Abstract
Recent advances in biomedical research in cancer immunotherapy have identified the use of an oxidative stress-based approach to treat cancers, which works by inducing immunogenic cell death (ICD) in cancer cells. Since the anti-cancer effects of non-thermal plasma (NTP) are largely attributed to the reactive oxygen and nitrogen species that are delivered to and generated inside the target cancer cells, it is reasonable to postulate that NTP would be an effective modality for ICD induction. NTP treatment of tumors has been shown to destroy cancer cells rapidly and, under specific treatment regimens, this leads to systemic tumor-specific immunity. The translational benefit of NTP for treatment of cancer relies on its ability to enhance the interactions between NTP-exposed tumor cells and local immune cells which initiates subsequent protective immune responses. This review discusses results from recent investigations of NTP application to induce immunogenic cell death in cancer cells. With further optimization of clinical devices and treatment protocols, NTP can become an essential part of the therapeutic armament against cancer.
Cancer and conventional therapies:
Cancer development is partly attributed to cells acquiring multiple mutations that remain unrecognized by the immune system due to low immunogenicity of these mutated cells. Failure of the immune system to recognize cancer cells contributes to cancer progression by selecting for tumor cells that can survive in an immunocompetent host. Unchecked growth is further facilitated within the tumor microenvironment by the production of immunosuppressive factors by these cancerous cells, stromal cells, and local immune cells. [1–3]. To successfully check the development and progression of cancer, the innate and adaptive immune system must act together to identify and destroy mutated cells [1]. The innate arm of the immune system generates a rapid, non-specific inflammatory response on recognition of a “foreign” cell (mutated) which serves to initiate development of specific immune responses. The innate cells have the difficult job of differentiating between normal cells (self) and mutated cells (altered mutated self), often a key step in the failure of immune recognition. The adaptive arm of the immune system, on the other hand, allows for specific immunity to mutated cells via T cells, but takes days to weeks to develop. It is the T lymphocytes that selectively destroy cancerous cells that carry the specific mutation throughout the body. Furthermore, immunologic memory, the hallmark of the adaptive immune system, allows for a more rapid immune response upon re-exposure to a specific antigen that defines its “foreignness” [4].
Modern cancer treatment typically entails a multimodal approach involving a combination of systemic chemotherapies, surgery, and radiation; it is associated with a broad spectrum of inherent toxicities and in many cases, only modest patient survival. The evolution of immunotherapeutic treatment strategies has offered newer treatment modalities for cancer patients with better clinical outcomes. These strategies are based on methods that break immune tolerance of self-antigens (or ‘altered mutated self’) to overcome ignorance and tolerance to potential antigenic targets on cancer cells as a way to destroy tumor cells by immunological mechanisms, hence improving selectivity and reducing toxicity[5, 6]. Recent historic advances in immunotherapy highlighted in this review underscore our continued quest for novel approaches to the treatment of cancer [7–11].
Non-thermal plasma in cancer therapy
Non-thermal plasma (NTP), also known as cold atmospheric plasma or non-equilibrium, atmospheric pressure plasma, has emerged as a promising anti-cancer approach. NTP is formed by the application of an electric field to ionize surrounding gas (ambient air or a specific gas), creating a discharge consisting of charged molecules, ultraviolet light, reactive oxygen species (ROS), and neutral molecules [12, 13] Mounting evidence demonstrates that NTP can change the oxidative status of cells through stimulation of intracellular ROS production [14, 15]. ROS are known to influence multiple signaling pathways regulating cell processes including proliferation, differentiation, and cell death [16]. Because NTP has the advantage of being a controlled source of incremental ROS, it is therefore being employed for the treatment of various diseases, including cancer. Two distinct methods are used to generate NTP [13, 15, 17–19]. Dielectric barrier discharge (DBD) devices generate plasma directly on the treatment target while plasma jets generate the bulk of the plasma remotely and utilize a gas, such as helium or argon, for transport of plasma components to the treatment target [13, 20–23]. The two main strategies employed for experimental, biomedical delivery of plasma involve (1) direct exposure of cultured cells in vitro or tissue in vivo or, more recently, (2) indirect exposure to plasma components using plasma-activated liquids in standard cell culture for in vitro studies or via perfusion of affected organs [13]. The anti-cancer effects of indirectly and directly applied NTP appear to be similar. Direct treatment exposes the target tissue to all the components of plasma. Yan et al demonstrated that directly applied NTP induced an “activation” state, resulting in increased sensitivity of cells to ROS and RNS [24]. With indirect treatment, active species dissolve into a liquid when exposed to plasma from different sources. The “treatment dose” is determined by time of plasma-exposure of the liquid as well as the period for which the cells or tissue is allowed to remain in contact with this liquid. While the differences between the interaction of NTP with liquid media (indirect) or the skin (direct) are complex and not fully clear, an important distinction between the two treatment modalities is the presence of short lived reactive species such as OH during direct treatment [25]. These short lived species may trigger production of intracellular, long lived reactive species such as H2O2, potentially resulting in H2O2-mediated lymphocyte activation[26]. In a study comparing indirect and direct application of NTP against metastatic melanoma, Saadati et al demonstrated significantly greater cancer cell death and reduction in tumor growth with direct treatment [25]. In the same study, they recognized the potential advantage of plasma activated liquids for the treatment of tumors deeper than skin level in combination with chemotherapy [25]. Other studies have demonstrated that application of plasma-treated medium decreases peritoneal tumor burden in mice and prolongs survival [27–29]. Although direct treatment with NTP is more effective than indirect treatment, indirect treatment is believed to be associated with less toxicity [24–26, 30].
NTP, whether generated from DBD or jet-based devices, results in somewhat selective toxicity to cancer cells both in vitro and in vivo (Table 1), although the subject of selectivity is open to question. In vitro, anticancer efficacy of NTP has been demonstrated in several distinct, established cancer cell lines: brain cancer, skin cancer, breast cancer, colorectal cancer, lung cancer, cervical cancer, leukemia, head and neck cancer, and hepatoma (Table 1) [13, 31–44]. The majority (70%) of these studies employed jet devices [13]. In a pioneering study demonstrating cancer growth inhibition in vivo, Vandamme et al. directly applied NTP to subcutaneous U87-Luc malignant glioma tumors in nude mice. They reported safety of their treatment protocol as well as anti-tumor activity [17, 45, 46]. Subsequent in vivo investigations looking at NTP for treatment of subcutaneous xenograft tumors or orthotopic melanomas in mice have consistently demonstrated tumor cell death/growth inhibition and increased animal survival [13, 37, 45–53]. Other mouse model studies have demonstrated anticancer capacity of plasma in bladder cancer [37], head and neck cancer [48], ovarian cancer [54], pancreatic cancer [47, 55], neuroblastoma [49], melanoma [51, 52], and breast cancer [50]. Local tumor regression was in seen in all cases, and in some cases, improved animal survival was documented. Since these studies employed different NTP devices, it is difficult to judge if the inconsistent animal survival is because of differences in tumor types or the nature and delivery of plasma generated. Furthermore, studies variously used immunocompetent or immunocompromised animals making it difficult to delineate the contribution of immune pathways in NTP efficacy. Finally, the mechanism of tumor cell death or the organismal level pathways activated were not investigated in depth in the aforementioned studies. These topics are subjects of ongoing studies in many labs around the world.
Table 1: Studies Investigating Anti-Cancer Potential of Non-Thermal Plasma (NTP).
A list of in vivo and in vitro studies reviewed, including type of cancer investigated in each study.
| In Vivo | In Vitro | Plasma Source Used | Article |
|---|---|---|---|
| Benign Melanocytic Tumors | Ar Plasma Jet | Yajima et al | |
| Bladder Ca | He Plasma Jet | Keidar et al | |
| Breast | He Plasma Jet | Mirpour et al | |
| Breast | He Plasma Jet | Mirpour et al | |
| Breast | He Plasma Jet | Wang et al | |
| Cervical | Microplasma Jet Device | Tan et al | |
| Cervical | Plasma Jet with Air Flow | Ahn et al | |
| Colorectal | Ar Plasma Jet | Bekeschus et al | |
| Colorectal | FE-DBD | Vandamme et al | |
| Colorectal | He Plasma Jet | Ishaq et al | |
| Colorectal | Colorectal | DBD | Lin et al* |
| Glioblastoma | FE-DBD | Vandamme et al | |
| Glioblastoma | DBD | Kaushik et al | |
| Glioblastoma | Indirect PAM treated with Ar gas flow | Tanaka et al | |
| Glioblastoma | Glioblastoma | FE-DBD | Vandamme et al |
| Hepatoma | Microplasma Jet Device | Tan et al | |
| Hepatoma | He Plasma Jet | Zhao et al | |
| HNSCC | He Plasma Jet | Guerrero-Preston et al | |
| HNSCC | HNSCC | He/O2 Plasma Jet | Kang et al |
| Leukemia | Atmospheric Air Plasma System | Thiyagarajan et al | |
| Leukemia | DBD | Thiyagarajan et al | |
| Lung | DBD | Lin et al* | |
| Lung | He Plasma Jet | Keidar et al | |
| Lung | Microplasma Jet Device | Kim, JY et al | |
| Melanoma | DBD | Chernets et al | |
| Melanoma | DBD | Lin et al* | |
| Melanoma | N2/O2 Plasma Jet | Mizuno et al | |
| Melanoma | Ar Plasma Jet | Bekeschus et al | |
| Melanoma | DBD | Kim, G et al | |
| Melanoma | FE-DBD | Fridman et al | |
| Melanoma | Melanoma | He Plasma Jet | Keidar et al |
| Neuroblastoma | He Plasma Jet | Walk et al | |
| Ovarian | Ovarian | Indirect PAM treated Ar Plasma Jet | Utsumi et al |
| Pancreatic | He Plasma Jet | Brulle et al | |
| Pancreatic | He Plasma Jet | Brulle et al | |
| Pancreatic | Pancreatic | Direct Ar Plasma Jet, Indirect PAM treated with Ar Plasma Jet | Liedtke et al* |
| Pancreatic | Pancreatic | Ar Plasma Jet | Pertecke et al |
indicated studies investigating NTP-induced immunogenic cell death (ICD).
Anti-cancer mechanism of non-thermal plasma
It is fair to state that the anti-cancer mechanism of NTP is incompletely understood. The effects and importance of individual NTP components (radiation, electromagnetic fields, and reactive species) in cancer cell death continue to be investigated and it seems unlikely that any single component will emerge as the critical factor. Nevertheless, the direct anti-cancer potential of NTP has been ascribed to a rise in reactive species inside cancer cells, changing the cellular redox balance. Although there is evidence that other components of NTP contribute to its therapeutic role, the cocktail of RONS, both originating from NTP and those produced by NTP-exposed cells, are postulated to be the most important active constituent for the effects on cancer cells [56]. The resulting challenge to the cancer cell’s anti-oxidant response ultimately pushes the cell toward cell cycle arrest and apoptosis [13]. While other therapies function through a similar pathway, one of the primary advantages of NTP is selective toxicity against cancer cells by the ability to controllably deliver ROS such that there is relatively little impact on non-cancerous cells in the tumor microenvironment and the surrounding tissues. Although defining the mechanism of selectivity against cancer cells continues to be a key challenge in the field, it has been attributed to greater rise in ROS in cancer cells compared to normal cells [13, 57, 58].
Another challenge in the understanding of NTP mechanism of action is the penetration of plasma components in treated tissue. The physical effects of plasma are believed to be only a few hundred micrometers, but biological effects been observed several millimeters deep within tissue. The biological effects of NTP extend beyond the superficial layers, evidenced by significant shrinkage of subcutaneous tumors with diameters of ~ 1 cm following surface NTP treatment [17, 47, 49]. However, the specific plasma-produced RONS delivered to biological targets and the depth to which they are delivered remain uncertain. Furthermore, there remain questions as to how RONS interact with various components of the target tissue [59]. Experimental and simulation studies have shown that deeper in the tissue (greater than millimeters below the surface), most plasma-derived RONS are converted to more stable RONS with longer half-lives [60–64]. Since the initial interaction between plasma-derived RONS and the tissue remains largely unknown, the reasons why biological effects permeate beyond physical reach of NTP components remain a key area of research interest.
Recent studies suggest that immune activation may also contribute to NTP’s anti-cancer effects [1, 65–67]. In that context, the efficacy of plasma treatment, with respect to life-expectancy, was greater in studies performed on immunocompetent mice compared to those using immunodeficient mice. These particular findings warrant further investigations of the immunogenic capacity of plasma [1, 37, 49]. More recently, studies have demonstrated macrophage augmentation and immunogenic cell death in response to NTP-induced ROS and reactive nitrogen species (RNS) [65–70]. Importantly, Mizuno et. al. demonstrated abscopal effects of NTP on B16-F10 melanoma growth. This was evidenced by tumor growth suppression at sites distal to site of NTP treatment in CD2F1 and C57BL/6 murine models [71], further endorsing the immunogenic potential of NTP. It is important to reiterate that more in depth longitudinal studies of plasma interaction with cancers, especially in animal models, are critically needed to define the anti-cancer mechanism of NTP.
Recent advances in immunotherapy for cancer:
Cancer immunotherapy has gained popularity and momentum in recent years. In fact, the 2018 Nobel Prize in Physiology or Medicine was awarded to cancer immunotherapy researchers James P. Allison and Tasuku Honjo for their historic work on immune check-point inhibitors. Indeed, the ultimate goal of cancer immunotherapy is to induce immunologic memory against tumor cells. However, it remains challenging to induce immunologic memory against a tumor cell that continues to acquire mutations, thus changing its antigenic nature and allowing for immune escape. Allison and Honjo established that by inhibiting cellular “off” signals [i.e., cytotoxic T lymphocyte associated protein 4 (CTLA-4), which is vital for maintaining homeostasis during normal immune responses, and programmed death protein 1 (PD-1) of T lymphocytes (the foot soldiers of the immune system)], the T lymphocyte response would be unleashed to eliminate tumor cells. Today, ipilimumab, the antibody to CTLA-4, is an FDA approved treatment for melanoma [9, 10] (Figure 1a). Furthermore PD-1 blocking agents (nivolumab, pembrolizumab, durvalumab, atezolizumab and avelumab) remove the brakes on T cells, allowing them to effect tumor regression and improved survival in patients with advanced and metastatic tumors [11] (Figure 1b).
Fig 1: Current Immunotherapy Approaches:
When the T cell receptor encounters a foreign antigen presented in the MHC cleft of an APC, several checks and balances are put in place to avoid inappropriate immune responses. T cell proliferation and activation occurs only if the next step of CD28 (a co-stimulatory molecule) binding with B7 ensues. CTLA4 is another natural ligand for B7 but this binding does results in T cell inhibition. It is the fine-tuned balance between CD28/B7 and CTLA4/B7 that determines the activated (T cell on) and inactivated (T cell off) status and hence the nature of immune responses. PD1 is another negative regulator of T cell function that functions later during an immune response. Another approach being used in the clinic is the production of genetically engineered T cells (CAR T cells) that are designed to attack specific tumor antigens. (MHC, major histocompatibility complex; APC, antigen presenting cell)
1a: Cytotoxic T Lymphocyte Associated Protein 4 (CTLA4) Blockade
CTLA4 functions as a checkpoint during immune responses to foreign antigens by sending inhibitory signals and down regulating T cell responses. This function is necessary for preventing autoimmunity. Normally, T-cell activation is attenuated by the regulatory mechanism of CTLA4/B7 interaction (T cell OFF). Release of negative regulation by using anti-CTLA4 antibodies is achieved by making CTLA4 unavailable for B7 interaction (T cell ON).
1b: Programmed Death 1/Programmed Death – Ligand 1 Blockade
T-cell proliferation and activation is attenuated by the normal regulatory mechanisms of PD-1/PD-L1 interaction (T cell OFF). PD1 expression is a hallmark of T cell exhaustion. When this interaction is blocked by antibodies, these inhibitory signals are overcome and the T cell becomes functional again (T cell ON).
1c: Chimeric Antigen Receptor-T Cell Production
CARs are fusion proteins consisting of antigen recognition domain and T cell signaling domain. T cells can be made to express CARs, resulting in a targeted anti-cancer immune response. The antigen-recognizing variable regions (scFVs) are from monoclonal antibodies (mAB) with an intracellular T-cell signaling domain. The CD3 ζ chain of the T-cell receptor (TCR) was used in first generation receptors. Second and third generation receptors utilize costimulatory signals such as CD28 or 4–1 BB (which is derived from the TNF family). CARs are then inserted into T-cells via a retrovirus-mediated gene transfer.
Chimeric antigen receptors (CARs) are another novel class of drugs in the field of cancer immunotherapy. CAR is a fusion protein consisting of an antigen recognition domain and a T cell signaling domain. It is the antigen recognition domain that recognizes and binds tumor proteins to provide the specificity against the tumor being targeted. T cells can be genetically engineered in the laboratory to express specific CARs against cancer cells, resulting in direct, targeted anti-cancer immune response. The signaling domain initiates killing pathways once the antigen recognition domain is bound to the antigen – in this case, the cancer cell expressing the antigen. Two CAR T cell therapy agents are now approved for the treatment of hematologic malignancies for pediatric acute lymphoblastic leukemia (ALL) and adult diffuse large B-cell lymphoma (DLBCL) [72, 73] (Figure 1c).
While there have been significant breakthroughs made in the field of cancer immunotherapy, these agents have, at times, been subject to side effects and lack of success against all tumors. Many patients are refractory to these treatments and others develop resistance. They are also extremely expensive and time intensive to generate. Importantly, these advances provide strong support for the importance of harnessing the immune system for cancer control as a vital part of cancer treatment regimens. They provide a platform for continued development of novel immunotherapeutic strategies.
Rationale for use of physical methods in immunotherapy
In addition to these immunotherapeutic agents, more general, non-specific physical methods known to activate the immune system, including photodynamic therapy (PDT) and radiation therapy (RT), have been employed to treat cancer [1]. The anti-cancer properties of PDT are attributed to the combination of non-toxic photosensitizers and visible light, which, upon interaction with oxygen, produce cytotoxic ROS that kill malignant cells via apoptosis/necrosis, shut down tumor vasculature, and subsequently stimulate the immune system [74]. Immunogenicity of PDT is attributed to induction of acute inflammation and the release of cytokines/stress response proteins, with the resultant leukocyte influx contributing to tumor destruction and, importantly, immune memory [1, 74, 75]. RT is part of the traditional anti-cancer armament being used for tumor regression through direct cytotoxicity. In recent years, the immuno-stimulatory potential of RT, another redox stress-based therapy, in conjunction with other immunotherapeutic agents has been and continues to be investigated in ongoing clinical trials involving several types of cancer [1, 76, 77].
Immunogenic cell death-based immunotherapy and rationale for plasma-based immunotherapy
Immunogenic cell death (ICD), initially described by the Kroemer/Zitvogel model, is a form of programmed cell death that has the potential to stimulate the adaptive immune system [67, 78, 79]. Figure 2a demonstrates the proposed role of NTP in induction of ICD. Various anti-cancer agents, including select systemic chemotherapies, PDT, RT, high hydrostatic pressure, small molecules, and oncolytic viruses, have been described as having the capacity to elicit ICD. When ICD occurs, immunostimulatory molecules collectively called damage associated molecular patterns (DAMPs) are released from or displayed by the dying cell [78]. Figure 2b demonstrates the proposed mechanism of NTP-induced ICD. Several surrogate markers suggestive of ICD can be detected both in vitro and in vivo [78, 79]. These surrogate markers, or critical DAMPs, include surface-expressed calreticulin (ecto-CRT), secreted ATP, high mobility group box 1 (HMGB1), heat shock protein 70 (HSP70), heat shock protein 90 (HSP90), and type I interferons (IFNs) [78, 80]. These molecules are harmless when inside the cell and perform vital functions for maintaining cellular integrity and function. They become immunogenic only when outside the cell, whether membrane-bound or released. Once externalized, DAMPs initiate an immunologic response by attracting innate cells, also called antigen presenting cells (APCs) (e.g., macrophages, dendritic cells) to the tumor. ATP and HMGB1 act as “find me” signals for APCs and chemotactically recruit APCs to the area of DAMP emission. Ecto-CRT acts as an “eat me” signal for APCs, promoting phagocytosis of DAMP-emitting cells and thus causing activation of APCs. The activated APCs travel to lymphatic organs and present tumor antigens to T cells resulting in the expansion of effector and memory T cells that are specific for that tumor [78, 80].
Figure 2a: Potential Action of Non-thermal Plasma (NTP) in Cancer Immunotherapy.
NTP via proposed exposure both directly stimulates immune cells and induces immunogenic cell death. Immunogenic cell death (Ref 28, 67, 85) results in recruitment and stimulation of APC (antigen presenting cells) (Ref 28, 67, 85), memory cell formation, and T-cell development (Ref 67, 85). These circulating cells can then target other non-NTP exposed metastatic tumors of the same origin.
Figure 2b: Proposed Mechanism of Non-thermal Plasma Induced Immunogenic Cell Death (ICD).
Immunogenic cell death is a specific cell death mechanism that stimulates cells of the adaptive immune system. When ICD occurs, damage associated molecular patterns (DAMPs) are emitted; calreticulin is externalized on the cell membrane and ATP, HMGB1, and HSPs (heat shock proteins 70 and 90) are released by the dying cell (Ref 65, 67, 68, 69, 70). These DAMPs then initiate an immunologic response by attracting innate dendritic cells to the tumor. DAMPs also aid in the maturation of antigen carrying dendritic cells, resulting in a tumor-specific immune response by activated T cells (Ref 28, 67).
In vitro emission of DAMPs serves as a useful indicator of ICD. However, the gold standard for verifying the functional ICD notably requires a vaccination protocol involving immunocompetent animal models and syngeneic cancer cells [78] (Figure 3). This vaccination protocol involves in vitro exposure of malignant cells to an ICD inducer under evaluation. Following thorough washing of malignant cells to remove the ICD-inducing agent but not the DAMPs, these dying malignant cells are inoculated subcutaneously into a flank of immunocompetent mice. One week later, the mice are challenged with subcutaneous inoculation of the same living malignant cells in the opposite flank. The mice are then monitored for development of subcutaneous tumors at the challenge site. The proportion of mice that do not develop tumors at the challenge site reflects the immunogenicity of cell death induced by the ICD agent being tested [78]. NTP has now emerged as a bona fide ICD inducer. [67, 81]
Figure 3: Validation of Non-Thermal Plasma-Induced (NTP) ICD Using a Vaccination Protocol.
Vaccination protocol demonstrating protective immunity against a tumor challenge in mice immunized with non-thermal plasma (NTP) treated tumor cells. Vaccination with tumor cells undergoing ICD causes development of a robust, protective immune response against the specific tumor in the animals. When challenged with live tumor cells at a different site, none or smaller tumor nodes should be observed, if the vaccination was successful. In mice immunized with NTP treated tumor cells, several animals developed no tumors. Of those that did, a significantly smaller mean tumor volume at the distal challenge site was observed as compared to the control group.
Existing inducers of ICD have drawbacks as well as advantages. Although ICD-mediated immuno-stimulatory effects of radiation have been identified, there is little evidence of direct immune cell stimulation. To the contrary, immune cells are highly radiosensitive [1, 82]. Additionally, there is mounting evidence suggesting that RT can contribute to reversion of the tumor suppressive barriers of the tumor microenvironment [83]. Similarly, while PDT is an ICD inducer, it has also been linked to immunosuppression in mice and occurrence of late metastases in patients [84].
Similar to RT and PDT, NTP is an attractive candidate as an ICD inducer with the added benefit of stimulating, or at the very least, preserving immune cells. NTP for cancer treatment would compete in this landscape and the comparison is summarized in Table 2. NTP is unique in that it induces ICD in tumor cells and simultaneously stimulates immune cells at the same treatment regimen. Studies have demonstrated enhanced migratory activity in macrophages exposed to NTP in vitro [1, 66]. Furthermore, increased macrophage-mediated, tumor cell killing of radioresistant, human CNE-1 nasopharyngeal carcinoma cells subsequent to NTP exposure has been demonstrated [1, 65]. Thus, NTP may confer a significant advantage over RT, PDT, and other ICD inducers that have demonstrated concomitant immunosuppression. Further work is required to confirm and characterize this direct immunostimulatory effect of NTP within plasma-exposed tumors.
Table 2:
Comparison of Nonthermal Plasma vs Radiation Therapy vs Photodynamic Therapy
| NTP | RT | PDT | |
|---|---|---|---|
| Targeted | Yes | Yes | Yes |
| May require Invasive procedure for application | Yes | No | Yes |
| Side effects | Unknown No major side effects reported |
Skin changes, second cancer, site specific side effects caused by damage to nearby organs |
Skin changes |
| Mechanism of action | Oxidative stress [13] | DNA breakage [82] | Oxidative stress, damage to tumor blood vessels [74] |
| Depth of effect | Superficial | Deep | Superficial |
| Causes ICD | Yes [66] | Yes [76] | Yes [74] |
| Direct effects on immune cells | Preservation/stimulatory [1] | Suppressive [82] | Suppressive [84] |
NTP- non-thermal plasma, RT- radiation therapy, PDT- photodynamic therapy
In vitro and In vivo data demonstrating NTP-induced ICD
NTP-induced ICD has been investigated in vitro in lung cancer and nasopharyngeal carcinoma via detection of DAMPs, which are surrogate markers of ICD. Specifically, intracellular ROS-induced expression of DAMPs (ecto-CRT and ATP) was shown in human A549 lung carcinoma cells and human CNE-1 nasopharyngeal carcinoma cells [65, 68]. In addition, there was simultaneous activation of anti-tumor effects of macrophages. This was evidenced by trans-well experiments demonstrating increased CNE-1 cell loss when CNE-1 cells were co-cultured with plasma-exposed macrophages [65]. Tumor cell toxicity and immunogenicity were also demonstrated following exposure to NTP in cultured murine metastatic B16F10 melanoma cells [70]. In this study, NTP treatment resulted in decreased metabolic activity, decelerated cell growth, and increased cell death. Immunogenicity was confirmed by expression of surrogate ICD markers, including major histocompatibility complex I, ecto-CRT, and melanocortin receptor I [70]. ICD resulting from NTP treatment has also been demonstrated in vitro in CT26 colon cancer cells. These cells were found to undergo apoptosis accompanied by calreticulin externalization upon exposure to NTP [69]. Studies with newer plasma devices and diverse treatment regimens are ongoing in many laboratories around the world to induce ICD in different tumor cell types.
In vivo investigations of the anticancer capacity of NTP have been undertaken for brain cancer, skin cancer, breast cancer, colorectal cancer, lung cancer, cervical cancer, leukemia, head and neck cancer, and hepatoma [12]. The role of ICD in plasma-mediated cancer immunotherapy has been specifically investigated in multiple studies using murine models. These studies have introduced a novel paradigm of plasma onco-immunotherapy to engage the immune system via ICD induction. In the seminal study demonstrating NTP-induced ICD in vivo, a CT26 murine colorectal tumor model was employed [67]. The generation of DAMPs (ATP and externalized-CRT) along with recruitment of APCs to the tumor area was clearly demonstrated. Most importantly, the capacity of NTP to function as a bona fide ICD inducer using the prescribed vaccination protocol was demonstrated. This vaccination assay demonstrated protective immunity against CT26 tumor challenge in Balb/c mice immunized with NTP-treated CT26 cells. Ninety percent of the mice in the plasma-immunized group had tumor volumes smaller than the mean tumor volume of the control group. Furthermore, three of the ten mice in the plasma-immunized group did not develop subcutaneous tumors at the challenge site, signifying complete protection [67].
This vaccination protocol has also been successfully employed in a melanoma mouse model [85]. C57BL/6 mice were vaccinated with melanoma cells treated with DBD plasma. Of the eight mice in the NTP-treated group, five were completely protected against challenge with live tumor cells, comparable to cells treated with a known ICD-inducer (Mitoxantrone) [85]. Furthermore, it was demonstrated that the observed ICD-inducing effects of DBD plasma were from the short-lived ROS (e.g. OH, NO, O/O) produced, and not from more persistent species (H2O2, NO2−, NO3−, ONOO−). Vaccines created with equivalent amounts of persistent species did not significantly improve protection of mice against tumor challenge (37.5%) compared to the negative control group (25%) [85]. These two in vivo studies offer conclusive evidence that direct application of NTP induces ICD in multiple cancer cell lines of different tissue origins. Similar observations have been reported with the perfusion of plasma-activated liquids in the abdominal cavities of mice bearing metastatic pancreatic cancer masses [27, 53]. Here, the effect is largely due to the more stable species, in contrast to the observations of Lin et al [85]. In-depth, mechanistic studies and longitudinal studies to determine the effect on disease progression/remission and animal survival are needed. Further investigations into plasma delivery and administration protocols with other cancer-cell lines are also warranted for potential clinical translation.
Clinical translation of NTP
Modern approaches to cancer treatment include multimodality strategies that typically involve combinations of surgery, radiation, systemic chemotherapy, and more recently, immunotherapy. These therapeutic modalities are frequently associated with minimal or incomplete treatment responses, disease progression/recurrence, inherent toxicity, and limited patient survival. NTP provides an important platform, rationale, and applicability in the arena of cancer treatment. We propose several avenues for using NTP alone as a cancer therapy or in combination with currently available therapeutic strategies.
Previous applications of NTP in oncology have focused on direct tumor cell killing, ignoring NTP’s impact on host immunity. Future studies will extend recent observations to further elucidate the immunogenic cascade and therapeutic response induced in the host in response to NTP treatment. If successful, NTP can provide a safer alternative or adjunct to existing ICD mediators, such as RT, chemotherapy, and PDT. NTP used in conjunction with current therapeutic strategies may synergistically increase antitumor responses, hence decreasing therapeutic levels of chemotherapy administered (dose sparing) while converting ICD transforming “cold” metastatic lesions into inflammatory “hot” spots prone to immune recognition and tumor immunity. The therapeutic potential of NTP is particularly promising in the treatment of cancers refractory to conventional therapies due to development of resistance.
Another challenge faced by the clinician is the all-too-often clinical scenario of patients with unresectable or borderline resectable locally advanced tumors. We propose that NTP can potentially transition/downstage these tumors to resectable tumors making patients better candidates for operative removal. To accomplish this, NTP can be directly applied to tumors both by laparoscopic and robotic as well as open laparotomy approaches for in situ delivery and treatment. Additionally, NTP may be used to directly treat microscopic positive surgical tumor margins in order to prevent local disease recurrence. Moreover, after surgical resection, NTP may also be applied indirectly to treat clinically occult micro metastatic peritoneal and serosal disease bearing surfaces to halt significant disease progression. Likewise, indirect application may be used in the case of peritoneal metastases whereby NTP-treated peritoneal fluid is infused into the peritoneal cavity by placing indwelling catheters, similar to application frequently used for delivery of intraperitoneal chemotherapy. This expanding treatment paradigm may also be applied similarly to treating recurrent disease. As our understanding of mechanism of NTP action improves, so too will the clinical applications available for the use of NTP in cancer therapy.
Conclusion
Herein we have summarized the therapeutic potential of NTP application for cancer immunotherapy via ICD. Previous applications of NTP in oncology have focused on direct anti-cancer (ablative) approaches, ignoring the potential impact of plasma therapy on host immunity. NTP-induced ICD can promote lasting immunity against tumor antigens. Several animal studies have, in part, elucidated the mechanisms and potential efficacy of this approach. Further evaluation of mechanism of action of NTP, optimization of NTP devices, clinical delivery systems, and treatment protocols, will facilitate application of NTP-induced ICD to the clinical setting.
References
- [1].Miller V, Lin A, Fridman A, Why Target Immune Cells for Plasma Treatment of Cancer, 36 (2016) 259–268 [Google Scholar]
- [2].Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, Deng W, Wang L, Shifrin N, Raulet DH, Recognition of tumors by the innate immune system and natural killer cells, Advances in immunology, 122 (2014) 91–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Schreiber RD, Old LJ, Smyth MJ, Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion, Science (New York, N.Y.), 331 (2011) 1565–1570. [DOI] [PubMed] [Google Scholar]
- [4].Charles PT Janeway A Jr, Walport Mark, Shlomchik Mark J, Immunobiology: The Immune System in Health and Disease, Garland Science, New York, 2001. [Google Scholar]
- [5].Weber LW, Bowne WB, Wolchok JD, Srinivasan R, Qin J, Moroi Y, Clynes R, Song P, Lewis JJ, Houghton AN, Tumor immunity and autoimmunity induced by immunization with homologous DNA, The Journal of Clinical Investigation, 102 (1998) 1258–1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Bowne WB, Srinivasan R, Wolchok JD, Hawkins WG, Blachere NE, Dyall R, Lewis JJ, Houghton AN, Coupling and Uncoupling of Tumor Immunity and Autoimmunity, The Journal of Experimental Medicine, 190 (1999) 1717–1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Yasuda H, Solid tumor physiology and hypoxia-induced chemo/radio-resistance: novel strategy for cancer therapy: nitric oxide donor as a therapeutic enhancer, Nitric oxide : biology and chemistry, 19 (2008) 205–216. [DOI] [PubMed] [Google Scholar]
- [8].Sun CC, Bodurka DC, Weaver CB, Rasu R, Wolf JK, Bevers MW, Smith JA, Wharton JT, Rubenstein EB, Rankings and symptom assessments of side effects from chemotherapy: insights from experienced patients with ovarian cancer, Supportive Care in Cancer, 13 (2005) 219–227 [DOI] [PubMed] [Google Scholar]
- [9].Allison JP, Checkpoints, Cell, 162 (2015) 1202–1205. [DOI] [PubMed] [Google Scholar]
- [10].Allison JP, Immune checkpoint blockade in cancer therapy: The 2015 lasker-debakey clinical medical research award, JAMA, 314 (2015) 1113–1114. [DOI] [PubMed] [Google Scholar]
- [11].Chowdhury PS, Chamoto K, Honjo T, Combination therapy strategies for improving PD-1 blockade efficacy: a new era in cancer immunotherapy, Journal of internal medicine, 283 (2018) 110–120. [DOI] [PubMed] [Google Scholar]
- [12].Keidar M, Yan D, Beilis II, Trink B, Sherman JH, Plasmas for Treating Cancer: Opportunities for Adaptive and Self-Adaptive Approaches, Trends in biotechnology, 36 (2018) 586–593. [DOI] [PubMed] [Google Scholar]
- [13].Yan D, Sherman JH, Keidar M, Cold atmospheric plasma, a novel promising anti-cancer treatment modality, Oncotarget, 8 (2017) 15977–15995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Lu X, Naidis GV, Laroussi M, Reuter S, Graves DB, Ostrikov K, Reactive species in non-equilibrium atmospheric-pressure plasmas: Generation, transport, and biological effects, Physics Reports, 630 (2016) 1–84. [Google Scholar]
- [15].Steinbeck M, Chernets N, Zhang J, S Kurpad D, Fridman G, Fridman A, Freeman T, Skeletal Cell Differentiation Is Enhanced by Atmospheric Dielectric Barrier Discharge Plasma Treatment, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Sarsour EH, Kumar MG, Chaudhuri L, Kalen AL, Goswami PC, Redox control of the cell cycle in health and disease, Antioxidants & redox signaling, 11 (2009) 2985–3011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Vandamme M, Robert E, Pesnel S, Barbosa E, Dozias S, Sobilo J, Lerondel S, Le Pape A, Pouvesle J-M, Antitumor Effect of Plasma Treatment on U87 Glioma Xenografts: Preliminary Results, Plasma Processes and Polymers, 7 (2010) 264–273. [Google Scholar]
- [18].Miller V, Lin A, Kako F, Gabunia K, Kelemen S, Brettschneider J, Fridman G, Fridman A, Autieri M, Microsecond-pulsed dielectric barrier discharge plasma stimulation of tissue macrophages for treatment of peripheral vascular disease, Physics of Plasmas, 22 (2015) 122005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Heinlin J, Morfill G, Landthaler M, Stolz W, Isbary G, Zimmermann JL, Shimizu T, Karrer S, Plasma medicine: possible applications in dermatology, Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG, 8 (2010) 968–976. [DOI] [PubMed] [Google Scholar]
- [20].Wagner HE, Brandenburg R, Kozlov KV, Sonnenfeld A, Michel P, Behnke JF, The barrier discharge: basic properties and applications to surface treatment, Vacuum, 71 (2003) 417–436. [Google Scholar]
- [21].Laroussi M, Akan T, Arc-Free Atmospheric Pressure Cold Plasma Jets: A Review, Plasma Processes and Polymers, 4 (2007) 777–788. [Google Scholar]
- [22].Weltmann K-D, Kindel E, von Woedtke T, Hähnel M, Stieber M, Brandenburg R, Atmospheric-pressure plasma sources: Prospective tools for plasma medicine, 2010. [Google Scholar]
- [23].Kogelschatz U, Dielectric-Barrier Discharges: Their History, Discharge Physics, and Industrial Applications, Plasma Chemistry and Plasma Processing, 23 (2003) 1–46. [Google Scholar]
- [24].Yan D, Xu W, Yao X, Lin L, Sherman JH, Keidar M, The Cell Activation Phenomena in the Cold Atmospheric Plasma Cancer Treatment, Scientific Reports, 8 (2018) 15418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Saadati F, Mahdikia H, Abbaszadeh H-A, Abdollahifar M-A, Khoramgah MS, Shokri B, Comparison of Direct and Indirect cold atmospheric-pressure plasma methods in the B16F10 melanoma cancer cells treatment, Scientific Reports, 8 (2018) 7689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Yan D, Cui H, Zhu W, Talbot A, Zhang LG, Sherman JH, Keidar M, The Strong Cell based Hydrogen Peroxide Generation Triggered by Cold Atmospheric Plasma, Scientific reports, 7 (2017) 10831–10831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Liedtke KR, Freund E, Hackbarth C, Heidecke C-D, Partecke L-I, Bekeschus S, A myeloid and lymphoid infiltrate in murine pancreatic tumors exposed to plasma-treated medium, Clinical Plasma Medicine, 11 (2018) 10–17. [Google Scholar]
- [28].Takeda S, Yamada S, Hattori N, Nakamura K, Tanaka H, Kajiyama H, Kanda M, Kobayashi D, Tanaka C, Fujii T, Fujiwara M, Mizuno M, Hori M, Kodera Y, Intraperitoneal Administration of Plasma-Activated Medium: Proposal of a Novel Treatment Option for Peritoneal Metastasis From Gastric Cancer, Annals of surgical oncology, 24 (2017) 1188–1194. [DOI] [PubMed] [Google Scholar]
- [29].Nakamura K, Peng Y, Utsumi F, Tanaka H, Mizuno M, Toyokuni S, Hori M, Kikkawa F, Kajiyama H, Novel Intraperitoneal Treatment With Non-Thermal Plasma-Activated Medium Inhibits Metastatic Potential of Ovarian Cancer Cells, Scientific Reports, 7 (2017) 6085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Privat-Maldonado A, Gorbanev Y, Dewilde S, Smits E, Bogaerts A, Reduction of Human Glioblastoma Spheroids Using Cold Atmospheric Plasma: The Combined Effect of Short- and Long-Lived Reactive Species, Cancers, 10 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Tanaka H, Mizuno M, Ishikawa K, Nakamura K, Kajiyama H, Kano H, Kikkawa F, Hori M, Plasma-Activated Medium Selectively Kills Glioblastoma Brain Tumor Cells by Down-Regulating a Survival Signaling Molecule, AKT Kinase, 1 (2011) 265–277. [Google Scholar]
- [32].Kaushik NK, Attri P, Kaushik N, Choi EH, A preliminary study of the effect of DBD plasma and osmolytes on T98G brain cancer and HEK non-malignant cells, Molecules (Basel, Switzerland), 18 (2013) 4917–4928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Fridman G, Shereshevsky A, Jost MM, Brooks AD, Fridman A, Gutsol A, Vasilets V, Friedman G, Floating Electrode Dielectric Barrier Discharge Plasma in Air Promoting Apoptotic Behavior in Melanoma Skin Cancer Cell Lines, Plasma Chemistry and Plasma Processing, 27 (2007) 163–176. [Google Scholar]
- [34].Kim G, Kim G, Park S, Jeon S, Seo H, Iza F, Lee JK, Air plasma coupled with antibody-conjugated nanoparticles: a new weapon against cancer, Journal of Physics D: Applied Physics, 42 (2008) 032005. [Google Scholar]
- [35].Wang M, Holmes B, Cheng X, Zhu W, Keidar M, Zhang LG, Cold atmospheric plasma for selectively ablating metastatic breast cancer cells, PloS one, 8 (2013) e73741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Ishaq M, Evans MD, Ostrikov KK, Atmospheric pressure gas plasma-induced colorectal cancer cell death is mediated by Nox2-ASK1 apoptosis pathways and oxidative stress is mitigated by Srx-Nrf2 anti-oxidant system, Biochimica et biophysica acta, 1843 (2014) 2827–2837. [DOI] [PubMed] [Google Scholar]
- [37].Keidar M, Walk R, Shashurin A, Srinivasan P, Sandler A, Dasgupta S, Ravi R, Guerrero-Preston R, Trink B, Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy, British journal of cancer, 105 (2011) 1295–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Kim JY, Ballato J, Foy P, Hawkins T, Wei Y, Li J, Kim SO, Apoptosis of lung carcinoma cells induced by a flexible optical fiber-based cold microplasma, Biosensors & bioelectronics, 28 (2011) 333–338. [DOI] [PubMed] [Google Scholar]
- [39].Ahn HJ, Kim KI, Hoan NN, Kim CH, Moon E, Choi KS, Yang SS, Lee JS, Targeting cancer cells with reactive oxygen and nitrogen species generated by atmospheric-pressure air plasma, PloS one, 9 (2014) e86173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Tan X, Zhao S, Lei Q, Lu X, He G, Ostrikov K, Single-cell-precision microplasma-induced cancer cell apoptosis, PloS one, 9 (2014) e101299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Thiyagarajan M, Waldbeser L, Whitmill A, THP-1 leukemia cancer treatment using a portable plasma device, Studies in health technology and informatics, 173 (2012) 515–517. [PubMed] [Google Scholar]
- [42].Thiyagarajan M, Anderson H, Gonzales XF, Induction of apoptosis in human myeloid leukemia cells by remote exposure of resistive barrier cold plasma, Biotechnology and bioengineering, 111 (2014) 565–574. [DOI] [PubMed] [Google Scholar]
- [43].Zhao S, Xiong Z, Mao X, Meng D, Lei Q, Li Y, Deng P, Chen M, Tu M, Lu X, Yang G, He G, Atmospheric pressure room temperature plasma jets facilitate oxidative and nitrative stress and lead to endoplasmic reticulum stress dependent apoptosis in HepG2 cells, PloS one, 8 (2013) e73665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Guerrero-Preston R, Ogawa T, Uemura M, Shumulinsky G, Valle BL, Pirini F, Ravi R, Sidransky D, Keidar M, Trink B, Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells, International journal of molecular medicine, 34 (2014) 941–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Vandamme M, Robert E, Dozias S, Sobilo J, Lerondel S, Le Pape A, Pouvesle J-M, Response of Human Glioma U87 Xenografted on Mice to Non Thermal Plasma Treatment, 1 (2011) 27–43. [Google Scholar]
- [46].Vandamme M, Robert E, Lerondel S, Sarron V, Ries D, Dozias S, Sobilo J, Gosset D, Kieda C, Legrain B, Pouvesle JM, Pape AL, ROS implication in a new antitumor strategy based on non-thermal plasma, International journal of cancer, 130 (2012) 2185–2194. [DOI] [PubMed] [Google Scholar]
- [47].Brulle L, Vandamme M, Ries D, Martel E, Robert E, Lerondel S, Trichet V, Richard S, Pouvesle JM, Le Pape A, Effects of a non thermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model, PLoS One, 7 (2012) e52653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Kang SU, Cho JH, Chang JW, Shin YS, Kim KI, Park JK, Yang SS, Lee JS, Moon E, Lee K, Kim CH, Nonthermal plasma induces head and neck cancer cell death: the potential involvement of mitogen-activated protein kinase-dependent mitochondrial reactive oxygen species, Cell death & disease, 5 (2014) e1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Walk RM, Snyder JA, Srinivasan P, Kirsch J, Diaz SO, Blanco FC, Shashurin A, Keidar M, Sandler AD, Cold atmospheric plasma for the ablative treatment of neuroblastoma, Journal of pediatric surgery, 48 (2013) 67–73. [DOI] [PubMed] [Google Scholar]
- [50].Mirpour S, Piroozmand S, Soleimani N, Jalali Faharani N, Ghomi H, Fotovat Eskandari H, Sharifi AM, Mirpour S, Eftekhari M, Nikkhah M, Utilizing the micron sized non-thermal atmospheric pressure plasma inside the animal body for the tumor treatment application, Sci Rep, 6 (2016) 29048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Yajima I, Iida M, Kumasaka MY, Omata Y, Ohgami N, Chang J, Ichihara S, Hori M, Kato M, Non-equilibrium atmospheric pressure plasmas modulate cell cycle-related gene expressions in melanocytic tumors of RET-transgenic mice, Experimental dermatology, 23 (2014) 424–425. [DOI] [PubMed] [Google Scholar]
- [52].Chernets N, Kurpad DS, Alexeev V, Rodrigues DB, Freeman TA, Reaction Chemistry Generated by Nanosecond Pulsed Dielectric Barrier Discharge Treatment is Responsible for the Tumor Eradication in the B16 Melanoma Mouse Model, Plasma processes and polymers (Print), 12 (2015) 1400–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Liedtke KR, Bekeschus S, Kaeding A, Hackbarth C, Kuehn J-P, Heidecke C-D, von Bernstorff W, von Woedtke T, Partecke LI, Non-thermal plasma-treated solution demonstrates antitumor activity against pancreatic cancer cells in vitro and in vivo, Scientific reports, 7 (2017) 8319–8319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Utsumi F, Kajiyama H, Nakamura K, Tanaka H, Mizuno M, Ishikawa K, Kondo H, Kano H, Hori M, Kikkawa F, Effect of indirect nonequilibrium atmospheric pressure plasma on anti-proliferative activity against chronic chemo-resistant ovarian cancer cells in vitro and in vivo, PloS one, 8 (2013) e81576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Partecke LI, Evert K, Haugk J, Doering F, Normann L, Diedrich S, Weiss FU, Evert M, Huebner NO, Guenther C, Heidecke CD, Kramer A, Bussiahn R, Weltmann KD, Pati O, Bender C, von Bernstorff W, Tissue Tolerable Plasma (TTP) induces apoptosis in pancreatic cancer cells in vitro and in vivo, BMC Cancer, 12 (2012) 473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Graves DB, Oxy-nitroso shielding burst model of cold atmospheric plasma therapeutics, Clinical Plasma Medicine, 2 (2014) 38–49. [Google Scholar]
- [57].Yan D, Talbot A, Nourmohammadi N, Sherman JH, Cheng X, Keidar M, Toward understanding the selective anticancer capacity of cold atmospheric plasma--a model based on aquaporins (Review), Biointerphases, 10 (2015) 040801. [DOI] [PubMed] [Google Scholar]
- [58].Ja Kim S, Min Joh H, Chung TH, Production of intracellular reactive oxygen species and change of cell viability induced by atmospheric pressure plasma in normal and cancer cells, Applied Physics Letters, 103 (2013) 153705. [Google Scholar]
- [59].Szili EJ, Hong SH, Oh JS, Gaur N, Short RD, Tracking the Penetration of Plasma Reactive Species in Tissue Models, Trends in biotechnology, 36 (2018) 594–602. [DOI] [PubMed] [Google Scholar]
- [60].Traylor MJ, Pavlovich MJ, Karim S, Hait P, Sakiyama Y, Clark DS, Graves DB, Long-term antibacterial efficacy of air plasma-activated water, Journal of Physics D: Applied Physics, 44 (2011) 472001. [Google Scholar]
- [61].Oh J-S, Szili EJ, Gaur N, Hong S-H, Furuta H, Kurita H, Mizuno A, Hatta A, Short RD, How to assess the plasma delivery of RONS into tissue fluid and tissue, Journal of Physics D: Applied Physics, 49 (2016) 304005. [Google Scholar]
- [62].Naïtali M, Kamgang-Youbi G, Herry J-M, Bellon-Fontaine M-N, Brisset J-L, Combined Effects of Long-Living Chemical Species during Microbial Inactivation Using Atmospheric Plasma-Treated Water, Applied and Environmental Microbiology, 76 (2010) 7662–7664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Gaur N, Szili EJ, Oh J-S, Hong S-H, Michelmore A, Graves DB, Hatta A, Short RD, Combined effect of protein and oxygen on reactive oxygen and nitrogen species in the plasma treatment of tissue, Applied Physics Letters, 107 (2015) 103703. [Google Scholar]
- [64].Szili EJ, Oh J-S, Hong S-H, Hatta A, Short RD, Probing the transport of plasma-generated RONS in an agarose target as surrogate for real tissue: dependency on time, distance and material composition, Journal of Physics D: Applied Physics, 48 (2015) 202001. [Google Scholar]
- [65].Lin A, Truong B, Pappas A, Kirifides L, Oubarri A, Chen S, Lin S, Dobrynin D, Fridman G, Fridman A, Sang N, Miller V, Uniform Nanosecond Pulsed Dielectric Barrier Discharge Plasma Enhances Anti-Tumor Effects by Induction of Immunogenic Cell Death in Tumors and Stimulation of Macrophages, Plasma Processes and Polymers, 12 (2015) 1392–1399. [Google Scholar]
- [66].Miller V, Lin A, Fridman G, Dobrynin D, Fridman A, Plasma Stimulation of Migration of Macrophages, Plasma Processes and Polymers, 11 (2014) 1193–1197. [Google Scholar]
- [67].Lin AG, Xiang B, Merlino DJ, Baybutt TR, Sahu J, Fridman A, Snook AE, Miller V, Non-thermal plasma induces immunogenic cell death in vivo in murine CT26 colorectal tumors, Oncoimmunology, 7 (2018) e1484978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Lin A, Truong B, Patel S, Kaushik N, Choi EH, Fridman G, Fridman A, Miller V, Nanosecond-Pulsed DBD Plasma-Generated Reactive Oxygen Species Trigger Immunogenic Cell Death in A549 Lung Carcinoma Cells through Intracellular Oxidative Stress, International journal of molecular sciences, 18 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Bekeschus S, Mueller A, Miller V, Gaipl U, Weltmann K, Physical Plasma Elicits Immunogenic Cancer Cell Death and Mitochondrial Singlet Oxygen, IEEE Transactions on Radiation and Plasma Medical Sciences, 2 (2018) 138–146. [Google Scholar]
- [70].Bekeschus S, Rodder K, Fregin B, Otto O, Lippert M, Weltmann KD, Wende K, Schmidt A, Gandhirajan RK, Toxicity and Immunogenicity in Murine Melanoma following Exposure to Physical Plasma-Derived Oxidants, Oxidative medicine and cellular longevity, 2017 (2017) 4396467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Mizuno KYK, Shirakawa Y, Akiyama T, Ono R, Anti-tumor immune response induced by nanosecond pulsed streamer sicharge in mice, Journal of Physics D: Applied Physics, 50 (2017). [Google Scholar]
- [72].Brudno JN, Kochenderfer JN, Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management, Blood reviews, DOI 10.1016/j.blre.2018.11.002(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Huang Y, Liang D, Liu J, Zeng J, Zeng Y, The Breakthroughs in Cancer Immune Checkpoint Based Therapy: A Review of Development in Immune Checkpoint Study and its Application, Combinatorial chemistry & high throughput screening, 20 (2017) 430–439. [DOI] [PubMed] [Google Scholar]
- [74].Castano AP, Mroz P, Hamblin MR, Photodynamic therapy and anti-tumour immunity, Nature reviews. Cancer, 6 (2006) 535–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Castano AP, Demidova TN, Hamblin MR, Mechanisms in photodynamic therapy: Part three-Photosensitizer pharmacokinetics, biodistribution, tumor localization and modes of tumor destruction, Photodiagnosis and photodynamic therapy, 2 (2005) 91–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Crittenden M, Kohrt H, Levy R, Jones J, Camphausen K, Dicker A, Demaria S, Formenti S, Current clinical trials testing combinations of immunotherapy and radiation, Seminars in radiation oncology, 25 (2015) 54–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Kalbasi A, June CH, Haas N, Vapiwala N, Radiation and immunotherapy: a synergistic combination, The Journal of Clinical Investigation, 123 (2013) 2756–2763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, Breckpot K, Brough D, Buqué A, Castro MG, Cirone M, Colombo MI, Cremer I, Demaria S, Dini L, Eliopoulos AG, Faggioni A, Formenti SC, Fučíková J, Gabriele L, Gaipl US, Galon J, Garg A, Ghiringhelli F, Giese NA, Guo ZS, Hemminki A, Herrmann M, Hodge JW, Holdenrieder S, Honeychurch J, Hu H-M, Huang X, Illidge TM, Kono K, Korbelik M, Krysko DV, Loi S, Lowenstein PR, Lugli E, Ma Y, Madeo F, Manfredi AA, Martins I, Mavilio D, Menger L, Merendino N, Michaud M, Mignot G, Mossman KL, Multhoff G, Oehler R, Palombo F, Panaretakis T, Pol J, Proietti E, Ricci J-E, Riganti C, Rovere-Querini P, Rubartelli A, Sistigu A, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Sukkurwala AQ, Tartour E, Thorburn A, Thorne SH, Vandenabeele P, Velotti F, Workenhe ST, Yang H, Zong W-X, Zitvogel L, Kroemer G, Galluzzi L, Consensus guidelines for the detection of immunogenic cell death, Oncoimmunology, 3 (2014) e955691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Green DR, Ferguson T, Zitvogel L, Kroemer G, Immunogenic and tolerogenic cell death, Nature reviews. Immunology, 9 (2009) 353–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].van Vloten JP, Workenhe ST, Wootton SK, Mossman KL, Bridle BW, Critical Interactions between Immunogenic Cancer Cell Death, Oncolytic Viruses, and the Immune System Define the Rational Design of Combination Immunotherapies, Journal of immunology (Baltimore, Md. : 1950), 200 (2018) 450–458. [DOI] [PubMed] [Google Scholar]
- [81].Lin A, Gorbanev Y, De Backer J, Van Loenhout J, Van Boxem W, Lemière F, Cos P, Dewilde S, Smits E, Bogaerts A, Non-Thermal Plasma as a Unique Delivery System of Short-Lived Reactive Oxygen and Nitrogen Species for Immunogenic Cell Death in Melanoma Cells, Advanced Science, DOI 10.1002/advs.201802062(2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Manda K, Glasow A, Paape D, Hildebrandt G, Effects of ionizing radiation on the immune system with special emphasis on the interaction of dendritic and T cells, Frontiers in oncology, 2 (2012) 102–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Formenti SC, Demaria S, Systemic effects of local radiotherapy, Lancet Oncol, 10 (2009) 718–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Kohoutova D, Haidry R, Banks M, Bown S, Sehgal V, Butt MA, Graham D, Thorpe S, Novelli M, Rodriguez-Justo M, Lovat L, Esophageal neoplasia arising from subsquamous buried glands after an apparently successful photodynamic therapy or radiofrequency ablation for Barrett’s associated neoplasia, Scandinavian journal of gastroenterology, 50 (2015) 1315–1321. [DOI] [PubMed] [Google Scholar]
- [85].Lin A, Gorbanev Y, De Backer J, Van Loenhout J, Van Boxem W, Lemière F, Cos P, Dewilde S, Smits E, Bogaerts A, Non-Thermal Plasma as a Unique Delivery System of Short-Lived Reactive Oxygen and Nitrogen Species for Immunogenic Cell Death in Melanoma Cells, Advanced Science, 0 1802062. [DOI] [PMC free article] [PubMed] [Google Scholar]






