Fact-checked by Grok 2 weeks ago

Cryoablation

Cryoablation is a minimally invasive that destroys abnormal or diseased tissue by applying extreme cold, typically through a probe that delivers freezing temperatures to induce direct cellular damage via formation, rupture, and , as well as indirect effects like vascular stasis and . This technique, also known as or , has evolved since its origins in the mid-19th century when Dr. James Arnott first used salt-ice mixtures at -18°C to -24°C for pain relief and , and it now employs modern cryoprobes utilizing gases like or via the Joule-Thomson effect to achieve temperatures as low as -140°C. Cryoablation is particularly valued for its ability to preserve surrounding healthy tissue through real-time imaging guidance with , , or MRI, resulting in reduced pain, minimal scarring, and fewer complications compared to surgical alternatives. In , cryoablation serves as a primary or palliative treatment for various solid tumors, including those in the , , liver, , and , where it effectively freezes and eliminates small or inoperable lesions while potentially stimulating an against residual cancer cells. For cardiac applications, it is commonly used to treat by isolating pulmonary veins through catheter-based freezing, offering a safer profile than heat-based with lower risks to adjacent structures and high success rates in restoring normal rhythm. Despite its benefits, potential risks include , , nerve damage, and rare events like cryoshock or organ fracture, necessitating careful patient selection and procedural monitoring.

Fundamentals

Definition and Overview

Cryoablation is a minimally invasive that employs extreme cold temperatures, typically ranging from -20°C to -196°C, to induce cellular and in targeted abnormal tissues, such as tumors or foci of cardiac arrhythmias. The technique utilizes cryoprobes—thin, insulated needles inserted into or near the target area—to deliver cryogens like , gas, or , which rapidly cool the surrounding tissue. The process involves the formation of an ice ball, a visible frozen zone that encompasses the , achieved through repeated freeze-thaw cycles that disrupt cellular integrity and lead to tissue destruction. This real-time visualization of the ice ball via imaging modalities like or enhances procedural precision, allowing clinicians to monitor the ablation zone directly during treatment. Compared to heat-based alternatives like , which uses to coagulate , cryoablation better preserves adjacent structures such as and blood vessels due to its anesthetic cooling effect and reduced risk of thermal spread. Unlike embolization techniques that block blood supply to induce ischemia, cryoablation directly targets cellular destruction through freezing, often resulting in lower bleeding risk and outpatient feasibility. Its advantages include minimal invasiveness, shorter recovery times, and less post-procedural pain relative to surgical options. Originating in the with the development of cryoprobes initially for dermatological applications, cryoablation has since evolved into a versatile tool for internal therapeutic use across various medical fields.

Cryoablation induces destruction through direct and indirect biophysical mechanisms triggered by extreme . At the cellular level, rapid freezing leads to extracellular formation, which sequesters free and increases extracellular osmolarity, causing osmotic of cells and subsequent shrinkage. This imbalance disrupts cellular , leading to inactivation and damage via protein denaturation. Intracellular crystals form during sufficiently rapid cooling, mechanically rupturing cell and organelles, which results in immediate of targeted . Vascular effects contribute significantly to the overall tissue by compromising supply in the freeze zone. Freezing damages endothelial cells in microvasculature, promoting formation and vascular upon thawing, which induces ischemia and . This indirect injury extends beyond the initial ice-affected area, as reduced prevents recovery and exacerbates through oxygen deprivation and metabolite accumulation. The procedure typically employs two freeze-thaw cycles to maximize destruction: the first forms an ball encompassing the target, while the second enhances lethality by promoting further intracellular growth during thawing. The ball features distinct thermal zones, with a central region below -40°C where immediate predominates due to intracellular formation, an intermediate zone from -20°C to -40°C prone to osmotic and vascular damage, and a peripheral sub-lethal zone above -20°C where delayed may occur. Tissue death generally requires exposure to temperatures below -40°C with rapid cooling rates to induce , as achieved through gas expansion effects like the Joule-Thomson , promoting intracellular formation and amplifying damage compared to slower cooling. occurs centrally via mechanical and ischemic pathways, while the periphery undergoes apoptosis triggered by protein denaturation, , and mitochondrial dysfunction, leading to over hours to days. Post-procedure, a local inflammatory response facilitates debris clearance and may enhance immune recognition of antigens released from destroyed cells. Neutrophils and macrophages infiltrate the ablation zone within hours, releasing cytokines that increase vascular permeability and promote edema, followed by gradual resolution of necrosis over weeks through phagocytosis and fibrosis, while preserving structural collagen. This inflammation can stimulate adaptive immunity by exposing tumor antigens, potentially aiding systemic anti-tumor effects.

Cryogens and Equipment

Cryoablation relies on specialized cryogens to achieve the extreme cold required for tissue destruction, with argon gas, , and being the most commonly employed agents. Argon gas, utilized in pressurized closed-loop systems, cools via the Joule-Thomson effect during rapid expansion, reaching temperatures as low as -120°C to -135°C at the probe tip, though some advanced systems achieve -160°C to -170°C. Its is -185.8°C, enabling efficient heat extraction without direct liquid contact, and it expands rapidly to form balls while minimizing vascular damage compared to open systems. , with a of -196°C, provides the coldest temperatures among these cryogens, operating at probe tips near -196°C and offering superior for faster cooling rates near the applicator. , boiling at -88.5°C, achieves probe temperatures around -76°C and is favored in balloon-based or endoscopic applications due to its moderate expansion properties, though it produces smaller lesions (e.g., 493 ± 197 mm³ after 1 minute of freezing) than (826 ± 163 mm³ under similar conditions). Carbon dioxide, boiling at -78.5°C, is used in some low-cost systems for superficial or limited applications. Cryoprobes serve as the delivery mechanism for these cryogens, typically featuring single or multi-needle arrays with diameters ranging from 1.5 (17-gauge) to 3 (14-gauge) to facilitate percutaneous insertion under imaging guidance such as or . These probes, often constructed from biocompatible metals with insulated shafts, generate balls with radii of 1.5 to 2.5 (diameters of 3 to 5 ), depending on the probe gauge, cryogen type, and freeze duration; for instance, larger 10-gauge needles can produce diameters up to 4.3 at 37°C isotherms. Multi-needle arrays allow for overlapping zones to treat tumors up to several centimeters, with designs like the IceRod or V-Probe enabling adjustable ball shapes for precise targeting near critical structures. Cryoablation systems integrate these components into controlled platforms, predominantly using closed-loop configurations with for freezing and for active thawing to accelerate the freeze-thaw cycle essential for cellular . Console-based units, such as the FDA-cleared Visual ICE system (approved in 2012), support up to 20 needle ports and operate with argon/helium gases, incorporating safety features like real-time pressure regulators, multi-point thermal sensors, and automatic alarms to prevent over-pressurization or incomplete freezing. Portable variants, often -based like the ProSense system, offer flexibility for outpatient use with low-pressure to reduce logistical demands, though they require periodic refills. These systems evolved from early 20th-century liquid nitrogen spray devices, which were limited to superficial applications, to modern needles introduced in the , enabling minimally invasive tumor treatments with enhanced precision and reduced complication rates. Despite advancements, cryoablation equipment carries limitations, including the risk of cryogen leakage, which can occur if probes are not pre-tested in saline for gas , potentially leading to inefficient cooling or under-treatment. migration during insertion or freezing may also cause unintended shifts in the ice ball position, risking damage to adjacent organs or from organ if excessive is applied.

Procedural Techniques

Percutaneous Approach

The approach to cryoablation involves the insertion of thin cryoprobes through the skin to target internal tumors, guided by real-time imaging such as , computed tomography (CT), or (MRI), making it a minimally invasive alternative to open . This method is particularly suited for lesions in organs like the or liver that are accessible without surgical exposure. The procedure typically begins with or to ensure patient comfort, followed by a small skin puncture to insert one or more cryoprobes directly into the target tissue under guidance for precise placement. gas is then circulated through the probes to rapidly freeze the tissue, forming an ice ball that encompasses the tumor; this is followed by thawing using gas or passive warming. The process usually consists of 2-3 freeze-thaw cycles, each lasting 10-15 minutes, with the ice ball monitored intermittently via to confirm adequate coverage. After completion, the probes are removed, and patients undergo post-procedure to assess for immediate complications, followed by a short period. Indications for cryoablation include superficial or deep-seated tumors, such as small renal cell carcinomas (<4 cm) or unresectable liver lesions, where the approach avoids the need for and is suitable for patients unfit for more invasive procedures. Advantages of this technique include its outpatient feasibility, with most patients discharged the same day or after an overnight stay, low morbidity due to the absence of large incisions, and rapid recovery allowing resumption of normal activities within 1-2 days. The major complication rate is generally below 5%, with common issues limited to minor hemorrhage or infection. Technical considerations emphasize the use of 1-5 cryoprobes depending on tumor size (typically for lesions <3-5 cm), positioned to ensure overlapping balls that extend 0.5-1 cm beyond the tumor margins for complete . Proper probe spacing (≤1.5-2 cm apart) and real-time imaging verification of the lethal isotherm (around -40°C) are critical to achieving efficacy while minimizing damage to adjacent structures. Contraindications include coagulopathy, which increases bleeding risk, and lesions that are inaccessible percutaneously due to location or anatomical barriers.

Surgical Approach

Surgical cryoablation involves open or laparoscopic access to the target tissue, allowing for direct visualization and precise placement of cryoprobes in scenarios where percutaneous methods may be inadequate. The procedure typically begins with general anesthesia to ensure patient comfort and immobility. In the laparoscopic approach, small incisions (usually 3-4 trocars of 5-12 mm) are made in the abdomen to establish pneumoperitoneum with CO2 insufflation at 15-20 mmHg, followed by minimal dissection to expose the organ, such as mobilizing the colon and opening Gerota's fascia for renal tumors. For open surgery, a larger incision provides direct access to the target site. Intraoperative ultrasound guides cryoprobe insertion, with probes (typically 1-3, sized 1.7-2.1 mm) positioned 1 cm from the tumor edge and extending 5-10 mm beyond the deep margin to ensure complete coverage. Once probes are secured, the ablation cycle commences using argon gas for rapid freezing to -20°C to -40°C, forming an ice ball visible via for real-time monitoring and adjustment. A standard double freeze-thaw protocol is employed: an initial 6-10 minute freeze, followed by passive thawing (8-10 minutes) and active thawing (1-3 minutes) with gas, then a second freeze cycle of similar duration. Direct allows confirmation of ice ball margins and protection of adjacent structures, such as suturing the or applying hemostatic agents like Flowseal post-thaw. The procedure concludes with closure of incisions or using absorbable sutures, typically lasting 60-120 minutes depending on tumor complexity. Indications for surgical cryoablation include larger tumors exceeding 5 cm (such as T1b renal cell carcinomas up to 7 cm), anatomically challenging locations near vital structures, or cases requiring integration with partial resection in hybrid procedures to preserve organ function. It is particularly suited for patients with comorbidities where full is risky, or for multifocal lesions amenable to simultaneous treatment. Historically, surgical cryoablation was the predominant method for in the 1960s through 1980s, using via open perineal access, before transitioning to techniques in the 1990s with advancements in transrectal ultrasound and smaller probes that reduced invasiveness. Advantages of the surgical approach encompass enhanced precise control through direct exposure and visualization, enabling accurate placement and immediate management of or adjacent tissue protection, which is beneficial for addressing multiple lesions in a single session or combining with . Compared to methods, offers the ability to cover probe tracts and inspect for ice ball cracks, potentially lowering hemorrhage risk in complex cases. Complications are generally higher than in cryoablation due to the invasive access, with wound-related issues such as or dehiscence occurring in approximately 10-15% of cases, alongside a median stay of 3-5 days for and . Major complications, including significant requiring or ureteral obstruction from clots, affect about 1-6% of patients, though overall morbidity remains lower than traditional open partial .

Catheter-Based Approach

The catheter-based approach to cryoablation involves the endovascular delivery of extreme cold through specialized s to ablate targeted cardiac tissue, primarily for treating arrhythmias without the need for open surgery. This method utilizes transvenous access to reach endocardial sites, enabling precise lesion formation through cryogen expansion at the catheter tip or balloon. It is particularly suited for intracardiac targets, offering a minimally invasive alternative to . The procedure typically begins with vascular access via the using guidance to minimize complications. A steerable is advanced under fluoroscopic guidance to the right atrium, followed by transseptal puncture for left atrial access in cases like . For focal , a catheter identifies the arrhythmogenic site, after which the cryo catheter is positioned; cryogen () is delivered to the tip, cooling it to -75°C for 240 seconds per application, often repeated in 2-4 cycles with thawing intervals. In cryoballoon , the balloon is advanced over a guidewire to the , inflated to achieve , and frozen at -40°C to -60°C for 180-240 seconds per , with confirmed via a circular catheter; 2-4 applications per are standard. Post-, catheters are withdrawn, and is secured at the access site. Indications for catheter-based cryoablation center on cardiac arrhythmias, including paroxysmal and persistent for pulmonary vein isolation, atrioventricular nodal reentrant , , and septal accessory pathways. It is recommended as a second-line after antiarrhythmic drug failure, with emerging first-line use for paroxysmal based on trials showing superior arrhythmia-free survival. Key equipment includes focal cryoablation catheters, such as the 6-8 mm tip Freezor Xtra (), which features a flexible for and delivers cryogen through an ultrafine lumen for homogeneous freezing. For pulmonary vein isolation, cryoballoon systems like the Arctic Front Advance () use a 28 mm balloon with eight injection ports for uniform cooling, deployed via a deflectable like FlexCath, and integrated with the Achieve for electrophysiological . These devices connect to a console that regulates cryogen flow and monitors temperature. Advantages of this approach include the ability to target endocardial sites without , reducing recovery time and procedural complexity compared to surgical methods. Cryoablation provides stability through during freezing, enables reversible cryomapping to confirm sites without permanent , and minimizes risks like due to lack of char formation. Success rates for range from 70% to 85% freedom from recurrence at one year, with acute isolation exceeding 98%; for atrioventricular nodal reentrant , long-term success reaches 89%. Risks include vascular access-site complications such as or (1-2%), and procedure-specific issues like in cryoballoon cases (up to 6%, often transient and monitored via pacing). Other potential adverse events are (0.4%) and rare atrioesophageal (0.003-0.25%), though overall major complication rates remain low at 2-4%.

Pre-Procedure Preparation

For applications, pre-procedure preparation for cryoablation varies from other uses, such as cardiac arrhythmias, which may involve , anticoagulation management, and different imaging.

Patient Selection

Patient selection for cryoablation in applications is a critical step to ensure optimal outcomes, balancing oncologic efficacy with procedural safety and patient suitability. Ideal candidates typically include those with small, localized tumors, with size limits varying by organ (e.g., up to 4 cm for renal tumors, 3.5 cm for lung metastases), as larger lesions may reduce complete ablation rates and increase recurrence risk. Tumor location is also pivotal, with eligibility influenced by proximity to critical structures. Cryoablation is often suitable for lesions near major vessels or airways due to preservation of adjacent , but distances such as at least 5 mm from and 3 mm from neurovascular bundles or nerves are preferred in contexts like soft-tissue sarcomas or tumors to minimize . Patient comorbidities play a key role, with suitable individuals often exhibiting good , such as an Eastern Cooperative Oncology Group (ECOG) score of 0-2, and a exceeding 3 months to justify the intervention. Pre-procedure diagnostic confirmation is essential, involving histopathological to verify and TNM to assess disease extent, ensuring cryoablation targets clinically significant lesions rather than benign or indolent ones. This step helps stratify patients, particularly for early-stage cancers where ablation serves as a curative intent alternative to . Absolute contraindications include uncorrectable , which heightens bleeding risk. Relative contraindications include proximity to vital structures requiring protective techniques, and general risks like active for invasive procedures. Multidisciplinary evaluation is standard, involving collaboration among oncologists, interventional radiologists, and surgeons to review , fitness, and alternatives such as surgical resection or , followed by thorough discussing efficacy, risks, and recurrence potential. For specific sites like renal tumors, scoring systems such as the R.E.N.A.L. nephrometry score or the modified (MC2) aid in assessing procedural complexity and predicting complications, guiding suitability for approaches.

Imaging and Site Testing

Pre-procedure imaging plays a crucial role in planning cryoablation by mapping tumor location, size, and relationship to adjacent structures. Computed tomography (), magnetic resonance imaging (), and are commonly employed for this purpose, with contrast-enhanced providing detailed anatomical visualization of renal or hepatic lesions, while offers superior soft tissue contrast for prostate or tumors. serves as an initial screening tool for superficial lesions but is limited by operator dependence and acoustic interference. Positron emission tomography- (PET-) may be integrated to assess metabolic activity in metabolically active tumors, such as phosphaturic mesenchymal tumors, aiding in precise localization. Intraoperative guidance modalities ensure accurate probe placement and monitor treatment progression during cryoablation. is the most common real-time guidance method due to its portability and lack of , though it can be hindered by patient body habitus or gas. fluoroscopy provides excellent visualization for deeper lesions, allowing frequent imaging during freeze cycles. MRI guidance, when available, excels in soft tissue delineation and enables dynamic monitoring with MR-compatible probes, though contraindicated for patients with certain implanted devices like non-MRI-conditional pacemakers due to interference. The ice ball, formed during freezing, appears hypoechoic with posterior shadowing on , as a low-attenuation region on , and as a signal void on T1- and T2-weighted MRI sequences, facilitating assessment of tumor coverage. Site testing involves direct measurement to verify the extent of lethal freezing zones. Temperature probes or thermocouples are inserted near the tumor margin to monitor temperatures in , confirming that isotherms below -30°C to -40°C encompass the with an adequate margin (typically 0.5-1 cm). These devices help adjust probe activation to avoid incomplete or damage to surrounding structures. Advances in fusion imaging enhance precision by overlaying pre-procedure MRI or PET- data onto real-time or , improving targeting accuracy for complex lesions. MRI- fusion, for instance, facilitates better delineation of tumor margins during probe placement, reducing the risk of recurrence as evidenced by lower local tumor progression rates with adequate ablative margins.

Oncology Applications

Prostate Cancer

Cryoablation is employed as a minimally invasive for localized , particularly in cases where preserving is prioritized alongside oncologic control. The procedure targets cancerous tissue within the gland using extreme cold to induce through formation and vascular disruption, often as an alternative to or for suitable candidates. This approach is adaptable for both primary of early-stage and salvage following prior interventions like . The technique typically involves transperineal insertion of 6-8 cryoprobes under transrectal ultrasound (TRUS) guidance to ensure precise placement and real-time monitoring of the ice ball formation. For whole-gland , probes are arranged to cover the entire , while focal targets only the index lesion, sparing surrounding healthy tissue to minimize side effects. A urethral warmer is commonly used to protect the during the dual freeze-thaw cycles, with the procedure performed on an outpatient basis under general or spinal . Indications for cryoablation include low- to intermediate-risk , defined by Gleason scores of ≤7 (typically 3+3 or 3+4), (PSA) levels <10 ng/mL, and clinical stage T1-T2a without significant extraprostatic extension. It is also indicated for salvage treatment in patients with biochemical recurrence after , where repeat irradiation may not be feasible. Patient selection emphasizes those seeking to avoid the morbidity of radical , with multiparametric MRI often used to confirm unilateral or focal disease for targeted . Oncologic outcomes demonstrate biochemical recurrence-free survival (BRFS) rates of 80-90% at 5 years for low- to intermediate-risk patients undergoing primary cryoablation, with higher rates (up to 94%) in low-risk cases and slightly lower (around 84%) in intermediate-risk. Functional preservation is a key advantage, with potency rates maintained in 50-80% of patients post-focal cryoablation, compared to lower rates after radical prostatectomy. These results are supported by nadir levels post-treatment, where values ≤0.5 ng/mL correlate with sustained BRFS. Common complications include in approximately 40% of cases, in about 5%, and requiring intervention in a smaller subset. These rates vary with technique, with focal approaches showing lower incidence of sexual and urinary dysfunction compared to whole-gland . Serious adverse events like recto-urethral are rare (less than 1%) with modern protective measures. Cryoablation is recognized in NCCN guidelines (Version 2.2025) as a category 2B option for salvage therapy in patients with biochemical recurrence after , without evidence of metastatic disease. For primary treatment of low- to intermediate-risk , it is considered an emerging option in clinical trials or for select patients, though long-term data are limited compared to standard therapies like active surveillance, surgery, or radiation. Focal therapy trials, such as those evaluating intermediate endpoints in (e.g., ICECaP collaborative analyses), support its investigational role in intermediate-risk cases, emphasizing metastasis-free survival as a surrogate for overall survival while gathering long-term data.

Renal Cancer

Cryoablation serves as a minimally invasive, nephron-sparing for (RCC), particularly valued for its to preserve in patients with small renal masses. It is especially beneficial for individuals with T1a stage tumors (≤4 cm, confined to the ), those with a solitary , or comorbidities that make surgical options like partial risky. The American Urological Association (AUA) endorses thermal , including cryoablation, as an alternative to for cT1a solid renal masses smaller than 3 cm, prioritizing approaches to reduce morbidity. The R.E.N.A.L. nephrometry score is commonly used to assess tumor complexity and guide procedural planning, with lower scores indicating simpler lesions more amenable to . The procedure typically involves or laparoscopic access, utilizing 1 to 3 cryoprobes inserted under or (US) guidance to target polar tumors less than 4 cm in diameter. Probes are positioned approximately 0.5 cm from the margins, with a double freeze-thaw cycle (10 minutes freeze, followed by passive and active thawing) to form an ice ball extending 0.5–1 cm beyond the tumor for complete . This approach allows real-time monitoring via contrast-enhanced scans every 5 minutes during freezing, ensuring efficacy while minimizing damage to surrounding structures. For more complex cases, laparoscopic assistance may be employed, though methods predominate due to lower invasiveness. Oncologic outcomes demonstrate robust local control, with 5-year local tumor progression-free survival rates of 94.9% in image-guided percutaneous cryoablation series. Renal function is well-preserved, showing no significant decline in estimated (eGFR) over 2 years post-procedure (pre: 72.4 mL/min/1.73 m²; post: 69.7 mL/min/1.73 m²), often exceeding 90% preservation compared to partial in matched cohorts. Long-term data confirm comparable overall survival and cancer-specific survival to surgical for T1 RCC, with no increased risk of . Complications are infrequent and generally minor, including hemorrhage in approximately 3% of cases and ureteral in about 2%, often managed conservatively. Track seeding remains rare, occurring in less than 1% of procedures. Overall morbidity is low, supporting cryoablation's role in patients requiring renal function preservation.

Breast Cancer

Cryoablation for involves the percutaneous, ultrasound-guided insertion of cryoprobes to freeze and destroy targeted tumors, typically performed as an outpatient procedure under . This minimally invasive technique is suitable for small lesions measuring less than 1.5 in diameter, allowing precise ablation while preserving surrounding healthy tissue. Indications for cryoablation in are limited to low-risk, early-stage (T1) tumors that are estrogen receptor-positive (ER+), primarily in women aged 70 years and older who may not be ideal candidates for due to comorbidities or preferences for less invasive options. In 2025, the U.S. granted De Novo marketing authorization to IceCure Medical's ProSense cryoablation system specifically for the local treatment of such low-risk, early-stage s in this patient population, marking the first approval of its kind for this indication. Clinical outcomes from pivotal trials demonstrate high , with the ICE3 reporting a complete rate of approximately 94% and an ipsilateral breast tumor recurrence rate of 4.3% at five years, alongside a breast cancer-specific of 96.7%. Compared to traditional , cryoablation offers superior cosmetic results, including minimal scarring and no need for postoperative , leading to higher in physical well-being and as measured by validated questionnaires like BREAST-Q. Complications associated with breast cryoablation are generally minor and infrequent, including bruising, localized , and skin changes such as dimpling in about 5% of cases, with infection rates below 1% and no serious device-related adverse events reported in key trials. Long-term follow-up from the ICE3 study confirms low recurrence rates under 5% at five years, supporting its safety profile for eligible patients. Ongoing research is expanding cryoablation's applicability, with trials investigating its use in younger patients under 70 and in HER2-positive subtypes, including evaluations of complete in triple-negative breast cancers to broaden indications beyond current low-risk criteria.

Bone Tumors

Cryoablation serves as a minimally invasive option for treating primary and metastatic tumors, particularly for pain palliation in patients with osteolytic lesions. This technique involves freezing tumor tissue to induce through formation and vascular disruption, offering targeted while preserving surrounding structures due to the visible ice ball on . It is especially valuable for patients who are to or systemic therapies, providing rapid symptom relief and local tumor control. The procedure typically employs CT-guided cryoablation, where cryoprobes are inserted through the skin into the under to ensure precise targeting. It is frequently combined with cementoplasty, such as vertebroplasty, to stabilize the treated and prevent fractures by injecting polymethylmethacrylate into the cavity post-freeze-thaw cycles. This hybrid approach enhances structural integrity in weight-bearing sites like the or . Indications for cryoablation include painful osteolytic metastases in the spine or pelvis, with lesions greater than 1 cm in diameter that are refractory to prior radiation therapy. It is suitable for patients with advanced cancers, such as those originating from breast or lung primaries, where pain significantly impacts quality of life and alternative interventions are limited. Clinical outcomes demonstrate substantial pain reduction, with studies reporting 70-90% improvement in pain scores within one month post-procedure, often measured via numeric rating scales. Local tumor control rates reach approximately 80% at one year, reducing the need for opioids and improving overall function. For instance, in metastatic bone disease, mean pain scores decreased by over 2 points within the first week, with durable effects observed up to six months. These benefits are particularly evident in breast and lung metastases, where cryoablation acts as an adjuvant to systemic therapies like bisphosphonates, enhancing palliation without delaying chemotherapy. Complications are generally low, with major adverse events occurring in about 2.5% of cases. The most common include pathologic fractures (around 3-5%), transient nerve damage or neuropathy (2-3%), and, in spinal procedures, rare from cement leakage (less than 1%). Minor issues like temporary pain or affect up to 2-3% of patients but resolve without intervention. Careful patient selection and imaging guidance minimize these risks.

Liver Tumors

Cryoablation is utilized for treating liver tumors, particularly (HCC) in patients with and colorectal liver metastases (CRLM), as a minimally invasive alternative to for unresectable lesions. It is indicated for early-stage HCC classified as BCLC 0 or A, typically involving solitary tumors ≤3 cm or up to three nodules <3 cm, in patients with preserved liver function (Child-Pugh A or B) and no vascular invasion or extrahepatic spread. For CRLM, it is suitable for patients who are poor surgical candidates due to comorbidities or multifocal disease, often as part of . The procedure addresses liver-specific challenges, such as high vascularity and proximity to biliary structures, which can complicate thermal ablation. The technique primarily involves or laparoscopic approaches under (US) or computed (CT) guidance to ensure precise probe placement and avoid major vessels or bile ducts. Multiple cryoprobes (typically 14-17 , 1-3 per lesion for those <3 cm) are inserted to create an ice ball encompassing the tumor with a 5-10 mm margin, using 2-3 freeze-thaw cycles at temperatures of -20°C to -60°C, with total time around 30-40 minutes for small lesions. Track along the probe path is performed to minimize or tumor . In cirrhotic livers, careful assessment of is essential to mitigate risks during probe insertion. Clinical outcomes demonstrate efficacy comparable to (RFA) for small HCC, with 3-year overall survival rates of 60-70% in early-stage cases and local recurrence rates of 10-20%. For CRLM, 3-year survival exceeds 50% in selected patients, with lower local progression near vessels compared to heat-based ablations. Recurrence rates range from 20-30% within 3 years, often distant rather than local. Complications occur in approximately 5% of cases, including bleeding (5-10% in due to ), biloma from biliary injury, and rare cryoshock syndrome (systemic inflammatory response with multi-organ failure, <1%). Major adverse events like hemorrhage are managed conservatively in most instances. The European Association for the Study of the Liver (EASL) recommends cryoablation for inoperable early HCC when is unsuitable, such as perivascular locations, with evidence level III and grade B. It is often combined with transarterial chemoembolization (TACE) to enhance outcomes in multifocal or larger tumors.

Other Medical Applications

Cardiac Arrhythmias

Cryoablation is widely utilized in the management of cardiac arrhythmias, particularly for (PVI) in patients with (AF), where it employs a cryoballoon to deliver controlled freezing to disrupt abnormal electrical pathways. The typically involves transseptal access through the , allowing the cryoballoon to be advanced into the left atrium and positioned at the ostia of the pulmonary veins. Once occlusion is confirmed via contrast , a single freeze of approximately 4 minutes is applied to achieve , often guided by mapping catheters like the Achieve for real-time verification of electrical block. This single-shot approach simplifies the process compared to point-by-point methods, reducing procedural time while targeting the arrhythmogenic foci around the pulmonary veins. Indications for cryoablation in cardiac arrhythmias primarily include symptomatic paroxysmal refractory or intolerant to at least one class I or III antiarrhythmic drug, serving as a first-line rhythm control strategy in selected patients. It is also applied for atrioventricular nodal reentrant (AVNRT), where the cryothermic energy minimizes risk to the node due to its reversible formation, allowing for safer and near critical structures. Additionally, cryoablation addresses (VT) in patients with structural heart disease, particularly when endocardial substrates are perivalvular or intramural, leveraging deeper creation with ultra-low temperatures for durable block. Clinical outcomes demonstrate high efficacy, with freedom from AF recurrence ranging from 70% to 80% at 1 year post-PVI using second-generation cryoballoons, often assessed via continuous monitoring or symptomatic reporting. Compared to , cryoablation shows comparable long-term arrhythmia control but with potentially lower rates of repeat procedures due to fewer reconnections in isolated veins and shorter overall treatment durations. The Arctic Front system, a leading cryoballoon platform, received FDA approval for initial treatment of recurrent symptomatic paroxysmal in 2021, supporting its role in early intervention. According to the 2023 //ACCP/HRS guidelines, cryoablation for PVI holds a class I recommendation for rhythm control in symptomatic paroxysmal after drug failure. Complications are generally low, with phrenic nerve palsy occurring in about 6% of cases—predominantly transient and resolving within months through monitoring and cessation of freezing upon detection via diaphragmatic . Cardiac affects approximately 1% of procedures, often managed percutaneously, while other risks like or remain rare with modern protocols. These rates underscore cryoablation's favorable safety profile, particularly in avoiding permanent esophageal or damage compared to alternatives.

Benign Breast Lesions

Cryoablation for benign lesions primarily targets fibroadenomas, which are common non-cancerous tumors that can cause pain, discomfort, or cosmetic concerns. This uses extreme cold to destroy the targeted tissue, offering an alternative to surgical excision, particularly for women seeking to preserve appearance and avoid general . The involves ultrasound-guided insertion of a cryoprobe directly into the , following confirmation of its benign nature via core needle biopsy. For fibroadenomas smaller than 2 cm, a single probe is typically sufficient, with freezing cycles that form an ice ball encompassing the to induce through ice crystal formation and vascular disruption. The procedure is performed under in an outpatient setting and usually lasts less than an hour. Indications include symptomatic fibroadenomas causing pain or palpable masses that affect , as well as cases where patients prefer a non-surgical option to avoid excision-related scarring or recovery time. It is especially suitable for younger women prioritizing cosmetic outcomes, as the method aligns with patient goals for minimal intervention without compromising breast integrity. Clinical outcomes demonstrate significant lesion volume reduction, with studies reporting mean decreases of 73-89% by 12 months post-procedure, often rendering the mass non-palpable. relief is achieved in the majority of cases, with patients experiencing of symptoms shortly after and sustained over time. Follow-up confirms the ablated area's contraction, supporting long-term efficacy. Complications are minimal and primarily minor, such as transient bruising or occurring in less than 7% of cases, with no reported major adverse events like or significant scarring. The procedure's low risk profile contributes to its tolerability, allowing rapid recovery without downtime. for cryoablation in benign lesions is supported by FDA clearance of systems for treating fibroadenomas, first granted in 2002 (e.g., Her Option™ system), based on multicenter studies showing high success rates and avoidance of in eligible patients. This underscores its established role as a safe, effective option for managing these conditions without oncologic concerns.

Vascular Malformations

Cryoablation is employed as a minimally invasive treatment for congenital and acquired vascular malformations, particularly low-flow lesions such as venous malformations (VMs) and fibroadipose vascular anomalies (FAVA), as well as residual high-flow arteriovenous malformations (AVMs) following embolization. Indications typically include symptomatic cases presenting with focal pain, swelling, bleeding, or functional impairment, often in patients who have not responded adequately to prior interventions like sclerotherapy or surgery. This approach is suitable for focal lesions in the head, neck, and extremities, with increasing application in pediatric patients aged 10 years and older. The technique involves image-guided cryoablation, where 17-gauge cryoprobes are inserted directly into the malformation nidus under (US), computed (CT), or magnetic resonance (MR) guidance, often incorporating Doppler for flow assessment. Procedures typically utilize argon-based systems for two freeze-thaw cycles, with real-time monitoring of the ice ball formation to ensure complete coverage of the target while sparing adjacent structures via hydrodissection if needed. In select cases, cryoablation may follow or complement to enhance efficacy, particularly for , and is performed on an outpatient basis with and post-procedural pain management. Clinical outcomes demonstrate high , with volume reductions ranging from 70% to 92% across treated cases, and symptom or marked improvement in approximately 80-94% of patients, including complete relief in over 60%. For FAVA and residual AVMs, scores decrease by an average of 77-78%, with sustained benefits observed at 6-12 months follow-up. Technical success rates approach 100%, as the ice ball reliably encompasses the symptomatic region. Complications are predominantly minor and self-limiting, occurring in about 15-19% of procedures, including transient numbness, skin blisters or breakdown, bruising, and swelling that resolve within 2-8 weeks. Major adverse events, such as persistent or , are rare at around 3-5%, with no reports of or significant recanalization in reviewed series; however, diffuse lesions may experience worsening symptoms requiring alternative therapies like . Evidence from systematic reviews and case series supports cryoablation's role as a safe and effective option for head and neck , extremity FAVAs, and post-embolization AVM residuals, particularly in cases, though long-term data remain limited and further prospective studies are recommended. Pediatric applications are expanding, with successful outcomes in adolescents and young adults demonstrating feasibility without increased risk.

Emerging Therapies

Cryoimmunotherapy

Cryoimmunotherapy leverages cryoablation to induce immunogenic cell death, releasing tumor antigens and danger-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1) and (ATP), which serve as signals to activate the . These DAMPs promote the maturation of dendritic cells, which then process and present tumor antigens to T-cells, initiating a systemic anti-tumor . In addition to local effects, this process can trigger the , where untreated distant metastases regress due to activated cytotoxic T-cells targeting similar antigens elsewhere in the body. Combining cryoablation with inhibitors, such as PD-1 blockers, enhances this immune activation by overcoming inhibitory signals in the . Preclinical studies in mouse models of lung have demonstrated that cryoablation plus anti-PD-1 therapy significantly inhibits tumor growth, prolongs survival, and increases infiltration of + T-cells compared to either treatment alone. For instance, in bilateral tumor models, the combination reduced distant tumor progression and improved median survival rates beyond monotherapy levels. Clinical evidence supports the potential of cryoimmunotherapy, particularly in advanced cancers. In a phase II trial for metastatic , cryoablation followed by post-progression inhibition achieved an objective response rate (ORR) of 23.5% and a control rate of 41%, with manageable . Similarly, a pilot study in non-small cell combining cryoactivation with PD-1 inhibitors reported a 25% ORR and median overall survival of 13 months. Despite these advances, challenges persist in optimizing cryoimmunotherapy. The immunosuppressive , characterized by upregulation of PD-1/ and CTLA-4 pathways, can dampen the post-ablation, leading to insufficient T-cell activation and risk of recurrence. Additionally, determining optimal dosing for adjuvants like checkpoint inhibitors remains complex, as trials often prioritize safety over precise regimens, with variations in timing and sequence affecting efficacy. Looking ahead, cryoimmunotherapy holds promise for integration with personalized vaccines derived from post-ablation tumor antigens. Preclinical and early-phase studies suggest that cryoablation-released autologous antigens can form the basis for vaccines, enhancing dendritic cell-based therapies tailored to individual tumor profiles and potentially amplifying long-term immunity. Such approaches may address current limitations by boosting antigen-specific T-cell responses in a patient-specific manner.

Recent Advances

In 2025, the U.S. (FDA) granted marketing authorization to IceCure Medical's ProSense cryoablation system for the local treatment of early-stage, low-risk in women aged 70 and older, marking the first such approval for cryoablation in this indication when combined with standard endocrine therapy. This approval was supported by demonstrating effective tumor with minimal invasiveness, addressing a need for alternatives to in elderly patients. Additionally, focal cryoablation for has seen expanded adoption, with recent advancements in multiparametric MRI (mpMRI) enhancing precision and functional outcomes like erectile function preservation, as evidenced by 2025 studies reporting favorable short- to intermediate-term oncologic results. Technological innovations have improved cryoablation's accuracy and integration with imaging. Boston Scientific's Visual ICE MRI Cryoablation System, introduced in recent years, enables real-time visualization of the ablation zone during MRI-guided procedures, facilitating precise tumor targeting in organs like the and liver. AI-assisted planning has advanced with models, such as 3D-Unet-based systems, that predict iceball boundaries for focal cryoablation, allowing clinicians to optimize probe placement and coverage pre-procedure. Nanoparticles have emerged to enhance cryoablation efficacy, with research showing improved tumor penetration and immune activation when integrated into ablation protocols, particularly for solid tumors. Ongoing clinical trials underscore cryoablation's evolving role in combination therapies. Clinical studies and meta-analyses of cryoablation combined with transarterial chemoembolization (TACE) for advanced (HCC) have demonstrated improved safety and efficacy, with meta-analyses confirming low major complication rates under 5%. Nanotechnology-enhanced approaches are being explored in trials to boost cryogen penetration in dense tissues, potentially expanding applications beyond traditional limits. The cryoablation devices market, valued at approximately USD 477 million in , is projected to reach over USD 1 billion by 2030, driven by rising demand for minimally invasive treatments and integrations with to leverage cryoablation-induced antigen release. Recent long-term data from breast cancer cohorts indicate sustained tumor control with low recurrence rates up to five years post-procedure, filling evidence gaps for non-surgical options. Robotics-assisted probe positioning has reduced procedural complications in image-guided cryoablation, with systems like MRI coil-mounted positioners minimizing insertion errors and tissue trauma.

History

Early Development

The concept of using extreme cold to treat medical conditions dates back to the mid-19th century, with early precursors laying the groundwork for modern cryoablation. In the , British physician James Arnott pioneered the therapeutic application of cold by using mixtures of and to achieve temperatures as low as -24°C, primarily for palliation of pain and treatment of lesions and advanced cancers, including breast and uterine tumors. By the early 1900s, experiments with emerged as a more potent cryogenic agent; in 1899, dermatologist Campbell White applied via sprays or swabs to treat various conditions such as , , and carcinomas, reporting rapid and effective lesion destruction with minimal scarring. These rudimentary methods, often limited to surface applications, highlighted cold's potential for tissue destruction but suffered from imprecise control and logistical challenges in cryogen handling. A pivotal advancement occurred in 1961 when American neurosurgeon Irving S. Cooper, in collaboration with engineer and cryobiologist Arthur Rinfret, invented the first closed-system cryosurgical probe utilizing to reach temperatures of -196°C. This device, designed for precise intracerebral application, enabled targeted freezing of neural tissue and was initially employed in to create lesions in the for treating like and to ablate brain tumors. Cooper's probe marked a shift from superficial sprays to invasive, controlled cryoablation, allowing for deeper tissue access while minimizing surrounding damage through reversible freezing effects. Following its introduction, cryoablation saw rapid adoption in several medical fields during the early . In , liquid nitrogen sprays and probes effectively treated benign skin lesions such as viral warts and seborrheic keratoses, building on earlier liquid air techniques with improved efficacy and reduced pain. benefited from cryotherapy's precision in procedures like retinal cryopexy for , where probes facilitated adhesion of the to the underlying without invasive . Similarly, in gynecology, cryosurgical probes using were applied to erosions and intraepithelial neoplasias starting around , offering a minimally invasive alternative to traditional excision for abnormal . A notable milestone in came in 1967, when researchers reported the first transurethral cryoablation of the for both (BPH) and cancer, employing a liquid nitrogen probe inserted via the to freeze obstructive tissue. This approach, inspired by Cooper's probe, aimed to relieve urinary symptoms while preserving function, though early outcomes varied due to procedural limitations. Despite these innovations, initial cryoablation faced significant challenges, including the bulkiness of equipment—such as large flasks for storage and cumbersome delivery systems—that restricted portability and operating room integration. Additionally, the absence of real-time imaging modalities like meant reliance on manual guidance, leading to inconsistent freeze zones and higher risks of incomplete treatment or collateral damage.

Key Milestones and Modern Evolution

The 1970s marked the beginning of modern cryoablation's shift toward minimally invasive techniques, with early percutaneous applications emerging for liver tumors using liquid nitrogen probes during open procedures that transitioned to image-guided insertions by the late 1980s. This evolution facilitated safer access for inoperable liver malignancies, reducing the need for laparotomy while leveraging intraoperative ultrasound for probe placement. By the early 1990s, percutaneous cryoablation extended to prostate cancer, enabling transperineal probe insertion under ultrasound guidance to target localized lesions with improved functional preservation compared to radical surgery. The introduction of argon gas-based cryoprobes in the 1990s revolutionized the field by enabling smaller, more precise devices through the Joule-Thomson effect, which allowed rapid cooling to -140°C and facilitated approaches for both liver and applications. In 1998, transcatheter cryoablation was first applied clinically for cardiac arrhythmias, such as atrioventricular nodal reentrant , marking a pivotal advancement in endovascular use and paving the way for standardized guidance protocols that enhanced real-time monitoring of ice ball formation across tumor ablations. The U.S. (FDA) later formalized approvals for cardiac devices in the early 2000s, building on these foundations. The 2000s saw the emergence of focal therapy paradigms, where cryoablation targeted specific tumor foci rather than whole organs, particularly in , to minimize side effects like incontinence and while achieving comparable oncologic control. Multi-probe arrays, introduced during this decade, allowed simultaneous deployment of up to 10 cryoprobes for larger ablation zones, improving efficacy for irregular tumors in the liver and . Applications expanded to renal and cancers, with cryoablation demonstrating feasibility for small renal masses (<4 cm) and peripheral lesions, often under guidance, yielding local control rates exceeding 90% in select cohorts. In the , concepts of cryoimmunotherapy gained traction, as cryoablation's release of tumor antigens was shown to prime systemic immune responses, prompting combinations with checkpoint inhibitors like anti-PD-1 to enhance antitumor immunity in preclinical and early-phase trials. The FDA approved cryoablation for benign fibroadenomas in , offering a minimally invasive alternative that achieved over 90% volume reduction in lesions up to 3 cm, with guidance standardizing outpatient procedures. The have integrated advanced technologies, including AI-driven for probe placement and ball prediction to optimize margins, as seen in developments enhancing precision for complex anatomies like tumors. In October 2025, the FDA granted marketing authorization for the ProSense cryoablation system to treat low-risk, early-stage in women aged 70 and older, expanding indications based on trials showing complete response rates of 94% with minimal complications. Concurrently, global phase II trials have advanced cryoimmunotherapy combinations, such as cryoablation with inhibition prior to for metastatic , demonstrating improved objective response rates of up to 23.5% and immune reprogramming in distant tumors.

References

  1. [1]
    Cryoablation: Mechanism of Action and Devices - PMC - NIH
    Cryoablation refers to all methods of destroying tissue by freezing. Cryoablation causes cellular damage, death, and necrosis of tissues by direct mechanisms.Cryoablation: Mechanism Of... · Mechanism Of Action · Why Cryoablation?
  2. [2]
    Cryoablation: What It Is, Used For, Benefits & Risks - Cleveland Clinic
    Cryoablation is treatment for some common cancers like lung and prostate cancer. It works by freezing cancer cells and eliminating tumors.Overview · Treatment Details · Risks / Benefits
  3. [3]
    Cryoablation for the Treatment of Atrial Fibrillation
    Doctors perform cryoablation to restore normal heart rhythm by disabling heart cells that create an irregular heartbeat.
  4. [4]
    Definition of cryoablation - NCI Dictionary of Cancer Terms
    A procedure in which an extremely cold liquid or an instrument called a cryoprobe is used to freeze and destroy abnormal tissue.
  5. [5]
    Lethal isotherms of cryoablation in a phantom study: Effects of heat ...
    Cryoablation uses lethal cold temperatures of −20°C to −40°C depending upon the tissue type (12, 13), and has been applied since 1845 (14).Missing: range | Show results with:range
  6. [6]
    Ultra-Low–Temperature Cryoablation for Ventricular Tachycardia
    Ultra-low–temperature cryoablation (ULTC) uses near-critical nitrogen and can generate temperatures as low as –196 °C.
  7. [7]
    Measurement Analysis of Ice Ball Size during CT-Guided ... - NIH
    Freezing the target tissue will result in the formation of an ice ball around the cryoprobe with its size determined by four factors: rate of gas infusion, time ...
  8. [8]
    Percutaneous Cryoablation of Renal Lesions With Radiographic Ice ...
    The radiographic ice ball was defined as the hypoattenuating region surrounding the cryoprobes that had a clearly defined margin. The margin of the renal sinus ...Missing: process | Show results with:process
  9. [9]
    Cryoablation vs. Radiofrequency Ablation for Small Renal Masses
    One major advantage of cryoablation over RFA is the real-time monitoring of the ablation zone 23, 26. The basis of this ablative technique is the destruction ...
  10. [10]
    Radiofrequency Ablation versus Cryoablation in the Treatment of ...
    Radiofrequency ablation (RFA) causes tissue damage using heat while cryoablation causes tissue damage by freezing the target region. RFA has been considered to ...
  11. [11]
    What is Cryoablation? - Tampa General Hospital
    Cryoablation is a less-invasive, interventional radiology treatment used to treat advanced or metastatic renal cell carcinoma and other tumors,
  12. [12]
    Cryoablation for cancer - Mayo Clinic
    Jan 3, 2025 · Cryoablation for cancer is a treatment that uses extreme cold directly on cancer cells, causing them to die.
  13. [13]
    What is Cryoablation? Risks & Benefits - MedStar Health
    Read about cryoablation, a minimally invasive interventional radiology procedure that freezes and kills abnormal cancerous and precancerous cells.Reasons · What to Expect · Risk & Benefits · Preparation
  14. [14]
    Cryotherapy for Cancer - Penn Medicine
    As a minimally invasive procedure, it typically results in less pain, swelling, and damage to healthy tissue, while allowing for a shorter recovery time.
  15. [15]
    The history of cryosurgery - PMC - NIH
    Temple Fay, in Philadelphia, applied both local and general cold treatment for cervical tumours during the 1940s and in 1964 Cahan developed the liquid nitrogen ...
  16. [16]
    [PDF] Percutaneous cryoablation techniques and clinical applications
    Dec 8, 2009 · The de- velopment of liquid nitrogen-containing cryoprobes in the 1960s has allowed deeper tissues to be treated with cryoablation (1).
  17. [17]
    Cryoablation: Physical and Molecular Basis with Putative ... - NIH
    Cryoablation provides more complex, sequenced effects on targeted cells than initially recognized [1]. Initial freeze rupture occurs during the freeze process ...
  18. [18]
    Mechanisms of Cryoablation: Clinical Consequences on Malignant ...
    The alcohol draws water from the cell, producing dehydration and cell destruction. Multiple injections may be needed, depending upon the size of the tumor [64].
  19. [19]
    Use of cryoablation beyond the prostate - PMC - PubMed Central - NIH
    An emerging indication of cryoablation is breast cancer. In the last decade, percutaneous US-guided ablation has been used successfully in the treatment of ...Missing: steps | Show results with:steps
  20. [20]
    Renal cryoablation – a practical guide for interventional radiologists
    Renal cryoablation is a treatment option for early stage renal cell carcinomas with excellent oncological outcomes and low morbidity.
  21. [21]
    Percutaneous CT-Guided Renal Cryoablation - NIH
    Mar 20, 2021 · Cryoablation is a safe and effective technique for the treatment of small renal lesions, with no major complications when performed by expert interventional ...Missing: steps | Show results with:steps
  22. [22]
    Percutaneous Cryoablation in the Liver: A Meta-Analysis and ...
    Oct 15, 2024 · Analysis of currently available evidence indicates that cryoablation has a relative safe profile with a pooled incidence of major complications below 5%.Materials And Methods · Cryoablation Technique · Complications<|control11|><|separator|>
  23. [23]
    Percutaneous Cryoablation for Liver Cancer - PMC - NIH
    There are several advantages to using cryoablation for the treatment of liver cancer: it can be performed percutaneously, intraoperatively, and ...Missing: contraindications | Show results with:contraindications
  24. [24]
    Laparoscopic Renal Cryoablation - PMC - PubMed Central - NIH
    Cryosurgery, also known as cryoablation, offers benefits in preserving renal function with fewer complications, shorter hospitalization times, and allows for ...Missing: steps indications advantages
  25. [25]
    [PDF] How I do it: laparoscopic renal cryoablation (LRC)
    Cryoablation is performed using a third generation machine. Flowseal is then applied to the cryoablation site as an adjunct step for hemostasis. The Gerota's.
  26. [26]
    [PDF] CRYOABLATION TREATMENT GUIDE | Boston Scientific
    Cryoablation offers clinicians a number of unique advantages: • Treatment zone visibility and control. • Reduced pain due to analgesic effect of ice.
  27. [27]
    History and Development of Prostate Cryoablation - Abdominal Key
    Jan 29, 2018 · The major complications that were frequently reported in the early periods of cryosurgery for prostate cancer in the 1960s through the 1980s ...
  28. [28]
    Prostate cryoablation: Update 1998 - Schmidt
    In 1966, Gonder et al performed prostate cryosurgery in humans, treating bladder neck obstruction from both cancer and benign prostatic hyperplasia. They used a ...
  29. [29]
    Comparison of Percutaneous and Laparoscopic Cryoablation for the ...
    With laparoscopic cryoablation, the kidney can be directly visualized and the puncture site and any cracks in the ice ball can be filled and covered to reduce ...Missing: steps | Show results with:steps
  30. [30]
    Cryoablation - Medical Clinical Policy Bulletins - Aetna
    Although cryoablation is less invasive than surgical incision, a key disadvantage of cryoablation is the lack of a tissue sample to examine histologically to ...
  31. [31]
    Long-term outcomes of cryoablation for biopsy-proven T1 stage ...
    The median postoperative hospital stay was 3 days (1~6 days). Compared with LCA, procedure time of PCA was shortened, the volume of bleeding was reduced ...Missing: wound issues
  32. [32]
    Catheter Cryoablation | Circulation: Arrhythmia and Electrophysiology
    Whereas partial damage and delayed recovery will occur with longer exposures to tissue temperatures in the −5°C to −15°C range, permanent AV block can be ...
  33. [33]
    Best Practice Guide for Cryoballoon Ablation in Atrial Fibrillation - NIH
    In this most basic technique, the catheter sheath is used to apply pressure to the balloon, leading to vein occlusion. Care must be taken to avoid pushing the ...
  34. [34]
    Catheter Cryoablation of Cardiac Arrhythmias | USC Journal
    Catheter-based cryotherapy includes two complementary procedures - cryomapping, which allows for the confirmation of target sites, and cryoablation, which ...The Biology Of Cryotherapy · Cryomapping And Cryoablation... · How The Cryotherapy System...Missing: steps | Show results with:steps
  35. [35]
    Lung Cryoablation: Patient Selection, Techniques, and Postablation ...
    May 22, 2025 · Recent studies have established an upper size limit of 3.5 cm for treating OMD lesions with LCA (17–19). Currently, no definitive guidelines ...
  36. [36]
    Standardization of selection criteria for percutaneous image-guided ...
    Eligibility for cryoablation was assessed by two radiologists according to predefined criteria: maximal diameter size of the tumor ≤ 10 cm, distance to skin >5 ...
  37. [37]
    Ablation of Small Renal Masses - ScienceDirect.com
    Contraindications. Uncorrectable coagulopathy is the only absolute contraindication to percutaneous thermal ablation. Proximity of the ablation zone to ...
  38. [38]
    Society of Interventional Radiology Quality Improvement Standards ...
    Jan 5, 2020 · The MC2 score was shown to be superior to the R.E.N.A.L. nephrometry score in predicting major complications after renal cryoablation in 1 ...
  39. [39]
    An Overview of Image-Guided Percutaneous Ablation of Renal Tumors
    IMAGING GUIDANCE. Percutaneous image-guided ablation can be performed under ultrasound, computed tomography (CT), or magnetic resonance imaging (MRI) guidance.
  40. [40]
    Multimodality Image-Guided Cryoablation for Inoperable Tumor ...
    Multi-modality imaging guides the treatment needles in real-time. Ultrasound and CT guidance are augmented by fusion of MRI, FDG-PET/CT, or DOTATATE-PET/CT ...
  41. [41]
    Percutaneous cryoablation in soft tissue tumor management
    Nov 18, 2024 · PCA is efficient in treating soft tissue tumors, including desmoid tumors, vascular malformations, and abdominal wall endometriosis.
  42. [42]
    Image-Guided Prostate Cryoablation: State-of-the-Art - PMC - NIH
    Given the lack of MRI guidance platforms readily available, all procedures are performed with CT and ultrasound guidance under general anesthesia.
  43. [43]
  44. [44]
    Cryotherapy, HIFU, and Other Ablative Treatments for Prostate Cancer
    Dec 10, 2024 · Cryotherapy (also called cryosurgery or cryoablation) is the use of very cold temperatures to freeze and kill prostate cancer cells. Even though ...
  45. [45]
    Prostate Cryoablation - Urologic Surgeons of Washington
    Prostate cryoablation is a primary treatment for prostate cancer in which extremely low temperatures are precisely introduced to the prostate gland.
  46. [46]
    What are the treatments for prostate Cancer? - Koelis
    To do this, cryoablation needles (generally 4-8) are inserted into the prostate transperineally. The needles produce cold, which destroys the cells while ...
  47. [47]
    Transrectal Ultrasound-Guided Transperineal Cryoablation in the ...
    Transrectal ultrasound-guided transperineal percutaneous cryoablation of the prostate produces few major complications and appears at 3 months to be ...
  48. [48]
    Focal Cryotherapy for Clinically Unilateral, Low-Intermediate Risk ...
    Conclusions. Primary focal cryoablation for low-intermediate risk unilateral cancer affords encouraging oncologic and functional outcomes over a median 3.7-yr ...
  49. [49]
    Comparing the Oncological Outcomes of Cryoablation vs. Radical ...
    Aug 25, 2020 · Purpose: To compare the oncologic outcomes of cryoablation (CA) and radical prostatectomy (RP) in patients with low- and intermediate-risk ...Missing: indications | Show results with:indications
  50. [50]
    Current Status of Cryoablation in Prostate Cancer Management - NIH
    Jul 3, 2025 · The purpose of this narrative review is to evaluate the current evidence on cryoablation as both focal and whole-gland treatment for PCa, ...Missing: verification biomarkers
  51. [51]
    Primary Whole-gland Cryoablation for Prostate Cancer - PubMed - NIH
    Sep 28, 2018 · The 5-yr clinical recurrence-free survival was 83% overall and 94% for low-, 84% for intermediate-, and 69% for high-risk PCa (p=0.046). Failure ...
  52. [52]
    Focal prostate cryoablation: initial results show cancer ... - PubMed
    Focal cryoablation can provide biochemical and local control of prostate cancer while preserving potency and continence. Further investigation is needed.
  53. [53]
    Cryotherapy for prostate cancer - Mayo Clinic
    Apr 30, 2025 · Side effects of cryotherapy for prostate cancer can include: Erectile dysfunction; Pain and swelling of the scrotum and penis; Blood in the ...Overview · Why It's Done · What You Can Expect
  54. [54]
    Treatment profile and complications associated with cryotherapy for ...
    Twelve months following cryotherapy, lower urinary tract obstruction, erectile dysfunction, urinary incontinence, and bowel bleeding rose to 28.7%, 20.1%, 9.8%, ...
  55. [55]
    [PDF] NCCN Guidelines for Patients: Early-Stage Prostate Cancer
    These NCCN Guidelines for Patients are based on the NCCN. Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Prostate Cancer, Version 2.2025 — ...
  56. [56]
    Intermediate clinical endpoints for surrogacy in localised prostate ...
    Mar 19, 2024 · We sought to comprehensively assess aggregate trial-level performance of commonly reported ICEs across all randomized trials in localized PCa.Missing: cryoablation | Show results with:cryoablation
  57. [57]
    Ultrasound-Guided Breast Cancer Cryoablation - PubMed
    Percutaneous ultrasound-guided breast cryoablation is a minimally invasive technique that kills targeted tissue with extreme cold.
  58. [58]
    Percutaneous ultrasound-guided cryoablation for early-stage ...
    Apr 27, 2024 · Cryoablation is a nonsurgical minimally invasive technique using extreme cold temperatures to destroy benign or malignant tumors that meet those ...
  59. [59]
    IceCure Receives FDA Marketing Authorization for ProSense ...
    Oct 3, 2025 · IceCure Receives FDA Marketing Authorization for ProSense® Cryoablation for the Treatment of Low-Risk Breast Cancer in Women Aged 70 and Above.
  60. [60]
    Cryoablation Regimen Receives FDA Marketing Authorization for ...
    Oct 7, 2025 · IceCure Medical's ProSense cryoablation granted FDA marketing authorization for treatment of low-risk breast cancer in women aged 70 and above: ...
  61. [61]
    ICE3 Trial 5-Year Follow-Up on Ipsilateral Breast Tumor Recurrence
    Sep 16, 2024 · This study demonstrates cryoablation to be safe for early-stage breast cancer ≤ 1.5 cm, with minor adverse events (such as bruising, minor ...
  62. [62]
    IceCure's ICE3 5-Year Trial Results Published in the Peer Reviewed ...
    Sep 24, 2024 · ... ICE3 trial, the IBTR rate at 5 years was 4.3% and breast cancer survival was 96.7%. Of the 124 patients who received cryoablation and ...
  63. [63]
    Cryoablation Allows the Ultimate De-escalation of Surgical Therapy ...
    Sep 28, 2023 · Cryoablation is emerging as a cost-effective ablative option because the procedure can be performed in an office setting under ultrasound ...Missing: indications | Show results with:indications
  64. [64]
    ICE3 Trial 5-Year Follow-Up on Ipsilateral Breast Tumor Recurrence
    Sep 16, 2024 · The conducted meta-analysis (n = 1266 patients) showed an 85% complete ablation rate in the cryoablation-specific cohort with the lowest ...
  65. [65]
    Breast Cryoablation Inhibits Recurrence, Enhances Survival in ...
    Feb 11, 2025 · Breast cryoablation elicited low recurrence and high survival rates in women 60 years or older with low-risk, early-stage breast cancers.
  66. [66]
    IceCure's ProSense® Cryoablation Featured in Six Studies ...
    Mar 18, 2025 · The study found that cryoablation with ProSense® can completely ablate T1 breast cancer, including triple negative breast cancer, and younger ...
  67. [67]
    IceCure's ProSense Cryoablation System Evaluated for Breast ...
    Sep 17, 2025 · Cryoablation with ProSense achieved the highest complete ablation rate with no complications, supporting its selection for a phase 3 trial.
  68. [68]
    CIRSE Standards of Practice on Thermal Ablation of Bone Tumours
    Cryoablation is a safe treatment of bone tumours with a very low rate of major complications (2.5%), the most common of which is secondary fracture (1.2%). ...
  69. [69]
    Role of Percutaneous Cryoablation in Bone and Soft Tissue Tumors
    Jul 17, 2023 · This review reports the most important literature on CT-guided cryoablation's application in musculoskeletal oncology, including sarcoma, bone ...
  70. [70]
    Cementoplasty to cryoablation: review and current status - PMC
    Percutaneous cementoplasty is a minimally invasive technique comprising of cannula placement within a bone lesion with subsequent cement injection. The cement, ...
  71. [71]
    Percutaneous image-guided cryoablation of painful metastases ...
    Mar 1, 2013 · Conclusions: Percutaneous cryoablation is a safe, effective, and durable method for palliation of pain due to metastatic disease involving bone.Missing: reduction outcomes<|separator|>
  72. [72]
    Efficacy of Cryoablation to Control Cancer Pain: A Systematic Review
    Cryoablation decreased mean pain scores by 62.5% at 24 hours post-cryoablation, by 70% at 3 months, and by 80.9% at 6 months.Missing: reduction | Show results with:reduction
  73. [73]
    Cryoablation for Palliation of Painful Bone Metastases: The MOTION ...
    Feb 12, 2021 · Cryoablation of metastatic bone tumors provided rapid and durable pain palliation, improved quality of life, and offered an alternative to ...
  74. [74]
    Minimally Invasive Interventional Procedures for Metastatic Bone ...
    The results demonstrated that cryoablation plus zoledronic acid regimen induced a significant drop in the worst and average pain between week 1 and week 4 ...
  75. [75]
    Complications of Percutaneous Bone Tumor Cryoablation
    Feb 26, 2019 · Bone tumor cryoablation is safe, with a 2.5% rate of major complications, most commonly secondary fracture (1.2%).
  76. [76]
    Complications of cryoprobe cryoablation as a surgical adjuvant for ...
    Jul 17, 2023 · These complications included postoperative fracture (5/148, 3.4%), nerve palsy (2/148, 2.7%), wound complications (11/148, 7.4%), and infection ...
  77. [77]
    complication rates for lung, bone, and soft tissue tumors cryoablation
    Sep 3, 2024 · The overall major complication rate for cryoablation in bone lesions ranges from 0% to 7.4%. A retrospective study by Auloge P et al described a ...
  78. [78]
    Local Ablation for Hepatocellular Carcinoma: 2024 Expert ... - NIH
    Sep 3, 2024 · Approximately 5% of patients develop complications after local ablation,16,19,70 Major complications include liver abscesses, hemoperitoneum, ...Patient Selection · 1) Us-Ct/mri Fusion... · 5. Local Ablation Techniques
  79. [79]
    Survival outcomes and quality of life after percutaneous cryoablation ...
    Aug 16, 2023 · Cryoablation is an effective procedure for the treatment of liver metastases, especially in cases that are poor candidates for liver resection.Missing: indications | Show results with:indications
  80. [80]
    Outcomes of ultrasound-guided percutaneous argon-helium ... - NIH
    Indeed, two recent studies have described the outcomes of percutaneous cryoablation for HCC under CT or MRI guidance, and showed that it was safe and effective ...
  81. [81]
    Long-Term Outcomes of Percutaneous Cryoablation for Patients ...
    Percutaneous cryoablation is an effective therapy for patients with HCC within Milan criteria, with comparable efficacy, safety and long-term survival
  82. [82]
    Hepatocellular Carcinoma Locoregional Therapies - AASLD
    May 28, 2023 · Cryoablation induces tumor necrosis by ... EASL guidelines recommend surgery as first line for single tumor with compensated liver disease.
  83. [83]
    [PDF] EASL Clinical Practice Guidelines: Management of hepatocellular ...
    The following Clinical Practice Guidelines will give up-to-date advice for the clinical management of patients with hepatocel- lular carcinoma, as well as ...
  84. [84]
  85. [85]
    Single 3-Minute versus Double 4-Minute Freeze Strategy ... - PubMed
    A "single 3-minute strategy" showed equal efficacy as compared to the conventional 4-minute plus bonus freeze approach at 2-year follow-up, providing shorter ...Missing: protocol | Show results with:protocol
  86. [86]
    2023 ACC/AHA/ACCP/HRS Guideline for the Diagnosis and ...
    Nov 30, 2023 · The 2023 ACC/AHA/ACCP/HRS Guideline for the Diagnosis and Management of Atrial Fibrillation provides recommendations to guide clinicians in the treatment of ...
  87. [87]
    Cryoablation Versus Radiofrequency Energy for the Ablation of ...
    Nov 15, 2010 · Our findings indicate that cryoablation is as effective as RF ablation in the short term but is associated with a significantly higher ...
  88. [88]
    Ultra-low-temperature cryoablation for ventricular tachycardia ... - NIH
    Cryoablation is a safe and effective treatment option for nonischemic cardiomyopathy patients suffering from ventricular tachycardia. •. Preprocedural cardiac ...
  89. [89]
    Cryoballoon pulmonary vein isolation as a standard approach for ...
    This single-shot technique makes the pulmonary vein isolation procedure easier and has the potential to expand access to the interventional treatment of atrial ...
  90. [90]
    Cryoballoon or Radiofrequency Ablation for Atrial Fibrillation ...
    Oct 21, 2019 · Cryoballoon ablation and contact force–guided radiofrequency ablation are similarly efficacious in preventing atrial fibrillation (AF) ...
  91. [91]
    Medtronic Receives FDA Expanded Approval for Arctic Front ...
    Jun 21, 2021 · First and Only Approval to Indicate Cryoablation as an Initial Rhythm Control Strategy. - Offers Safe, Effective Alternative to ...
  92. [92]
    Complications of Atrial Fibrillation Cryoablation - PMC - NIH
    Major complications of percutaneous catheter ablation for AF include pericardial effusion/tamponade, embolic events, PV stenosis, atrioesophageal fistula (AEF), ...
  93. [93]
    Complications of Cryoballoon Pulmonary Vein Isolation | AER Journal
    Mar 8, 2019 · CB ablation is associated with a significant risk of phrenic nerve injury (PNI) due to the limited balloon size, and right PNI is the most common complication.
  94. [94]
    Cryosurgery for breast fibroadenomas - PMC - NIH
    With use of ultrasound-guided cryoablation for breast fibroadenomas, there is little or no pain, targeted lesions are reduced in size or eliminated, scarring ...Missing: IceSeed | Show results with:IceSeed
  95. [95]
    Ultrasound-Guided Breast Cancer Cryoablation | AJR
    Cryoablation has been used in the breast to treat fibroadenomas and cancers for over a decade. Treated fibroadenomas show mean volume reduction of 73–89%, with ...
  96. [96]
    Cryoablation and Breast Disease | Current Breast Cancer Reports
    Jun 10, 2024 · Cryoablation remains an effective and well-tolerated non-surgical option for women with benign breast fibroadenomas. Cryobalation for breast ...
  97. [97]
    Fibroadenoma Cryoablation - IceCure
    Cryoablation gives fibroadenoma patients the choice of less invasive treatment without surgery and minimal scarring or change to breast shape.Missing: complications IceSeed
  98. [98]
    Percutaneous Image-Guided Ablation of Breast Tumors: An Overview
    Cryoablation can be performed in the office with only local anesthesia, whereas surgery is more expensive and may have poor cosmetic outcome. Reimbursement for ...
  99. [99]
    Cryosurgery for breast fibroadenomas - Niu - Gland Surgery
    A median volume reduction of 99% was observed with ultrasound. Ninety ... Interim results from the FibroAdenoma Cryoablation Treatment Registry. Am J ...
  100. [100]
    Breast cryoablation for management of benign fibroadenomas
    The pooled minor complication rate from cryoablation of fibroadenomas was found to be 6.9 % (95 % CI: 3.1 %–14.8 %). Minor complications included edema, ...
  101. [101]
    Cryoablation Is Option In Breast Fibroadenomas - The Hospitalist
    Jan 17, 2019 · The complication rate was low: a 0.8% infection rate and a 2.9% incidence of hematoma. Transient ecchymosis was observed in a total of 41 ...
  102. [102]
    Percutaneous Image-Guided Cryoablation in Vascular Anomalies
    Targeted cryoablation destroys the malformation by causing effective cell death within the malformation. With argon-based cryoablation systems, the Joule ...
  103. [103]
  104. [104]
    Cryoablation of low-flow vascular malformations
    Cryoablation is safe and effective in the treatment of low-flow vascular malformations, either after sclerotherapy or as primary treatment.Missing: catheter- based
  105. [105]
    Cryoablation as Second-line Therapy for Fibroadipose Vascular ...
    Mar 2, 2021 · Cryoablation has recently emerged as a promising treatment. Our experience suggests FAVA with failed prior interventions may respond favorably to cryoablation.
  106. [106]
    Self-amplifying cryo-immunotherapy enabled by Ga-MOF ...
    Nov 1, 2025 · This process releases danger signals such as HMGB1 and ATP and upregulates CRT exposure, thereby fully activating the innate immune system [22].
  107. [107]
    Progress of Immune‐Inducible Biomaterials for Post‐Ablation Cancers
    Jun 17, 2025 · Increase releasing tumor antigens, elevating dendritic cell (DC) numbers in ablated tumors, and promoting a significant CD4+ and CD8+ T‐cell ...
  108. [108]
    Radiotherapy, photodynamic therapy, and cryoablation‐induced ...
    Abscopal effects of cryoablation. Cryoablation has a distinct advantage over other therapies. It causes severe necrosis by disrupting the cell membrane and ...
  109. [109]
    Cryoablation synergizes with anti-PD-1 immunotherapy induces an ...
    Nov 2, 2024 · Cryoablation in combination with anti-PD-1 antibody inhibited distant tumors growth and improved mouse survival. Mechanistically, this ...Fig. 1 · Fig. 4 · Cryoablation Can Exert An...
  110. [110]
    Modern cancer therapy: cryoablation meets immune checkpoint ...
    Cryoablation not only directly destroys tumor cells at the local treatment site but also activates the body's immune system, impacting metastatic tumors at ...
  111. [111]
    Cryoablation and post-progression immune checkpoint inhibition in ...
    Aug 27, 2024 · The study met its primary endpoints with the combination strategy found to be safe and feasible with an objective response rate of 23.5% and ...
  112. [112]
    A Pilot Study of Lung Cancer Cryoactivation in Combination ... - NIH
    Cryoactivation, a technique inducing cell death by cycles of freezing and thawing, has the potential to augment tumor responses when combined with ICIs.
  113. [113]
    Size selection of intrahepatic lesions for cryoablation contributes to ...
    Mar 2, 2024 · Size selection of intrahepatic lesions for cryoablation contributes to abscopal effect and long-term survival in patients with liver metastatic melanoma
  114. [114]
    Cryoablation for breast cancer: a narrative review of advances ... - NIH
    Feb 26, 2025 · Additionally, it explores the challenges associated with combining cryoablation with immunotherapy to prevent tumor recurrence. As technological ...Tumor Selection · Cryoablation For Breast... · Cryoablation And...
  115. [115]
    A phase I prospective, non-randomized trial of autologous dendritic ...
    Mar 20, 2023 · By combining cryoablation with intratumoral DC injection, cryoimmunotherapy (CryoIT) exposes the entire individualized collection of tumor ...
  116. [116]
    IceCure Receives FDA Marketing Authorization for ProSense ...
    Oct 3, 2025 · IceCure Receives FDA Marketing Authorization for ProSense® Cryoablation for the Treatment of Low-Risk Breast Cancer in Women Aged 70 and Above.
  117. [117]
    FDA Grants De Novo Approval for Cryoablation Treatment of Breast ...
    Oct 6, 2025 · The ProSense cryoablation system recently garnered De Novo approval from the FDA for the local treatment of low-risk, early-stage breast cancer ...<|control11|><|separator|>
  118. [118]
    VISUAL ICE™ MRI Cryoablation System - Boston Scientific
    Visual ICE MRI Cryoablation System offers precise and effective tumor treatment by combining cryoablation zone visibility with high-resolution MRI imaging.
  119. [119]
    AI-Based Isotherm Prediction for Focal Cryoablation of Prostate ...
    This paper presents a convolutional neural network model based on 3D-Unet to predict the frozen isotherm boundaries (iceball) resultant from a given a cryo- ...
  120. [120]
    Recent progress in cryoablation cancer therapy and nanoparticles ...
    Cellular death by cryoablation is accomplished through multiple mechanisms, including physical damage of cellular membrane by ice formation, initiation of ...
  121. [121]
    Cryoablation Devices Market Size | Industry Report, 2030
    The global cryoablation devices market size was valued at USD 477.1 million in 2024 and is projected to reach USD 1036.7 million by 2030, growing at a CAGR ...
  122. [122]
    Cryoablation in the treatment of early breast cancer - PubMed Central
    May 20, 2025 · The fundamental principle involves the application of freezing temperatures to induce cellular injury and death (29, 30). This process is ...<|control11|><|separator|>
  123. [123]
    An MRI Coil-Mounted Multi-Probe Robotic Positioner for Cryoablation
    This paper presents an interventional guidance device for faster and more accurate alignment and insertion of multiple probes during cryoablation performed in ...Missing: nanotechnology | Show results with:nanotechnology
  124. [124]
    [PDF] History of Cryosurgery
    6 The first physician to use liquid air was Dr. Campbell White from New York, who in 1899 used it to treat various skin conditions.7 The liquid air was applied ...
  125. [125]
    Irving S. Cooper (1922–1985): a pioneer in functional neurosurgery in
    In 1960, with the help of Robert Johnson, an engineer, and Arthur Rinfret, a cryobiologist, Cooper designed the first cryosurgical probe for use in the brain.
  126. [126]
    Cryotherapy of benign and neoplastic tumors of the prostate - PubMed
    Cryotherapy of benign and neoplastic tumors of the prostate. Surg Gynecol Obstet. 1967 Dec;125(6):1265-8. Authors. W P Jordan Jr, D Walker, G H Miller Jr, ...
  127. [127]
    Interventional oncology of liver tumors: how it all started and where ...
    The history of cryoablation for the treatment of liver malignancies started in the late 80s as “cryosurgery”. Liquid nitrogen was initially placed directly on ...
  128. [128]
    Percutaneous cryoablation for hepatocellular carcinoma - PMC - NIH
    Joule-Thompson coefficients of gases vary with pressure and temperature. For argon gas, cooling effect becomes negligible when the gas expands under lower ...
  129. [129]
    US-guided percutaneous cryoablation of prostate cancer - PubMed
    A tailored approach to cryosurgery of prostate cancer is presented. The authors have performed 214 cryoprocedures to date with use of this protocol.Missing: shift liver 1980s standardization 1990s multi- arrays
  130. [130]
    Cryoablation: History, Mechanism of Action, and Guidance Modalities
    Cryoablation has been applied for the treatment of cancer in various nonaerated organs; recently it has been adapted to the treatment of lung tumors.Missing: FDA approvals, cryoimmunotherapy
  131. [131]
    Cryoballoon ablation for atrial fibrillation - Open Access Journals
    Cryoballoon ablation (CBA) emerged as a potential 'single shot' tool for pulmonary vein isolation. Lesion formation develops in three phases with cryoablation: ...Practical Considerations · Cba Efficacy & Safety · Indications For Cryoablation...
  132. [132]
    [PDF] U.S. Premarket Approval (PMA) Application Number
    Apr 17, 2003 · The 7F Freezor® Cardiac Cryoablation Catheter and CCT.2 CryoConsole System is indicated for the cryoablation of the conducting tissues of ...
  133. [133]
    Focal therapy in prostate cancer: Development, application and ...
    Feb 20, 2025 · First used in the early 2000s, focal cryotherapy appeared to be comparable in oncological control over 5 years compared to whole-gland ...
  134. [134]
    Progress in the cryoablation and cryoimmunotherapy for tumor - PMC
    Cryoablation may lead to tumor cell necrosis through a variety of mechanisms, including mechanical damage to the cell membrane after the formation of ice ...
  135. [135]
    Cryoablation and immunotherapy: an overview of evidence on its ...
    May 20, 2019 · It is hypothesised that cell death by cryoablation leaves the intracellular contents of the cancer cells intact for the immune system to induce ...Results · Cryoablation And The Immune... · Breast Cancer
  136. [136]
    Cryoablation for Benign Breast Tumor Treatment | Breastlink
    Aug 21, 2014 · Approved in 2010 by the FDA, the procedure is widely regarded as safe and effective as a fibroadenoma treatment. The American Society of ...
  137. [137]
    Techsomed and Medtronic Collaborate on AI-Guided Ablation Therapy
    Oct 22, 2025 · The collaboration will combine Techsomed's BioTraceIO360 software and Medtronic's Emprint microwave ablation system for image-guided ablation ...
  138. [138]
    IceCure Medical's ProSense® Cryoablation Granted FDA Marketing ...
    Oct 3, 2025 · Breast cancer cryoablation with ProSense®, is a simple, quick, out-patient procedure that can have a notably positive impact on the patient ...
  139. [139]
    Immune Checkpoint Inhibition in Combination with Cryoablation for ...
    This phase II trial tests how well immune checkpoint inhibition in combination with cryoablation prior to standard of care surgery works in treating ...
  140. [140]
    Cryoablation and post-progression immune checkpoint inhibition in ...
    Aug 27, 2024 · The study met its primary endpoints with the combination strategy found to be safe and feasible with an objective response rate of 23.5% and ...<|separator|>