensity ultrasound. Int J Hyperthermia 2007; 23:153–163. [DOI] [PubMed] [Google Scholar]
  • 27.Rosenberg C, Puls R, Hegenscheid K, et al. Laser ablation of metastatic lesions of the lung: long-term outcome. AJR Am J Roentgenol 2009; 192: 785–792. [DOI] [PubMed] [Google Scholar]
  • 28.Vogl TJ, Dommermuth A, Heinle B, et al. Colorectal cancer liver metastases: long-term survival and progression-free survival after thermal ablation using magnetic resonance-guided laser-induced interstitial thermotherapy in 594 patients: analysis of prognostic factors. Invest Radiol 2014; 49:48–56. [DOI] [PubMed] [Google Scholar]
  • 29.Callstrom MR, Dupuy DE, Solomon SB, et al. Percutaneous image-guided cryoablation of painful metastases involving bone: multicenter trial. Cancer 2013; 119:1033–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Georgiades CS, Hong K, Bizzell C, Geschwind JF, Rodriguez R. Safety and efficacy of CT-guided percutaneous cryoablation for renal cell carcinoma. J Vasc Interv Radiol 2008; 19:1302–1310. [DOI] [PubMed] [Google Scholar]
  • 31.Rubinsky B, Lee CY, Bastacky J, Onik G. The process of freezing and the mechanism of damage during hepatic cryosurgery. Cryobiology 1990; 27:85–97. [DOI] [PubMed] [Google Scholar]
  • 32.Ben-David E, Appelbaum L, Sosna J, Nissenbaum I, Goldberg SN. Characterization of irreversible electroporation ablation in in vivo porcine liver. AJR Am J Roentgenol 2012; 198:W62–W68. [DOI] [PubMed] [Google Scholar]
  • 33.Lencioni R, Crocetti L, Cioni D, et al. Single-session percutaneous ethanol ablation of early-stage hepatocellular carcinoma with a multipronged injection needle: results of a pilot clinical study. J Vasc Interv Radiol 2010; 21:1533–1538. [DOI] [PubMed] [Google Scholar]
  • 34.Goldberg SN, Gazelle GS, Dawson SL, Rittman WJ, Mueller PR, Rosenthal DI. Tissue ablation with radiofrequency: effect of probe size, gauge, duration, and temperature on lesion volume. Acad Radiol 1995; 2:399–404. [DOI] [PubMed] [Google Scholar]
  • 35.Stafford RJ, Fuentes D, Elliott AA, Weinberg JS, Ahrar K. Laser-induced thermal therapy for tumor ablation. Crit Rev Biomed Eng 2010; 38:79–100. [DOI] [PubMed] [Google Scholar]
  • 36.Cabassa P, Donato F, Simeone F, Grazioli L, Romanini L. Radiofrequency ablation of hepatocellular carcinoma: long-term experience with expandable needle electrodes. AJR Am J Roentgenol 2006; 186:S316–S321. [DOI] [PubMed] [Google Scholar]
  • 37.Shock SA, Meredith K, Warner TF, et al. Microwave ablation with loop antenna: in vivo porcine liver model. Radiology 2004; 231:143–149. [DOI] [PubMed] [Google Scholar]
  • 38.Goldberg SN, Solbiati L, Hahn PF, et al. Large-volume tissue ablation with radio frequency by using a clustered, internally cooled electrode technique: laboratory and clinical experience in liver metastases. Radiology 1998; 209:371–379. [DOI] [PubMed] [Google Scholar]
  • 39.Lubner MG, Hinshaw JL, Andreano A, Sampson L, Lee FT Jr, Brace CL. High-powered microwave ablation with a small-gauge, gascooled antenna: initial ex vivo and in vivo results. J Vasc Interv Radiol 2012; 23:405–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kim SK, Gu MS, Hong HP, Choi D, Chae SW. CT findings after radiofrequency ablation in rabbit livers: comparison of internally cooled electrodes, perfusion electrodes, and internally cooled perfusion electrodes. J Vasc Interv Radiol 2007; 18:1417–1427. [DOI] [PubMed] [Google Scholar]
  • 41.Cua IH, Lin CC, Lin CJ, et al. Treatment of hepatocellular carcinoma using internally cooled electrodes: a prospective comparison of modified automated vs. manual pulsed radiofrequency algorithms. Oncology 2007; 72:76–82. [DOI] [PubMed] [Google Scholar]
  • 42.Park MJ, Kim YS, Rhim H, Lim HK, Lee MW, Choi D. A comparison of USguided percutaneous radiofrequency ablation of medium-sized hepatocellular carcinoma with a cluster electrode or a single electrode with a multiple overlapping ablation technique. J Vasc Interv Radiol 2011; 22:771–779. [DOI] [PubMed] [Google Scholar]
  • 43.Chen MH, Yang W, Yan K, et al. Large liver tumors: protocol for radiofrequency ablation and its clinical application in 110 patients—mathematic model, overlapping mode, and electrode placement process. Radiology 2004; 232:260–271. [DOI] [PubMed] [Google Scholar]
  • 44.Dodd GD III, Frank MS, Aribandi M, Chopra S, Chintapalli KN. Radiofrequency thermal ablation: computer analysis of the size of the thermal injury created by overlapping ablations. AJR Am J Roentgenol 2001; 177:777–782. [DOI] [PubMed] [Google Scholar]
  • 45.Laeseke PF, Sampson LA, Haemmerich D, et al. Multiple-electrode radiofrequency ablation creates confluent areas of necrosis: in vivo porcine liver results. Radiology 2006; 241:116–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Brace CL, Sampson LA, Hinshaw JL, Sandhu N, Lee FT Jr. Radiofrequency ablation: simultaneous application of multiple electrodes via switching creates larger, more confluent ablations than sequential application in a large animal model. J Vasc Interv Radiol 2009; 20:118–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Brace CL, Laeseke PF, Sampson LA, Frey TM, van der Weide DW, Lee FT Jr. Microwave ablation with multiple simultaneously powered small-gauge triaxial antennas: results from an in vivo swine liver model. Radiology 2007; 244:151–156. [DOI] [PubMed] [Google Scholar]
  • 48.Brace CL. Radiofrequency and microwave ablation of the liver, lung, kidney, and bone: what are the differences? Curr Probl Diagn Radiol 2009; 38:135–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dodd GD III, Dodd NA, Lanctot AC, Glueck DA. Effect of variation of portal venous blood flow on radiofrequency and microwave ablations in a blood-perfused bovine liver model. Radiology 2013; 267:129–136. [DOI] [PubMed] [Google Scholar]
  • 50.Lu DS, Raman SS, Vodopich DJ, Wang M, Sayre J, Lassman C. Effect of vessel size on creation of hepatic radiofrequency lesions in pigs: assessment of the “heat sink” effect. AJR Am J Roentgenol 2002; 178:47–51. [DOI] [PubMed] [Google Scholar]
  • 51.Oshima F, Yamakado K, Akeboshi M, et al. Lung radiofrequency ablation with and without bronchial occlusion: experimental study in porcine lungs. J Vasc Interv Radiol 2004; 15:1451–1456. [DOI] [PubMed] [Google Scholar]
  • 52.Livraghi T, Goldberg SN, Lazzaroni S, et al. Hepatocellular carcinoma: radio-frequency ablation of medium and large lesions. Radiology 2000; 214:761–768. [DOI] [PubMed] [Google Scholar]
  • 53.Goldberg SN, Hahn PF, Tanabe KK, et al. Percutaneous radiofrequency tissue ablation: does perfusion-mediated tissue cooling limit coagulation necrosis? J Vasc Interv Radiol 1998; 9:101–111. [DOI] [PubMed] [Google Scholar]
  • 54.Ginat DT, Saad WE. Bowel displacement and protection techniques during percutaneous renal tumor thermal ablation. Tech Vasc Interv Radiol 2010; 13:66–74. [DOI] [PubMed] [Google Scholar]
  • 55.Callstrom MR, York JD, Gaba RC, et al. Research reporting standards for image-guided ablation of bone and soft tissue tumors. J Vasc Interv Radiol 2009; 20:1527–1540. [DOI] [PubMed] [Google Scholar]
  • 56.Pelayo-Alvarez M, Perez-Hoyos S, Agra-Varela Y. Reliability and concurrent validity of the Palliative Outcome Scale, the Rotterdam Symptom Checklist, and the Brief Pain Inventory. J Palliat Med 2013; 16:867–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kaufman CS, Bachman B, Littrup PJ, et al. Office-based ultrasound-guided cryoablation of breast fibroadenomas. Am J Surg 2002; 184:394–400. [DOI] [PubMed] [Google Scholar]
  • 58.Mendiratta-Lala M, Brennan DD, Brook OR, et al. Efficacy of radiofrequency ablation in the treatment of small functional adrenal neoplasms. Radiology 2011; 258:308–316. [DOI] [PubMed] [Google Scholar]
  • 59.Rosenthal DI, Hornicek FJ, Torriani M, Gebhardt MC, Mankin HJ. Osteoid osteoma: percutaneous treatment with radiofrequency energy. Radiology 2003; 229:171–175. [DOI] [PubMed] [Google Scholar]
  • 60.Ahmed M, Moussa M, Goldberg SN. Synergy in cancer treatment between liposomal chemotherapeutics and thermal ablation. Chem Phys Lipids 2012; 165:424–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Spalding AC, Lawrence TS. New and emerging radiosensitizers and radioprotectors. Cancer Invest 2006; 24:444–456. [DOI] [PubMed] [Google Scholar]
  • 62.Grieco CA, Simon CJ, Mayo-Smith WW, DiPetrillo TA, Ready NE, Dupuy DE. Percutaneous image-guided thermal ablation and radiation therapy: outcomes of combined treatment for 41 patients with inoperable stage I/II non-small-cell lung cancer. J Vasc Interv Radiol 2006; 17:1117–1124. [DOI] [PubMed] [Google Scholar]
  • 63.Hamamoto S, Okuma T, Yamamoto A, et al. Radiofrequency ablation and immunostimulant OK-432: combination therapy enhances systemic antitumor immunity for treatment of VX2 lung tumors in rabbits. Radiology 2013; 267:405–413. [DOI] [PubMed] [Google Scholar]
  • 64.Solomon SB, Silverman SG. Imaging in interventional oncology. Radiology 2010; 257:624–640. [DOI] [PubMed] [Google Scholar]
  • 65.Claudon M, Dietrich CF, Choi BI, et al. Guidelines and good clinical practice recommendations for contrast enhanced ultrasound (CEUS) in the liver—update 2012: a WFUMB-EFSUMB initiative in cooperation with representatives of AFSUMB, AIUM, ASUM, FLAUS and ICUS. Ultraschall Med 2013; 34:11–29. [DOI] [PubMed] [Google Scholar]
  • 66.Chen MH, Yang W, Yan K, et al. The role of contrast-enhanced ultrasound in planning treatment protocols for hepatocellular carcinoma before radiofrequency ablation. Clin Radiol 2007; 62:752–760. [DOI] [PubMed] [Google Scholar]
  • 67.Clasen S, Pereira PL. Magnetic resonance guidance for radiofrequency ablation of liver tumors. J Magn Reson Imaging 2008; 27:421–433. [DOI] [PubMed] [Google Scholar]
  • 68.Abi-Jaoudeh N, Kruecker J, Kadoury S, et al. Multimodality image fusion-guided procedures: technique, accuracy, and applications. Cardiovasc Intervent Radiol 2012; 35:986–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wood BJ, Kruecker J, Abi-Jaoudeh N, et al. Navigation systems for ablation. J Vasc Interv Radiol 2010; 21:S257–S263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang X, Sofocleous CT, Erinjeri JP, et al. Margin size is an independent predictor of local tumor progression after ablation of colon cancer liver metastases. Cardiovasc Intervent Radiol 2013; 36:166–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rendon RA, Kachura JR, Sweet JM, et al. The uncertainty of radio frequency treatment of renal cell carcinoma: findings at immediate and delayed nephrectomy. J Urol 2002; 167:1587–1592. [PubMed] [Google Scholar]
  • 72.Sofocleous CT, Nascimento RG, Petrovic LM, et al. Histopathologic and immunohistochemical features of tissue adherent to multitined electrodes after RF ablation of liver malignancies can help predict local tumor progression: initial results. Radiology 2008; 249:364–374. [DOI] [PubMed] [Google Scholar]
  • 73.Goldberg SN, Gazelle GS, Compton CC, Mueller PR, Tanabe KK. Treatment of intrahepatic malignancy with radiofrequency ablation: radiologic-pathologic correlation. Cancer 2000; 88:2452–2463. [PubMed] [Google Scholar]
  • 74.Coad JE, Kosari K, Humar A, Sielaff TD. Radiofrequency ablation causes ‘thermal fixation’ of hepatocellular carcinoma: a post-liver transplant histopathologic study. Clin Transplant 2003; 17:377–384. [DOI] [PubMed] [Google Scholar]
  • 75.Motoyoshi A, Noguchi M, Earashi M, Zen Y, Fujii H. Histopathological and immunohistochemical evaluations of breast cancer treated with radiofrequency ablation. J Surg Oncol 2010; 102:385–391. [DOI] [PubMed] [Google Scholar]
  • 76.Thomsen S Pathologic analysis of photothermal and photomechanical effects of laser-tissue interactions. Photochem Photobiol 1991; 53:825–835. [DOI] [PubMed] [Google Scholar]
  • 77.Lee EW, Loh CT, Kee ST. Imaging guided percutaneous irreversible electroporation: ultrasound and immunohistological correlation. Technol Cancer Res Treat 2007; 6:287–294. [DOI] [PubMed] [Google Scholar]
  • 78.Sainani NI, Gervais DA, Mueller PR, Arellano RS. Imaging after percutaneous radiofrequency ablation of hepatic tumors. I. Normal findings. AJR Am J Roentgenol 2013; 200:184–193. [DOI] [PubMed] [Google Scholar]
  • 79.Tsivian M, Kim CY, Caso JR, Rosenberg MD, Nelson RC, Polascik TJ. Contrast enhancement on computed tomography after renal cryoablation: an evidence of treatment failure? J Endourol 2012; 26:330–335. [DOI] [PubMed] [Google Scholar]
  • 80.Kim YS, Lee WJ, Rhim H, Lim HK, Choi D, Lee JY. The minimal ablative margin of radiofrequency ablation of hepatocellular carcinoma (. 2 and, 5 cm) needed to prevent local tumor progression: 3D quantitative assessment using CT image fusion. AJR Am J Roentgenol 2010; 195:758–765. [DOI] [PubMed] [Google Scholar]
  • 81.Levinson AW, Su LM, Agarwal D, et al. Long-term oncological and overall outcomes of percutaneous radio frequency ablation in high risk surgical patients with a solitary small renal mass. J Urol 2008; 180: 499–504, (discussion 504). [DOI] [PubMed] [Google Scholar]
  • 82.Kawamoto S, Solomon SB, Bluemke DA, Fishman EK. Computed tomography and magnetic resonance imaging appearance of renal neoplasms after radiofrequency ablation and cryoablation. Semin Ultrasound CT MR 2009; 30:67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.yer RS, Timm BA, Mitsumori LM, Kolokythas O. Image fusion as a new postprocessing method to evaluate the radiofrequency ablation zone after treatment of malignant liver tumors. J Comput Assist Tomogr 2010; 34:226–228. [DOI] [PubMed] [Google Scholar]
  • 84.Brace CL, Diaz TA, Hinshaw JL, Lee FT Jr. Tissue contraction caused by radiofrequency and microwave ablation: a laboratory study in liver and lung. J Vasc Interv Radiol 2010; 21:1280–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Georgiades C, Rodriguez R, Azene E, et al. Determination of the nonlethal margin inside the visible “ice-ball” during percutaneous cryoablation of renal tissue. Cardiovasc Intervent Radiol 2013; 36:783–790. [DOI] [PubMed] [Google Scholar]
  • 86.Forner A, Ayuso C, Varela M, et al. Evaluation of tumor response after locoregional therapies in hepatocellular carcinoma: are response evaluation criteria in solid tumors reliable? Cancer 2009; 115:616–623. [DOI] [PubMed] [Google Scholar]
  • 87.Laeseke PF, Lee FT Jr, Sampson LA, van der Weide DW, Brace CL. Microwave ablation versus radiofrequency ablation in the kidney: high-power triaxial antennas create larger ablation zones than similarly sized internally cooled electrodes. J Vasc Interv Radiol 2009; 20:1224–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Sacks D, McClenny TE, Cardella JF, Lewis CA. Society of Interventional Radiology clinical practice guidelines. J Vasc Interv Radiol 2003; 14: S199–S202. [DOI] [PubMed] [Google Scholar]
  • 89.Clavien PA, Barkun J, de Oliveira ML, et al. The Clavien-Dindo classification of surgical complications: five-year experience. Ann Surg 2009; 250:187–196. [DOI] [PubMed] [Google Scholar]
  • 90.Common Terminology Criteria for Adverse Events (CTCAE) v4.0. National Cancer Institute; http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5×7.pdf Accessed May 26, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Wang YB, Chen MH, Yan K, Yang W, Dai Y, Yin SS. Quality of life after radiofrequency ablation combined with transcatheter arterial chemoembolization for hepatocellular carcinoma: comparison with transcatheter arterial chemoembolization alone. Qual Life Res 2007; 16:389–397. [DOI] [PubMed] [Google Scholar]
  • 92.Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis 2010; 30:52–60. [DOI] [PubMed] [Google Scholar]
  • 93.Steele G Jr, Bleday R, Mayer RJ, Lindblad A, Petrelli N, Weaver D. A prospective evaluation of hepatic resection for colorectal carcinoma metastases to the liver: Gastrointestinal Tumor Study Group Protocol 6584. J Clin Oncol 1991; 9:1105–1112. [DOI] [PubMed] [Google Scholar]
  • 94.Mulier S, Ni Y, Jamart J, Michel L, Marchal G, Ruers T. Radiofrequency ablation versus resection for resectable colorectal liver metastases: time for a randomized trial? Ann Surg Oncol 2008; 15:144–157. [DOI] [PubMed] [Google Scholar]
  • 95.Sacks D, Marinelli DL, Martin LG, Spies JB, Society of Interventional Radiology Technology Assessment Committee. General principles for evaluation of new interventional technologies and devices. J Vasc Interv Radiol 2003; 14:S391–S394. [DOI] [PubMed] [Google Scholar]
  • 96.Grunkemeier GL, Starr A. Alternatives to randomization in surgical studies. J Heart Valve Dis 1992; 1:142–151. [PubMed] [Google Scholar]
  • 97.Alexander ES, Machan JT, Ng T, Breen LD, DiPetrillo TA, Dupuy DE. Cost and effectiveness of radiofrequency ablation versus limited surgical resection for stage I non-small-cell lung cancer in elderly patients: is less more? J Vasc Interv Radiol 2013; 24:476–482. [DOI] [PubMed] [Google Scholar]
  • 98.Shetty SK, Rosen MP, Raptopoulos V, Goldberg SN. Cost-effectiveness of percutaneous radiofrequency ablation for malignant hepatic neoplasms. J Vasc Interv Radiol 2001; 12:823–833. [DOI] [PubMed] [Google Scholar]
  • 99.Guyatt GH, Feeny DH, Patrick DL. Measuring health-related quality of life. Ann Intern Med 1993; 118:622–629. [DOI] [PubMed] [Google Scholar]

RESOURCES