Next Article in Journal
Double Mutation of Days to Heading 2 and CONSTANS 3 Improves Agronomic Performance of Japonica Rice under Short Daylight Conditions in Southern China
Previous Article in Journal
Topical Application of Deglycating Enzymes as an Alternative Non-Invasive Treatment for Presbyopia
Previous Article in Special Issue
COVID-19: Has the Liver Been Spared?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends

1
Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
2
Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
3
Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
4
Division of Image-Guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem 12000, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(8), 7344; https://doi.org/10.3390/ijms24087344
Submission received: 27 March 2023 / Revised: 7 April 2023 / Accepted: 13 April 2023 / Published: 16 April 2023

Abstract

:
Personalized cancer treatments help to deliver tailored and biologically driven therapies for cancer patients. Interventional oncology techniques are able to treat malignancies in a locoregional fashion, with a variety of mechanisms of action leading to tumor necrosis. Tumor destruction determines a great availability of tumor antigens that can be recognized by the immune system, potentially triggering an immune response. The advent of immunotherapy in cancer care, with the introduction of specific immune checkpoint inhibitors, has led to the investigation of the synergy of these drugs when used in combination with interventional oncology treatments. The aim of this paper is to review the most recent advances in the field of interventional oncology locoregional treatments and their interactions with immunotherapy.

1. Introduction

In recent years, we have witnessed the exponential expansion and impact of interventional radiology in oncology. Cross-sectional imaging techniques play a crucial role in the diagnosis, treatment planning, and follow-up of cancer patients, but also provide the ability to perform minimally invasive approaches to procure tissue for histological diagnosis, including the genetic material necessary to develop better tailored and biologically driven treatments. This permits personalized medicine, which potentially maximizes therapeutic effects [1]. Moreover, there is increasing attention in the scientific and medical community on the development of interventional oncology techniques and procedures as locoregional approaches to be employed in cancer treatment in a multidisciplinary cancer management setting.
Currently, percutaneous interventional approaches are being performed for the treatment of a wide range of both primary and secondary malignancies as an alternative or in combination with surgery and other treatment modalities [2]. Indeed, multidisciplinary guidelines for the treatment of HCC and RCC now incorporate their use [3,4,5].
Interventional oncology has the unique capability to treat malignancy in a locoregional fashion, enabling curative (ablative treatments), disease control (intra-arterial chemo- or radio-embolization), and palliative treatment [6]. Locoregional eradication therapy involves the application of different energy sources that, despite a variety of mechanisms of action (such as heat, freezing, or electricity), induce effective necrosis of the tumor core and apoptosis of the adjacent tissue, with a substantial preservation of healthy parenchyma [7].
The destruction of the tumor, along with the release of necrotic material, creates in situ availability of antigens that may be recognized by the immune system as a threat and potentially trigger an immune response throughout the body, bringing to the so-called abscopal effect [8,9].
With the advent of immunotherapy in cancer care and the introduction of immune checkpoint inhibitors, several efforts have been made to investigate the synergy of such treatment in combination with interventional radiology treatments, as this new class of drug influences the immunologic microenvironment of the tumor-acting on several key target molecules, restoring immune system function against malignancy [10].
Although preliminary evidence from immune checkpoint inhibitors monotherapy is promising, the greatest potential of these treatments is likely to be achieved in their combination with other treatments that can trigger an immune response [11]. In this manuscript, we review the most recent advances in locoregional interventional oncology treatments and their interactions with immune checkpoint inhibitors. Figure 1 depicts the most important interventional oncology techniques and their mode of action.

2. Percutaneous Treatment and Immuno-Oncology

Based upon multi-disciplinary guidelines, percutaneous interventional techniques are currently considered a possible therapeutic strategy to treat both primary and secondary tumors in multiple anatomical sites [12,13,14,15]. However, therapeutic outcomes of interventional techniques are frequently limited by recurrence and distant metastasis. Recent pre-clinical and clinical studies have suggested that percutaneous ablative therapies lead to an alteration of the patient’s immuno-profile [16]. Among these immunological effects, for some therapies such as cryoablation and IRE, the central area of necrosis caused by the percutaneous ablation induces antigenic release that leads to an antigenic presentation by dendritic cells, increase in serum cytokines level, activation of the CTLA4 cascade and T cell response [17,18]. On the other hand, the peripheral area of apoptosis induced by ablation downregulates the immunological system [17]. These interactions produce both local and systemic effects, including occasionally the abscopal effect of distant tumor shrinkage [6,11,17,19,20].
While the immune response induced by ablation alone appears to be transient, there is strong evidence that it could potentially enhance the effect of immunotherapies [16,19,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39]. In this section, we review the evidence regarding the most common percutaneous interventional techniques and their interaction with immunotherapies in cancer treatment.

2.1. Radiofrequency Ablation (RFA)

A sufficiently high thermal insult induces coagulation necrosis in the target tissue and cytokines and antigens blood release, leading to both local and systemic effects [40]. Slovak et al. demonstrated that, in a VX-2 rabbit liver cancer model, the combination of RFA plus CpG-B (a factor that stimulates innate immunity) increased the presence of activating lymphocyte and the rabbit survival compared with either RFA or CpG-B alone [41].
Schneider et al. analyzed the ablated area in non-small cell lung cancer (NSCLC), demonstrating a surge of CD4+ and CD8+ lymphocytes in the peripheral zone and an intensification of pro-inflammatory cytokines [42].
Mizukoshi et al. investigated the immune responses before and after RFA in 69 HCC patients and highlighted that there was a significant increase in tumor-associated antigen (TAA)-specific T cells in the peripheral blood of 62.3% of patients. Moreover, the number of TAA-specific T cells after RFA was predictive of HCC recurrence after ablation by univariate and multivariate analyses [43].
A comparative study involving patients with intermediate to advanced stage HCC investigated the efficacy of RFA plus monoclonal antibody (131I-chTNT) as a combination therapy [44]. This study demonstrated that such combination therapy is significantly more effective than RFA alone, as demonstrated by the longer survival time of patients who received RFA plus 131I-chTNT compared to those who received the RFA alone (p = 0.052) [44,45].
Regarding the systemic abscopal effects of radiofrequency ablation, it has been demonstrated in a colon-cancer murine model that the combination of RFA with a vaccine encoding CEA produces regression of distal metastasis and a significant increase in CEA-specific CD4+ T cells compared with RFA or vaccine alone (p < 0.0001; p = 0.0003, respectively) [46]. Nakagawa et al. demonstrated that the administration of dendritic cells stimulated by OK-432 (a clinical bacterial product that can induce DC maturation) after RFA increased the number of CD8+ T cells infiltrating untreated secondary tumors as compared to RFA alone (p < 0.001) [47]. However, the abscopal effect achieved by the RFA alone is weak, transient, or even occasionally counterproductive [41]. In fact, there is the risk of inducing an immunologically tolerogenic state if the RFA is not supported by immunotherapy. Indeed, it has been shown in rat breast cancer that RFA alone stimulates hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF), leading to unwanted effects, such as an increased cell replication (evaluated by Ki-67) and microvascular density in distant tumors [48]. Therefore, the deactivation of HGF (using PHA-665752) and VEGF (using semaxanib) pathways may improve clinical outcomes of RFA ablation [48]. This opposite tumorigenic effect of RFA may also explain a worse prognosis of ablated HCC compared with surgical resection. Other clinical consequences of this pro-tumorigenic effect are the evidence that an incomplete radiofrequency ablation enhances neo-angiogenesis in HCC and tumoral growth in non-small cell lung cancer [49].

2.2. Cryoablation

Cryoablation is based on a cycle of freezing and thawing that causes intra- and extracellular ice crystal formation, damage to the cell membrane, osmotic pressure changes, and, thus, cellular dehydration. The use of cooling energy makes cryoablation suitable for lesions close to vital structures [50]. While the central area of the ablation is composed of necrotic tissue, the peripheral boundary is largely composed of apoptotic cells [51].
Despite all ablation techniques releasing tumor antigens, cryoablation avoids protein denaturation and preserves native antigen structures [52,53]. As a consequence, serum levels of interleukin-1 (IL-1), IL-6, NF-κβ, and TNF-α are significantly higher after cryoablation compared to other ablative therapies, suggesting a stronger immunostimulatory response [19].
In a renal cell carcinoma model, the combination of cryoablation and anti-PDL1 drug lead to anti-tumor immune responses and delayed tumor growth of distant untreated tumors [54]. Furthermore, in a melanoma model, den Brok et al. demonstrated that the combination of cryoablation and CpG-B induced the regression of the existing secondary tumors in 40% of cryoablation-treated mice, suggesting strong abscopal effects [19].
In metastatic liver cancer patients, Niu et al. demonstrated that the combination of cryoablation and immunotherapy leads to a significantly increased median overall survival (OS) compared with cryoablation or immunotherapy alone (32 vs. 17.5 vs. 3 months; p < 0.05) [25]. Similar results in terms of OS and immune responses were reported in patients with lung, renal cell, and hepatocellular cancers treated with cryoablation and allogeneic NK cell transfers and in patients with breast cancer treated with cryoablation plus anti-CTLA4 and anti-PD1 [55,56,57,58]. Despite the volume and the level of evidence being lower for cryoablation with respect to RFA, the combination of cryoablation with immunotherapy appears to offer promising results representing an optimistic basis for further investigations.

2.3. Irreversible Electroporation (IRE)

IRE is a novel non-thermal ablation technology based on the application of pulsatile and targeted high-voltage electric energy that alters the current potential of the cellular membrane, leading to permanent nanopore formation within the lipid bilayer membrane. This membranous disruption results in loss of homeostasis with subsequent cellular apoptosis and death.
The first significant evidence of the immunological effects of IRE was reported in 2016 by Bulvik et al., who demonstrated a greater lymphocyte infiltration and tumor size reduction for IRE compared to RFA in an HCC murine model [59]. Furthermore, in preclinical models of hepatocellular carcinoma, Vivas et al. showed that the administration of an immunostimulant drug (Poly-ICLC) before IRE was able to increase the immunogenic response and reduce tumor growth compared to both IRE and Poly-ICLC alone (40%, p < 0.05) [60]. These findings were confirmed by Alnagger et al., who reported an increased median overall survival (10.1 months of the IRE-NK group vs. 8.9 months of the IRE alone group, p = 0.0078) and a decrease in alpha-fetoprotein expression in patients with metastatic liver tumor (IV stage) treated with IRE plus allogeneic NK cell immunotherapy [61]. The same strategy (IRE plus NK vs. IRE alone) was investigated by Yang et al., who demonstrated longer median progression-free survival (PFS) and overall survival (OS) (PFS 15.1 vs. 10.6 months, p < 0.05, OS 17.9 vs. 23.2 months, p < 0.05) with a reduction of circulating tumor cells in patients who received combination therapy [62].
IRE has also been evaluated in other clinical contexts, as classical systemic immunotherapy has only limited efficacy against pancreatic ductal adenocarcinoma (PDAC) due to the presence of an immunosuppressive tumor-associated stroma, and the rationale of studies on IRE is that ablative therapies could destroy the pancreatic immunosuppressive microenvironment, leading to a greater response to systemic immunotherapy [63,64]. Zhao et al. utilized a mouse model of PDAC to demonstrate that the association of IRE and systemic anti-PD1 treatment promotes CD8+ T cell infiltration and increases overall survival when compared to both IRE and anti-PD1 as monotherapy [64]. Narayanan et al. also utilized a mouse model of PDAC. They combined IRE with systemic anti-PD1 and an intra-tumoral TLR7 agonist. This triple strategy improved local response compared to IRE alone and promoted regression of untreated concomitant metastases [65]. These encouraging results have brought to first preliminary human studies, showing that IRE combined with NK cells or allogenic Vγ9Vδ2 T cell infusion has prolonging effects on progression-free survival rates (11 versus 8.5 months), overall response rates at 1 month, and overall survival rates (14.5 versus 11 months) when compared to IRE alone in PDAC patients [66,67,68]. The soon-to-be-conducted PANFIRE-III trial (NCT04612530) will also combine IRE, systemic anti-PD1, and an intra-tumoral TLR9 agonist in metastasized PDAC human patients.

2.4. Microwave Ablation (MWA)

The association of MWA with immunotherapy is weaker, as preliminary studies suggested that MWA is less immunogenic compared to RFA and cryoablation [69]. However, Leutche et al. uncovered de-novo or enhanced tumor-specific T-cell responses in 30% of patients with hepatocellular carcinoma (HCC) treated with MWA alone. The T-cell response was associated with longer progression-free survival (27.5 vs. 10.0 months) [6]. In the same study, the analysis of HCC samples (n = 18) of patients receiving combined MWA and resection revealed superior disease-free survival in patients with high T-cell sample infiltration at the time of thermal ablation (37.4 vs. 13.1 months).
Regarding the synergic effect of MWA and immunotherapy, Chen et al. demonstrated that the combination of MW and GM-CSF significantly increased the free tumor survival and decreased the tumor volume in a murine hepatoma model [70]. Similar results were obtained in human patients with HCC, although in this initial study, the increase was not statistically significant [71]. Additionally, in a pilot study by Zhou P et al., the application of adoptive immunotherapy in association with MWA for HCC patients was demonstrated to be safe and capable of increasing the percentage of peripheral lymphocytes [72].

2.5. High-Intensity Focal Ultrasound (HIFU) and Laser-Induced Thermotherapy (LiTT)

Less evidence can be found in the literature for other ablation techniques. HIFU has been used for primary and secondary malignancy of the breast, soft tissue, bone, pancreas, kidney, and liver [6]. Yet, although HIFU can induce cytokine release and stress response with an augmented CD4+/CD8+ ratio, it appears to be less immunogenic compared with RFA and cryoablation [69].
LiTT has been reported to increase the level of cytokines (IL-6, TNFRI, and CRP levels) in liver malignancies [73]. Moreover, Vogl et al. highlighted that the levels of CD3+, CD4+, and CD8+ were increased after LiTT (12.73 ± 4.83 vs. 92.09 ± 12.04; 4.36 ± 3.32 vs. 42.92 ± 16.68; 3.64 ± 1.77 vs. 47.54 ± 15.68; p < 0.05) with an associated improvement in cytotoxic effects (RLU = 1493 ± 1954.68 vs. 7260 ± 3929.76; p < 0.001) [74].
Ablative techniques associated with immunotherapy seem to obtain a synergistic effect, as ablative therapies alone can increase neoangiogenesis when complete ablation is not achieved, also leading to immune tolerance. Most of the literature studies were made on RFA, whereas the combination of immunotherapy with cryoablation, MWA, IRE, and HIFU is emerging as a promising alternative to RFA on a great variety of target lesions. Table 1 summarizes the pros and cons of the current practice of locoregional percutaneous interventional oncology treatments when associated with immunotherapy, also reporting results of preclinical studies.
Table 2 describes in what lesions the clinical trials investigated the role of immunotherapy associated with ablative treatments.

3. Endovascular Treatments and Immuno-Oncology

In current practice, interventional intra-arterial treatments of tumors include the use of a wide variety of active tumoricidal agents, including conventional transcatheter arterial chemoembolization (cTACE), drug-eluting beads transcatheter arterial chemoembolization (DEB-TACE) and transarterial radioembolization (TARE) [75,76,77].
Chemoembolization (both cTACE and DEB-TACE) is the treatment of choice in patients with intermediate-stage HCC (Barcelona Clinic Liver Cancer—BCLC stage B), while radioembolization can potentially be used as an alternative based on level 2 of evidence in these patients [78,79,80]. Moreover, there is the ever-increasing use of these techniques for the treatment of hepatic metastases, including colorectal and neuroendocrine cancer [81,82]. In this section, we review the evidence of interaction between immunotherapy and the most common endovascular interventional techniques.

3.1. Transarterial Chemoembolization (TACE)

TACE is commonly used in patients with unresectable HCC with preserved liver function [3,78,83]. Conventional TACE (cTACE) commonly delivers an emulsion of lipiodol and chemotherapeutic agent (most often doxorubicin or cisplatin) followed by gelatine sponge as the embolic agent, whereas the most recent drug-eluting beads (DEB) TACE utilizes drug-eluting beads preloaded with the chemotherapeutic agent, with a reported reduction of drug-related side effects due to a better pharmacokinetic profile [83,84].
Both types of TACE induce local tumor necrosis by the occlusion of feeding arteries, leading secondarily to the release of tumor antigens, which activate the immune response [10]. Moreover, TACE can potentially modify the cytokine spectrum and the activation level of T cells [85]. TACE stimulates the secretion interleukines (IL) as IL-1 and IL-10, and of Interferon-γ, with activation of T helper-17 and T helper-1 cells [8,86,87]. TACE also leads to a modulation of immunosuppressive factors such as T-regulatory cells, PD1/PDL1, and HIF-1α, potentially bringing immune tolerance [87,88,89].
The combination of TACE and immunotherapy may amplify the antitumoral effect. In a pilot study, Sangro et al. used a CTLA4 inhibitor (tremelimumab) combined with TACE in 21 advanced HCC patients, showing promising results, with a good safety profile and a median survival of 8 months. Tremelimumab was administered intravenously at a dose of 15 mg/kg every 90 days until progression or intolerable toxicity [90,91].
Duffy et al. showed the efficacy of combined treatment of TACE/ablation and tremelimumab in a group of 32 patients with advanced HCC (75% of patients had the progressive disease). The median overall survival was 12.3 months, and most patients showed a reduction of tumor load, tumor reduction in non-ablated or non-embolized areas, and intratumoral infiltration of CD8+ T cells [92].
A phase-I clinical trial (NCT 03143270) in patients with advanced HCC treated with nivolumab (a PD1 inhibitor) combined with DEB-TACE is ongoing. All patients are scheduled to receive 24 mg of nivolumab intravenously for up to 1 year every 2 weeks.
Another open-label single-arm phase II study (NCT 03572582) combines nivolumab with TACE in patients with intermediate HCC is ongoing. Nivolumab treatment will start 2–3 days after the initial TACE and will be administered intravenously (240 mg, fixed-dose) every 2 weeks for up to two years until progression. A second TACE will be performed 8 weeks after the first one.
Other investigations using durvalumab (a PDL1 inhibitor) plus tremelimumab combined with TACE are underway, including a phase-II clinical trial (NCT02821754) and a clinical study (NCT03638141).

3.2. Transarterial Radioembolization (TARE)

TARE is emerging as a “multi-purpose” treatment in patients with HCC, as it can represent an effective alternative to both TACE and, as more recent studies demonstrate, ablative treatments. Under ideal conditions, it can lead to possible tumor downstaging and also act as a bridge to surgical resection and liver transplantation in selected patients [3,93,94].
TARE is usually performed with Yttrium-90 (90Y) in resin or glass microspheres (although there are ever-increasing reports regarding the effectiveness of Holmium-166 (166Ho) in poly(L-lactic acid) (PLLA) microspheres) [95]. As opposed to TACE, TARE utilizes local beta radiation to achieve tumor necrosis instead of artery occlusion. It is the following two-step treatment: where the first part is comprised of pre-treatment angiography with an injection of macroaggregated albumin (MAA) marked with Technetium-99m and a scintigraphy in order to evaluate lung shunt fraction (to avoid the risk of radiation pneumonitis) and to identify arteries that supply the gastrointestinal tract (to avoid ulcerations). The second part is the actual treatment with 90Y-loaded microspheres [96]. When judiciously delivered, TARE has a minimal embolic effect, so the post-embolization syndrome is reduced compared to TACE [97].
Recent studies underline that the immunocompetence of the tumor microenvironment is elevated after 90Y TARE. This effect is possibly explained by the expression of TNF-α by CD4+ T cells for the upregulation of CD8+ T cells, and the APCs ratio is increased [98].
A recent retrospective study of 26 patients with aggressive intermediate-stage or advanced HCC showed that the combination of nivolumab (or nivolumab plus ipilimumab) plus TARE is safe (little treatment toxicity) and has promising results, with a median overall survival of 16.5 months and progression-free survival of 5.7 months. One patient even achieved a complete response [99].
Moreover, a case report has been published of a patient affected by advanced HCC with a macrovascular invasion that was treated with nivolumab plus TARE, obtaining a downstaging and was amenable to surgery (with surgery confirming a complete response) [100].
These results are confirmed by a phase-II trial with 36 patients with an advanced HCC treated with TARE plus nivolumab, showing an overall response rate (31%), and an overall survival of 15.1 months [101].
Other trials are still evaluating TARE plus Nivolumab (NCT03380130, NCT03033446, NCT02837029) and TARE plus Pembrolizumab (NCT03099564).
The low volume and level of evidence of studies on combination treatments of immunotherapy and TACE or TARE make it difficult to discuss its safety and long-term efficacy, even though phase-I and -II clinical trials are ongoing and will certainly shed light on this promising combination technique. Table 3 summarizes the pros and cons of the current practice of locoregional endovascular interventional oncology treatments when associated with immunotherapy, also reporting results of preclinical studies.
Table 4 describes in what lesions the clinical trials investigated the role of immunotherapy associated with endovascular treatments.

4. Conclusions

The combination of interventional radiology treatments with immunotherapies in oncology is growing rapidly. The results, albeit currently with only a low level of evidence, are encouraging, suggesting that more evidence could better define the role of combination therapies in this field. In particular, the greatest evidence is focused on the combination of immunotherapy with RFA or cryoablation, whereas the combination of IRE and immunotherapy may play a greater role in the future, particularly in patients with pancreatic malignancies, where other methods have shown reduced efficacy.
The evidence from the literature is still rather limited for TACE and TARE, composed mostly of small monocentric studies. Further research in this field is required, ideally with randomized trials, to better understand how to achieve the desired immunogenic/abscopal effects while limiting unwanted pro-tumorigenic phenomena.

Author Contributions

Conceptualization, A.P., A.C., S.N.G. and R.I.; methodology, A.P., A.C., S.N.G. and R.I.; investigation, A.P. and A.C.; resources, E.P., A.S., G.M. and F.R.P.; writing—original draft preparation, E.P., A.S., G.M. and F.R.P.; writing—review and editing, A.P., A.C., E.S. and R.I.; visualization, M.P., S.N.G. and R.I.; supervision, A.G., L.N. and E.S.; project administration, R.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Erinjeri, J.P.; Fine, G.C.; Adema, G.J.; Ahmed, M.; Chapiro, J.; Brok, M.D.; Duran, R.; Hunt, S.J.; Johnson, D.T.; Ricke, J.; et al. Immunotherapy and the Interventional Oncologist: Challenges and Opportunities—A Society of Interventional Oncology White Paper. Radiology 2019, 292, 25–34. [Google Scholar] [CrossRef] [PubMed]
  2. Helmberger, T. The evolution of interventional oncology in the 21st century. Br. J. Radiol. 2020, 93, 20200112. [Google Scholar] [CrossRef]
  3. Reig, M.; Forner, A.; Rimola, J.; Ferrer-Fàbrega, J.; Burrel, M.; Garcia-Criado, Á.; Kelley, R.K.; Galle, P.R.; Mazzaferro, V.; Salem, R.; et al. BCLC strategy for prognosis prediction and treatment recommendation Barcelona Clinic Liver Cancer (BCLC) staging system: The 2022 update. J. Hepatol. 2021, 76, 681–693. [Google Scholar] [CrossRef]
  4. Campbell, S.C.; Clark, P.E.; Chang, S.S.; Karam, J.A.; Souter, L.; Uzzo, R.G. Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow-Up: AUA Guideline: Part I. J. Urol. 2021, 206, 199–208. [Google Scholar] [CrossRef] [PubMed]
  5. Campbell, S.C.; Uzzo, R.G.; Karam, J.A.; Chang, S.S.; Clark, P.E.; Souter, L. Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow-up: AUA Guideline: Part II. J. Urol. 2021, 206, 209–218. [Google Scholar] [CrossRef] [PubMed]
  6. Lucatelli, P.; Iezzi, R.; De Rubeis, G.; Goldberg, S.N.; Bilbao, J.I.; Sami, A.; Akhan, O.; Giuliante, F.; Pompili, M.; Tagliaferri, L.; et al. Immuno-oncology and interventional oncology: A winning combination. The latest scientific evidence. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5343–5350. [Google Scholar]
  7. Ridouani, F.; Srimathveeravalli, G. Percutaneous image-guided ablation: From techniques to treatments. Presse Med. 2019, 48, e219–e231. [Google Scholar] [CrossRef]
  8. Liao, J.; Xiao, J.; Zhou, Y.; Liu, Z.; Wang, C. Effect of transcatheter arterial chemoembolization on cellular immune function and regulatory T cells in patients with hepatocellular carcinoma. Mol. Med. Rep. 2015, 12, 6065–6071. [Google Scholar] [CrossRef]
  9. Leuchte, K.; Staib, E.; Thelen, M.; Gödel, P.; Lechner, A.; Zentis, P.; Garcia-Marquez, M.; Waldschmidt, D.; Datta, R.R.; Wahba, R.; et al. Microwave ablation enhances tumor-specific immune response in patients with hepatocellular carcinoma. Cancer Immunol. Immunother. 2020, 70, 893–907. [Google Scholar] [CrossRef]
  10. Zeng, P.; Shen, D.; Zeng, C.-H.; Chang, X.-F.; Teng, G.-J. Emerging Opportunities for Combining Locoregional Therapy with Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. Curr. Oncol. Rep. 2020, 22, 76. [Google Scholar] [CrossRef]
  11. Hickey, R.M.; Kulik, L.M.; Nimeiri, H.; Kalyan, A.; Kircher, S.; Desai, K.; Riaz, A.; Lewandowski, R.J.; Salem, R. Immuno-oncology and Its Opportunities for Interventional Radiologists: Immune Checkpoint Inhibition and Potential Synergies with Interventional Oncology Procedures. J. Vasc. Interv. Radiol. 2017, 28, 1487–1494. [Google Scholar] [CrossRef] [PubMed]
  12. Krokidis, M.E.; Orsi, F.; Katsanos, K.; Helmberger, T.; Adam, A. CIRSE Guidelines on Percutaneous Ablation of Small Renal Cell Carcinoma. Cardiovasc. Interv. Radiol. 2016, 40, 177–191. [Google Scholar] [CrossRef] [PubMed]
  13. Forner, A.; Reig, M.; de Lope, C.R.; Bruix, J. Current Strategy for Staging and Treatment: The BCLC Update and Future Prospects. Semin. Liver Dis. 2010, 30, 61–74. [Google Scholar] [CrossRef] [PubMed]
  14. Pereira, P.L.; Salvatore, M.; Cardiovascular and Interventional Radiological Society of Europe (CIRSE). Standards of Practice: Guidelines for Thermal Ablation of Primary and Secondary Lung Tumors. Cardiovasc. Interv. Radiol. 2012, 35, 247–254. [Google Scholar] [CrossRef] [PubMed]
  15. Gangi, A.; Tsoumakidou, G.; Buy, X.; Quoix, E. Quality Improvement Guidelines for Bone Tumour Management. Cardiovasc. Interv. Radiol. 2010, 33, 706–713. [Google Scholar] [CrossRef]
  16. Rangamuwa, K.; Leong, T.; Weeden, C.; Asselin-Labat, M.-L.; Bozinovski, S.; Christie, M.; John, T.; Antippa, P.; Irving, L.; Steinfort, D. Thermal ablation in non-small cell lung cancer: A review of treatment modalities and the evidence for combination with immune checkpoint inhibitors. Transl. Lung Cancer Res. 2021, 10, 2842–2857. [Google Scholar] [CrossRef] [PubMed]
  17. Takahashi, Y.; Matsutani, N.; Nakayama, T.; Dejima, H.; Uehara, H.; Kawamura, M. Immunological effect of local ablation combined with immunotherapy on solid malignancies. Chin. J. Cancer 2017, 36, 49. [Google Scholar] [CrossRef]
  18. Vanlangenakker, N.; Berghe, T.V.; Krysko, D.; Festjens, N.; Vandenabeele, P. Molecular Mechanisms and Pathophysiology of Necrotic Cell Death. Curr. Mol. Med. 2008, 8, 207–220. [Google Scholar] [CrossRef]
  19. Brok, M.H.M.G.M.D.; Sutmuller, R.P.M.; van der Voort, R.; Bennink, E.J.; Figdor, C.G.; Ruers, T.J.M.; Adema, G.J. In Situ Tumor Ablation Creates an Antigen Source for the Generation of Antitumor Immunity. Cancer Res. 2004, 64, 4024–4029. [Google Scholar] [CrossRef]
  20. Takahashi, Y.; Izumi, Y.; Matsutani, N.; Dejima, H.; Nakayama, T.; Okamura, R.; Uehara, H.; Kawamura, M. Optimized magnitude of cryosurgery facilitating anti-tumor immunoreaction in a mouse model of Lewis lung cancer. Cancer Immunol. Immunother. 2016, 65, 973–982. [Google Scholar] [CrossRef]
  21. Liang, S.; Niu, L.; Xu, K.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Lin, M. Tumor cryoablation in combination with natural killer cells therapy and Herceptin in patients with HER2-overexpressing recurrent breast cancer. Mol. Immunol. 2017, 92, 45–53. [Google Scholar] [CrossRef] [PubMed]
  22. McArthur, H.L.; Diab, A.; Page, D.B.; Yuan, J.; Solomon, S.B.; Sacchini, V.; Comstock, C.; Durack, J.C.; Maybody, M.; Sung, J.; et al. A Pilot Study of Preoperative Single-Dose Ipilimumab and/or Cryoablation in Women with Early-Stage Breast Cancer with Comprehensive Immune Profiling. Clin. Cancer Res. 2016, 22, 5729–5737. [Google Scholar] [CrossRef]
  23. Si, T.; Guo, Z.; Hao, X. Combined Cryoablation and GM-CSF Treatment for Metastatic Hormone Refractory Prostate Cancer. J. Immunother. 2009, 32, 86–91. [Google Scholar] [CrossRef]
  24. Yuanying, Y.; Lizhi, N.; Feng, M.; Xiaohua, W.; Jianying, Z.; Fei, Y.; Feng, J.; Lihua, H.; Jibing, C.; Jialiang, L.; et al. Therapeutic outcomes of combining cryotherapy, chemotherapy and DC-CIK immunotherapy in the treatment of metastatic non-small cell lung cancer. Cryobiology 2013, 67, 235–240. [Google Scholar] [CrossRef] [PubMed]
  25. Niu, L.-Z.; Li, J.-L.; Zeng, J.-Y.; Mu, F.; Liao, M.-T.; Yao, F.; Li, L.; Liu, C.-Y.; Chen, J.-B.; Zuo, J.-S.; et al. Combination treatment with comprehensive cryoablation and immunotherapy in metastatic hepatocellular cancer. World J. Gastroenterol. 2013, 19, 3473–3480. [Google Scholar] [CrossRef] [PubMed]
  26. Niu, L.; Chen, J.; He, L.; Liao, M.; Yuan, Y.; Zeng, J.; Li, J.; Zuo, J.; Xu, K. Combination Treatment With Comprehensive Cryoablation and Immunotherapy in Metastatic Pancreatic Cancer. Pancreas 2013, 42, 1143–1149. [Google Scholar] [CrossRef]
  27. Benzon, B.; Glavaris, S.A.; Simons, B.W.; Hughes, R.M.; Ghabili, K.; Mullane, P.; Miller, R.; Nugent, K.; Shinder, B.; Tosoian, J.; et al. Combining immune check-point blockade and cryoablation in an immunocompetent hormone sensitive murine model of prostate cancer. Prostate Cancer Prostatic Dis. 2018, 21, 126–136. [Google Scholar] [CrossRef]
  28. Zhang, M.; Yin, T.; Lu, Y.; Feng, H. The Application of Cytidyl Guanosyl Oligodeoxynucleotide Can Affect the Antitumor Immune Response Induced by a Combined Protocol of Cryoablation and Dendritic Cells in Lewis Lung Cancer Model. Med. Sci. Monit. 2016, 22, 1309–1317. [Google Scholar] [CrossRef] [PubMed]
  29. Lin, C.; Wang, Q.; Lu, G.; Yin, Z.; He, X.; Xu, H.; Pan, J.; Zhang, S. In-situ administration of dendritic cells following argon–helium cryosurgery enhances specific antiglioma immunity in mice. Neuroreport 2014, 25, 900–908. [Google Scholar] [CrossRef]
  30. Li, F.; Guo, Z.; Yu, H.; Zhang, X.; Si, T.; Liu, C.; Yang, X.; Qi, L. Anti-tumor immunological response induced by cryoablation and anti-CTLA-4 antibody in an in vivo RM-1 cell prostate cancer murine model. Neoplasma 2014, 61, 659–671. [Google Scholar] [CrossRef]
  31. Alteber, Z.; Azulay, M.; Cafri, G.; Vadai, E.; Tzehoval, E.; Eisenbach, L. Cryoimmunotherapy with local co-administration of ex vivo generated dendritic cells and CpG-ODN immune adjuvant, elicits a specific antitumor immunity. Cancer Immunol. Immunother. 2014, 63, 369–380. [Google Scholar] [CrossRef]
  32. Machlenkin, A.; Goldberger, O.; Tirosh, B.; Paz, A.; Volovitz, I.; Bar-Haim, E.; Lee, S.-H.; Vadai, E.; Tzehoval, E.; Eisenbach, L. Combined Dendritic Cell Cryotherapy of Tumor Induces Systemic Antimetastatic Immunity. Clin. Cancer Res. 2005, 11, 4955–4961. [Google Scholar] [CrossRef]
  33. Xu, H.; Wang, Q.; Lin, C.; Yin, Z.; He, X.; Pan, J.; Lu, G.; Zhang, S. Synergism between cryoablation and GM-CSF: Enhanced immune function of splenic dendritic cells in mice with glioma. Neuroreport 2015, 26, 346–353. [Google Scholar] [CrossRef]
  34. Brok, M.H.D.; Sutmuller, R.P.; Nierkens, S.; Bennink, E.J.; Toonen, L.W.; Figdor, C.G.; Ruers, T.J.; Adema, G.J. Synergy between In situ Cryoablation and TLR9 Stimulation Results in a Highly Effective In vivo Dendritic Cell Vaccine. Cancer Res. 2006, 66, 7285–7292. [Google Scholar] [CrossRef]
  35. Waitz, R.; Solomon, S.B.; Petre, E.N.; Trumble, A.E.; Fassò, M.; Norton, L.; Allison, J.P. Potent Induction of Tumor Immunity by Combining Tumor Cryoablation with Anti–CTLA-4 Therapy. Cancer Res. 2012, 72, 430–439. [Google Scholar] [CrossRef] [PubMed]
  36. Hamamoto, S.; Okuma, T.; Yamamoto, A.; Kageyama, K.; Takeshita, T.; Sakai, Y.; Nishida, N.; Matsuoka, T.; Miki, Y. Radiofrequency Ablation and Immunostimulant OK-432: Combination Therapy Enhances Systemic Antitumor Immunity for Treatment of VX2 Lung Tumors in Rabbits. Radiology 2013, 267, 405–413. [Google Scholar] [CrossRef] [PubMed]
  37. Habibi, M.; Kmieciak, M.; Graham, L.; Morales, J.K.; Bear, H.D.; Manjili, M.H. Radiofrequency thermal ablation of breast tumors combined with intralesional administration of IL-7 and IL-15 augments anti-tumor immune responses and inhibits tumor development and metastasis. Breast Cancer Res. Treat. 2008, 114, 423–431. [Google Scholar] [CrossRef] [PubMed]
  38. Zhu, J.; Yu, M.; Chen, L.; Kong, P.; Li, L.; Ma, G.; Ge, H.; Cui, Y.; Li, Z.; Pan, H.; et al. Enhanced antitumor efficacy through microwave ablation in combination with immune checkpoints blockade in breast cancer: A pre-clinical study in a murine model. Diagn. Interv. Imaging 2018, 99, 135–142. [Google Scholar] [CrossRef]
  39. Kuang, M.; Liu, S.Q.; Saijo, K.; Uchimura, E.; Huang, L.; Leong, K.; Lu, M.D.; Huang, J.F.; Ohno, T. Microwave tumour coagulation plus in situ treatment with cytokine-microparticles: Induction of potent anti-residual tumour immunity. Int. J. Hyperth. 2005, 21, 247–257. [Google Scholar] [CrossRef]
  40. Vanagas, T.; Gulbinas, A.; Pundzius, J.; Barauskas, G. Radiofrequency ablation of liver tumors (I): Biological background. Medicina 2010, 46, 13. [Google Scholar] [CrossRef]
  41. Slovak, R.; Ludwig, J.M.; Gettinger, S.N.; Herbst, R.S.; Kim, H.S. Immuno-thermal ablations—Boosting the anticancer immune response. J. Immunother. Cancer 2017, 5, 78. [Google Scholar] [CrossRef]
  42. Schneider, T.; Hoffmann, H.; Dienemann, H.; Herpel, E.; Heussel, C.P.; Enk, A.H.; Ring, S.; Mahnke, K. Immune Response After Radiofrequency Ablation and Surgical Resection in Nonsmall Cell Lung Cancer. Semin. Thorac. Cardiovasc. Surg. 2016, 28, 585–592. [Google Scholar] [CrossRef]
  43. Mizukoshi, E.; Yamashita, T.; Arai, K.; Sunagozaka, H.; Ueda, T.; Arihara, F.; Kagaya, T.; Yamashita, T.; Fushimi, K.; Kaneko, S. Enhancement of tumor-associated antigen-specific T cell responses by radiofrequency ablation of hepatocellular carcinoma. Hepatology 2012, 57, 1448–1457. [Google Scholar] [CrossRef] [PubMed]
  44. Tu, J.; Ji, J.; Wu, F.; Wang, Y.; Zhang, D.; Zhao, Z.; Ying, X. Effectiveness of Combined 131I-chTNT and Radiofrequency Ablation Therapy in Treating Advanced Hepatocellular Carcinoma. Cell Biochem. Biophys. 2014, 71, 777–784. [Google Scholar] [CrossRef] [PubMed]
  45. Da Costa, A.C.; Sodergren, M.; Jayant, K.; Cruz, F.S.; Spalding, D.; Pai, M.; Habib, N. Radiofrequency combined with immunomodulation for hepatocellular carcinoma: State of the art and innovations. World J. Gastroenterol. 2020, 26, 2040–2048. [Google Scholar] [CrossRef]
  46. Gameiro, S.R.; Higgins, J.P.; Dreher, M.R.; Woods, D.L.; Reddy, G.; Wood, B.J.; Guha, C.; Hodge, J.W. Combination Therapy with Local Radiofrequency Ablation and Systemic Vaccine Enhances Antitumor Immunity and Mediates Local and Distal Tumor Regression. PLoS ONE 2013, 8, e70417. [Google Scholar] [CrossRef]
  47. Nakagawa, H.; Mizukoshi, E.; Iida, N.; Terashima, T.; Kitahara, M.; Marukawa, Y.; Kitamura, K.; Nakamoto, Y.; Hiroishi, K.; Imawari, M.; et al. In vivo immunological antitumor effect of OK-432-stimulated dendritic cell transfer after radiofrequency ablation. Cancer Immunol. Immunother. 2014, 63, 347–356. [Google Scholar] [CrossRef]
  48. Ahmed, M.; Kumar, G.; Moussa, M.; Wang, Y.; Rozenblum, N.; Galun, E.; Goldberg, S.N. Hepatic Radiofrequency Ablation–induced Stimulation of Distant Tumor Growth Is Suppressed by c-Met Inhibition. Radiology 2016, 279, 103–117. [Google Scholar] [CrossRef]
  49. Greten, T.F.; Mauda-Havakuk, M.; Heinrich, B.; Korangy, F.; Wood, B.J. Combined locoregional-immunotherapy for liver cancer. J. Hepatol. 2019, 70, 999–1007. [Google Scholar] [CrossRef] [PubMed]
  50. Iezzi, R.; Contegiacomo, A.; Posa, A.; Attempati, N.; Punzi, E.; Tanzilli, A.; Margaritora, S.; Congedo, M.T.; Cassano, A.; Bria, E.; et al. Cryoablation in Locoregional Management of Complex Unresectable Chest Neoplasms. Tomography 2021, 7, 688–696. [Google Scholar] [CrossRef] [PubMed]
  51. Mehta, A.; Oklu, R.; Sheth, R.A. Thermal Ablative Therapies and Immune Checkpoint Modulation: Can Locoregional Approaches Effect a Systemic Response? Gastroenterol. Res. Pract. 2016, 2016, 9251375. [Google Scholar] [CrossRef]
  52. Yakkala, C.; Denys, A.; Kandalaft, L.; Duran, R. Cryoablation and immunotherapy of cancer. Curr. Opin. Biotechnol. 2020, 65, 60–64. [Google Scholar] [CrossRef] [PubMed]
  53. Shao, Q.; O’Flanagan, S.; Lam, T.; Roy, P.; Pelaez, F.; Burbach, B.J.; Azarin, S.M.; Shimizu, Y.; Bischof, J.C. Engineering T cell response to cancer antigens by choice of focal therapeutic conditions. Int. J. Hyperth. 2019, 36, 130–138. [Google Scholar] [CrossRef] [PubMed]
  54. Zhu, C.; Lin, S.; Liang, J.; Zhu, Y. PD-1 blockade enhances the anti-tumor immune response induced by cryoablation in a murine model of renal cell carcinoma. Cryobiology 2019, 87, 86–90. [Google Scholar] [CrossRef] [PubMed]
  55. Lin, M.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L.; Xu, K. Cryoablation combined with allogenic natural killer cell immunotherapy improves the curative effect in patients with advanced hepatocellular cancer. Oncotarget 2017, 8, 81967–81977. [Google Scholar] [CrossRef] [PubMed]
  56. Lin, M.; Xu, K.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L. Prospective study of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced renal cell cancer. Immunol. Lett. 2017, 184, 98–104. [Google Scholar] [CrossRef] [PubMed]
  57. Lin, M.; Liang, S.-Z.; Wang, X.-H.; Liang, Y.-Q.; Zhang, M.-J.; Niu, L.-Z.; Chen, J.-B.; Li, H.-B.; Xu, K.-C. Clinical efficacy of percutaneous cryoablation combined with allogenic NK cell immunotherapy for advanced non-small cell lung cancer. Immunol. Res. 2017, 65, 880–887. [Google Scholar] [CrossRef]
  58. Comen, E.A.; Bryce, Y.; Page, D.B.; Solomon, S.B.; Rodine, M.; Abaya, C.D.; Morris, E.A.; Plitas, G.; El-Tamer, M.; Gemignani, M.; et al. Preoperative checkpoint inhibition (CPI) and cryoablation (Cryo) in women with early-stage breast cancer (ESBC). J. Clin. Oncol. 2019, 37, 592. [Google Scholar] [CrossRef]
  59. Bulvik, B.E.; Rozenblum, N.; Gourevich, S.; Ahmed, M.; Andriyanov, A.V.; Galun, E.; Goldberg, S.N. Irreversible Electroporation versus Radiofrequency Ablation: A Comparison of Local and Systemic Effects in a Small-Animal Model. Radiology 2016, 280, 413–424. [Google Scholar] [CrossRef]
  60. Vivas, I.; Iribarren, K.; Lozano, T.; Cano, D.; Lasarte-Cia, A.; Chocarro, S.; Gorraiz, M.; Sarobe, P.; Hervás-Stubbs, S.; Bilbao, J.I.; et al. Therapeutic Effect of Irreversible Electroporation in Combination with Poly-ICLC Adjuvant in Preclinical Models of Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2019, 30, 1098–1105. [Google Scholar] [CrossRef]
  61. Alnaggar, M.; Lin, M.; Mesmar, A.; Liang, S.; Qaid, A.; Xu, K.; Chen, J.; Niu, L.; Yin, Z. Allogenic Natural Killer Cell Immunotherapy Combined with Irreversible Electroporation for Stage IV Hepatocellular Carcinoma: Survival Outcome. Cell. Physiol. Biochem. 2018, 48, 1882–1893. [Google Scholar] [CrossRef] [PubMed]
  62. Yang, Y.; Qin, Z.; Du, D.; Wu, Y.; Qiu, S.; Mu, F.; Xu, K.; Chen, J. Safety and Short-Term Efficacy of Irreversible Electroporation and Allogenic Natural Killer Cell Immunotherapy Combination in the Treatment of Patients with Unresectable Primary Liver Cancer. Cardiovasc. Interv. Radiol. 2018, 42, 48–59. [Google Scholar] [CrossRef]
  63. Zhao, J.; Wen, X.; Tian, L.; Li, T.; Xu, C.; Wen, X.; Melancon, M.P.; Gupta, S.; Shen, B.; Peng, W.; et al. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat. Commun. 2019, 10, 899. [Google Scholar] [CrossRef] [PubMed]
  64. Timmer, F.E.; Geboers, B.; Nieuwenhuizen, S.; Schouten, E.A.; Dijkstra, M.; de Vries, J.J.; Tol, M.P.V.D.; de Gruijl, T.D.; Scheffer, H.J.; Meijerink, M.R. Locally Advanced Pancreatic Cancer: Percutaneous Management Using Ablation, Brachytherapy, Intra-arterial Chemotherapy, and Intra-tumoral Immunotherapy. Curr. Oncol. Rep. 2021, 23, 68. [Google Scholar] [CrossRef] [PubMed]
  65. Narayanan, J.S.S.; Ray, P.; Hayashi, T.; Whisenant, T.C.; Vicente, D.; Carson, D.A.; Miller, A.M.; Schoenberger, S.P.; White, R.R. Irreversible Electroporation Combined with Checkpoint Blockade and TLR7 Stimulation Induces Antitumor Immunity in a Murine Pancreatic Cancer Model. Cancer Immunol. Res. 2019, 7, 1714–1726. [Google Scholar] [CrossRef]
  66. Lin, M.; Alnaggar, M.; Liang, S.; Wang, X.; Liang, Y.; Zhang, M.; Chen, J.; Niu, L.; Xu, K. An important discovery on combination of irreversible electroporation and allogeneic natural killer cell immunotherapy for unresectable pancreatic cancer. Oncotarget 2017, 8, 101795–101807. [Google Scholar] [CrossRef]
  67. Pan, Q.; Hu, C.; Fan, Y.; Wang, Y.; Li, R.; Hu, X. Efficacy of irreversible electroporation ablation combined with natural killer cells in treating locally advanced pancreatic cancer. J. BUON 2020, 25, 1643–1649. [Google Scholar] [PubMed]
  68. Lin, M.; Zhang, X.; Liang, S.; Luo, H.; Alnaggar, M.; Liu, A.; Yin, Z.; Chen, J.; Niu, L.; Jiang, Y. Irreversible electroporation plus allogenic Vγ9Vδ2 T cells enhances antitumor effect for locally advanced pancreatic cancer patients. Signal Transduct. Target. Ther. 2020, 5, 215. [Google Scholar] [CrossRef]
  69. Haen, S.P.; Pereira, P.L.; Salih, H.R.; Rammensee, H.-G.; Gouttefangeas, C. More Than Just Tumor Destruction: Immunomodulation by Thermal Ablation of Cancer. Clin. Dev. Immunol. 2011, 2011, 160250. [Google Scholar] [CrossRef]
  70. Chen, Z.; Shen, S.; Peng, B.; Tao, J. Intratumoural GM-CSF microspheres and CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a subcutaneous murine hepatoma model. Int. J. Hyperth. 2009, 25, 374–382. [Google Scholar] [CrossRef]
  71. Yu, M.-A.; Liang, P.; Yu, X.-L.; Han, Z.-Y.; Dong, X.-J.; Wang, Y.; Cheng, C.; Li, X. Multiple courses of immunotherapy with different immune cell types for patients with hepatocellular carcinoma after microwave ablation. Exp. Ther. Med. 2015, 10, 1460–1466. [Google Scholar] [CrossRef]
  72. Zhou, P.; Liang, P.; Dong, B.; Yu, X.; Han, Z.; Xu, Y. Phase I clinical study of combination therapy with microwave ablation and cellular immunotherapy in hepatocellular carcinoma. Cancer Biol. Ther. 2011, 11, 450–456. [Google Scholar] [CrossRef] [PubMed]
  73. Kallio, R.; Sequeiros, R.; Surcel, H.-M.; Ohtonen, P.; Kiviniemi, H.; Syrjälä, H. Early cytokine responses after percutaneous magnetic resonance imaging guided laser thermoablation of malignant liver tumors. Cytokine 2006, 34, 278–283. [Google Scholar] [CrossRef] [PubMed]
  74. Vogl, T.J.; Wissniowski, T.T.; Naguib, N.N.N.; Hammerstingl, R.M.; Mack, M.G.; Münch, S.; Ocker, M.; Strobel, D.; Hahn, E.G.; Hänsler, J. Activation of tumor-specific T lymphocytes after laser-induced thermotherapy in patients with colorectal liver metastases. Cancer Immunol. Immunother. 2009, 58, 1557–1563. [Google Scholar] [CrossRef]
  75. Inchingolo, R.; Posa, A.; Mariappan, M.; Spiliopoulos, S. Locoregional treatments for hepatocellular carcinoma: Current evidence and future directions. World J. Gastroenterol. 2019, 25, 4614–4628. [Google Scholar] [CrossRef]
  76. Iezzi, R.; Pompili, M.; Posa, A.; Coppola, G.; Gasbarrini, A.; Bonomo, L. Combined locoregional treatment of patients with hepatocellular carcinoma: State of the art. World J. Gastroenterol. 2016, 22, 1935–1942. [Google Scholar] [CrossRef]
  77. Iezzi, R.; Pompili, M.; La Torre, M.F.; Campanale, M.C.; Montagna, M.; Saviano, A.; Cesario, V.; Siciliano, M.; Annicchiarico, E.; Agnes, S.; et al. Radiofrequency ablation plus drug-eluting beads transcatheter arterial chemoembolization for the treatment of single large hepatocellular carcinoma. Dig. Liver Dis. 2015, 47, 242–248. [Google Scholar] [CrossRef] [PubMed]
  78. Llovet, J.M.; Brú, C.; Bruix, J. Prognosis of hepatocellular carcinoma: The BCLC staging classification. In Seminars in Liver Disease; Thieme Medical Publishers, Inc.: New York, NY, USA, 1999; Volume 19, pp. 329–338. [Google Scholar]
  79. European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2018, 69, 182–236, Erratum in J. Hepatol. 2019, 70, 817. [Google Scholar]
  80. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [PubMed]
  81. Pereira, P.L.; Iezzi, R.; Manfredi, R.; Carchesio, F.; Bánsághi, Z.; Brountzos, E.; Spiliopoulos, S.; Echevarria-Uraga, J.J.; Gonçalves, B.; Inchingolo, R.; et al. The CIREL Cohort: A Prospective Controlled Registry Studying the Real-Life Use of Irinotecan-Loaded Chemoembolisation in Colorectal Cancer Liver Metastases: Interim Analysis. Cardiovasc. Interv. Radiol. 2020, 44, 50–62. [Google Scholar] [CrossRef] [PubMed]
  82. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; van Krieken, J.H.; Aderka, D.; Aguilar, E.A.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. 2016, 27, 1386–1422. [Google Scholar] [CrossRef] [PubMed]
  83. Lammer, J.; Malagari, K.; Vogl, T.; Pilleul, F.; Denys, A.; Watkinson, A.; Pitton, M.; Sergent, G.; Pfammatter, T.; Terraz, S.; et al. Prospective Randomized Study of Doxorubicin-Eluting-Bead Embolization in the Treatment of Hepatocellular Carcinoma: Results of the PRECISION V Study. Cardiovasc. Interv. Radiol. 2010, 33, 41–52. [Google Scholar] [CrossRef] [PubMed]
  84. Lencioni, R.; De Baere, T.; Burrel, M.; Caridi, J.G.; Lammer, J.; Malagari, K.; Martin, R.C.G.; O’Grady, E.; Real, M.I.; Vogl, T.J.; et al. Transcatheter Treatment of Hepatocellular Carcinoma with Doxorubicin-loaded DC Bead (DEBDOX): Technical Recommendations. Cardiovasc. Interv. Radiol. 2012, 35, 980–985. [Google Scholar] [CrossRef]
  85. Ayaru, L.; Pereira, S.P.; Alisa, A.; Pathan, A.A.; Williams, R.; Davidson, B.; Burroughs, A.K.; Meyer, T.; Behboudi, S. Unmasking of α-Fetoprotein-Specific CD4+ T Cell Responses in Hepatocellular Carcinoma Patients Undergoing Embolization. J. Immunol. 2007, 178, 1914–1922. [Google Scholar] [CrossRef] [PubMed]
  86. Liao, Y.; Wang, B.; Huang, Z.-L.; Shi, M.; Yu, X.-J.; Zheng, L.; Li, S.; Li, L. Increased Circulating Th17 Cells after Transarterial Chemoembolization Correlate with Improved Survival in Stage III Hepatocellular Carcinoma: A Prospective Study. PLoS ONE 2013, 8, e60444. [Google Scholar] [CrossRef]
  87. Lee, H.L.; Jang, J.W.; Lee, S.W.; Yoo, S.H.; Kwon, J.H.; Nam, S.W.; Bae, S.H.; Choi, J.Y.; Han, N.I.; Yoon, S.K. Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in patients treated with transarterial chemoembolization. Sci. Rep. 2019, 9, 3260. [Google Scholar] [CrossRef]
  88. Zhang, J.; Li, H.; Gao, D.; Zhang, B.; Zheng, M.; Lun, M.; Wei, M.; Duan, R.; Guo, M.; Hua, J.; et al. A prognosis and impact factor analysis of DC-CIK cell therapy for patients with hepatocellular carcinoma undergoing postoperative TACE. Cancer Biol. Ther. 2018, 19, 475–483. [Google Scholar] [CrossRef]
  89. Semaan, A.; Dietrich, D.; Bergheim, D.; Dietrich, J.; Kalff, J.C.; Branchi, V.; Matthaei, H.; Kristiansen, G.; Fischer, H.-P.; Goltz, D. CXCL12 expression and PD-L1 expression serve as prognostic biomarkers in HCC and are induced by hypoxia. Virchows Arch. 2016, 470, 185–196. [Google Scholar] [CrossRef]
  90. Sangro, B.; Gomez-Martin, C.; de la Mata, M.; Iñarrairaegui, M.; Garralda, E.; Barrera, P.; Riezu-Boj, J.I.; Larrea, E.; Alfaro, C.; Sarobe, P.; et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J. Hepatol. 2013, 59, 81–88. [Google Scholar] [CrossRef]
  91. Duffy, A.G.; Makarova-Rusher, O.V.; Kerkar, S.P.; Kleiner, D.E.; Fioravanti, S.; Walker, M.; Carey, S.; Figg, W.D.; Steinberg, S.M.; Anderson, V.; et al. A pilot study of tremelimumab—A monoclonal antibody against CTLA-4—In combination with either trans catheter arterial chemoembolization (TACE) or radiofrequency ablation (RFA) in patients with hepatocellular carcinoma (HCC). J. Clin. Oncol. 2015, 33, 4081. [Google Scholar] [CrossRef]
  92. Duffy, A.G.; Ulahannan, S.V.; Makorova-Rusher, O.; Rahma, O.; Wedemeyer, H.; Pratt, D.; Davis, J.L.; Hughes, M.S.; Heller, T.; ElGindi, M.; et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J. Hepatol. 2016, 66, 545–551. [Google Scholar] [CrossRef]
  93. Salem, R.; Mazzaferro, V.; Sangro, B. Yttrium 90 radioembolization for the treatment of hepatocellular carcinoma: Biological lessons, current challenges, and clinical perspectives. Hepatology 2013, 58, 2188–2197. [Google Scholar] [CrossRef] [PubMed]
  94. Salem, R.; Johnson, G.E.; Kim, E.; Riaz, A.; Bishay, V.; Boucher, E.; Fowers, K.; Lewandowski, R.; Padia, S.A. Yttrium-90 Radioembolization for the Treatment of Solitary, Unresectable HCC: The LEGACY Study. Hepatology 2021, 74, 2342–2352. [Google Scholar] [CrossRef] [PubMed]
  95. Stella, M.; Braat, A.J.A.T.; van Rooij, R.; de Jong, H.W.A.M.; Lam, M.G.E.H. Holmium-166 Radioembolization: Current Status and Future Prospective. Cardiovasc. Interv. Radiol. 2022, 45, 1634–1645. [Google Scholar] [CrossRef]
  96. Bilbao, J.L.; Iezzi, R.; Goldberg, S.N.; Sami, A.; Akhan, O.; Giuliante, F.; Pompili, M.; Crocetti, L.; Malagari, K.; Valentini, V.; et al. The ten commandments of hepatic radioembolization: Expert discussion and report from Mediterranean Interventional Oncology (MIOLive) congress 2017. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4014–4021. [Google Scholar] [PubMed]
  97. Riaz, A.; Lewandowski, R.J.; Kulik, L.M.; Mulcahy, M.F.; Sato, K.T.; Ryu, R.K.; Omary, R.A.; Salem, R. Complications Following Radioembolization with Yttrium-90 Microspheres: A Comprehensive Literature Review. J. Vasc. Interv. Radiol. 2009, 20, 1121–1130. [Google Scholar] [CrossRef]
  98. Chew, V.; Lee, Y.H.; Pan, L.; Nasir, N.J.M.; Lim, C.J.; Chua, C.; Lai, L.; Hazirah, S.N.; Lim, T.K.H.; Goh, B.K.P.; et al. Immune activation underlies a sustained clinical response to Yttrium-90 radioembolisation in hepatocellular carcinoma. Gut 2018, 68, 335–346. [Google Scholar] [CrossRef] [PubMed]
  99. Zhan, C.; Ruohoniemi, D.; Shanbhogue, K.P.; Wei, J.; Welling, T.H.; Gu, P.; Park, J.S.; Dagher, N.N.; Taslakian, B.; Hickey, R.M. Safety of Combined Yttrium-90 Radioembolization and Immune Checkpoint Inhibitor Immunotherapy for Hepatocellular Carcinoma. J. Vasc. Interv. Radiol. 2019, 31, 25–34. [Google Scholar] [CrossRef] [PubMed]
  100. Wehrenberg-Klee, E.; Goyal, L.; Dugan, M.; Zhu, A.X.; Ganguli, S. Y-90 Radioembolization Combined with a PD-1 Inhibitor for Advanced Hepatocellular Carcinoma. Cardiovasc. Interv. Radiol. 2018, 41, 1799–1802. [Google Scholar] [CrossRef]
  101. Tai, W.M.D.; Loke, K.S.H.; Gogna, A.; Tan, S.H.; Ng, D.C.E.; Hennedige, T.P.; Irani, F.; Lee, J.J.X.; Too, C.W.; Ng, M.C.; et al. A phase II open-label, single-center, nonrandomized trial of Y90-radioembolization in combination with nivolumab in Asian patients with advanced hepatocellular carcinoma: CA 209-678. J. Clin. Oncol. 2020, 38, 4590. [Google Scholar] [CrossRef]
Figure 1. Interventional oncology ablative and endovascular techniques and mode of action. IRE: irreversible electroporation; RFA: radiofrequency ablation; MWA: microwave ablation; TACE: transcatheter arterial chemoembolization; TARE: transcatheter arterial radioembolization.
Figure 1. Interventional oncology ablative and endovascular techniques and mode of action. IRE: irreversible electroporation; RFA: radiofrequency ablation; MWA: microwave ablation; TACE: transcatheter arterial chemoembolization; TARE: transcatheter arterial radioembolization.
Ijms 24 07344 g001
Table 1. Pros and cons of locoregional percutaneous interventional oncology treatments plus immunotherapy.
Table 1. Pros and cons of locoregional percutaneous interventional oncology treatments plus immunotherapy.
TechniquePROSCONS
RFARFA + CpG-B increase activating lymphocytes and OS [41]
RFA + monoclonal antibody increase OS [44,45]
RFA + a vaccine encoding CEA produce regression of distal metastasis and increases CEA-specific CD4+ T cells [46]
RFA + dendritic cells stimulated by OK-432 increase the number of CD8+ T cells infiltrating untreated secondary tumors [47]
RFA alone stimulates HGF and VEGF, increases microvascular density and tumor cell replication [48]
Incomplete ablation enhances neo-angiogenesis and tumor growth [49]
CryoablationCA + anti-PDL1 drugs lead to anti-tumor immune responses and delayed tumor growth of distant untreated tumors [54]
CA + CpG-B induces regression of existing secondary tumors [19]
CA + immunotherapy increases OS [25]
CA + allogeneic NK cells increase OS [55,56,57]
CA + anti-CTLA4 and anti-PD1 increase OS [58]
Low volume and level of literature evidence compared to RFA
IREIRE + poly-ICLC increases immunogenic response and reduces tumor growth [60]
IRE + allogeneic NK cell immunotherapy increases OS, PFS, and decreases AFP [61,62]
IRE + anti-PD1 drug promotes CD8+ T cell infiltration and increases OS [64]
IRE + anti-PD1 and TLR-7 agonist improves local response and regression of untreated lesions [65]
IRE + NK cells or allogenic Vγ9Vδ2 T cell infusion improves PFS, OR, OS rates [66,67,68]
Low volume and level of literature evidence compared to RFA
MWAMWA + GM-CSF increases DFS and decreases tumor volume [70]
MWA + adoptive immunotherapy increases peripheral lymphocytes [72]
MWA seems to be less immunogenic compared to RFA and CA [69]
HIFU/LiTTHIFU and LiTT increase cytokine levels [69,73,74]HIFU induces less immunogenic effect compared with RFA and cryoablation [69]
RFA: radiofrequency ablation; OS: overall survival; CEA: carcinoembryonic antigen; CA: cryoablation; NK: natural killer; IRE: irreversible electroporation; PFS: progression-free survival; AFP: alpha-fetoprotein; OR: overall response; MWA: microwave ablation; DFS: disease-free survival; HIFU: high-intensity focused ultrasound; LiTT: laser-induced thermal therapy.
Table 2. Ablative technique, immunotherapy agents, and target lesions.
Table 2. Ablative technique, immunotherapy agents, and target lesions.
TechniqueImmunotherapy AgentTarget Lesion and Study Type
RFAMonoclonal antibody [44]Intermediate- to advanced-stage HCC in liver cancer murine model study [44].
CEA-encoding vaccine [46] Distal colorectal cancer metastasis murine model study [46]
OK-432-stimulated dendritic cells transfer [47]In vivo untreated secondary tumors [47]
CAAnti-PDL1 [54]Distant untreated tumors in renal cell carcinoma murine model study [54]
CpG-B [19]Secondary tumors in melanoma murine model study [19]
ImmunotherapyMetastatic liver cancer patients [25]
Allogeneic NK cells [55,56,57]Lung cancer, renal cancer, or HCC patients [55,56,57]
Anti-CTLA4 and anti-PD1 [58]Breast cancer patients [58]
IREPoly-ICLC [60]Mice and rabbit HCC model study [60]
Allogeneic NK cells [61,62]Patients with metastatic liver tumor [61,62]
Anti-PD1 [64]Pancreatic ductal adenocarcinoma [64]
Anti-PD1 and TLR-7 agonist [65]Pancreatic ductal adenocarcinoma murine model [65]
NK cells or allogeneic Vγ9Vδ2 T cell infusion [66,67,68]Patients with pancreatic ductal adenocarcinoma [66,67,68]
MWAGM-CSF [70]Murine hepatoma model [70]
Adoptive immunotherapy [72]HCC patients [72]
RFA: radiofrequency ablation; HCC: hepatocellular carcinoma; CEA: carcinoembryonic antigen; CA: cryoablation; NK: natural killer; IRE: irreversible electroporation; MWA: microwave ablation.
Table 3. Pros and cons of locoregional endovascular interventional oncology treatments plus immunotherapy.
Table 3. Pros and cons of locoregional endovascular interventional oncology treatments plus immunotherapy.
TechniquePROSCONS
TACETACE + CTLA4 inhibitor increases OS [91,92]
TACE + ablation and tremelimumab increases OS, reduces tumor load, reduces non-ablated or non-embolized tumors [93]
Low volume and level of literature evidence
Phase-I and -II clinical trials ongoing
TARETARE + nivolumab (or nivolumab + ipilimumab) showed good results in terms of OS and PFS [100]Low volume and level of literature evidence
Phase-I and -II clinical trials ongoing
TACE: transarterial chemoembolization; OS: overall survival; TARE: transarterial radioembolization; PFS: progression-free survival.
Table 4. Endovascular technique, immunotherapy agents, and target lesions.
Table 4. Endovascular technique, immunotherapy agents, and target lesions.
TechniqueImmunotherapy AgentTarget Lesion and Study Type
TACECTLA4 inhibitor [91,92]Advanced HCC patients [91,92]
TACE + ablationTremelimumab [93]Advanced HCC patients [93]
TARENivolumab [100]Retrospective study on advanced HCC patients [100]
TACE: transarterial chemoembolization; HCC: hepatocellular carcinoma; TARE: transarterial radioembolization.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Posa, A.; Contegiacomo, A.; Ponziani, F.R.; Punzi, E.; Mazza, G.; Scrofani, A.; Pompili, M.; Goldberg, S.N.; Natale, L.; Gasbarrini, A.; et al. Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends. Int. J. Mol. Sci. 2023, 24, 7344. https://doi.org/10.3390/ijms24087344

AMA Style

Posa A, Contegiacomo A, Ponziani FR, Punzi E, Mazza G, Scrofani A, Pompili M, Goldberg SN, Natale L, Gasbarrini A, et al. Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends. International Journal of Molecular Sciences. 2023; 24(8):7344. https://doi.org/10.3390/ijms24087344

Chicago/Turabian Style

Posa, Alessandro, Andrea Contegiacomo, Francesca Romana Ponziani, Ernesto Punzi, Giulia Mazza, Annarita Scrofani, Maurizio Pompili, Shraga Nahum Goldberg, Luigi Natale, Antonio Gasbarrini, and et al. 2023. "Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends" International Journal of Molecular Sciences 24, no. 8: 7344. https://doi.org/10.3390/ijms24087344

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop