Review Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 7, 2022; 28(13): 1288-1303
Published online Apr 7, 2022. doi: 10.3748/wjg.v28.i13.1288
Locoregional therapies and their effects on the tumoral microenvironment of pancreatic ductal adenocarcinoma
Thomas Lambin, Cyril Lafon, Robert Andrew Drainville, LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, Lyon 69003, France
Thomas Lambin, Mathieu Pioche, Department of Gastroenterology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69008, France
Frédéric Prat, Service d’Endoscopie Digestive, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy 92110, France
Frédéric Prat, INSERM U1016, Institut Cochin, Université de Paris, Paris 75014, France
ORCID number: Thomas Lambin (0000-0002-9437-0154); Cyril Lafon (0000-0003-1550-970X); Robert Andrew Drainville (0000-0003-2922-9522); Mathieu Pioche (0000-0002-6482-2375); Frédéric Prat (0000-0002-6018-0491).
Author contributions: Lambin T and Prat F reviewed the literature and prepared the manuscript; Lafon C, Drainville RA, and Pioche M contributed to and revised the manuscript; all authors approved the final manuscript.
Supported by the Labex DEVweCan (Université de Lyon) and PCSI ITMO Cancer INSERM.
Conflict-of-interest statement: All authors declare no conflicts-of-interest related to this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Frédéric Prat, MD, Doctor, Service d’Endoscopie Digestive, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 100 Bd du Général Leclerc, Clichy 92110, France. Frederic.prat@aphp.fr
Received: December 7, 2021
Peer-review started: December 7, 2021
First decision: January 27, 2022
Revised: February 10, 2022
Accepted: February 27, 2022
Article in press: February 27, 2022
Published online: April 7, 2022
Processing time: 112 Days and 18.3 Hours

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second leading cause of death from cancer by 2030. Despite intensive research in the field of therapeutics, the 5-year overall survival is approximately 8%, with only 20% of patients eligible for surgery at the time of diagnosis. The tumoral microenvironment (TME) of the PDAC is one of the main causes for resistance to antitumoral treatments due to the presence of tumor vasculature, stroma, and a modified immune response. The TME of PDAC is characterized by high stiffness due to fibrosis, with hypo microvascular perfusion, along with an immunosuppressive environment that constitutes a barrier to effective antitumoral treatment. While systemic therapies often produce severe side effects that can alter patients’ quality of life, locoregional therapies have gained attention since their action is localized to the pancreas and can thus alleviate some of the barriers to effective antitumoral treatment due to their physical effects. Local hyperthermia using radiofrequency ablation and radiation therapy - most commonly using a local high single dose - are the two main modalities holding promise for clinical efficacy. Recently, irreversible electroporation and focused ultrasound-derived cavitation have gained increasing attention. To date, most of the data are limited to preclinical studies, but ongoing clinical trials may help better define the role of these locoregional therapies in the management of PDAC patients.

Key Words: Pancreatic ductal adenocarcinoma, Tumoral microenvironment, Stroma, Hyperthermia, Radiation therapy, High-intensity focused ultrasound

Core Tip: The prognosis of pancreatic ductal adenocarcinoma is poor, with a 5-year survival rate of approximately 8%. This is mainly due to an unfavorable tumoral microenvironment (tumor vasculature, stroma, and immune response). Locoregional therapies can alleviate some barriers to effective antitumoral treatment. This review explores the action of locoregional treatments on pancreatic cancer, with a specific focus on hyperthermia, radiation therapy, high-intensity focused ultrasound, and irreversible electroporation. After a description of the particularities of the tumoral microenvironment of pancreatic cancer, the effects of these treatments on the tumoral microenvironment and implications for future management of patients are discussed.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) is one of the most common types of pancreatic cancer[1]. Given the rapidly increasing incidence of PDAC in recent decades, it is expected to become the second leading cause of cancer death by 2030 in the United States and other industrialized countries[2]. The 5-year survival rate is approximately 8% following diagnosis[3], with only 20% of the patients eligible for surgery[4]. Among the patients who undergo surgery, the 5-year survival rate is only 20% due to rapid recurrence and metastasis development[5]. Systemic treatments are associated with various types of adverse events that result in a poor quality of life for patients[6] and are often inefficient due to the characteristics of the PDAC tumor microenvironment (TME), which protects tumor cells from chemotherapies and immunotherapies. The TME is increasingly being considered a potential target of choice to improve outcomes for PDAC. Locoregional treatments, such as hyperthermia (HT) with microwave ablation or radiofrequency ablation (RFA), radiation therapy (RT), irreversible electroporation (IRE), and high-intensity focused ultrasound (HIFU) therapy, are gaining increasing attention for their ability to specifically target the tumor while limiting deleterious systemic adverse events and may often be used in combination with anticancerous drugs. Among other effects, locoregional therapies can induce changes in the structure, components, and properties of the TME that may help alleviate some of the barriers to successful treatment. In this review, we will summarize the characteristics of the TME in PDAC and describe the effects of locoregional therapies on vasculature, stroma, and immune response. We will pay special attention on characterizing the TME for clinical applications (Table 1).

Table 1 Locoregional therapies and their main effects on the tumoral microenvironment of pancreatic ductal adenocarcinoma.

Vasculature
Stroma
Immune response
HyperthermiaIncreased blood flow and vascular permeability. Recruitment of bradykinin and histamin. Increased iNOS.Destructuration of collagen fibers. Reduction of CAF. Reduction of tumor stiffness.Promotes APC activation. Increased infiltrating CD8+. Increased pro-inflammatory cytokines. Abscopal effect (RFA).
Radiation therapyReduced blood perfusion. Destructuration of microvessels with thickening vessel walls. Platelet aggregation. Microthrombus formation. Increased HIF-1 and VEGF. Increased vascular permeability.Accumulation of extracellular matrix proteins. Increased stromal cells (fibroblasts). Thickened and stiffened tissue. Loss of hyaluronic acid. Collagen remodeling. Modification of CAF population.Release of tumor antigens (DAMPs) ≥ APC presentation and CD8+ activation. Increased peptide availability and T cell repertoire. Release of inflammatory cytokines, CD8+, and CD4+ cells. Increased adhesion molecules (VCAM-1, ICAM-1). T cells homing. Increased PDL-1.
HIFU (mechanical effect)Reduced blood perfusion and microvascular density.Disruption of the collagen matrix.Released DAMPs ≥ T cell activation. Induction of Th1 inflammation. Increased CD8+/Treg ratio.
PDAC MICROENVIRONMENT

Although most solid tumors develop specific interactions with their host via neoangiogenesis, the creation of a supporting network of cells and extracellular matrix, and some form of immunomodulation, PDAC remains one of the most stroma-rich cancers, with 90% of its tumor mass composed of fibroblasts and their products[7]. The PDAC stroma is composed of an acellular and cellular compartment. The acellular compartment consists of an extracellular matrix rich in collagen, fibronectin, laminin, integrins, glycosaminoglycan, matrix metalloproteinase and secreted protein acids and is rich in cysteine[1,8] with cytokines and growth factors[9]. The cellular component is composed of pancreatic stellate cells and immune cells, such as macrophages, mast cells, lymphocytes, and plasma cells[1]. Pancreatic stellate cells, which are resident cells of the pancreas, acquire an activated myofibroblast-like phenotype when activated and are assumed to be the main regulators of TME extracellular matrix production. With the tumoral microvasculature, all these elements form the TME. Interactions between acellular and cellular components of the TME are key factors in PDAC progression[10]. In PDAC, the TME has been associated with tumoral progression, metastasis dissemination, and resistance to chemotherapy by various mechanisms[8]. The abundant extracellular matrix increases intratumoral interstitial pressure[11] and acts as a barrier for drug delivery with compression of blood vessels. Tumor stiffness can also have a direct effect on chemosensitivity: in vitro, mechanically compressed PDAC spheroids (displaying high stiffness) are less sensitive to gemcitabine than free spheroids (without any mechanical compression), whereas there is no difference in Hoechst dye penetration between compressed and free spheroids, suggesting a therapeutic effect of compression independent of the sole penetration of gemcitabine into the spheroid cells[12]. One hypothesis to explain this phenomenon is that mechanical stress decreases cell proliferation, which may alter the efficacy of chemotherapies targeting proliferating cells[12]. Intratumoral stiffness itself can modify intracellular signaling pathways and promote epithelial-mesenchymal transition, leading to tumoral progression and chemoresistance[13]. In PDAC, the microvascular density is generally low and leaky. Combined with mechanical forces caused by the dense stroma and tumoral growth that compresses the vessels, limited perfusion can result, which is responsible for hypoxia and low nutrient availability along with low anticancer drug delivery[1,14]. On the immune response side, the TME in PDAC is characterized by a reduced number of cytotoxic T cells, along with an increase in M2 macrophages, N2 neutrophils, and T-regulatory cells at the tumor site, which all contribute to an immunosuppressive environment[15].

Current studies suggest that the TME is an attractive target in the management of PDAC. Provenzano et al[11] showed that the administration of an enzymatic agent allowing the deletion of stromal hyaluronic acid (PEGPH20) in a murine model of PDAC led to normalized intratumoral interstitial pressure, expansion of the tumoral microvasculature, and increased survival in vivo when combined with gemcitabine compared to gemcitabine alone. However, caution should be taken since other studies have shown that the presence of some TME elements serves to prevent cancer progression and should therefore not be suppressed indistinctively[16,17]. In a mouse model of PDAC, as well as in patients with pancreatic cancer, a decreased bulk of aSMA + myofibroblasts was associated with poor prognosis and reduced overall survival[17].

HT

HT is a therapeutic procedure used to increase the intratumoral temperature. There are various ways to increase temperature: for superficial tumors, local HT can be applied by means of antennas or applicators that emit microwaves (microwave ablation) or radio waves (radiofrequency ablation or RFA) placed at the surface of the tumor with an intervening medium. Interstitial and endocavitary HT are used for small tumors in which an intratumoral implantation of the antenna can deliver various types of waves: microwaves, radiofrequency, ultrasound, heat sources, or laser fibers. Regional HT and partial body HT are more suitable for deep seated tumors, such as PDAC, that can be heated by antennas placed in rings around the patient. Whole-body HT is dedicated to the treatment of metastatic tumors[18]. HT can be generated by an external source, such as hot air or infrared radiation, or an internal source, such as magnetic nanoparticles, which can be deposited in the region of interest and then exposed to a magnetic field, leading to an increase in temperature and allowing localized heating [magnetic hyperthermia (MHT)][14]. Nanoparticles can also be used for photothermal therapy (PTT), during which a laser can activate nanophotoabsorbers. MHT and PTT are both nanoparticle-based HT treatments[19,20].

Hyperthermia and tumoral vasculature

HT is thought to increase blood flow and vascular permeability with an increase in antitumor effects. A study by Miyamoto et al[21] evaluated the effects of mild HT on the efficacy and accumulation of an anti-EGFR agent (cetuximab) in various xenograft mouse models of pancreatic cancer. Using a water bath, a temperature of 37 °C or 41 °C was applied to the tumor and allowed to decrease the tumoral volume compared to mice exposed to a standard ambient temperature of 25 °C (control). This was accompanied by an increase in cetuximab accumulation in cancer cells by 2.5- to 5-fold, depending on the model studied. Of note, this effect was also observed when stromal dense tissue derived from surgically resected pancreatic cancer was transplanted into mice. This effect may be due to an increase in blood flow with an increase in tumor vessel permeability[22]. Indeed, another study showed that mild HT in a mouse model of pancreatic cancer could induce an 11-fold increase in blood perfusion in a reversible manner during heating and a 3-fold increase after the end of HT treatment along with an increase in vasculature permeability and an enhanced extravasation of macromolecules[23]. In a xenograft mouse model of breast cancer, non invasive radiofrequency (RF) produced increased transport and perfusion of fluorescent tracers into the tumors at temperatures below 41°C, whereas vessel deformation and blood coagulation were observed when the temperature reached 44°C[24]. The mechanism through which HT increases blood perfusion may be linked to a relaxation of smooth muscle following an increase in nitric oxide synthetized by endothelial cells. In a study by Song et al[25], the content of inducible nitric oxide synthase (iNOS) was evaluated in a murine model of fibrosarcoma following HT. No iNOS was detectable before HT treatment, while an increase in iNOS was observed 3 h after HT and remained detectable 24 h after treatment. Additionally, HT increases the recruitment of bradykinin and histamine molecules responsible for vessel dilation and the recruitment of capillaries[26].

Hyperthermia and stromal architecture

HT has been shown to disrupt the stromal architecture. In a study by Piehler et al[14], Achilles tendons (mainly composed of type I collagen) were exposed to various regimens of either extrinsic or ion-oxide nanoparticle-based MHT. The amount of intact collagen fibers decreased with the application of HT, with only 10% of collagen fibers intact after 1 h of the 42 °C regimen and almost complete degradation after 1 h of the 50 °C or 70 °C regimen. Mild HT applied to spheroids of pancreatic cancer cells (Panc1) and fibroblasts (WI 38) significantly decreased the amount of intact collagen fibers, with a coinciding decrease in spheroid volume and cell viability by apoptotic and necrotic processes[14]. Local HT through the use of a photothermal agent combined with a photothermal-chemotherapeutic agent (Abraxane@Mose2) and subsequently irradiated by a laser beam could disrupt tissue architecture and reduce the number of carcinoma-associated fibroblasts (CAFs), subsequently enhancing the efficacy of Abraxane in a mouse model of PDAC[27]. Similarly, in a mouse model of cholangiocarcinoma, a nanoheater used for PTT (multifunctional iron oxide nanoflowers decorated with gold particles) with high uptake by CAFs produced a significant depletion of CAFs as well as a reduction in tumor stiffness followed by significant tumor regression[28]. In another study by Marangon et al[29], tumor stiffness was monitored following PTT in squamous cell carcinoma in mice. Shear wave elastrography revealed a transient and reversible increase in tumor stiffness after thermal ablation or mild HT, followed by a return to its initial value within 24 h of laser exposure in the case of thermal ablation or a reduced level for mild HT. Additionally, while increased tumor stiffness was observed in untreated mice, the stiffness in the treated group was stable over time. In the same study, second harmonic generation was used to evaluate the effect of PTT on collagen structure and revealed a destructuration of collagen fibers in the vicinity of heated carbon nanotubes.

In several mouse models of pancreatic cancer displaying various levels of stroma formation, the concomitant application of mild hyperthermia and cetuximab induced a more significant antitumoral effect on stroma-rich models[21]. In vitro, noninvasive RF has been shown to affect molecular transport in a 3D model of PDAC with increased diffusion of DAPI fluorescence in spheroids following RF compared to no treatment[30]. In a xenograft mouse model of squamous cell carcinoma, the combination of MHT and doxorubicin demonstrated a more efficient reduction in tumor growth than doxorubicin alone[31]. In the same study, the space between collagen fibers was determined following MHT: while there were no differences between the control group and the group injected with nanocubes without exposure to a magnetic field (no HT), there was an increase in the interfibrillar space between the group injected and exposed to the magnetic field compared to the injected group without exposure to the magnetic field[31].

Hyperthermia and immune response

Data on the effect of HT on the immune response in PDAC are scarce, but the literature is abundant for other types of cancer. HT promotes antigen presenting cell (APC) activation and antigen-specific naïve CD8+ T cell differentiation, allows CD4+ T cells to shift towards the Th1 phenotype, and transforms regulatory T cells (Tregs) into Th17 cells[32]. In a mouse model of PDAC, RFA induced an increase in infiltrating CD8+ T cells and a decrease in Treg cells but showed no difference in the proportion of infiltrating CD4+ lymphocytes[33]. HT has been shown to induce chemokine production, such as CCL21, combined with adhesion factors (selectin, integrin, ICAM-1), thus allowing an increase in the interactions between lymphocytes and endothelial cells and the homing of lymphocytes[34-37]. In parallel, HT induces the production of various proinflammatory cytokines, such as IL6[38]. HT induces immunogenic cell death through various mechanisms[32], such as triggering DNA damage that produces mutations in tumor cell genes, which generate neoantigens that stimulate the T cell-based immune response[32,39]. HT can also generate damage-associated molecular patterns (DAMPs), of which heat shock proteins (HSP) are the most important but also include molecules such as calreticulin, HMGB1 or ATP. HSPs are chaperones that participate in the presentation of the chaperoned antigen to the MHC-1 complex of dendritic cells, thus allowing antigen-specific T-cell activation[34,35,40]. High levels of HSP are associated with poor prognosis in parallel to an enhanced immune response[32]. Membrane HSP has been found to be a tumor-specific target for natural killer cells, whereas extracellular HSP can be considered a potent adjuvant to facilitate tumor antigen presentation and the induction of antitumor immunity[32,41,42]. More specifically, HSP70 has been shown to induce tumor cell proliferation in a mouse model of PDAC by activating AKT-mTOR signaling[33]. HSP60 has been shown to induce IFNg secretion and T cell upregulation[43]. In a murine model of PDAC, a study by Lin et al[44] found that the maximum HSP synthesis was achieved at 43 °C, corresponding with an increased antitumor immune response. Beyond this temperature, both the release of HSP and the associated immune response decreased[34,44]. The accumulation of neoantigens, secondary to mutations and DAMPs, favors the activation of dendritic cells, allowing the transformation of the tumoral immunosuppressive microenvironment by inhibiting Treg cells and promoting tumor-infiltrating lymphocyte maturation[32].

The so-called “abscopal effect” has often been invoked to suggest that an immunomodulating mechanism had to take place when the local treatment of a malignant tumor - most commonly the use of RT - results in a response at a distant location[45,46]. In a PDAC mouse model implanted with tumors on both flanks, Fei et al[47] tried to determine whether RFA on one flank’s tumor could affect the untreated tumor located on the other side. After RFA on one side, the immune response on the opposite side showed an increase in CD8+/PD-1+ T cells, along with suppression of immunosuppressive components of the tumor microenvironment (i.e., Tregs, tumor-associated macrophages, and tumor-associated neutrophils). Additionally, immune checkpoints such as PD-1 and LAG3 were upregulated in distant (untreated) T cells after one-sided RFA. Similarly, Gameiro et al[48] found that RFA induced local immunogenic modulation at the tumor surface in a model of colon adenocarcinoma, and the combination of RFA with vaccine therapy eradicated both primary and secondary tumors. Finally, in a clinical study for 10 patients with locally advanced pancreatic cancer (LAPC) that evaluated the immune response following coagulation necrosis-inducing RFA ablation, an increase in CD4+, CD8+, and effector memory T cells along with IL 6 was seen[49].

RT

RT uses an ionizing radiation beam (X-rays) whose energy is deposited in water along its path, leading to the formation of free radicals (reactive oxygen species or reactive nitrogen species) that oxidize molecular targets, provoking a dysregulation of cellular functions. These free radicals target DNA, leading to single- or double-strand breaks[50]. Today, there is no consensus on the role of RT in PDAC. The LPA07 trial did not show any improvement to the tumor in a small number of fractions to minimize the impact on the surrounding organs.

Radiation therapy and tumor vasculature

The effect of RT on tumor vasculature has been widely explored in various types of cancers. RT has been shown to have many direct or indirect effects on endothelial cells[50], and these effects are dependent on the dose received and the radiation schedule[51]. High single doses of radiation have been shown to cause vascular damage with reduced blood perfusion and hypoxia[52]. RT induces changes in tumor vasculature by destructuring microvessels and thickening vessel walls, thus reducing vessel lumen, all of which favor atherosclerosis. RT also induces platelet aggregation and microthrombus formation with an increase in inflammatory cell adhesion to endothelial cells[53]. RT can regulate and stabilize the level of HIF-1, leading to the production of VEGF, which is responsible for endothelial cell proliferation and increased survival. RT can directly upregulate the expression of avb3 integrins[54] and adhesion proteins[50]. In pancreatic cells, HIF-1 has been shown to induce the sonic hedgehog protein, leading to the formation of a stroma-rich microenvironment[55]. In a rodent model of pancreatic tumor, a single high dose (SHD) of radiation led to temporary vascular dysfunction along with enhanced expression of HIF-1, which could be restored after 14 d. However, vascular permeability was higher in irradiated tumors 14 d after RT[56]. Similarly, a study by Lee et al[57] evaluated the effect of an SHD of radiation vs a fractionated regimen of radiation, which showed increased perfusion ability of tumor vessels following SHD, whereas fractionated RT had no effect. Mechanisms were further studied and showed that vessels treated with SHD-RT had lower pericyte coverage; increased vessel perfusion could therefore be due to an increased leakage of immature vessels, and the surviving vessels after SHD-RT might favor the penetration of small molecule drugs.

RT and stroma

RT induces chronic inflammation, leading to fibrosis through the accumulation of extracellular matrix proteins and an increase in stromal cells such as fibroblasts[58], which thicken and stiffen the tissue[51,59]. Fibrosis formation depends on the dose of radiation received. For example, in a 3D model of mammary cancer stroma, increasing RT doses resulted in a reduction in fibroblast proliferation and activation along with a modest increase in matrix stiffness[60]. RT induces a loss of hyaluronic acid along with a remodeling of collagen and a modification of CAF population[50]. Protease activity is also altered with an upregulation of MMP2[61], which is responsible for an increase in tumor invasiveness[58]. In vitro studies showed that human lung fibroblasts develop an irreversible senescent phenotype after exposure to a radiation dose higher than 10 Gy, while lower doses induced reversible DNA damage without growth arrest[51,62,63]. Senescent fibroblasts can release proteolytic enzymes, cytokines, growth factors, and ROS, creating a protumorogenic environment[49,57,64]. Similarly, in vitro, the coculture of ionizing radiation-exposed CAFs with pancreatic cancer cells enhanced the invasion-promoting capacity of CAFs, induced a high secretion of CXCL12 (a chemokine implicated in hematopoietic stem cell maintenance and cell migration) by CAFs, and promoted pancreatic cell migration, invasion, and epithelial-mesenchymal transition[65].

RT and immune response

RT has been shown to modulate the immune response by various mechanisms, the first of which is the release of tumor antigens, whereby DAMPs allow APC presentation and CD8+ activation followed by cell death, called immunogenic cell death. RT can also increase peptide availability and activate mTOR, leading to an increase in the MHC-1 protein subunit and an increase in the T cell repertoire[66]. RT induces the release of inflammatory cytokines such as IFN via the cGAS-STING pathway, which is activated by DNA damage caused by RT[67]. Adhesion molecules are also upregulated, with an increase in VCAM-1 and ICAM-1 leading to increased infiltration of lymphocytes to tumor cells and affinity binding to CD3+ cells[68]. Finally, RT facilitates homing of T cells to the TME by upregulating chemokines such as CXCL16[69]. In a murine orthotopic pancreatic cancer model, irradiated tumors displayed increased CD8+ and CD4+ cells, with a high single dose of RT being more efficient in recruiting CD8+ T lymphocytes than fractionated RT. However, fractionated RT induced more infiltration of myeloid-derived suppressor cells than high-dose RT[57]. In vitro, RT increased the expression of PDL-1 in a Jack/stat1-dependent manner[70]. Evidence to date suggests that immunotherapy such as anti-CTLA-4 or anti-PD-1 in PDAC has disappointing results or displays efficacy only in patients with PDAC who test positive for mismatch repair deficiency or microsatellite instability-high (MSI-h) due to the poorly immunogenic nature of PDAC[71-74]. Some data suggest that the combination of RT with immunotherapy could be a future approach to overcome this limitation. In a study by Lee et al[57], the combination of SHD-RT with anti-PD1 increased the delivery of anti-PD1 in a murine orthotopic mouse model of PDAC (UN-KC-6141), which is consistent with the increased tumor perfusion observed in vivo following RT. The survival of mice receiving a combined treatment of anti-PD1/SHD-RT was significantly improved compared to that of mice receiving anti-PD1 or SHD-RT alone. Splenocytes isolated from mice treated with the combination therapy showed increased cytotoxicity specifically toward UN-KC-6141 cells. In addition, while the combined group was free of peritoneal tumors, all of the control, SHD-RT, and anti-PDL1 alone groups bore metastases. This encouraging result is in accordance with another in vivo PDAC mouse model study by Fujiwara et al[75] reporting increased survival following a combination of anti-PD1 therapy and RT.

HIFU

HIFU is a noninvasive therapeutic technique using a focused ultrasound beam to create either thermal effects or a mechanical effect called cavitation at the focal point. With respect to thermal effects, due to the focal concentration of energy delivery, HIFU is capable of producing rapid coagulation necrosis with limited inflammatory response and minimal damage to the TME outside the focal zone, inside of which the TME is destroyed. Otherwise, the effects of HT on the TME have been described above in a dedicated section. With respect to the mechanical effect of HIFU, acoustic cavitation can be defined as the initiation, growth, oscillation, and collapse of gas bubbles inside a medium due to high tensile acoustic pressures that exceed cohesion forces between molecules. When exposed to an acoustic field, a bubble will oscillate radially (regime of stable cavitation) and possibly collapse (regime of inertial cavitation). At the tissue level, stable cavitation can stretch tight junctions and allow the extravasation of molecules from the vascular to interstitial space, making the plasma cell membrane transiently permeable and allowing for the internalization of molecules. Comparatively, inertial cavitation is more violent and may induce irreversible membrane disruption and cell implosion or hemorrhage in tissues[76]. HIFU is regularly used in prostate cancer or in the management of uterine fibroids[77,78] but also in the management of PDAC, although it is much less common. HIFU has been suggested to improve quality of life and alleviate pain in patients with a metastatic course of their disease[79].

To date, few preclinical studies have evaluated cavitation as a potentiator of chemotherapy with promising results. A previous study from our group evaluated the impact of various inertial cavitation intensities combined with gemcitabine on the viability of PDAC spheroids composed of both KPC pancreatic cancer cells and activated fibroblasts designed to mimic the tumor stroma[80]. Even if this model was far from a PDAC tumor, it possessed some of its essential features, including the presence of activated fibroblasts, the production of extracellular matrix and a dense intercellular arrangement. This work demonstrated that inertial cavitation decreased the viability of spheroids exposed to cavitation and gemcitabine compared to either cavitation alone or gemcitabine alone. Moreover, gemcitabine had no impact on fibroblast viability, whereas the effect of chemotherapy on the viability of PDAC cells was enhanced when combined with cavitation. Of note, the effects of gemcitabine toxicity were less evident in spheroids composed of both KPC cells and fibroblasts compared to those composed of KPC cells only, which is consistent with the protective effect of the TME and supports the benefit of the combination[80].

In 2015, Li et al[81]. showed in KPC mice that cavitation with pulsed HIFU enhanced the intratumoral concentration of doxorubicin by 4.5-fold compared to controls, with an increase in doxorubicin concentration when cavitation was high and sustained. Of note, there were no differences when pulsed HIFU was delivered during or before doxorubicin administration. On the pulsed HIFU-treated tumors, macroscopic evaluation revealed hemorrhagic areas, while microscopic evaluation showed disorientation and separation of the collagen matrix with fraying of collagen fibrils. A study by Huang et al[82] evaluated the impact of cavitation induced with an ultrasound contrast agent (microbubble) in a mouse model of pancreatic cancer. Blood perfusion evaluated by contrast-enhanced ultrasound imaging revealed a decrease in blood flow within the tumor after cavitation treatment compared to pretreatment measurements, whereas blood perfusion of nontumoral tissue was not impacted. Immunostaining of blood vessels also showed decreased expression of CD31 and reduced microvascular density in the cavitation group.

On the immunotherapy side, the mechanical effects of HIFU have been shown to induce subcellular fragmentation, leading to the release of DAMPs that are subsequently presented to dendritic cells[83] and trigger cytotoxic T cell activation[84]. Pulsed HIFU or low-intensity HIFU have been shown to drive Th1 inflammation, to stimulate localized cell recruitment factors and tumor cell surface immunogenic proteins, and increase the CD8+/Treg ratio[85]. However, these data come from non-PDAC tumor types.

IRREVERSIBLE ELECTROPORATION

IRE is a nonthermal ablative therapy using a direct high voltage current with a short pulse length to increase cell membrane permeability, resulting in permanent cell death with minimal thermal deposition[86-88]. IRE is applied by placing two or more electrodes in the tumor or around it[89] and can be used intraoperatively, laparoscopically, or percutaneously. IRE induces damage only to the cell membrane and has no effect on protein denaturation, blood flow, and connective tissue[88] and was first described for the treatment of human pancreatic cancer in 2012[90].

Studies on the specific effects of IRE on the PDAC stroma are scarce. One study by Bhutiani et al[91] described an increase in gemcitabine delivery to the tissue located in the electroporation area in mice treated by IRE compared to untreated mice. Even if mechanistic explanations were not explored, this effect may be attributable to IRE-related alteration of the stroma. TME modulation following IRE is also characterized by a transient increase in microvascular density and an increase in tumor blood vessel permeability along with a softening of the extracellular matrix can lead to an increase in T cell infiltration[92]. IRE can induce microvessel endothelial cell apoptosis with microvessel thrombosis in vivo[93]. In a xenograft mouse model of PDAC, alterations of tumor microstructure were described following IRE in which acute coagulative necrosis and thrombosis were visible throughout the treated tumor volume after IRE, whereas minimal thrombosis was observed in the control group (no treatment). Using transmission electron microscopy, microvessel endothelial apoptosis and microvessel thrombosis were visualized, and magnetic resonance imaging (MRI) analysis revealed a significant increase in water diffusion after IRE, with a reduction in diffusion-weighted MRI images reflecting an increase in diffusion (water mobility) in the tissue after IRE[93].

From an immunologic point of view, available data are limited: a mouse study by Yang et al[94] described an increase in calreticulin after IRE, suggesting an induction of immunogenic cell death, with an increase in the intratumoral expression of CD8+ cells and GrB (granules of enzymes expressed by cytotoxic lymphocytes) when IRE was combined with a dendritic cell vaccine. In the same study, stromal fibrosis formation was not modified following IRE. In another study by White et al[95], IRE was found to induce an increase in macrophage, T cell, and neutrophil infiltration within the tumor.

CLINICAL PERSPECTIVES
Hyperthermia

In accordance with mouse studies showing an increase in drug delivery with hyperthermia, a recent systematic review evaluated the clinical benefit of HT (regional, intraoperative, or whole-body HT) combined with chemotherapy, RT or both in 248 patients. Out of 14 studies, 6 showed a longer median overall survival in the HT group compared to the control group, with an 11.7 mo median survival vs 5.6 mo. The response rate was also higher in the HT groups[96]. These encouraging results have prompted randomized clinical trials to more clearly demonstrate any benefit of this therapeutic approach. A phase II study (HEATPAC-NCT02439593) is currently recruiting to compare deep locoregional HT administered with a microwave system (Aim 40-43 °C for 60 min) with chemotherapy vs chemotherapy alone in LAPC. The results from this study could provide a practical assessment of the efficacy of HT in PDAC[97]. Other current studies are summarized in Table 2.

Table 2 Ongoing studies in locoregional therapies used alone or in combination with chemotherapy or immunotherapy for pancreatic ductal adenocarcinoma.

Clinical trial number
Study name
Number of patients
Status
Country
HyperthermiaNCT04858009Hyperthermic intraperitoneal chemotherapy for the treatment of pancreatic cancer and peritoneal metastasis.40Not yet recruitingUnited States
NCT04889742HT enhanced reirradiation of loco-regional recurrent tumors (HETERERO).100RecruitingGermany
NCT04310111EUS-RFA for unresectable pancreatic cancer.18RecruitingChina
NCT03218345EUS-guided RFA for pancreatic neoplasms.30RecruitingChina
NCT02439593Concurrent HT and chemoradiotherapy in LAPC: phase II study (HEATPAC).78RecruitingSwitzerland
NCT04164992EUS-RFA of not-resectable pancreatic cancer.15Not yet recruitingItaly
NCT03690323Pancreatic locally advanced irresectable cancer ablation (PELICAN).228RecruitingNetherlands
NCT04156087Progression-free survival after microwave ablation plus durvalumab and tremelimumab for unresectable LAPC (MIMIPAC).20RecruitingBelgium
Radiation therapyNCT04361162Nivolumab + ipilimumab + radiation in microsatellite stable pancreatic cancer.30RecruitingUnited States
NCT01972919Magnetic resonance guided, dose-escalated RT + chemotherapy in pancreatic cancer.23RecruitingUnited States
NCT03374293Combination of radiation therapy and anti-PD-1 antibody in treating patients with pancreatic cancer.21RecruitingChina
NCT03492671Testing the combination of two approved chemotherapy drugs and radiation prior to surgery in localized pancreatic cancer.30RecruitingUnited States
NCT04975516Standard of care chemotherapy with or without SBRT for the treatment of oligometastatic pancreatic cancer.50Not yet recruitingUnited States
NCT04327986Immune checkpoint inhibitor m7824 and the immunocytokine m9241 in combination with SBRT in adults with advanced pancreas cancer.52RecruitingUnited States
NCT03991962Phase II study to evaluate modified folfirinox and SBRT in nonmetastatic unresectable pancreatic adenocarcinoma.28RecruitingUnited States
NCT02128100Effects of folfirinox and SBRT for advanced pancreatic cancer.28RecruitingUnited States
NCT04089150Mfolfirinox and SBRT for pancreatic cancer with high risk and locally advanced disease.120Not yet recruitingAustralia
NCT04172532Testing the addition of a new anticancer drug, m3814 (peposertib), to radiation therapy for localized pancreatic cancer.24RecruitingUnited States
HIFUNCT04146441Ultrasound-enhanced uptake of chemotherapy in patients with inoperable PDAC.30RecruitingNorway
NCT04852367PanDox: targeted doxorubicin in pancreatic tumors.18Not yet recruitingEngland
Irreversible elecroporationNCT03257150A study of the use of IRE in pancreatic ductal cancer.47RecruitingCanada
NCT03105921IRE (nanoknife) for the treatment of pancreatic adenocarcinoma.20RecruitingFrance
NCT03899636A pivotal study of safety and effectiveness of nanoknife IRE for stage 3 pancreatic cancer (direct).528RecruitingUnited States
NCT02822716IRE for inoperable hepatic and pancreatic malignancy.35RecruitingChina
NCT02343835Antitumor immunity induced by IRE of unresectable pancreatic cancer.20RecruitingChina
NCT03484299Chemotherapy and IRE in the treatment of advanced pancreatic adenocarcinoma.20RecruitingUnited States
NCT04835402Electroporation potentiated immunotherapy in cancer (EPIC-1).16RecruitingDenmark
NCT04276857Systemic therapy with a loco-regional treatment in patients with LAPC (smart).27Not yet recruitingCanada
NCT04212026IRE followed by nivolumab in patients with metastatic pancreatic cancer.15RecruitingSwitzerland
NCT04612530Panfire-3 trial: assessing safety and efficacy of IRE + nivolumab + CpG for metastatic pancreatic cancer.18RecruitingNetherlands
NCT02041936Outcomes of ablation of unresectable pancreatic cancer using the nanoknife IRE system.12RecruitingUnited States
NCT04310553An open-label, multicenter, prospective study of IRE (nanoknife) combined with RT and chemotherapy in patients with LAPC.240RecruitingChina
NCT04093141Chemotherapy followed by IRE in patients with unresectable LAPC (chemofire-2).30RecruitingDenmark
NCT03614910Ablation of unresectable LAPC with IRE system.30RecruitingUnited States

RFA in PDAC has been reported in small exploratory series for tumor debulking rather than for complete tumor ablation because safety margins are needed to avoid thermal damage to surrounding structures[98]. Following RFA combined with chemotherapy, overall survival ranges from 19 to 25.6 mo[98,99]. There is a lack of randomized studies assessing the place of RFA in the management of LAPC. The PELICAN trial (NCT03690323) is planned to evaluate whether the combination of chemotherapy and RFA improves overall survival compared to chemotherapy alone in patients with LAPC without any progression after 2 mo of systemic treatment[100] (Table 2).

Endoscopic application of RFA, which is already feasible, is an attractive approach because of its minimal invasiveness. The active component is a 19G needle that has a tip equipped with an electrode to be placed in the lesion under endoscopic ultrasound (EUS) guidance. This approach has been proven safe and feasible in small-sized studies of patients with unresectable PDAC[101-103]. Nevertheless, larger prospective studies are needed. EUS-RFA clinical trials are ongoing (Table 2), and it would be interesting to evaluate EUS-RFA as an alternative to RT in LAPC patients with an objective response to chemotherapy who retain criteria against surgical resection.

RT

Stereotactic body radiotherapy (SBRT) is increasingly being explored for the management of PDAC in combination with anticancerous drugs, especially for LAPC. An open-label phase 2 multicenter study by Herman et al. evaluated the combination of gemcitabine plus SBRT in patients with LAPC, showing a good safety profile[104]. Similarly, 39 patients who underwent FOLFIRINOX followed by SBRT seemed to have an increased chance of undergoing radical surgery[105]. SBRT in combination with immune therapy is also being studied. In a phase I study by Xie et al[106], a combination of immune therapy (durvalumab ± tremelimumab) with SBRT in metastatic PDAC showed a favorable safety profile but only a modest clinical efficacy. Of note, none of the responders were MSI-h. While these results are interesting, further exploration is required, and many clinical trials are underway to evaluate the combination of SBRT or RT with anticancerous drugs (Table 2). A challenge for the use of SBRT is the required placement of fiducials to facilitate the delivery of radiation, which can be made difficult by respiratory movements and the vicinity of other organs[107]. These fiducials can be placed percutaneously when not impeded by surrounding organs or in a more invasive fashion, surgically. EUS-guided placement of fiducial also appears to be a promising method with a high rate of technical success and a reasonable rate of adverse events[91,108], but randomized studies are needed.

HIFU

Cavitation generated by HIFU is a very attractive method with a high potential to disrupt the stroma, thus overcoming the barrier to efficient drug delivery and stimulating the immune response in preclinical works. To date, there are no published clinical trials. However, one upcoming clinical trial (NCT04146441) of HIFU combined with chemotherapy (FOLFIRINOX) will determine whether focused ultrasound can increase drug uptake and overcome chemoresistance (Table 2). In a minimalist approach of ultrasound-induced enhancement of chemotherapy, 10 patients were enrolled in a phase I clinical trial to receive gemcitabine combined with low intensity ultrasound and microbubbles as an ultrasound contrast agent programmed to favor sonoporation, with encouraging results in terms of the number of chemotherapy cycles tolerated and median overall survival when compared to 63 historical controls receiving only chemotherapy[109].

HIFU is also a very attractive approach to increase the intratumoral temperature and increase drug delivery. In a monocentric retrospective study among 523 patients, a combination of HIFU with gemcitabine appeared to produce better overall survival than standard CT in unresectable PDAC[110]. The PanDox study is a phase I study that plans to evaluate whether HIFU can increase the amount of drug delivery (doxorubicin or heat-sensitive doxorubicin) within the tumor in 18 patients with unresectable PDAC (NCT04852367, Table 2).

Challenges in the method of ultrasound delivery still need to be addressed, since extracorporeal delivery to the deeply seated pancreas with gas interposition could be challenging. We are currently working on an endoscopic device that could overcome these limitations and noninvasively deliver cavitation at any part of the pancreas. The endoscopic approach to HIFU delivery, foreseen by our team some time ago[111], has also been recently studied in a porcine model[112].

Irreversible electroporation

After the landmark study by Martin et al[90] and subsequent large series of intraoperative applications[86], less invasive percutaneous IRE has shown promising results in terms of efficacy. A nonrandomized prospective single-center case series by Ma et al[113] evaluated the efficacy of a combination of percutaneous IRE with gemcitabine compared with gemcitabine alone. The combination increased the overall survival from the time of diagnosis by 3-fold and nearly doubled the progression-free survival. In a post hoc comparison of data derived from a prospective IRE-FOLFIRINOX cohort and a retrospective FOLFIRINOX-only cohort, van Veldhuisen et al[114] found that the combination (30 LAPC patients) increased the time to progression compared to standard therapy (22 patients). Lin et al[115] showed promising results with IRE combined with allogenic natural killer cell immunotherapy with an increase in progression-free survival and overall survival. However, a multicenter prospective study by Ruarus et al[116] (PANFIRE II) described a high rate of adverse events in patients undergoing percutaneous IRE, with 29 out of 50 participants experiencing adverse events, 21 of which were major, and 2 deaths, including one clearly related to IRE. Thus, this procedure can be considered a high-risk procedure that requires the selection of patients who will benefit the most from the treatment. This high rate of adverse events, along with a relative cumbersomeness to set up, has limited the spread of this technique. New application methods are needed to overcome these issues. Many clinical trials are ongoing to better understand the benefits of combining IRE with chemotherapy or immunotherapy (Table 2).

CONCLUSION

The TME is one of the major causes of therapeutic resistance in PDAC. Fibrosis-related stiffness, hypomicrovascular perfusion, and an immune suppressive microenvironment are, within the limits of current knowledge, key determinants of this resistance. While systemic chemotherapies and immunotherapies have disappointing results and are responsible for adverse events resulting in poor quality of life, locoregional therapies can specifically target the tumor area with limited effects on surrounding tissues but significant impacts on the TME. Local HT using RFA and radiotherapy using local SHD are the two main modalities currently holding promise for clinical efficacy, but IRE and focused ultrasound-derived cavitation are also gaining increasing attention as treatments for PDAC. These techniques influence the tumor stroma, microvasculature, and immune environment and response (Table 1). To date, most of the data are preclinical with some promising results. Clinical trials are underway (Table 2) and will allow the scientific community to have a more precise idea of the interest in using these treatment options alone or in combination with systemic therapies.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: France

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Isaji S, Japan; Li L, China S-Editor: Zhang H L-Editor: A P-Editor: Zhang H

References
1.  Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:1039-1049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1500]  [Cited by in F6Publishing: 1584]  [Article Influence: 158.4]  [Reference Citation Analysis (0)]
2.  Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913-2921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3888]  [Cited by in F6Publishing: 4701]  [Article Influence: 470.1]  [Reference Citation Analysis (0)]
3.  Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66:7-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12135]  [Cited by in F6Publishing: 12760]  [Article Influence: 1595.0]  [Reference Citation Analysis (2)]
4.  Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1481]  [Cited by in F6Publishing: 1519]  [Article Influence: 76.0]  [Reference Citation Analysis (0)]
5.  He J, Ahuja N, Makary MA, Cameron JL, Eckhauser FE, Choti MA, Hruban RH, Pawlik TM, Wolfgang CL. 2564 resected periampullary adenocarcinomas at a single institution: trends over three decades. HPB (Oxford). 2014;16:83-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 205]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
6.  Spichiger E, Müller-Fröhlich C, Denhaerynck K, Stoll H, Hantikainen V, Dodd M. Prevalence of symptoms, with a focus on fatigue, and changes of symptoms over three months in outpatients receiving cancer chemotherapy. Swiss Med Wkly. 2011;141:w13303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
7.  Karagiannis GS, Poutahidis T, Erdman SE, Kirsch R, Riddell RH, Diamandis EP. Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue. Mol Cancer Res. 2012;10:1403-1418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 398]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
8.  Murakami T, Hiroshima Y, Matsuyama R, Homma Y, Hoffman RM, Endo I. Role of the tumor microenvironment in pancreatic cancer. Ann Gastroenterol Surg. 2019;3:130-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 98]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
9.  Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA. The pancreas cancer microenvironment. Clin Cancer Res. 2012;18:4266-4276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 841]  [Cited by in F6Publishing: 951]  [Article Influence: 86.5]  [Reference Citation Analysis (0)]
10.  Kota J, Hancock J, Kwon J, Korc M. Pancreatic cancer: Stroma and its current and emerging targeted therapies. Cancer Lett. 2017;391:38-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 122]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
11.  Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1408]  [Cited by in F6Publishing: 1495]  [Article Influence: 124.6]  [Reference Citation Analysis (0)]
12.  Rizzuti IF, Mascheroni P, Arcucci S, Ben-Mériem Z, Prunet A, Barentin C, Rivière C, Delanoë-Ayari H, Hatzikirou H, Guillermet-Guibert J, Delarue M. Mechanical Control of Cell Proliferation Increases Resistance to Chemotherapeutic Agents. Phys Rev Lett. 2020;125:128103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
13.  Rice AJ, Cortes E, Lachowski D, Cheung BCH, Karim SA, Morton JP, Del Río Hernández A. Matrix stiffness induces epithelial-mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis. 2017;6:e352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 331]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
14.  Piehler S, Wucherpfennig L, Tansi FL, Berndt A, Quaas R, Teichgraeber U, Hilger I. Hyperthermia affects collagen fiber architecture and induces apoptosis in pancreatic and fibroblast tumor hetero-spheroids in vitro. Nanomedicine. 2020;28:102183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
15.  Jiang B, Zhou L, Lu J, Wang Y, Liu C, You L, Guo J. Stroma-Targeting Therapy in Pancreatic Cancer: One Coin With Two Sides? Front Oncol. 2020;10:576399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
16.  Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, Westphalen CB, Kitajewski J, Fernandez-Barrena MG, Fernandez-Zapico ME, Iacobuzio-Donahue C, Olive KP, Stanger BZ. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1359]  [Cited by in F6Publishing: 1504]  [Article Influence: 150.4]  [Reference Citation Analysis (0)]
17.  Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719-734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1787]  [Cited by in F6Publishing: 1719]  [Article Influence: 171.9]  [Reference Citation Analysis (1)]
18.  Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, Felix R, Schlag PM. Hyperthermia in combined treatment of cancer. Lancet Oncol. 2002;3:487-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1439]  [Cited by in F6Publishing: 1105]  [Article Influence: 50.2]  [Reference Citation Analysis (0)]
19.  Kolosnjaj-Tabi J, Marangon I, Nicolas-Boluda A, Silva AKA, Gazeau F. Nanoparticle-based hyperthermia, a local treatment modulating the tumor extracellular matrix. Pharmacol Res. 2017;126:123-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
20.  Bañobre-López M, Teijeiro A, Rivas J. Magnetic nanoparticle-based hyperthermia for cancer treatment. Rep Pract Oncol Radiother. 2013;18:397-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 229]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
21.  Miyamoto R, Oda T, Hashimoto S, Kurokawa T, Inagaki Y, Shimomura O, Ohara Y, Yamada K, Akashi Y, Enomoto T, Kishimoto M, Yanagihara H, Kita E, Ohkohchi N. Cetuximab delivery and antitumor effects are enhanced by mild hyperthermia in a xenograft mouse model of pancreatic cancer. Cancer Sci. 2016;107:514-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
22.  Song CW, Park HJ, Lee CK, Griffin R. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia. 2005;21:761-767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 199]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
23.  Kirui DK, Koay EJ, Guo X, Cristini V, Shen H, Ferrari M. Tumor vascular permeabilization using localized mild hyperthermia to improve macromolecule transport. Nanomedicine. 2014;10:1487-1496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
24.  Corr SJ, Shamsudeen S, Vergara LA, Ho JC, Ware MJ, Keshishian V, Yokoi K, Savage DJ, Meraz IM, Kaluarachchi W, Cisneros BT, Raoof M, Nguyen DT, Zhang Y, Wilson LJ, Summers H, Rees P, Curley SA, Serda RE. A New Imaging Platform for Visualizing Biological Effects of Non-Invasive Radiofrequency Electric-Field Cancer Hyperthermia. PLoS One. 2015;10:e0136382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
25.  Song CW, Park H, Griffin RJ. Improvement of tumor oxygenation by mild hyperthermia. Radiat Res. 2001;155:515-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 133]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
26.  Song CW. Effect of local hyperthermia on blood flow and microenvironment: a review. Cancer Res. 1984;44:4721s-4730s.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Teng T, Lin R, Lin Z, Ke K, Lin X, Pan M, Zhang D, Huang H. Photothermal augment stromal disrupting effects for enhanced Abraxane synergy chemotherapy in pancreatic cancer PDX mode. Biomater Sci. 2020;8:3278-3285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
28.  Nicolás-Boluda A, Vaquero J, Laurent G, Renault G, Bazzi R, Donnadieu E, Roux S, Fouassier L, Gazeau F. Photothermal Depletion of Cancer-Associated Fibroblasts Normalizes Tumor Stiffness in Desmoplastic Cholangiocarcinoma. ACS Nano. 2020;14:5738-5753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 38]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
29.  Marangon I, Silva AA, Guilbert T, Kolosnjaj-Tabi J, Marchiol C, Natkhunarajah S, Chamming's F, Ménard-Moyon C, Bianco A, Gennisson JL, Renault G, Gazeau F. Tumor Stiffening, a Key Determinant of Tumor Progression, is Reversed by Nanomaterial-Induced Photothermal Therapy. Theranostics. 2017;7:329-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
30.  Ware MJ, Curtis LT, Wu M, Ho JC, Corr SJ, Curley SA, Godin B, Frieboes HB. Pancreatic adenocarcinoma response to chemotherapy enhanced with non-invasive radio frequency evaluated via an integrated experimental/computational approach. Sci Rep. 2017;7:3437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
31.  Kolosnjaj-Tabi J, Di Corato R, Lartigue L, Marangon I, Guardia P, Silva AK, Luciani N, Clément O, Flaud P, Singh JV, Decuzzi P, Pellegrino T, Wilhelm C, Gazeau F. Heat-generating iron oxide nanocubes: subtle "destructurators" of the tumoral microenvironment. ACS Nano. 2014;8:4268-4283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 130]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
32.  Li Z, Deng J, Sun J, Ma Y. Hyperthermia Targeting the Tumor Microenvironment Facilitates Immune Checkpoint Inhibitors. Front Immunol. 2020;11:595207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
33.  Gao S, Pu N, Yin H, Li J, Chen Q, Yang M, Lou W, Chen Y, Zhou G, Li C, Li G, Yan Z, Liu L, Yu J, Wang X. Radiofrequency ablation in combination with an mTOR inhibitor restrains pancreatic cancer growth induced by intrinsic HSP70. Ther Adv Med Oncol. 2020;12:1758835920953728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
34.  Mahmood J, Shukla HD, Soman S, Samanta S, Singh P, Kamlapurkar S, Saeed A, Amin NP, Vujaskovic Z. Immunotherapy, Radiotherapy, and Hyperthermia: A Combined Therapeutic Approach in Pancreatic Cancer Treatment. Cancers (Basel). 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
35.  Yagawa Y, Tanigawa K, Kobayashi Y, Yamamoto M. Cancer immunity and therapy using hyperthermia with immunotherapy, radiotherapy, chemotherapy, and surgery. J Cancer Metastasis Treat. 2017;3:218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
36.  Chen Q, Fisher DT, Clancy KA, Gauguet JM, Wang WC, Unger E, Rose-John S, von Andrian UH, Baumann H, Evans SS. Fever-range thermal stress promotes lymphocyte trafficking across high endothelial venules via an interleukin 6 trans-signaling mechanism. Nat Immunol. 2006;7:1299-1308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 164]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
37.  Vardam TD, Zhou L, Appenheimer MM, Chen Q, Wang WC, Baumann H, Evans SS. Regulation of a lymphocyte-endothelial-IL-6 trans-signaling axis by fever-range thermal stress: hot spot of immune surveillance. Cytokine. 2007;39:84-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
38.  Newton JM, Flores-Arredondo JH, Suki S, Ware MJ, Krzykawska-Serda M, Agha M, Law JJ, Sikora AG, Curley SA, Corr SJ. Non-Invasive Radiofrequency Field Treatment of 4T1 Breast Tumors Induces T-cell Dependent Inflammatory Response. Sci Rep. 2018;8:3474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
39.  Mantso T, Goussetis G, Franco R, Botaitis S, Pappa A, Panayiotidis M. Effects of hyperthermia as a mitigation strategy in DNA damage-based cancer therapies. Semin Cancer Biol. 2016;37-38:96-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
40.  Binder RJ, Srivastava PK. Peptides chaperoned by heat-shock proteins are a necessary and sufficient source of antigen in the cross-priming of CD8+ T cells. Nat Immunol. 2005;6:593-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 210]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
41.  Schildkopf P, Frey B, Ott OJ, Rubner Y, Multhoff G, Sauer R, Fietkau R, Gaipl US. Radiation combined with hyperthermia induces HSP70-dependent maturation of dendritic cells and release of pro-inflammatory cytokines by dendritic cells and macrophages. Radiother Oncol. 2011;101:109-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
42.  Zhu J, Zhang Y, Zhang A, He K, Liu P, Xu LX. Cryo-thermal therapy elicits potent anti-tumor immunity by inducing extracellular Hsp70-dependent MDSC differentiation. Sci Rep. 2016;6:27136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
43.  Breloer M, Dorner B, Moré SH, Roderian T, Fleischer B, von Bonin A. Heat shock proteins as "danger signals": eukaryotic Hsp60 enhances and accelerates antigen-specific IFN-gamma production in T cells. Eur J Immunol. 2001;31:2051-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
44.  Lin FC, Hsu CH, Lin YY. Nano-therapeutic cancer immunotherapy using hyperthermia-induced heat shock proteins: insights from mathematical modeling. Int J Nanomedicine. 2018;13:3529-3539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
45.  MOLE RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26:234-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 677]  [Cited by in F6Publishing: 743]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
46.  Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10:718-726.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 641]  [Cited by in F6Publishing: 709]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
47.  Fei Q, Pan Y, Lin W, Zhou Y, Yu X, Hou Z, Lin X, Lin R, Lu F, Guan H, Huang H. High-dimensional single-cell analysis delineates radiofrequency ablation induced immune microenvironmental remodeling in pancreatic cancer. Cell Death Dis. 2020;11:589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
48.  Gameiro SR, Higgins JP, Dreher MR, Woods DL, Reddy G, Wood BJ, Guha C, Hodge JW. Combination therapy with local radiofrequency ablation and systemic vaccine enhances antitumor immunity and mediates local and distal tumor regression. PLoS One. 2013;8:e70417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
49.  Giardino A, Innamorati G, Ugel S, Perbellini O, Girelli R, Frigerio I, Regi P, Scopelliti F, Butturini G, Paiella S, Bacchion M, Bassi C. Immunomodulation after radiofrequency ablation of locally advanced pancreatic cancer by monitoring the immune response in 10 patients. Pancreatology. 2017;17:962-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
50.  Leroi N, Lallemand F, Coucke P, Noel A, Martinive P. Impacts of Ionizing Radiation on the Different Compartments of the Tumor Microenvironment. Front Pharmacol. 2016;7:78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
51.  Arnold KM, Flynn NJ, Raben A, Romak L, Yu Y, Dicker AP, Mourtada F, Sims-Mourtada J. The Impact of Radiation on the Tumor Microenvironment: Effect of Dose and Fractionation Schedules. Cancer Growth Metastasis. 2018;11:1179064418761639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 98]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
52.  Song CW, Lee YJ, Griffin RJ, Park I, Koonce NA, Hui S, Kim MS, Dusenbery KE, Sperduto PW, Cho LC. Indirect Tumor Cell Death After High-Dose Hypofractionated Irradiation: Implications for Stereotactic Body Radiation Therapy and Stereotactic Radiation Surgery. Int J Radiat Oncol Biol Phys. 2015;93:166-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
53.  Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015;15:409-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1293]  [Cited by in F6Publishing: 1316]  [Article Influence: 146.2]  [Reference Citation Analysis (0)]
54.  Abdollahi A, Griggs DW, Zieher H, Roth A, Lipson KE, Saffrich R, Gröne HJ, Hallahan DE, Reisfeld RA, Debus J, Niethammer AG, Huber PE. Inhibition of alpha(v)beta3 integrin survival signaling enhances antiangiogenic and antitumor effects of radiotherapy. Clin Cancer Res. 2005;11:6270-6279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 184]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
55.  Katagiri T, Kobayashi M, Yoshimura M, Morinibu A, Itasaka S, Hiraoka M, Harada H. HIF-1 maintains a functional relationship between pancreatic cancer cells and stromal fibroblasts by upregulating expression and secretion of Sonic hedgehog. Oncotarget. 2018;9:10525-10535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
56.  Maeda A, Chen Y, Bu J, Mujcic H, Wouters BG, DaCosta RS. In Vivo Imaging Reveals Significant Tumor Vascular Dysfunction and Increased Tumor Hypoxia-Inducible Factor-1α Expression Induced by High Single-Dose Irradiation in a Pancreatic Tumor Model. Int J Radiat Oncol Biol Phys. 2017;97:184-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
57.  Lee YH, Yu CF, Yang YC, Hong JH, Chiang CS. Ablative Radiotherapy Reprograms the Tumor Microenvironment of a Pancreatic Tumor in Favoring the Immune Checkpoint Blockade Therapy. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
58.  Krisnawan VE, Stanley JA, Schwarz JK, DeNardo DG. Tumor Microenvironment as a Regulator of Radiation Therapy: New Insights into Stromal-Mediated Radioresistance. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
59.  Straub JM, New J, Hamilton CD, Lominska C, Shnayder Y, Thomas SM. Radiation-induced fibrosis: mechanisms and implications for therapy. J Cancer Res Clin Oncol. 2015;141:1985-1994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 345]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
60.  Qayyum MA, Insana MF. Stromal responses to fractionated radiotherapy. Int J Radiat Biol. 2012;88:383-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
61.  Qian LW, Mizumoto K, Urashima T, Nagai E, Maehara N, Sato N, Nakajima M, Tanaka M. Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin Cancer Res. 2002;8:1223-1227.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Hellevik T, Pettersen I, Berg V, Winberg JO, Moe BT, Bartnes K, Paulssen RH, Busund LT, Bremnes R, Chalmers A, Martinez-Zubiaurre I. Cancer-associated fibroblasts from human NSCLC survive ablative doses of radiation but their invasive capacity is reduced. Radiat Oncol. 2012;7:59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
63.  Baird JR, Friedman D, Cottam B, Dubensky TW Jr, Kanne DB, Bambina S, Bahjat K, Crittenden MR, Gough MJ. Radiotherapy Combined with Novel STING-Targeting Oligonucleotides Results in Regression of Established Tumors. Cancer Res. 2016;76:50-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 188]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
64.  Ghaly M, Gogineni E, Saif MW. The Evolving Field of Stereotactic Body Radiation Therapy in Pancreatic Cancer. Pancreas (Fairfax). 2019;3:9-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
65.  Li D, Qu C, Ning Z, Wang H, Zang K, Zhuang L, Chen L, Wang P, Meng Z. Radiation promotes epithelial-to-mesenchymal transition and invasion of pancreatic cancer cell by activating carcinoma-associated fibroblasts. Am J Cancer Res. 2016;6:2192-2206.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, Camphausen K, Luiten RM, de Ru AH, Neijssen J, Griekspoor A, Mesman E, Verreck FA, Spits H, Schlom J, van Veelen P, Neefjes JJ. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259-1271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1089]  [Cited by in F6Publishing: 1248]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]
67.  Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142-1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 928]  [Cited by in F6Publishing: 1265]  [Article Influence: 180.7]  [Reference Citation Analysis (0)]
68.  Rodriguez-Ruiz ME, Garasa S, Rodriguez I, Solorzano JL, Barbes B, Yanguas A, Teijeira A, Etxeberria I, Aristu JJ, Halin C, Melero I, Rouzaut A. Intercellular Adhesion Molecule-1 and Vascular Cell Adhesion Molecule Are Induced by Ionizing Radiation on Lymphatic Endothelium. Int J Radiat Oncol Biol Phys. 2017;97:389-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
69.  Menon H, Ramapriyan R, Cushman TR, Verma V, Kim HH, Schoenhals JE, Atalar C, Selek U, Chun SG, Chang JY, Barsoumian HB, Nguyen QN, Altan M, Cortez MA, Hahn SM, Welsh JW. Role of Radiation Therapy in Modulation of the Tumor Stroma and Microenvironment. Front Immunol. 2019;10:193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 92]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
70.  Azad A, Yin Lim S, D'Costa Z, Jones K, Diana A, Sansom OJ, Kruger P, Liu S, McKenna WG, Dushek O, Muschel RJ, Fokas E. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol Med. 2017;9:167-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 154]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
71.  Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, Sherry RM, Topalian SL, Yang JC, Lowy I, Rosenberg SA. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 701]  [Cited by in F6Publishing: 884]  [Article Influence: 68.0]  [Reference Citation Analysis (0)]
72.  Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3799]  [Cited by in F6Publishing: 4447]  [Article Influence: 635.3]  [Reference Citation Analysis (0)]
73.  Yoon JH, Jung YJ, Moon SH. Immunotherapy for pancreatic cancer. World J Clin Cases. 2021;9:2969-2982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (1)]
74.  Sohal DPS, Kennedy EB, Khorana A, Copur MS, Crane CH, Garrido-Laguna I, Krishnamurthi S, Moravek C, O'Reilly EM, Philip PA, Ramanathan RK, Ruggiero JT, Shah MA, Urba S, Uronis HE, Lau MW, Laheru D. Metastatic Pancreatic Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36:2545-2556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 176]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
75.  Fujiwara K, Saung MT, Jing H, Herbst B, Zarecki M, Muth S, Wu A, Bigelow E, Chen L, Li K, Jurcak N, Blair AB, Ding D, Wichroski M, Blum J, Cheadle N, Koenitzer J, Zheng L. Interrogating the immune-modulating roles of radiation therapy for a rational combination with immune-checkpoint inhibitors in treating pancreatic cancer. J Immunother Cancer. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
76.  Haar GT, Coussios C. High intensity focused ultrasound: physical principles and devices. Int J Hyperthermia. 2007;23:89-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 430]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
77.  Ziglioli F, Baciarello M, Maspero G, Bellini V, Bocchialini T, Cavalieri D, Bignami EG, Maestroni U. Oncologic outcome, side effects and comorbidity of high-intensity focused ultrasound (HIFU) for localized prostate cancer. A review. Ann Med Surg (Lond). 2020;56:110-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
78.  Liu L, Wang T, Lei B. High-intensity focused ultrasound (HIFU) ablation versus surgical interventions for the treatment of symptomatic uterine fibroids: a meta-analysis. Eur Radiol. 2022;32:1195-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
79.  Marinova M, Feradova H, Gonzalez-Carmona MA, Conrad R, Tonguc T, Thudium M, Becher MU, Kun Z, Gorchev G, Tomov S, Strassburg CP, Attenberger U, Schild HH, Dimitrov D, Strunk HM. Improving quality of life in pancreatic cancer patients following high-intensity focused ultrasound (HIFU) in two European centers. Eur Radiol. 2021;31:5818-5829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
80.  Leenhardt R, Camus M, Mestas JL, Jeljeli M, Abou Ali E, Chouzenoux S, Bordacahar B, Nicco C, Batteux F, Lafon C, Prat F. Ultrasound-induced Cavitation enhances the efficacy of Chemotherapy in a 3D Model of Pancreatic Ductal Adenocarcinoma with its microenvironment. Sci Rep. 2019;9:18916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
81.  Li T, Wang YN, Khokhlova TD, D'Andrea S, Starr F, Chen H, McCune JS, Risler LJ, Mashadi-Hossein A, Hingorani SR, Chang A, Hwang JH. Pulsed High-Intensity Focused Ultrasound Enhances Delivery of Doxorubicin in a Preclinical Model of Pancreatic Cancer. Cancer Res. 2015;75:3738-3746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
82.  Huang P, Zhang Y, Chen J, Shentu W, Sun Y, Yang Z, Liang T, Chen S, Pu Z. Enhanced antitumor efficacy of ultrasonic cavitation with up-sized microbubbles in pancreatic cancer. Oncotarget. 2015;6:20241-20251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
83.  Hu Z, Yang XY, Liu Y, Morse MA, Lyerly HK, Clay TM, Zhong P. Release of endogenous danger signals from HIFU-treated tumor cells and their stimulatory effects on APCs. Biochem Biophys Res Commun. 2005;335:124-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 65]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
84.  Xing Y, Lu X, Pua EC, Zhong P. The effect of high intensity focused ultrasound treatment on metastases in a murine melanoma model. Biochem Biophys Res Commun. 2008;375:645-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 56]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
85.  Maloney E, Khokhlova T, Pillarisetty VG, Schade GR, Repasky EA, Wang YN, Giuliani L, Primavera M, Hwang JH. Focused ultrasound for immuno-adjuvant treatment of pancreatic cancer: An emerging clinical paradigm in the era of personalized oncotherapy. Int Rev Immunol. 2017;36:338-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
86.  Martin RC 2nd, Kwon D, Chalikonda S, Sellers M, Kotz E, Scoggins C, McMasters KM, Watkins K. Treatment of 200 locally advanced (stage III) pancreatic adenocarcinoma patients with irreversible electroporation: safety and efficacy. Ann Surg. 2015;262:486-94; discussion 492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 272]  [Article Influence: 30.2]  [Reference Citation Analysis (0)]
87.  Tian G, Liu X, Zhao Q, Xu D, Jiang T. Irreversible Electroporation in Patients with Pancreatic Cancer: How Important Is the New Weapon? Biomed Res Int. 2018;2018:5193067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
88.  Martin RC 2nd. Use of irreversible electroporation in unresectable pancreatic cancer. Hepatobiliary Surg Nutr. 2015;4:211-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 9]  [Reference Citation Analysis (0)]
89.  Rai ZL, Feakins R, Pallett LJ, Manas D, Davidson BR. Irreversible Electroporation (IRE) in Locally Advanced Pancreatic Cancer: A Review of Current Clinical Outcomes, Mechanism of Action and Opportunities for Synergistic Therapy. J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
90.  Martin RC 2nd, McFarland K, Ellis S, Velanovich V. Irreversible electroporation therapy in the management of locally advanced pancreatic adenocarcinoma. J Am Coll Surg. 2012;215:361-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 184]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
91.  Bhutiani N, Agle S, Li Y, Li S, Martin RC 2nd. Irreversible electroporation enhances delivery of gemcitabine to pancreatic adenocarcinoma. J Surg Oncol. 2016;114:181-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
92.  Zhao J, Wen X, Tian L, Li T, Xu C, Melancon MP, Gupta S, Shen B, Peng W, Li C. Irreversible electroporation reverses resistance to immune checkpoint blockade in pancreatic cancer. Nat Commun. 2019;10:899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 149]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
93.  Zhang Z, Li W, Procissi D, Tyler P, Omary RA, Larson AC. Rapid dramatic alterations to the tumor microstructure in pancreatic cancer following irreversible electroporation ablation. Nanomedicine (Lond). 2014;9:1181-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
94.  Yang J, Eresen A, Shangguan J, Ma Q, Yaghmai V, Zhang Z. Irreversible electroporation ablation overcomes tumor-associated immunosuppression to improve the efficacy of DC vaccination in a mice model of pancreatic cancer. Oncoimmunology. 2021;10:1875638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
95.  White SB, Zhang Z, Chen J, Gogineni VR, Larson AC. Early Immunologic Response of Irreversible Electroporation versus Cryoablation in a Rodent Model of Pancreatic Cancer. J Vasc Interv Radiol. 2018;29:1764-1769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
96.  van der Horst A, Versteijne E, Besselink MGH, Daams JG, Bulle EB, Bijlsma MF, Wilmink JW, van Delden OM, van Hooft JE, Franken NAP, van Laarhoven HWM, Crezee J, van Tienhoven G. The clinical benefit of hyperthermia in pancreatic cancer: a systematic review. Int J Hyperthermia. 2018;34:969-979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 21]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
97.  Datta NR, Pestalozzi B, Clavien PA, Siebenhüner A, Puric E, Khan S, Mamot C, Riesterer O, Knuchel J, Reiner CS, Bodis S; members of the HEATPAC Trial Group. "HEATPAC" - a phase II randomized study of concurrent thermochemoradiotherapy versus chemoradiotherapy alone in locally advanced pancreatic cancer. Radiat Oncol. 2017;12:183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
98.  van Veldhuisen E, van den Oord C, Brada LJ, Walma MS, Vogel JA, Wilmink JW, Del Chiaro M, van Lienden KP, Meijerink MR, van Tienhoven G, Hackert T, Wolfgang CL, van Santvoort H, Groot Koerkamp B, Busch OR, Molenaar IQ, van Eijck CH, Besselink MG; Dutch Pancreatic Cancer Group and International Collaborative Group on Locally Advanced Pancreatic Cancer. Locally Advanced Pancreatic Cancer: Work-Up, Staging, and Local Intervention Strategies. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (2)]
99.  Ruarus A, Vroomen L, Puijk R, Scheffer H, Meijerink M. Locally Advanced Pancreatic Cancer: A Review of Local Ablative Therapies. Cancers (Basel). 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
100.  Walma MS, Rombouts SJ, Brada LJH, Borel Rinkes IH, Bosscha K, Bruijnen RC, Busch OR, Creemers GJ, Daams F, van Dam RM, van Delden OM, Festen S, Ghorbani P, de Groot DJ, de Groot JWB, Haj Mohammad N, van Hillegersberg R, de Hingh IH, D'Hondt M, Kerver ED, van Leeuwen MS, Liem MS, van Lienden KP, Los M, de Meijer VE, Meijerink MR, Mekenkamp LJ, Nio CY, Oulad Abdennabi I, Pando E, Patijn GA, Polée MB, Pruijt JF, Roeyen G, Ropela JA, Stommel MWJ, de Vos-Geelen J, de Vries JJ, van der Waal EM, Wessels FJ, Wilmink JW, van Santvoort HC, Besselink MG, Molenaar IQ; Dutch Pancreatic Cancer Group. Radiofrequency ablation and chemotherapy versus chemotherapy alone for locally advanced pancreatic cancer (PELICAN): study protocol for a randomized controlled trial. Trials. 2021;22:313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (1)]
101.  Scopelliti F, Pea A, Conigliaro R, Butturini G, Frigerio I, Regi P, Giardino A, Bertani H, Paini M, Pederzoli P, Girelli R. Technique, safety, and feasibility of EUS-guided radiofrequency ablation in unresectable pancreatic cancer. Surg Endosc. 2018;32:4022-4028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
102.  Song TJ, Seo DW, Lakhtakia S, Reddy N, Oh DW, Park DH, Lee SS, Lee SK, Kim MH. Initial experience of EUS-guided radiofrequency ablation of unresectable pancreatic cancer. Gastrointest Endosc. 2016;83:440-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 122]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
103.  Wang J, Wang Y, Zhao Y, Wu X, Zhang M, Hou W, Chen Q, Cheng B. Endoscopic ultrasound-guided radiofrequency ablation of unresectable pancreatic cancer with low ablation power and multiple applications: a preliminary study of 11 patients. Ann Palliat Med. 2021;10:1842-1850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
104.  Herman JM, Chang DT, Goodman KA, Dholakia AS, Raman SP, Hacker-Prietz A, Iacobuzio-Donahue CA, Griffith ME, Pawlik TM, Pai JS, O'Reilly E, Fisher GA, Wild AT, Rosati LM, Zheng L, Wolfgang CL, Laheru DA, Columbo LA, Sugar EA, Koong AC. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer. 2015;121:1128-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 332]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
105.  Teriaca MA, Loi M, Suker M, Eskens FALM, van Eijck CHJ, Nuyttens JJ. A phase II study of stereotactic radiotherapy after FOLFIRINOX for locally advanced pancreatic cancer (LAPC-1 trial): Long-term outcome. Radiother Oncol. 2021;155:232-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
106.  Xie C, Duffy AG, Brar G, Fioravanti S, Mabry-Hrones D, Walker M, Bonilla CM, Wood BJ, Citrin DE, Gil Ramirez EM, Escorcia FE, Redd B, Hernandez JM, Davis JL, Gasmi B, Kleiner D, Steinberg SM, Jones JC, Greten TF. Immune Checkpoint Blockade in Combination with Stereotactic Body Radiotherapy in Patients with Metastatic Pancreatic Ductal Adenocarcinoma. Clin Cancer Res. 2020;26:2318-2326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 53]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
107.  Moningi S, Abi Jaoude J, Kouzy R, Lin D, Nguyen ND, Garcia Garcia CJ, Phan JL, Avila S, Smani D, Cazacu IM, Singh BS, Smith GL, Holliday EB, Koay EJ, Das P, Bhutani MS, Herman JM, Minsky BD, Koong AC, Taniguchi CM. Impact of Fiducial Marker Placement Before Stereotactic Body Radiation Therapy on Clinical Outcomes in Patients With Pancreatic Cancer. Adv Radiat Oncol. 2021;6:100621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
108.  Patel JB, Revanur V, Forcione DG, Bechtold ML, Puli SR. Endoscopic ultrasound-guided fiducial marker placement in pancreatic cancer: A systematic review and meta-analysis. World J Gastrointest Endosc. 2020;12:231-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
109.  Dimcevski G, Kotopoulis S, Bjånes T, Hoem D, Schjøtt J, Gjertsen BT, Biermann M, Molven A, Sorbye H, McCormack E, Postema M, Gilja OH. A human clinical trial using ultrasound and microbubbles to enhance gemcitabine treatment of inoperable pancreatic cancer. J Control Release. 2016;243:172-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 288]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
110.  Ning Z, Xie J, Chen Q, Zhang C, Xu L, Song L, Meng Z. HIFU is safe, effective, and feasible in pancreatic cancer patients: a monocentric retrospective study among 523 patients. Onco Targets Ther. 2019;12:1021-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
111.  Prat F, Chapelon JY, Arefiev A, Cathignol D, Souchon R, Theillière Y. High-intensity focused ultrasound transducers suitable for endoscopy: feasibility study in rabbits. Gastrointest Endosc. 1997;46:348-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
112.  Li T, Khokhlova T, Maloney E, Wang YN, D'Andrea S, Starr F, Farr N, Morrison K, Keilman G, Hwang JH. Endoscopic high-intensity focused US: technical aspects and studies in an in vivo porcine model (with video). Gastrointest Endosc. 2015;81:1243-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
113.  Ma YY, Leng Y, Xing YL, Li HM, Chen JB, Niu LZ. Gemcitabine plus concurrent irreversible electroporation vs gemcitabine alone for locally advanced pancreatic cancer. World J Clin Cases. 2020;8:5564-5575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (1)]
114.  van Veldhuisen E, Vroomen LG, Ruarus AH, Derksen TC, Busch OR, de Jong MC, Kazemier G, Puijk RS, Sorgedrager NS, Vogel JA, Scheffer HJ, van Lienden KP, Wilmink JW, Besselink MG, Meijerink MR. Value of CT-Guided Percutaneous Irreversible Electroporation Added to FOLFIRINOX Chemotherapy in Locally Advanced Pancreatic Cancer: A Post Hoc Comparison. J Vasc Interv Radiol. 2020;31:1600-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
115.  Lin M, Liang S, Wang X, Liang Y, Zhang M, Chen J, Niu L, Xu K. Percutaneous irreversible electroporation combined with allogeneic natural killer cell immunotherapy for patients with unresectable (stage III/IV) pancreatic cancer: a promising treatment. J Cancer Res Clin Oncol. 2017;143:2607-2618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
116.  Ruarus AH, Vroomen LGPH, Geboers B, van Veldhuisen E, Puijk RS, Nieuwenhuizen S, Besselink MG, Zonderhuis BM, Kazemier G, de Gruijl TD, van Lienden KP, de Vries JJJ, Scheffer HJ, Meijerink MR. Percutaneous Irreversible Electroporation in Locally Advanced and Recurrent Pancreatic Cancer (PANFIRE-2): A Multicenter, Prospective, Single-Arm, Phase II Study. Radiology. 2020;294:212-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 70]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]