1
|
Vasiliauskas G, Žemaitė E, Skrodenienė E, Poškienė L, Maziliauskienė G, Mačionis A, Miliauskas S, Vajauskas D, Žemaitis M. Early Effects of Bronchoscopic Cryotherapy in Metastatic Non-Small Cell Lung Cancer Patients Receiving Immunotherapy: A Single-Center Prospective Study. Diagnostics (Basel) 2025; 15:201. [PMID: 39857085 PMCID: PMC11763732 DOI: 10.3390/diagnostics15020201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Cryotherapy is used for local tissue destruction through rapid freeze-thaw cycles. It induces cancer cell necrosis followed by inflammation in the treated tumor microenvironment, and it stimulates systemic adaptive immunity. Combining cryotherapy with immunotherapy may provide a sustained immune response by preventing T cell exhaustion. Methods: Fifty-five patients with metastatic non-small cell lung cancer who had received no prior treatment were randomized into two groups in a 1:1 ratio: the bronchoscopic cryotherapy group or the control group. Patients received up to four cycles of pembrolizumab as monotherapy or in combination with platinum-based chemotherapy. Immune-related adverse events (irAEs), complications, tumor size changes, overall response rate (ORR), and disease control rate (DCR) were evaluated. Results: Lung tumors, treated with cryotherapy, demonstrated continuous reduction from the baseline (22.4 cm2 vs. 14.4 cm2 vs. 10.2 cm2, p < 0.001). Similar changes were observed in pulmonary tumors in the control group (19.0 cm2 vs. 10.0 cm2, p < 0.001). The median change in pulmonary tumors between two groups was not significant (-42.9% vs. -27.7%, p = 0.175). No significant differences were observed in the ORR (28.6% vs. 23.1%, p = 0.461) or target lesion decrease (-24.0% vs. -23.4%, p = 0.296) between the groups. However, the DCR was significantly higher in the cryotherapy group (95.2% vs. 73.1%, p = 0.049). No cases of serious bleeding during cryotherapy or pneumothorax were observed. Six patients (25.0%) in the cryotherapy group and eight (26.7%) in the control group experienced irAEs. Conclusions: Our study demonstrated that combined bronchoscopic cryotherapy and immunotherapy with or without chemotherapy may reduce the rate of progressive disease in metastatic non-small cell lung cancer patients while maintaining a satisfactory safety profile.
Collapse
Affiliation(s)
- Gediminas Vasiliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (S.M.); (M.Ž.)
| | - Evelina Žemaitė
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.Ž.); (E.S.)
| | - Erika Skrodenienė
- Department of Laboratory Medicine, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (E.Ž.); (E.S.)
| | - Lina Poškienė
- Department of Pathology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Gertrūda Maziliauskienė
- Department of Radiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (G.M.); (A.M.); (D.V.)
| | - Aurimas Mačionis
- Department of Radiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (G.M.); (A.M.); (D.V.)
| | - Skaidrius Miliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (S.M.); (M.Ž.)
| | - Donatas Vajauskas
- Department of Radiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (G.M.); (A.M.); (D.V.)
| | - Marius Žemaitis
- Department of Pulmonology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (S.M.); (M.Ž.)
| |
Collapse
|
2
|
Yano H, Higashihara H, Kimura Y, Enchi Y, Nakamura S, Satomura H, Koretsune Y, Tanaka K, Ono Y, Tomiyama N. Unenhanced CT as an Alternative to Contrast-Enhanced CT in Evaluating Renal Cryoablation Zones. Cureus 2024; 16:e71295. [PMID: 39534811 PMCID: PMC11554433 DOI: 10.7759/cureus.71295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Background Advances in imaging technology and the increased use of abdominal imaging have led to a rise in renal cell carcinoma (RCC) detection. While surgery remains the primary treatment for small RCCs, minimally invasive procedures like cryoablation are gaining popularity, particularly for patients with comorbidities or renal dysfunction. CT-guided cryoablation offers advantages, including high spatial resolution and real-time visualization during the procedure. Post-procedure imaging is essential for assessing treatment success, with contrast-enhanced CT (CE-CT) typically considered vital. However, many patients, especially older individuals, have renal dysfunction that limits the use of contrast agents. In such cases, unenhanced CT (UE-CT) presents a viable alternative for post-procedural evaluation. This study explored the effectiveness of UE-CT in assessing cryoablation zones as a substitute for CE-CT. Materials and Methods This retrospective study included 54 patients (58 tumors) who underwent cryoablation at a single institution between 2014 and 2024. Only patients with available early follow-up CT (within three days post-cryoablation) and subsequent follow-up were included. Tumors marked with lipiodol prior to cryoablation and cases requiring transcatheter arterial embolization due to extravasation immediately after cryoablation were excluded. Percutaneous renal cryoablation was performed under CT fluoroscopy, and the ablation zone was assessed using a 64-channel multi-slice CT scanner. UE-CT was conducted before the procedure, followed by both UE-CT and CE-CT within three days after cryoablation. CT attenuation values were measured for pre-procedure UE-CT (kidneys and tumor), post-procedure UE-CT (kidneys, cryoablation zone, and tumor), and post-procedure CE-CT (kidneys, cryoablation zone, and tumor). Tumor volumes in the post-procedure regions were evaluated on both UE-CT and CE-CT. Statistical analyses were performed using Wilcoxon's signed-rank test and Spearman's rank correlation coefficient, with interobserver agreement determined by the intraclass correlation coefficient. Results The median tumor diameter was 1.56 cm (IQR: 1.33-2.00 cm). On UE-CT, the cryoablation zone exhibited high attenuation, while it showed low attenuation on CE-CT. The median attenuation values of the kidneys on UE-CT before and after cryoablation were not significantly different (33.6 Hounsfield unit (HU) vs. 34.3 HU, P = 0.17). However, on CE-CT, the median attenuation values of normal kidneys and the cryoablation zone significantly differed (171.7 HU vs. 55.7 HU, P < 0.0001). Similarly, on UE-CT, there was a significant difference in the median attenuation values between normal kidneys and the cryoablation zone (34.3 HU vs. 47.4 HU, P < 0.0001). The median renal volumes of the unenhanced regions on CE-CT and those with attenuation changes on UE-CT were not significantly different (26.52 cm³ vs. 28.83 cm³, P = 0.86). These values showed a strong correlation (r = 0.95; 95% CI: 0.91-0.97). Conclusions This study showed that UE-CT can reliably estimate the ablation zone in RCC patients post-cryoablation. While the contrast between the ablation zone and normal renal parenchyma was lower on UE-CT compared to CE-CT, the ablation zone was still detectable and highly correlated with CE-CT results. Further research with larger sample sizes is needed to validate the clinical utility of UE-CT and assess the reproducibility of these findings.
Collapse
Affiliation(s)
- Hiroki Yano
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| | - Hiroki Higashihara
- Department of High Precision Image-Guided Percutaneous Intervention, Osaka University Graduate School of Medicine, Suita, JPN
| | - Yasushi Kimura
- Department of High Precision Image-Guided Percutaneous Intervention, Osaka University Graduate School of Medicine, Suita, JPN
| | - Yukihiro Enchi
- Department of Radiology, Osaka University Hospital, Suita, JPN
| | - Soichi Nakamura
- Department of Radiology, Osaka University Hospital, Suita, JPN
| | - Hiroki Satomura
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| | - Yuji Koretsune
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| | - Kaishu Tanaka
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| | - Yusuke Ono
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| | - Noriyuki Tomiyama
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, JPN
| |
Collapse
|
3
|
Chen M, Liu W, Liu B. Cryoablation with KCl Solution Enhances Necrosis and Apoptosis of HepG2 Liver Cancer Cells. Ann Biomed Eng 2024; 52:2118-2133. [PMID: 38615077 DOI: 10.1007/s10439-024-03512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/04/2024] [Indexed: 04/15/2024]
Abstract
Cryoablation has become a valuable treatment modality for the management of liver cancer. However, one of the major challenges in cryosurgery is the incomplete cryodestruction near the edge of the iceball. This issue can be addressed by optimizing cryoablation parameters and administering thermotropic drugs prior to the procedure. These drugs help enhance tumor response, thereby strengthening the destruction of the incomplete frozen zone in liver cance. In the present study, the feasibility and effectiveness of a thermophysical agent, KCl solution, were investigated to enhance the cryodestruction of HepG2 human liver cancer cells. All cryoablation parameters were simultaneously optimized in order to significantly improve the effect of cryoablation, resulting in an increase in the lethal temperature from - 25 °C to - 17 °C. Subsequently, it was found that the application of KCl solution prior to freezing significantly decreased cell viability post-thaw compared to cryoablation treatment alone. This effect was attributed to the eutectic effect of KCl solution. Importantly, it was found that the combination of KCl solution and freezing was less effective when applied to LO2 human liver normal cells. The data revealed that the ratio of mRNA levels of Bcl-2 and bax decreased significantly more in HepG2 cells than in LO2 cells when cryoablation was used with KCl solution. In conclusion, the results of this study demonstrate the effectiveness of KCl solution in promoting cryoablation and describe a novel therapeutic model for the treatment of liver cancer that may distinguish between cancer and normal cells.
Collapse
Affiliation(s)
- Mu Chen
- Institute of Biothermal Science and Technology, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, 200093, China
- Shanghai Collaborative Innovation Center for Tumor Treatment with Energy, Shanghai, 200093, China
| | - Wei Liu
- Institute of Biothermal Science and Technology, University of Shanghai for Science and Technology, Shanghai, 200093, China
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, 200093, China
- Shanghai Collaborative Innovation Center for Tumor Treatment with Energy, Shanghai, 200093, China
| | - Baolin Liu
- Institute of Biothermal Science and Technology, University of Shanghai for Science and Technology, Shanghai, 200093, China.
- Shanghai Technical Service Platform for Cryopreservation of Biological Resources, Shanghai, 200093, China.
- Shanghai Collaborative Innovation Center for Tumor Treatment with Energy, Shanghai, 200093, China.
| |
Collapse
|
4
|
Campbell WA, Makary MS. Advances in Image-Guided Ablation Therapies for Solid Tumors. Cancers (Basel) 2024; 16:2560. [PMID: 39061199 PMCID: PMC11274819 DOI: 10.3390/cancers16142560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Image-guided solid tumor ablation methods have significantly advanced in their capability to target primary and metastatic tumors. These techniques involve noninvasive or percutaneous insertion of applicators to induce thermal, electrochemical, or mechanical stress on malignant tissue to cause tissue destruction and apoptosis of the tumor margins. Ablation offers substantially lower risks compared to traditional methods. Benefits include shorter recovery periods, reduced bleeding, and greater preservation of organ parenchyma compared to surgical intervention. Due to the reduced morbidity and mortality, image-guided tumor ablation offers new opportunities for treatment in cancer patients who are not candidates for resection. Currently, image-guided ablation techniques are utilized for treating primary and metastatic tumors in various organs with both curative and palliative intent, including the liver, pancreas, kidneys, thyroid, parathyroid, prostate, lung, breast, bone, and soft tissue. The invention of new equipment and techniques is expanding the criteria of eligible patients for therapy, as now larger and more high-risk tumors near critical structures can be ablated. This article provides an overview of the different imaging modalities, noninvasive, and percutaneous ablation techniques available and discusses their applications and associated complications across various organs.
Collapse
Affiliation(s)
- Warren A. Campbell
- Division of Vascular and Interventional Radiology, Department of Radiology, University of Virginia, Charlottesville, VA 22903, USA
| | - Mina S. Makary
- Division of Vascular and Interventional Radiology, Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Zhang K, Liu M, Cong L, He X, Xu Y, Wang Q, Li C. A Comparative Study of Antitumor Immunity Induced by Radiofrequency Microwave and Cryoablation in Hepatocellular Carcinoma. Appl Biochem Biotechnol 2024; 196:4088-4104. [PMID: 37889403 DOI: 10.1007/s12010-023-04760-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE This study aimed to compare the immune responses induced by microwave ablation (MWA), radiofrequency ablation (RFA), and cryoablation (CRYO) in hepatocellular carcinoma (HCC) and identify differences in immune responses and the timing of immune changes. MATERIALS AND METHODS A bilateral subcutaneous model was established in C57 mice, and the successfully modeled mice were divided into the microwave (n = 15), radiofrequency (n = 15), CRYO (n = 15), control (n = 9), and blank groups (n = 3). Mice in the control group were dissected before ablation, whereas mice in the three ablation groups underwent ultrasound-guided ablation of one axillary tumor. Three mice were sacrificed and dissected at 1-4 weeks after ablation. After tissue processing, flow cytometry was used to detect the levels of CD8 + T and regulatory T (Treg) cells in the tissue, and western blotting was used to assess the level of programmed cell death ligand 1 (PD-L1) protein in the tumor tissue. RESULTS The pattern of immune changes after the three types of ablation was consistent, with immune changes occurring at 3-4 weeks. CRYO induced the most significant increase in the percentage of CD8 + T cells. There were no significant differences in the levels of Treg cells and the level of PD-L1 protein among the three types of ablation (p > 0.05), but the decline in Treg cells and PD-L1 protein level caused by CRYO was the most pronounced. CONCLUSION In the HCC mouse model, the immune changes following the three types of ablation were consistent, with immune changes occurring at 3-4 weeks. Among them, CRYO elicited the strongest adaptive immune response, and RFA outperformed MWA.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ming Liu
- Department of Interventional MRI Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Lin Cong
- Department of Interventional Ultrasound Shandong Provincial Hospital Affiliated to Shandong, First Medical University, Jinan, Shandong, China
| | - Xiangmeng He
- Department of Interventional MRI Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Yujun Xu
- Department of Interventional MRI Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Qingwen Wang
- Department of Medical Image Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Chengli Li
- Department of Interventional MRI Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
6
|
Marchenko ES, Dubovikov KM, Kuzhelivskiy II, Pleshkov MO, Koroluk ES, Brazovskii KS, Volinsky AA. In vivo study of porous NiTi cryotweezers for bone tissue cryotherapy. Cryobiology 2024; 115:104894. [PMID: 38614237 DOI: 10.1016/j.cryobiol.2024.104894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
This study examined the effects of liquid nitrogen vapor on osteogenesis in the rabbit femur. Cryotweezers made of porous nickel titanium alloy (nitinol or NiTi) obtained by self-propagating high temperature synthesis were used in this experiment. The porous structure of the cryotweezers allows them to hold up to 10 g of liquid nitrogen after being immersed for 2 min, which completely evaporates after 160 s. To study the effects of liquid nitrogen evaporation on osteogenesis, a rabbit femur was perforated. The formed holes were subjected to cryotherapy with varying exposure times. It was found that a 3 s exposure time stimulates osteogenesis, which was manifested in a greater number of osteoblasts in the regenerate compared to the control sample without liquid nitrogen. It was observed that increasing the exposure to 6, 9 or 12 s had a destructive effect, to varying degrees. The most severe damage was exerted by a 12 s exposure, which resulted in the formation of osteonecrosis areas. In the samples exposed to 6 and 9 s of cryotherapy, destruction of the cytoplasm of osteocytes and osteoclasts was observed.
Collapse
Affiliation(s)
- Ekaterina S Marchenko
- Laboratory of Superelastic Biointerfaces, National Research Tomsk State University, 36 Lenin Ave., 634045, Tomsk, Russia
| | - Kirill M Dubovikov
- Laboratory of Superelastic Biointerfaces, National Research Tomsk State University, 36 Lenin Ave., 634045, Tomsk, Russia
| | - Ivan I Kuzhelivskiy
- Department of Pediatric Surgical Diseases, Siberian State Medical University, 72 Olega Koshevogo Str., 634050, Tomsk, Russia
| | - Maksim O Pleshkov
- Laboratory "Bionic Digital Platforms", Siberian State Medical University, 2c7 Moskovsky Trakt, 634050, Tomsk, Russia
| | - Evgeniy S Koroluk
- Laboratory "Bionic Digital Platforms", Siberian State Medical University, 2c7 Moskovsky Trakt, 634050, Tomsk, Russia
| | - Konstantin S Brazovskii
- Division of Medical and Biological Cybernetics, Department of Biomedicine, Siberian State Medical University, 2 Moskovsky Trakt, 634050, Tomsk, Russia; Research School of Chemistry & Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Alex A Volinsky
- Laboratory of Superelastic Biointerfaces, National Research Tomsk State University, 36 Lenin Ave., 634045, Tomsk, Russia; Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave. ENG030, Tampa, FL, 33620, USA.
| |
Collapse
|
7
|
Baust JM, Corcoran A, Robilotto A, Katz A, Santucci K, Van Buskirk RG, Baust JG, Snyder KK. Evaluation of a New Cystoscopic Cryocatheter and Method for the In Situ Destruction of Bladder Cancer: Preliminary In Vivo Study. J Endourol 2024; 38:513-520. [PMID: 38279787 PMCID: PMC11057528 DOI: 10.1089/end.2023.0635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024] Open
Abstract
Purpose: To assess the ability to deliver full-thickness bladder wall cryoablation through a cystoscopic approach using a new closed-loop 6F cryocatheter and thermal dose-controlled protocol. Materials and Methods: Evaluations were conducted using a chronic porcine model wherein 10 lesions/animal were created throughout the bladder (bladder wall, trigone region, ureteral orifice, and distal ureter). A 6F cryocatheter was passed through the working channel of a flexible cystoscope. Single 1- and 1.5-minute freeze protocols in a saline environment were evaluated and resultant lesion size was determined. A laparoscopic approach was utilized to observe the transmural extension of the ice propagation. Results: Studies demonstrated the generation of transmural lesions characterized by full-thickness histologic necrosis after freezing for 1.5 minutes regardless of tissue thickness (range 2-12 mm). All animals were found to have good overall health (maintained weight, appetite, mobility, and energy levels) throughout the recovery period. No significant deviations were noted in complete blood count and serum chemistry bloodwork except for elevated creatine kinase levels. Importantly, no fistulas or perforations were noted. Conclusions: The cryocatheter was able to rapidly and effectively freeze the bladder wall through a cystoscopic approach. The results showed the ability to consistently ablate an ∼1 cm diameter and up to 1.2 cm deep using a single 1.5-minute freeze protocol. Analysis of the ablation efficacy revealed ∼80% destruction within the frozen mass. Although further testing and refinement are needed, these studies demonstrate the potential of this new approach to provide a next-generation strategy for the treatment of bladder cancer.
Collapse
Affiliation(s)
- John M. Baust
- CPSI Biotech, Research & Development, Owego, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| | - Anthony Corcoran
- Department of Urology, NYU Langone Hospital Long Island, Mineola, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| | - Anthony Robilotto
- CPSI Biotech, Research & Development, Owego, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| | - Aaron Katz
- Department of Urology, NYU Langone Hospital Long Island, Mineola, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| | - Kimberly Santucci
- CPSI Biotech, Research & Development, Owego, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| | - Robert G. Van Buskirk
- CPSI Biotech, Research & Development, Owego, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, New York, USA
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| | - John G. Baust
- Vesana, Inc., Research & Development, Owego, New York, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, New York, USA
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
| | - Kristi K. Snyder
- CPSI Biotech, Research & Development, Owego, New York, USA
- Vesana, Inc., Research & Development, Owego, New York, USA
| |
Collapse
|
8
|
Baust JM, Robilotto A, Raijman I, Santucci KL, Van Buskirk RG, Baust JG, Snyder KK. The Assessment of a Novel Endoscopic Ultrasound-Compatible Cryocatheter to Ablate Pancreatic Cancer. Biomedicines 2024; 12:507. [PMID: 38540120 PMCID: PMC10968037 DOI: 10.3390/biomedicines12030507] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 11/11/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease that may be treated utilizing thermal therapies. Cryoablation is an effective, minimally invasive therapy that has been utilized for the treatment of various cancers, offering patients a quicker recovery and reduced side effects. Cryoablation has been utilized on a limited basis for the treatment of PDAC. With the recent reports on the success of cryoablation, there is a growing interest in the use of cryoablation as a standalone, minimally invasive procedure to treat PDAC. While offering a promising path, the application of cryoablation to PDAC is limited by current technologies. As such, there is a need for the development of new devices to support advanced treatment strategies for PDAC. To this end, this study investigated the performance of a new endoscopic ultrasound-compatible cryoablation catheter technology, FrostBite. We hypothesized that FrostBite would enable the rapid, effective, minimally invasive delivery of ultra-cold temperatures to target tissues, resulting in effective ablation via an endoscopic approach. Thermal properties and ablative efficacy were evaluated using a heat-loaded gel model, tissue-engineered models (TEMs), and an initial in vivo porcine study. Freeze protocols evaluated included single and repeat 3 and 5 min applications. Isotherm assessment revealed the generation of a 2.2 cm diameter frozen mass with the -20 °C isotherm reaching a diameter of 1.5 cm following a single 5 min freeze. TEM studies revealed the achievement of temperatures ≤ -20 °C at a diameter of 1.9 cm after a 5 min freeze. Fluorescent imaging conducted 24 h post-thaw demonstrated a uniformly shaped ellipsoidal ablative zone with a midline diameter of 2.5 cm, resulting in a total ablative volume of 6.9 cm3 after a single 5 min freeze. In vivo findings consistently demonstrated the generation of ablative areas measuring 2.03 cm × 3.2 cm. These studies demonstrate the potential of the FrostBite cryocatheter as an endoscopic ultrasound-based treatment option. The data suggest that FrostBite may provide for the rapid, effective, controllable freezing of cancerous pancreatic and liver tissues. This ablative power also offers the potential of improved safety margins via the minimally invasive nature of an endoscopic ultrasound-based approach or natural orifice transluminal endoscopic surgery (NOTES)-based approach. The results of this pre-clinical feasibility study show promise, affirming the need for further investigation into the potential of the FrostBite cryocatheter as an advanced, minimally invasive cryoablative technology.
Collapse
Affiliation(s)
- John M. Baust
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| | | | - Isaac Raijman
- Department of Medicine-Gastroenterology, Baylor College of Medicine, Houston, TX 77030, USA
- GI Alliance, Houston, TX 77030, USA
| | | | - Robert G. Van Buskirk
- CPSI Biotech, Owego, NY 13827, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - John G. Baust
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Kristi K. Snyder
- CPSI Biotech, Owego, NY 13827, USA
- Phase Therapeutics, Inc., Owego, NY 13827, USA
| |
Collapse
|
9
|
Liu Q, Zhang C, Chen X, Han Z. Modern cancer therapy: cryoablation meets immune checkpoint blockade. Front Oncol 2024; 14:1323070. [PMID: 38384806 PMCID: PMC10881233 DOI: 10.3389/fonc.2024.1323070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cryoablation, as a minimally invasive technology for the treatment of tumors, destroys target tumors with lethal low temperatures. It simultaneously releases a large number of tumor-specific antigens, pro-inflammatory cytokines, and nucleoproteins, known as "danger signals", activating the body's innate and adaptive immune responses. However, tumor cells can promote the inactivation of immune effector cells by reprogramming immune checkpoints, leading to the insufficiency of these antigens to induce an immune response capable of eradicating the tumor. Immune checkpoint blockers rejuvenate exhausted T cells by blocking immune checkpoints that induce programmed death of T cells, and are therefore considered a promising therapeutic strategy to enhance the immune effects of cryoablation. In this review, we provide a detailed explanation of the immunological mechanisms of cryoablation and articulate the theoretical basis and research progress of the treatment of cancer with cryoablation combined with immune checkpoint blockers. Preliminary data indicates that this combined treatment strategy exhibits good synergy and has been proven to be safe and effective.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
10
|
Thai JN, Sevrukov AB, Ward RC, Monticciolo DL. Cryoablation Therapy for Early-Stage Breast Cancer: Evidence and Rationale. JOURNAL OF BREAST IMAGING 2023; 5:646-657. [PMID: 38141236 DOI: 10.1093/jbi/wbad064] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Indexed: 12/25/2023]
Abstract
Recent advances in breast cancer research and treatment propel a paradigm shift toward less aggressive and less invasive treatment for some early-stage breast cancer. Select patients with small, low-risk tumors may benefit from a less aggressive approach with de-escalated local therapy. Cryoablation of breast cancer is an emerging nonsurgical treatment alternative to breast-conserving surgery. Advantages of cryoablation over surgery include the use of local anesthesia, faster recovery, improved cosmesis, and cost savings. Proper patient selection and meticulous technique are keys to achieving successful clinical outcomes. The best candidates for cryoablation have unifocal invasive ductal carcinoma tumors that are low grade, hormone receptor positive, and ≤1.5 cm in size. In this review, we outline the current evidence, patient selection criteria, procedural technique, pre- and postablation imaging, and the advantages and limitations of cryoablation therapy.
Collapse
Affiliation(s)
- Janice N Thai
- Massachusetts General Hospital, Harvard Medical School, Department of Radiology, Division of Breast Imaging, Boston, MA, USA
| | - Alexander B Sevrukov
- Sidney Kimmel College of Medicine, Thomas Jefferson University, Department of Radiology, Division of Breast Imaging, Philadelphia, PA, USA
| | - Robert C Ward
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital and Women and Infants Hospital, Department of Diagnostic, Imaging, Division of Breast Imaging, Providence, RI, USA
| | - Debra L Monticciolo
- Massachusetts General Hospital, Harvard Medical School, Department of Radiology, Division of Breast Imaging, Boston, MA, USA
| |
Collapse
|
11
|
Lou W, Xie L, Xu L, Xu M, Xu F, Zhao Q, Jiang T. Present and future of metal nanoparticles in tumor ablation therapy. NANOSCALE 2023; 15:17698-17726. [PMID: 37917010 DOI: 10.1039/d3nr04362b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Cancer is an important factor affecting the quality of human life as well as causing death. Tumor ablation therapy is a minimally invasive local treatment modality with unique advantages in treating tumors that are difficult to remove surgically. However, due to its physical and chemical characteristics and the limitation of equipment technology, ablation therapy cannot completely kill all tumor tissues and cells at one time; moreover, it inevitably damages some normal tissues in the surrounding area during the ablation process. Therefore, this technology cannot be the first-line treatment for tumors at present. Metal nanoparticles themselves have good thermal and electrical conductivity and unique optical and magnetic properties. The combination of metal nanoparticles with tumor ablation technology, on the one hand, can enhance the killing and inhibiting effect of ablation technology on tumors by expanding the ablation range; on the other hand, the ablation technology changes the physicochemical microenvironment such as temperature, electric field, optics, oxygen content and pH in tumor tissues. It helps to stimulate the degree of local drug release of nanoparticles and increase the local content of anti-tumor drugs, thus forming a synergistic therapeutic effect with tumor ablation. Recent studies have found that some specific ablation methods will stimulate the body's immune response while physically killing tumor tissues, generating a large number of immune cells to cause secondary killing of tumor tissues and cells, and with the assistance of metal nanoparticles loaded with immune drugs, the effect of this anti-tumor immunotherapy can be further enhanced. Therefore, the combination of metal nanoparticles and ablative therapy has broad research potential. This review covers common metallic nanoparticles used for ablative therapy and discusses in detail their characteristics, mechanisms of action, potential challenges, and prospects in the field of ablation.
Collapse
Affiliation(s)
- Wenjing Lou
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
| | - Liting Xie
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
| | - Lei Xu
- Department of Ultrasound Medicine, Affiliated Jinhua Hospital Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Min Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
| | - Fan Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
| | - Qiyu Zhao
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
| | - Tianan Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 31000, P. R. China.
- Zhejiang University Cancer Center, Zhejiang, Hangzhou, China
| |
Collapse
|
12
|
Liao Y, Chen Y, Liu S, Wang W, Fu S, Wu J. Low-dose total body irradiation enhances systemic anti-tumor immunity induced by local cryotherapy. J Cancer Res Clin Oncol 2023; 149:10053-10063. [PMID: 37261526 DOI: 10.1007/s00432-023-04928-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Strategies that restore the immune system's ability to recognize malignant cells have yielded clinical benefits but only in some patients. Tumor cells survive cryotherapy and produce a vast amount of antigens to trigger innate and adaptive responses. However, because tumor cells have developed immune escape mechanisms, cryotherapy alone may not be enough to induce a significant immune response. METHODS The mice were randomly divided into four groups: Group A: low-dose total body irradiation combined with cryotherapy (L-TBI+cryo); Group B: cryotherapy (cryo); Group C: low-dose total body irradiation(L-TBI); Group D: control group (Control). The tumor growth, recurrence, and survival time of mice in each group were compared and the effects of different treatments on systemic anti-tumor immunity were explored. RESULTS L-TBI in conjunction with cryotherapy can effectively control tumor regrowth, inhibit tumor lung metastasis, extend the survival time of mice, and stimulate a long-term protective anti-tumor immune response to resist the re-challenge of tumor cells. The anti-tumor mechanism of this combination therapy may be related to the stimulation of inflammatory factors IFN-γ and IL-2, as well as an increase in immune effector cells (CD8+ T cells) and a decrease in immunosuppressive cells (MDSC, Treg cells) in the spleen or tumor tissue. CONCLUSIONS We present unique treatment options for enhancing the immune response caused by cryotherapy, pointing to the way forward for cancer treatment.
Collapse
Affiliation(s)
- Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Weizhou Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China.
| |
Collapse
|
13
|
Argirò R, Ciccarese G, Vattermoli L, Crociati S, Iannibelli V, Forte V, Morosetti D, Floris R. Combined treatments with microballoon catheters and multiple cryoablation probes for shoulder-subclavian soft tissue hemangiopericytoma: A case report. Radiol Case Rep 2023; 18:3346-3350. [PMID: 37520395 PMCID: PMC10372045 DOI: 10.1016/j.radcr.2023.06.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
We describe a case of a 65-year-old woman affected by hemangiopericytoma/solitary fibrous tumor of the right shoulder-subclavian region. Hemangiopericytoma/solitary fibrous tumor is a rare tumor of uncertain malignancy. She reports shoulder pain and inability to abduct the arm and elevate the shoulder. Imaging showed erosion of the scapula. The patient underwent 5 sessions of "on demand" embolization in the previous 2 years scheduled for recurrence of symptoms-swelling of tissues. Further 2 treatments were achieved through embolization via 2 different microballoon catheter combined with percutaneous cryoablation with 5 probes. Images after the treatment demonstrate a marked reduction in the hypervascularized area and an increase in the necrosis area. So, this combined treatment is safety and reproducible also in extrahepatic tissue.
Collapse
Affiliation(s)
- Renato Argirò
- Interventional Radiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, Rome, 00133, Italy
| | - Giorgio Ciccarese
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| | - Leonardo Vattermoli
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| | - Sara Crociati
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| | - Vincenzo Iannibelli
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| | - Valentina Forte
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| | - Daniele Morosetti
- Interventional Radiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, Rome, 00133, Italy
| | - Roberto Floris
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, Rome, 00133, Italy
- Neurology Unit, Department of Neurology, University of Rome Tor Vergata, Viale Oxford 81, Rome, 00133, Italy
| |
Collapse
|
14
|
Alshebremi M, Tomchuck SL, Myers JT, Kingsley DT, Eid S, Abiff M, Bonner M, Saab ST, Choi SH, Huang AYC. Functional tumor cell-intrinsic STING, not host STING, drives local and systemic antitumor immunity and therapy efficacy following cryoablation. J Immunother Cancer 2023; 11:e006608. [PMID: 37553183 PMCID: PMC10414127 DOI: 10.1136/jitc-2022-006608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Despite its potential utility in delivering direct tumor killing and in situ whole-cell tumor vaccination, tumor cryoablation produces highly variable and unpredictable clinical response, limiting its clinical utility. The mechanism(s) driving cryoablation-induced local antitumor immunity and the associated abscopal effect is not well understood. METHODS The aim of this study was to identify and explore a mechanism of action by which cryoablation enhances the therapeutic efficacy in metastatic tumor models. We used the subcutaneous mouse model of the rhabdomyosarcoma (RMS) cell lines RMS 76-9STINGwt or RMS 76-9STING-/-, along with other murine tumor models, in C57BL/6 or STING-/- (TMEM173-/- ) mice to evaluate local tumor changes, lung metastasis, abscopal effect on distant tumors, and immune cell dynamics in the tumor microenvironment (TME). RESULTS The results show that cryoablation efficacy is dependent on both adaptive immunity and the STING signaling pathway. Contrary to current literature dictating an essential role of host-derived STING activation as a driver of antitumor immunity in vivo, we show that local tumor control, lung metastasis, and the abscopal effect on distant tumor are all critically dependent on a functioning tumor cell-intrinsic STING signaling pathway, which induces inflammatory chemokine and cytokine responses in the cryoablated TME. This reliance extends beyond cryoablation to include intratumoral STING agonist therapy. Additionally, surveys of gene expression databases and tissue microarrays of clinical tumor samples revealed a wide spectrum of expressions among STING-related signaling components. CONCLUSIONS Tumor cell-intrinsic STING pathway is a critical component underlying the effectiveness of cryoablation and suggests that expression of STING-related signaling components may serve as a potential therapy response biomarker. Our data also highlight an urgent need to further characterize tumor cell-intrinsic STING pathways and the associated downstream inflammatory response evoked by cryoablation and other STING-dependent therapy approaches.
Collapse
Affiliation(s)
- Mohammad Alshebremi
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Suzanne L Tomchuck
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Jay T Myers
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daniel T Kingsley
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Saada Eid
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Muta Abiff
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Melissa Bonner
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Shahrazad T Saab
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Sung Hee Choi
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Alex Yee-Chen Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Center for Pediatric Immunotherapy, Angie Fowler AYA Cancer Institute, UH Rainbow Babies & Children's Hospital, Cleveland, Ohio, USA
| |
Collapse
|
15
|
Young S, Hannallah J, Goldberg D, Sanghvi T, Arshad J, Scott A, Woodhead G. Friend or Foe? Locoregional Therapies and Immunotherapies in the Current Hepatocellular Treatment Landscape. Int J Mol Sci 2023; 24:11434. [PMID: 37511193 PMCID: PMC10380625 DOI: 10.3390/ijms241411434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Over the last several decades, a number of new treatment options for patients with hepatocellular carcinoma (HCC) have been developed. While treatment decisions for some patients remain clear cut, a large numbers of patients have multiple treatment options, and it can be hard for multidisciplinary teams to come to unanimous decisions on which treatment strategy or sequence of treatments is best. This article reviews the available data with regard to two treatment strategies, immunotherapies and locoregional therapies, with a focus on the potential of locoregional therapies to be combined with checkpoint inhibitors to improve outcomes in patients with locally advanced HCC. In this review, the available data on the immunomodulatory effects of locoregional therapies is discussed along with available clinical data on outcomes when the two strategies are combined.
Collapse
Affiliation(s)
- Shamar Young
- Department of Medical Imaging, Division of Interventional Radiology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, USA
| | - Jack Hannallah
- Department of Medical Imaging, Division of Interventional Radiology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, USA
| | - Dan Goldberg
- Department of Medical Imaging, Division of Interventional Radiology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, USA
| | - Tina Sanghvi
- Department of Radiology, Southern Arizona VA, Tucson, AZ 85723, USA
| | - Junaid Arshad
- Department of Medicine, Division of Hematology and Oncology, University of Arizona, Tucson, AZ 85724, USA
| | - Aaron Scott
- Department of Medicine, Division of Hematology and Oncology, University of Arizona, Tucson, AZ 85724, USA
| | - Gregory Woodhead
- Department of Medical Imaging, Division of Interventional Radiology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, USA
| |
Collapse
|
16
|
Dulu A, Tayban Y, Delaleu J, Cornelis FH, Pastores SM. Disseminated intravascular coagulation after cryoablation for metastatic pancreatic cancer: a case report. AME MEDICAL JOURNAL 2023; 8:17. [PMID: 38827122 PMCID: PMC11141771 DOI: 10.21037/amj-23-13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Background Pancreatic cancer is the fourth most common cause of cancer-related death in the United States. Despite advancements in surgery and chemoradiation therapies, pancreatic cancer has a 5-year survival rate of only 11% in the United States. Cryoablation is emerging as a new and effective therapy for locally advanced pancreatic cancer and symptom palliation in metastatic disease. To our knowledge, the occurrence of disseminated intravascular coagulation (DIC) after cryoablation is rare. Case Description A 47-year-old woman with no significant past medical history was diagnosed with pancreatic cancer and underwent a Whipple procedure followed by chemotherapy with gemcitabine and paclitaxel. Due to the abdominal lymph nodes, peritoneum, right femur, and surrounding soft tissue metastases, she received systemic palliative chemotherapy with gemcitabine and paclitaxel and underwent right femur tumor excision, open reduction, and internal fixation, followed by radiation therapy. She continued to have persistent pain and underwent palliative percutaneous cryoablation of the metastatic tumor under computed tomography (CT) and ultrasound guidance. Immediately post procedure, she developed slow but continuous blood oozing at the ablation site, which was difficult to control despite compression dressings, reinforcement sutures, and local thrombin powder. The patient was transferred to the intensive care unit where she was noted to be hypotensive and tachycardic, with petechiae in both lower extremities. Laboratory studies were consistent with DIC and peripheral blood smear revealed multiple schistocytes. CT angiogram of the right lower extremity did not show any bleeding vessel amenable to embolization. She was transfused red blood cells, platelets, fresh frozen plasma, and cryoprecipitate. Despite multiple daily transfusions, she continued to have pain and remained persistently thrombocytopenic and coagulopathic. After discussion with the patient and her family, she chose to transition to comfort care measures and died. Conclusions DIC is an unusual but life-threatening complication of advanced pancreatic cancer.
Collapse
Affiliation(s)
- Alina Dulu
- Critical Care Center, Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yekaterina Tayban
- Critical Care Center, Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joanne Delaleu
- Critical Care Center, Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francois H. Cornelis
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen M. Pastores
- Critical Care Center, Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
17
|
Wang Y, Wang GZ, Chen C, Huang HZ, Wang YH, He XH, Xu LX, Xu LC, Li WT. Exploration of the impact of multimode thermal therapy versus radiofrequency ablation on CD8 + T effector cells of liver malignancies based on single cell transcriptomics. Front Immunol 2023; 14:1172362. [PMID: 37334386 PMCID: PMC10272448 DOI: 10.3389/fimmu.2023.1172362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Multimode thermal therapy (MTT) is an innovative interventional therapy developed for the treatment of liver malignancies. When compared to the conventional radiofrequency ablation (RFA), MTT typically offers improved prognosis for patients. However, the effect of MTT on the peripheral immune environment and the mechanisms underlying the enhanced prognosis have yet to be explored. The aim of this study was to further investigate the mechanisms responsible for the difference in prognosis between the two therapies. Methods In this study, peripheral blood samples were collected from four patients treated with MTT and two patients treated with RFA for liver malignancies at different time points before and after the treatment. Single cell sequencing was performed on the blood samples to compare and analyze the activation pathways of peripheral immune cells following the MTT and RFA treatment. Results There was no significant effect of either therapy on the composition of immune cells in peripheral blood. However, the differential gene expression and pathway enrichment analysis demonstrated enhanced activation of T cells in the MTT group compared to the RFA group. In particular, there was a remarkable increase in TNF-α signaling via NF-κB, as well as the expression of IFN-α and IFN-γ in the CD8+ effector T (CD8+ Teff) cells subpopulation, when compared to the RFA group. This may be related to the upregulation of PI3KR1 expression after MTT, which promotes the activation of PI3K-AKT-mTOR pathway. Conclusion This study confirmed that MTT could more effectively activate peripheral CD8+ Teff cells in patients compared with RFA and promote the effector function, thus resulting in a better prognosis. These results provide a theoretical basis for the clinical application of MTT therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guang-Zhi Wang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Department of Medical Imaging Center, Affiliated Hospital, Weifang Medical University, Weifang, Shandong, China
| | - Chao Chen
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao-Zhe Huang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yao-Hui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin-Hong He
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lisa X. Xu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Chao Xu
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wen-Tao Li
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
18
|
Drogin J, Abdelbaki A, Lendermon J, Pereira K, Mendez-Castillo A. Percutaneous Image-Guided Cryoablation of Metastatic Colorectal Adenocarcinoma to the Abdominal Wall. THE ARAB JOURNAL OF INTERVENTIONAL RADIOLOGY 2023. [DOI: 10.1055/s-0043-1763497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
AbstractAbdominal wall metastases are rarely seen in primary colorectal cancer; the majority of cases are attributed to seeding from previous procedures. We report a case of percutaneous cryoablation of metastatic colorectal cancer to the abdominal wall with no evidence of recurrence 1 year later.
Collapse
Affiliation(s)
- Jillian Drogin
- Department of Interventional Radiology, Saint Louis University, Saint Louis, Missouri, United States
| | - Ahmed Abdelbaki
- Department of Interventional Radiology, Saint Louis University, Saint Louis, Missouri, United States
| | - Justin Lendermon
- Department of Interventional Radiology, Saint Louis University, Saint Louis, Missouri, United States
| | - Keith Pereira
- Department of Interventional Radiology, Saint Louis University, Saint Louis, Missouri, United States
| | - Alejandro Mendez-Castillo
- Department of Interventional Radiology, Saint Louis University, Saint Louis, Missouri, United States
| |
Collapse
|
19
|
McDaniel A, Freimark B, Navarro C, Von Rothstein K, Gonzalez D, Linder K, Nuccitelli R. Nano-pulse stimulation™ therapy (NPS™) is superior to cryoablation in clearing murine melanoma tumors. Front Oncol 2023; 12:948472. [PMID: 36844920 PMCID: PMC9945337 DOI: 10.3389/fonc.2022.948472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/30/2022] [Indexed: 02/11/2023] Open
Abstract
Background Nano-Pulse Stimulation™ Therapy (NPS™) is a new, bioelectric modality that applies ultrashort pulses of electric energy to trigger regulated cell death in treated tissues. Instead of initiating necrosis by heating or freezing, NPS therapy permeabilizes intracellular organelles to activate the cell's own self-destruct pathway of programmed or regulated cell death. Unlike cryotherapies that can both damage structural tissues and diffuse into the periphery beyond the margins of the lesion, NPS only affects cells within the treated zone leaving surrounding tissue and acellular components unaffected. Methods We generated melanoma tumors in mice by injecting B16-F10 cells intradermally and compared the efficacy and resulting skin damage from Nano-Pulse Stimulation Therapy with that of cryoablation in clearing these tumors. Results The results of the study demonstrate that NPS is superior at clearing B16-F10 melanoma lesions. NPS permanently eliminated up to 91% of all tumor lesions with a single treatment compared to cryoablation that only eliminated up to 66%. Importantly, NPS permanently eliminated these lesions with no recurrence and with minimal dermal fibrosis, underlying muscle atrophy, permanent hair follicle loss or other markers of permanent skin damage. Conclusions These findings suggest that NPS is a promising new modality for the clearance of melanoma tumors and is a more efficacious, less damaging approach than cryoablative methods for the treatment of aggressive malignant tumors.
Collapse
Affiliation(s)
- Amanda McDaniel
- Department of Biology, Pulse Biosciences, Hayward, CA, United States,*Correspondence: Amanda McDaniel, ; Richard Nuccitelli,
| | - Bruce Freimark
- Department of Biology, Pulse Biosciences, Hayward, CA, United States
| | - Cebrina Navarro
- Department of Biology, Pulse Biosciences, Hayward, CA, United States
| | | | - Dacia Gonzalez
- Department of Biology, Pulse Biosciences, Hayward, CA, United States
| | - Keith Linder
- Department of Dermatopathology, Linder Pathology Services, Raleigh, NC, United States
| | - Richard Nuccitelli
- Department of Biology, Pulse Biosciences, Hayward, CA, United States,*Correspondence: Amanda McDaniel, ; Richard Nuccitelli,
| |
Collapse
|
20
|
3D-Printed Biomaterial Testing in Response to Cryoablation: Implications for Surgical Ventricular Tachycardia Ablation. J Clin Med 2023; 12:jcm12031036. [PMID: 36769681 PMCID: PMC9918061 DOI: 10.3390/jcm12031036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/22/2022] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Background: The lack of thermally and mechanically performant biomaterials represents the major limit for 3D-printed surgical guides, aimed at facilitating complex surgery and ablations. Methods: Cryosurgery is a treatment for cardiac arrhythmias. It consists of obtaining cryolesions, by freezing the target tissue, resulting in selective and irreversible damage. MED625FLX and TPU95A are two biocompatible materials for surgical guides; however, there are no data on their response to cryoenergy delivery. The study purpose is to evaluate the biomaterials' thermal properties, examining the temperature changes on the porcine muscle samples (PMS) when the biomaterials are in place during the cryoablation. Two biomaterials were selected, MED625FLX and TPU95A, with two thicknesses (1.0 and 2.5 mm). To analyze the biomaterials' behavior, the PMS temperatures were measured during cryoablation, firstly without biomaterials (control) and after with the biomaterials in place. To verify the biomaterials' suitability, the temperatures under the biomaterial samples should not exceed a limit of -30.0 °C. Furthermore, the biomaterials' geometry after cryoablation was evaluated using the grid paper test. Results: TPU95A (1.0 and 2.5 mm) successfully passed all tests, making this material suitable for cryoablation treatment. MED625FLX of 1.0 mm did not retain its shape, losing its function according to the grid paper test. Further, MED625FLX of 2.5 mm is also suitable for use with a cryoenergy source. Conclusions: TPU95A (1.0 and 2.5 mm) and MED625FLX of 2.5 mm could be used in the design of surgical guides for cryoablation treatment, because of their mechanical, geometrical, and thermal properties. The positive results from the thermal tests on these materials and their thickness prompt further clinical investigation.
Collapse
|
21
|
Chen Z, Meng L, Zhang J, Zhang X. Progress in the cryoablation and cryoimmunotherapy for tumor. Front Immunol 2023; 14:1094009. [PMID: 36761748 PMCID: PMC9907027 DOI: 10.3389/fimmu.2023.1094009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
With the rapid advancement of imaging equipment and minimally invasive technology, cryoablation technology is being used more frequently in minimally invasive treatment of tumors, primarily for patients with early tumors who voluntarily consent to ablation as well as those with advanced tumors that cannot be surgically removed or cannot be tolerated. Cryoablation is more effective and secure for target lesions than other thermal ablation methods like microwave and radiofrequency ablation (RFA). The study also discovered that cryoablation, in addition to causing tumor tissue necrosis and apoptosis, can facilitate the release of tumor-derived autoantigens into the bloodstream and activate the host immune system to elicit beneficial anti-tumor immunological responses against primary. This may result in regression of the primary tumor and distant metastasis. The additional effect called " Accompanying effects ". It is the basis of combined ablation and immunotherapy for tumor. At present, there is a lot of research on the mechanism of immune response induced by cryoablation. Trying to solve the question: how positively induce immune response. In this review, we focus on: 1. the immune effects induced by cryoablation. 2. the effect and mechanism of tumor immunotherapy combined with cryoablation. 3.The clinical research of this combination therapy in the treatment of tumors.
Collapse
Affiliation(s)
- Zenan Chen
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Liangliang Meng
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Radiology, Chinese People's Armed Police (PAP) Force Hospital of Beijing, Beijing, China
| | - Jing Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiao Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
22
|
Pal K, Sheth RA. Engineering the Tumor Immune Microenvironment through Minimally Invasive Interventions. Cancers (Basel) 2022; 15:196. [PMID: 36612192 PMCID: PMC9818918 DOI: 10.3390/cancers15010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is a unique landscape that poses several physical, biochemical, and immune barriers to anti-cancer therapies. The rapidly evolving field of immuno-engineering provides new opportunities to dismantle the tumor immune microenvironment by efficient tumor destruction. Systemic delivery of such treatments can often have limited local effects, leading to unwanted offsite effects such as systemic toxicity and tumor resistance. Interventional radiologists use contemporary image-guided techniques to locally deliver these therapies to modulate the immunosuppressive TME, further accelerating tumor death and invoking a better anti-tumor response. These involve local therapies such as intratumoral drug delivery, nanorobots, nanoparticles, and implantable microdevices. Physical therapies such as photodynamic therapy, electroporation, hyperthermia, hypothermia, ultrasound therapy, histotripsy, and radiotherapy are also available for local tumor destruction. While the interventional radiologist can only locally manipulate the TME, there are systemic offsite recruitments of the immune response. This is known as the abscopal effect, which leads to more significant anti-tumoral downstream effects. Local delivery of modern immunoengineering methods such as locoregional CAR-T therapy combined with immune checkpoint inhibitors efficaciously modulates the immunosuppressive TME. This review highlights the various advances and technologies available now to change the TME and revolutionize oncology from a minimally invasive viewpoint.
Collapse
Affiliation(s)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
23
|
Zhang C, Zhu L, Tang S, Wang J, Li Y, Chen X, Bian C, Liu D, Ao G, Luo T. Feasibility and safety of trans-biliary cryoablation: Preclinical evaluation of a novel flexible cryoprobe. Cryobiology 2022:S0011-2240(22)00354-6. [PMID: 36414042 DOI: 10.1016/j.cryobiol.2022.11.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Cryoablation, as a well-characterized technology, has multifarious clinical applications in solid malignancy. However, trans-biliary cryoablation for malignant biliary obstruction has not been reported yet. Thus, this study aimed to determine the efficacy and safety of trans-biliary cryoablation with a novel CO2 gas-based flexible cryoprobe in standardized preclinical settings. For fresh porcine liver ex vivo, the freezing efficacy of cryoablation was evaluated by using fresh porcine liver. The real-time CO2 flow rate, freezing temperature and freezing range were examined and the frozen appearance was visualized. In vivo study, acute and chronical effects were investigated by using the models of canine bile duct. Histopathology and laboratory examination were performed. The lowest temperature that the electrode could deliver to the tissue was -60.7 °C. At 60s after freezing, the tissue temperature dropped to -22.6 °C and -4.3 °C at 0.1 and 0.2 cm from the electrode center, respectively. The frozen size was greater in liver tissue ex vivo than that in bile duct tissue in vivo. No biliary hemorrhage, perforation, stricture, obstruction, and adjacent organ injury were observed. With histopathologic examination, acute intercellular vacuoles were observed in the lamina propria adjacent to the lumen. Chronic changes, including uneven coagulative necrosis, fibro-proliferation, inflammatory infiltration and connective tissue thickening were observed in the lamina propria of the all biliary samples. The results demonstrated CO2 gas-based trans-biliary cryoablation is safe and efficacious. These findings may provide a potential new modality for primary malignant biliary obstruction and malignant obstruction within a biliary stent and contribute to cryoablation of clinical practice.
Collapse
Affiliation(s)
- Chao Zhang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Linzhong Zhu
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Shousheng Tang
- CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100080, 100091, PR China
| | - Jukun Wang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Yu Li
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Xin Chen
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Chunjing Bian
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Dongbin Liu
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Guokun Ao
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, 100053, PR China.
| |
Collapse
|
24
|
Yin L, Li XY, Zhu LL, Chen GL, Xiang Z, Wang QQ, Bi JW, Wang Q. Clinical application status and prospect of the combined anti-tumor strategy of ablation and immunotherapy. Front Immunol 2022; 13:965120. [PMID: 36131929 PMCID: PMC9483102 DOI: 10.3389/fimmu.2022.965120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Image-guided tumor ablation eliminates tumor cells by physical or chemical stimulation, which shows less invasive and more precise in local tumor treatment. Tumor ablation provides a treatment option for medically inoperable patients. Currently, clinical ablation techniques are widely used in clinical practice, including cryoablation, radiofrequency ablation (RFA), and microwave ablation (MWA). Previous clinical studies indicated that ablation treatment activated immune responses besides killing tumor cells directly, such as short-term anti-tumor response, immunosuppression reduction, specific and non-specific immune enhancement, and the reduction or disappearance of distant tumor foci. However, tumor ablation transiently induced immune response. The combination of ablation and immunotherapy is expected to achieve better therapeutic results in clinical application. In this paper, we provided a summary of the principle, clinical application status, and immune effects of tumor ablation technologies for tumor treatment. Moreover, we discussed the clinical application of different combination of ablation techniques with immunotherapy and proposed possible solutions for the challenges encountered by combined therapy. It is hoped to provide a new idea and reference for the clinical application of combinate treatment of tumor ablation and immunotherapy.
Collapse
Affiliation(s)
- Li Yin
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Xing-yu Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lin-lin Zhu
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Gui-lai Chen
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Zhuo Xiang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qing-qing Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Jing-wang Bi
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
| | - Qiang Wang
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, China
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- *Correspondence: Qiang Wang,
| |
Collapse
|
25
|
Mikhail M, Chua KJ, Khizir L, Tabakin A, Singer EA. Role of metastasectomy in the management of renal cell carcinoma. Front Surg 2022; 9:943604. [PMID: 35965871 PMCID: PMC9372304 DOI: 10.3389/fsurg.2022.943604] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022] Open
Abstract
Treatment of metastatic renal cell carcinoma (mRCC) has evolved with the development of a variety of systemic agents; however, these therapies alone rarely lead to a complete response. Complete consolidative surgery with surgical metastasectomy has been associated with improved survival outcomes in well-selected patients in previous reports. No randomized control trial exists to determine the effectiveness of metastasectomy. Therefore, reviewing observational studies is important to best determine which patients are most appropriate for metastasectomy for mRCC and if such treatment continues to be effective with the development of new systemic therapies such as immunotherapy. In this narrative review, we discuss the indications for metastasectomies, outcomes, factors associated with improved survival, and special considerations such as location of metastasis, number of metastases, synchronous metastases, and use of systemic therapy. Additionally, alternative treatment options and trials involving metastasectomy will be reviewed.
Collapse
Affiliation(s)
| | | | | | | | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
26
|
Pusceddu C, Mascia L, Ninniri C, Ballicu N, Zedda S, Melis L, Deiana G, Porcu A, Fancellu A. The Increasing Role of CT-Guided Cryoablation for the Treatment of Liver Cancer: A Single-Center Report. Cancers (Basel) 2022; 14:cancers14123018. [PMID: 35740682 PMCID: PMC9221264 DOI: 10.3390/cancers14123018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/06/2022] [Accepted: 06/14/2022] [Indexed: 02/06/2023] Open
Abstract
Purpose: Cryoablation (CrA) is a minimally invasive treatment that can be used in primary and metastatic liver cancer. The purpose of this study was to assess the effectiveness of CrA in patients with hepatocellular carcinoma (HCC) and liver metastases. Methods: We retrospectively evaluated the patients who had CrA for HCC or liver metastases between 2015 and 2020. Technical success, complete ablation, CrA-related complications, local tumor progression, local recurrences, and distant metastases were evaluated in the study population. In patients with HCC, the median survival was also estimated. Results: Sixty-four liver tumors in 49 patients were treated with CrA (50 metastases and 14 HCC). The mean tumor diameter was 2.15 cm. The mean follow-up was 19.8 months. Technical success was achieved in the whole study population. Complete tumor ablation was observed after one month in 92% of lesions treated with CrA (79% and 96% in the HCC Group and metastases Group, respectively, p < 0.001). Local tumor progression occurred in 12.5 of lesions, with no difference between the study groups (p = 0.105). Sixteen patients (33%) developed local recurrence (45% and 29% in the HCC Group and metastases Group, respectively, p = 0.477). Seven patients (14%) developed distant metastases in the follow-up period. Ten patients (20.8%) underwent redo CrA for local recurrence or incomplete tumor ablation. Minor complications were observed in 14% of patients. In patients with HCC, the median survival was 22 months. Conclusions: CrA can be safely used for treatment of HCC and liver metastases not amenable of surgical resection. Further studies are necessary to better define the role of CrA in the multidisciplinary treatment of liver malignancies.
Collapse
Affiliation(s)
- Claudio Pusceddu
- Department of Oncological and Interventional Radiology, Oncological Hospital A, Businco, I-09121 Cagliari, Italy; (C.P.); (N.B.); (S.Z.); (L.M.)
| | - Luigi Mascia
- Department of Medical Oncology, Oncological Hospital A, Businco, I-09121 Cagliari, Italy;
| | - Chiara Ninniri
- Unit of General Surgery 2, Clinica Chirurgica, Department of Medical, Surgical, and Experimental Sciences, University of Sassari, I-07100 Sassari, Italy; (C.N.); (G.D.); (A.P.)
| | - Nicola Ballicu
- Department of Oncological and Interventional Radiology, Oncological Hospital A, Businco, I-09121 Cagliari, Italy; (C.P.); (N.B.); (S.Z.); (L.M.)
| | - Stefano Zedda
- Department of Oncological and Interventional Radiology, Oncological Hospital A, Businco, I-09121 Cagliari, Italy; (C.P.); (N.B.); (S.Z.); (L.M.)
| | - Luca Melis
- Department of Oncological and Interventional Radiology, Oncological Hospital A, Businco, I-09121 Cagliari, Italy; (C.P.); (N.B.); (S.Z.); (L.M.)
| | - Giulia Deiana
- Unit of General Surgery 2, Clinica Chirurgica, Department of Medical, Surgical, and Experimental Sciences, University of Sassari, I-07100 Sassari, Italy; (C.N.); (G.D.); (A.P.)
| | - Alberto Porcu
- Unit of General Surgery 2, Clinica Chirurgica, Department of Medical, Surgical, and Experimental Sciences, University of Sassari, I-07100 Sassari, Italy; (C.N.); (G.D.); (A.P.)
| | - Alessandro Fancellu
- Unit of General Surgery 2, Clinica Chirurgica, Department of Medical, Surgical, and Experimental Sciences, University of Sassari, I-07100 Sassari, Italy; (C.N.); (G.D.); (A.P.)
- Correspondence: ; Tel.: +39-079-228432; Fax: +39-079-228394
| |
Collapse
|
27
|
An In Vitro Investigation into Cryoablation and Adjunctive Cryoablation/Chemotherapy Combination Therapy for the Treatment of Pancreatic Cancer Using the PANC-1 Cell Line. Biomedicines 2022; 10:biomedicines10020450. [PMID: 35203660 PMCID: PMC8962332 DOI: 10.3390/biomedicines10020450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
As the incidence of pancreatic ductal adenocarcinoma (PDAC) continues to grow, so does the need for new strategies for treatment. One such area being evaluated is cryoablation. While promising, studies remain limited and questions surrounding basic dosing (minimal lethal temperature) coupled with technological issues associated with accessing PDAC tumors and tumor proximity to vasculature and bile ducts, among others, have limited the use of cryoablation. Additionally, as chemotherapy remains the first-line of attack for PDAC, there is limited information on the impact of combining freezing with chemotherapy. As such, this study investigated the in vitro response of a PDAC cell line to freezing, chemotherapy, and the combination of chemotherapy pre-treatment and freezing. PANC-1 cells and PANC-1 tumor models were exposed to cryoablation (freezing insult) and compared to non-frozen controls. Additionally, PANC-1 cells were exposed to varying sub-clinical doses of gemcitabine or oxaliplatin alone and in combination with freezing. The results show that freezing to −10 °C did not affect viability, whereas −15 °C and −20 °C resulted in a reduction in 1 day post-freeze viability to 85% and 20%, respectively, though both recovered to controls by day 7. A complete cell loss was found following a single freeze below −25 °C. The combination of 100 nM gemcitabine (1.1 mg/m2) pre-treatment and a single freeze at −15 °C resulted in near-complete cell death (<5% survival) over the 7-day assessment interval. The combination of 8.8 µM oxaliplatin (130 mg/m2) pre-treatment and a single −15 °C freeze resulted in a similar trend of increased PANC-1 cell death. In summary, these in vitro results suggest that freezing alone to temperatures in the range of −25 °C results in a high degree of PDAC destruction. Further, the data support a potential combinatorial chemo/cryo-therapeutic strategy for the treatment of PDAC. These results suggest that a reduction in chemotherapeutic dose may be possible when offered in combination with freezing for the treatment of PDAC.
Collapse
|
28
|
Masroor S. Can Epicardial Ablation Create Optimal Cryolesions. Ann Thorac Surg 2021; 114:1522-1523. [PMID: 34883084 DOI: 10.1016/j.athoracsur.2021.10.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Saqib Masroor
- University of Toledo Medical Center, 3000 Arlington Road, Toledo, OH, 43614.
| |
Collapse
|
29
|
An Exploratory Analysis of Changes in Circulating Plasma Protein Profiles Following Image-Guided Ablation of Renal Tumours Provides Evidence for Effects on Multiple Biological Processes. Cancers (Basel) 2021; 13:cancers13236037. [PMID: 34885149 PMCID: PMC8656737 DOI: 10.3390/cancers13236037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 01/20/2023] Open
Abstract
Simple Summary Ablation techniques use extremely hot or cold temperatures to kill small cancers. It is now known that in addition to killing the cancer cells, the ablation treatment may stimulate an immune response in patients against the cancer cells, acting like a vaccine. As a result, there is now interest in combining ablation together with drugs that target the immune system of patients with cancer to enhance the effects of both treatments. In order to develop such combined treatments and test them in clinical trials, we need to understand more about their effects so trials can be optimally designed to be most effective. We have analysed 164 circulating proteins in the blood from patients with small renal tumours undergoing ablation treatment to understand more about the effects of ablation on the patient, both at the level of the effects on the cancer cells and the response of the patients. Abstract Further biological understanding of the immune and inflammatory responses following ablation is critical to the rational development of combination ablation-immunotherapies. Our pilot exploratory study evaluated the circulating plasma protein profiles after image-guided ablation (IGA) of small renal masses to determine the resultant systemic effects and provide insight into impact both on the tumour and immune system. Patients undergoing cryotherapy (CRYO), radiofrequency ablation (RFA) or microwave ablation (MWA) for small renal tumours were recruited. Blood samples were obtained at four timepoints; two baselines prior to IGA and at 24 h and 1–3 months post-IGA, and a panel of 164 proteins measured. Of 55 patients recruited, 35 underwent ablation (25 CRYO, 8 RFA, 2 MWA) and biomarker measurements. The most marked changes were 24 h post-CRYO, with 29 proteins increasing and 18 decreasing significantly, principally cytokines and proteins involved in regulating inflammation, danger-associated molecular patterns (DAMPs), cell proliferation, hypoxic response, apoptosis and migration. Intra-individual variation was low but inter-individual variation was apparent, for example all patients showed increases in IL-6 (1.7 to 29-fold) but only 50% in CD27. Functional annotation analysis highlighted immune/inflammation and cell proliferation/angiogenesis-related clusters, with interaction networks around IL-6, IL-10, VEGF-A and several chemokines. Increases in IL-8, IL-6, and CCL23 correlated with cryoprobe number (p = 0.01, rs = 0.546; p = 0.009, rs = 0.5515; p = 0.005, rs = 0.5873, respectively). This initial data provide further insights into ablation-induced biological changes of relevance in informing trial design of immunotherapies combined with ablation.
Collapse
|
30
|
Heger U, Hackert T. Can local ablative techniques replace surgery for locally advanced pancreatic cancer? J Gastrointest Oncol 2021; 12:2536-2546. [PMID: 34790414 DOI: 10.21037/jgo-20-379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/04/2020] [Indexed: 12/25/2022] Open
Abstract
In the treatment of pancreatic ductal adenocarcinoma (PDAC) the best chance at long term survival or cure has to date always included the complete surgical removal of the tumor. However, locally advanced pancreatic cancer (LAPC), about 25% of all newly diagnosed PDAC, is defined by its primary technical unresectability due to infiltration of visceral arteries and absence of metastasis. Induction therapies, especially FOLFIRINOX treatment, together with technical surgical advancement have increased the numbers for conversion to secondary resectability. Recent data on resections after induction therapy show promising, almost doubled survival compared to palliative treatment. Yet, around 70% of LAPC remain unresectable after induction therapy, often due to persistent local invasion. As locally ablative techniques are becoming more widely available this review examines their possible applicability to substitute for surgery in these cases which we propose to group under the new term "Inconvertible LAPC". The need for defining this novel subgroup who might benefit from ablative treatment is based on the findings in our review that high-level evidence on ablative techniques for PDAC is largely lacking and the latest effective, harmonized treatment guidelines for LAPC are not often incorporated in these studies. The "inconvertible LAPC" label requires persistent unresectability after staging and induction therapy of LAPC according to current guidelines followed by liberal indication for aggressive surgical exploration at a center equipped for extended pancreatic resections. Ideally, this specification of a new, distinct patient group will also put it in the spotlight more, hopefully prompt more trials designed to generate robust evidence and optimize transferability of study results.
Collapse
Affiliation(s)
- Ulrike Heger
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
31
|
Lopez O, Chevallier O, Guillen K, Comby PO, Pellegrinelli J, Tinel C, Falvo N, Midulla M, Mourey E, Loffroy R. Selective Arterial Embolization with N-Butyl Cyanoacrylate Prior to CT-Guided Percutaneous Cryoablation of Kidney Malignancies: A Single-Center Experience. J Clin Med 2021; 10:jcm10214986. [PMID: 34768506 PMCID: PMC8584960 DOI: 10.3390/jcm10214986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
The study’s purpose was to assess the safety, feasibility and efficiency of selective arterial embolization (SAE) using N-butyl cyanoacrylate (NBCA) glue before percutaneous cryoablation (PCA) of renal malignancies in patients whose tumor characteristics and/or comorbidities resulted in an unacceptable risk of bleeding. In this single-center retrospective study of 19 consecutive high-risk patients (median age, 74 years) with renal malignancies managed in 2017–2020 by SAE with NBCA followed by PCA, data about patients, tumor and procedures characteristics, complications, renal function and hemoglobin concentration before and after treatment, as well as recurrence were collected. Charlson comorbidity index was ≥4 in 89.5% of patients. Ten patients were treated by antiplatelet and/or anticoagulant therapy. Median tumor largest diameter was 3.75 cm (range, 1–6.5 cm) and R.E.N.A.L. nephrometry score was ≥7 in 80%, indicating substantial tumor complexity. No major complications were recorded and minor complications occurred in 7 patients. No residual tumor was found at 6-week imaging follow-up in 18/19 patients. Tumor recurrence was visible in 1/16 patients at 6-month imaging follow-up. No significant difference was found for renal function after treatment (p = 0.07), whereas significant decrease in hemoglobin concentration was noted (p = 0.00004), although it was relevant for only one patient who required only blood transfusion and no further intervention. SAE prior to PCA is safe and effective for managing renal malignancies in high-risk patients.
Collapse
Affiliation(s)
- Olivier Lopez
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
| | - Olivier Chevallier
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
- Imaging and Artificial Vision (ImViA) Laboratory-EA 7535, University of Bourgogne/Franche-Comté, 9 Avenue Alain Savary, BP 47870, 21078 Dijon, France;
| | - Kévin Guillen
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
- Imaging and Artificial Vision (ImViA) Laboratory-EA 7535, University of Bourgogne/Franche-Comté, 9 Avenue Alain Savary, BP 47870, 21078 Dijon, France;
| | - Pierre-Olivier Comby
- Imaging and Artificial Vision (ImViA) Laboratory-EA 7535, University of Bourgogne/Franche-Comté, 9 Avenue Alain Savary, BP 47870, 21078 Dijon, France;
- Department of Neuroradiology and Emergency Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France
| | - Julie Pellegrinelli
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
| | - Claire Tinel
- Department of Nephrology and Renal Transplantation, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France;
| | - Nicolas Falvo
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
| | - Marco Midulla
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
| | - Eric Mourey
- Department of Urology and Andrology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France;
| | - Romaric Loffroy
- Image-Guided Therapy Center, Department of Vascular and Interventional Radiology, François-Mitterrand University Hospital, 14 Rue Paul Gaffarel, BP 77908, 21079 Dijon, France; (O.L.); (O.C.); (K.G.); (J.P.); (N.F.); (M.M.)
- Imaging and Artificial Vision (ImViA) Laboratory-EA 7535, University of Bourgogne/Franche-Comté, 9 Avenue Alain Savary, BP 47870, 21078 Dijon, France;
- Correspondence: ; Tel.: +33-380-293-677
| |
Collapse
|
32
|
Alkhalifa H, Mohammed F, Taurin S, Greish K, Taha S, Fredericks S. Inhibition of aquaporins as a potential adjunct to breast cancer cryotherapy. Oncol Lett 2021; 21:458. [PMID: 33907568 PMCID: PMC8063341 DOI: 10.3892/ol.2021.12719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 03/02/2021] [Indexed: 12/20/2022] Open
Abstract
Cryoablation is an emerging type of treatment for cancer. The sensitization of tumors using cryosensitizing agents prior to treatment enhances ablation efficiency and may improve clinical outcomes. Water efflux, which is regulated by aquaporin channels, contributes to cancer cell damage achieved through cryoablation. An increase in aquaporin (AQP) 3 is cryoprotective, whereas its inhibition augments cryodamage. The present study aimed to investigate aquaporin (AQP1, AQP3 and AQP5) gene expression and cellular localization in response to cryoinjury. Cultured breast cancer cells (MDA-MB-231 and MCF-7) were exposed to freezing to induce cryoinjury. RNA and protein extracts were then analyzed using reverse transcription-quantitative PCR and western blotting, respectively. Localization of aquaporins was studied using immunocytochemistry. Additionally, cells were transfected with small interfering RNA to silence aquaporin gene expression and cell viability was assessed using the Sulforhodamine B assay. Cryoinjury did not influence gene expression of AQPs, except for a 4-fold increase of AQP1 expression in MDA-MD-231 cells. There were no clear differences in AQP protein expression for either cell lines upon exposure to frozen and non-frozen temperatures, with the exception of fainter AQP5 bands for non-frozen MCF-7 cells. The exposure of cancer cells to freezing temperatures altered the localization of AQP1 and AQP3 proteins in both MCF-7 and MDA-MD-231 cells. The silencing of AQP1, AQP3 and AQP5 exacerbated MDA-MD-231 cell damage associated with freezing compared with control siRNA. This was also observed with AQP3 and AQP5 silencing in MCF-7 cells. Inhibition of aquaporins may potentially enhance cryoinjury. This cryosensitizing process may be used as an adjunct to breast cancer cryotherapy, especially in the border area targeted by cryoablation where freezing temperatures are not cold enough to induce cellular damage.
Collapse
Affiliation(s)
- Haifa Alkhalifa
- Department of Basic Medical Sciences, Royal College of Surgeons in Ireland, Medical University of Bahrain, Adliya 15503, Kingdom of Bahrain
- Department of Science, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Fatima Mohammed
- Department of Basic Medical Sciences, Royal College of Surgeons in Ireland, Medical University of Bahrain, Adliya 15503, Kingdom of Bahrain
| | - Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al-Jawhara Centre for Molecular Medicine, Arabian Gulf University, Segaya, Manama 328, Kingdom of Bahrain
| | - Khaled Greish
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al-Jawhara Centre for Molecular Medicine, Arabian Gulf University, Segaya, Manama 328, Kingdom of Bahrain
| | - Safa Taha
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al-Jawhara Centre for Molecular Medicine, Arabian Gulf University, Segaya, Manama 328, Kingdom of Bahrain
| | - Salim Fredericks
- Department of Basic Medical Sciences, Royal College of Surgeons in Ireland, Medical University of Bahrain, Adliya 15503, Kingdom of Bahrain
| |
Collapse
|
33
|
Snyder KK, Van Buskirk RG, Baust JG, Baust JM. Breast Cancer Cryoablation: Assessment of the Impact of Fundamental Procedural Variables in an In Vitro Human Breast Cancer Model. Breast Cancer (Auckl) 2020; 14:1178223420972363. [PMID: 33239880 PMCID: PMC7672727 DOI: 10.1177/1178223420972363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Breast cancer is the most prominent form of cancer and the second leading cause of death in women behind lung cancer. The primary modes of treatment today include surgical excision (lumpectomy, mastectomy), radiation, chemoablation, anti-HER2/neu therapy, and/or hormone therapy. The severe side effects associated with these therapies suggest a minimally invasive therapy with fewer quality of life issues would be advantageous for treatment of this pervasive disease. Cryoablation has been used in the treatment of other cancers, including prostate, skin, and cervical, for decades and has been shown to be a successful minimally invasive therapeutic option. To this end, the use of cryotherapy for the treatment of breast cancer has increased over the last several years. Although successful, one of the challenges in cryoablation is management of cancer destruction in the periphery of the ice ball as the tissue within this outer margin may not experience ablative temperatures. In breast cancer, this is of concern due to the lobular nature of the tumors. As such, in this study, we investigated the level of cell death at various temperatures associated with the margin of a cryogenic lesion as well as the impact of repetitive freezing and thawing methods on overall efficacy. METHODS Human breast cancer cells, MCF-7, were exposed to temperatures of -5°C, -10°C, -15°C, -20°C, or -25°C for 5-minute freeze intervals in a single or repeat freeze-thaw cycle. Samples were thawed with either passive or active warming for 5 or 10 minutes. Samples were assessed at 1, 2, and 3 days post-freeze to assess cell survival and recovery. In addition, the modes of cell death associated with freezing were assessed over the initial 24-hour post-thaw recovery period. RESULTS Exposure of MCF-7 cells to -5°C and -10°C resulted in minimal cell death regardless of the freeze/thaw conditions. Freezing to a temperature of -25°C resulted in complete cell death 1 day post-thaw with no cell recovery in all freeze/thaw scenarios evaluated. Exposure to a single freeze event resulted in a gradual increase in cell death at -15°C and -20°C. Application of a repeat freeze-thaw cycle (dual 5-minute freeze) resulted in an increase in cell death with complete destruction at -20°C and near complete death at -15°C (day 1 survival: single -15°C freeze/thaw = 20%; repeated -15°C freeze/thaw = 4%). Analysis of thaw interval time (5 vs 10 minute) demonstrated that the shorter 5-minute thaw interval between freezes resulted in increased cell destruction. Furthermore, investigation of thaw rate (active vs passive thawing) demonstrated that active thawing resulted in increased cell survival thereby less effective ablation compared with passive thawing (eg, -15°C 5/10/5 procedure survival, passive thaw: 4% vs active thaw: 29%). CONCLUSIONS In summary, these in vitro findings suggest that freezing to temperatures of 25°C results in a high degree of breast cancer cell destruction. Furthermore, the data demonstrate that the application of a repeat freeze procedure with a passive 5-minute or 10-minute thaw interval between freeze cycles increases the minimal lethal temperature to the -15°C to -20°C range. The data also demonstrate that the use of an active thawing procedure between freezes reduces ablation efficacy at temperatures associated with the iceball periphery. These findings may be important to improving future clinical applications of cryoablation for the treatment of breast cancer.
Collapse
Affiliation(s)
| | - Robert G Van Buskirk
- CPSI Biotech, Owego, NY, USA
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - John G Baust
- Center for Translational Stem Cell and Tissue Engineering, Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | | |
Collapse
|
34
|
Baust JM, Robilotto A, Santucci KL, Snyder KK, Van Buskirk RG, Katz A, Corcoran A, Baust JG. Evaluation of a Novel Cystoscopic Compatible Cryocatheter for the Treatment of Bladder Cancer. Bladder Cancer 2020. [DOI: 10.3233/blc-200321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND: As the acceptance of cryoablative therapies for the treatment of non-metastatic cancers continues to grow, avenues for novel cryosurgical technologies and approaches have opened. Within the field of genitourinary tumors, cryosurgical treatments of bladder cancers remain largely investigational. Current modalities employ percutaneous needles or transurethral cryoballoons or sprays, and while results have been promising, each technology is limited to specific types and stages of cancers. OBJECTIVE: This study evaluated a new, self-contained transurethral cryocatheter, FrostBite-BC, for its potential to treat bladder cancer. METHODS: Thermal characteristics and ablative capacity were assessed using calorimetry, isothermal analyses, in vitro 3-dimensional tissue engineered models (TEMs), and a pilot in vivo porcine study. RESULTS: Isotherm assessment revealed surface temperatures below – 20°C within 9 sec. In vitro TEMs studies demonstrated attainment of ≤– 20°C at 6.1 mm and 8.2 mm in diameter following single and double 2 min freezes, respectively. Fluorescent imaging 24 hr post-thaw revealed uniform, ablative volumes of 326.2 mm3 and 397.9 mm3 following a single or double 2 min freeze. In vivo results demonstrated the consistent generation of ablative areas. Lesion depth was found to correlate with freeze time wherein 15 sec freezes resulted in ablation confined to the sub-mucosa and ≥30 sec full thickness ablation of the bladder wall. CONCLUSIONS: These studies demonstrate the potential of the FrostBite-BC cryocatheter as a treatment option for bladder cancer. Although preliminary, the outcomes of these studies were encouraging, and support the continued investigation into the potential of the FrostBite-BC cryocatheter as a next generation, minimally invasive cryoablative technology.
Collapse
Affiliation(s)
| | | | | | | | - Robert G. Van Buskirk
- CPSI Biotech, Owego, NY, USA
- Center for Translational Stem Cell and Tissue Engineering Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Aaron Katz
- Department of Urology, NYU Winthrop Hospital, Mineola, NY, USA
| | | | - John G. Baust
- Center for Translational Stem Cell and Tissue Engineering Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
35
|
Santucci KL, Baust JM, Snyder KK, Van Buskirk RG, Katz A, Corcoran A, Baust JG. Investigation of Bladder Cancer Cell Response to Cryoablation and Adjunctive Cisplatin Based Cryo/Chemotherapy. CLINICAL RESEARCH (MILPITAS, CALIF.) 2020; 6. [PMID: 35128225 PMCID: PMC8813088 DOI: 10.16966/2469-6714.154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Due to a rising annual incidence of bladder cancer, there is a growing need for development of new strategies for treatment. In 2018, the World Cancer Research Fund and other groups reported that there were ~550,000 new cases worldwide of bladder cancer. It has been further estimated that >200,000 individuals die annually from bladder cancer worldwide. Various treatment options exist. However, many if not all remain suboptimal. While the preferred chemotherapeutic options have changed in the past few years there have been few advances in the bladder cancer medical device field. Cryoablation is now being evaluated as a new option for the treatment of bladder cancer. While several studies have shown cryoablation to be promising for the treatment of bladder cancer, a lack of basic information pertaining to dosing (minimal lethal temperature) necessary to destroy bladder cancer has limited its use as a primary therapeutic option. Concerns with bladder wall perforation and other side effects have also slowed adoption. In an effort to detail the effects of freezing on bladder cancer, two human bladder cancer cell lines, SCaBER and UMUC3, were evaluated in vitro. SCaBER, a basal subtype of muscle invasive bladder cancer, and UMUC3, an intermediate transitional cell carcinoma, are both difficult to treat but are reportedly responsive to most conventional treatments. SCaBER and UMUC3 cells were exposed to a range of freezing temperatures from −10 to −25°C and compared to non-frozen controls. The data show that a single 5 minute freeze to −10°C did not affect cell viability, whereas −15°C and −20°C results in a significant reduction in viability 1 day post freeze to <20%. These populations, however, were able to recover in culture. A complete loss of cell viability was found following a single freeze at −25°C. Application of a repeat (double) freeze resulted in complete cell death at −20°C. In addition to freezing alone, studies investigating the impact of adjunctive low dose (1 μM) cisplatin pre-treatment (30 minutes and 24 hours) in combination with freezing were conducted. The combination of 30 minute cisplatin pre-treatment and mild (−15°C) freezing resulted in complete cell death. This suggests that subclinical doses of cisplatin may be synergistically effective when combined with freezing. In summary, these in vitro results suggest that freezing to temperatures in the range of −20 to 25°C results in a high degree of bladder cancer cell destruction. Further, the data describe a potential combinatorial chemo/cryo therapeutic strategy for the treatment of bladder cancer.
Collapse
Affiliation(s)
| | | | | | - Robert G Van Buskirk
- CPSI Biotech, Owego, USA.,Center for Translational Stem Cell and Tissue Engineering Binghamton University, USA.,Department of Biological Sciences, Binghamton University, USA
| | - Aaron Katz
- Department of Urology, NYU Winthrop Hospital, US
| | | | - John G Baust
- Center for Translational Stem Cell and Tissue Engineering Binghamton University, USA.,Department of Biological Sciences, Binghamton University, USA
| |
Collapse
|