Hyperbaric Oxygen: a Multifaceted Approach in Cancer Therapy
Qingyuan Deng,Xiangliang Yang,Zifu Li
DOI: https://doi.org/10.4103/mgr.medgasres-d-23-00028
2024-01-01
Medical Gas Research
Abstract:As one of the fundamental characteristics of solid tumors, hypoxia promotes tumor initiation and progression, induces tumor sternness, and leads to an abundant extracellular matrix (ECM), immunosuppression, and abnormal oxidative stress levels, rendering tumor cells insensitive to treatments such as chemotherapy, radiotherapy, and photodynamic therapy. Therefore, overcoming hypoxia to sensitize cancer therapy has become a current research hotspot. Hyperbaric oxygen (HBO) has emerged as a promising adjuvant therapy in various medical conditions, and there are 14 approved indications for HBO including "Air or gas embolism," "Acute thermal burn injury," "Carbon monoxide poisoning," and to name a few.[1-3] Apart from the listed applications, there are further conditions in which HBO may be useful, which is cancer treatment. HBO delivers pure oxygen with increased atmospheric pressure, which helps tumors overcome hypoxia. In this perspective, we delve into the multifaceted mechanisms by which HBO could potentially revolutionize cancer treatment (Figure 1). Based on our previous works, we summarized four aspects in which HBO could significantly influence cancer therapy: (1) HBO normalizes tumor mechanics and blood vessels by dramatically reducing fibronectin and collagen I within tumor area, which benefits the penetration depth and drug accumulation in solid tumors.[4,5] (2) HBO directly suppresses cancer stem cells (CSCs)[4-7] and cancer metastasis, prominently decreasing both intestinal metastatic nodules in pancreatic cancer mice model and lung metastasis in breast cancer bearing mice.[4,5] (3) HBO could disrupt hypoxia-mediated immunosuppression, which helps programmed death-1 (PD-1) antibody trigger cytotoxic T lymphocytes and long-lasting immunological memory to inhibit tumor recurrences.[8] (4) HBO facilitates an elevation in oxidative stress within cancer cells, working in conjunction with ROS-inducing nanoparticles to promote the accumulation of oxidative damage specifically in cancerous cells.[7,9,10] This process ultimately triggers apoptosis, leading to the demise of cancer cells.Figure 1:: The mechanisms by which HBO empowers tumor treatment.Note: There are four aspects listed above by which HBO boost tumor treatment: (1) Reshape immunosuppressive microenvironment; (2) Suppress ECM and CAF; (3) Eliminate CSCs and reduce metastasis; (4) Increase oxygen concentration and benefit anti-cancer drug therapy. Created with Microsoft PowerPoint 365MSO (version 2308 Build 16.0.16731.20496). CAF: Cancer associated fibroblast; CSC: cancer stem cells; ECM: extracellular matrix.Taking into account all these aspects, HBO has the potential to collaborate effectively with commercialized nanomedicine or immune checkpoint blockade inhibitors, yielding a range of benefits such as enhanced penetration, increased accumulation in tumor tissues, reduced recurrence rates, and more. This synergy opens up new and promising possibilities in the ongoing fight against cancers. Normalizing tumor mechanics and blood vessels through cancer-associated fibroblasts (CAFs) and ECM suppression: The tumor microenvironment plays a crucial role in tumor progression and cancer therapy. CAFs and ECM surrounding tumors contribute to tumor growth, invasiveness, and the development of abnormal tumor blood vessels. These factors create a hostile tumor microenvironment that hinders drug delivery and immune cell infiltration. HBO has been shown to directly suppress CAFs and remodel the ECM, leading to the normalization of tumor mechanics and blood vessels.[4,5] Additionally, HBO-induced normalization of the ECM can improve the homogeneous distribution of therapeutic agents within the tumor, augmenting their efficacy.[4] HBO regulates the hypoxia-inducible factor 1α/connective tissue growth factor/collagen I pathway in tumors to reduce the deposition of collagen fibrils.[6] On the other hand, HBO can modulate genes associated with collagen biogenesis pathways, such as C-X-C chemokine receptor type 4 and transforming growth factor-β. It also regulates the expression of matrix metalloproteinase 7.[8] Due to the remodeling of the ECM by HBO, reduced solid stress within the tumor contributes to the normalization of tumor vascular structure and function. Using various techniques, including fluorescent staining of tissue sections, western blotting, quantitative polymerase chain reaction, and flow cytometric analysis, many studies observed significant decreases in several ECM and CAF-related markers such as fibronectin, collagen I, α-smooth muscle actin, and fibroblast activation protein-α in tumors following HBO treatment.[4-6] Additionally, Liu et al.[5] used atomic force microscopy to measure the Young's modulus of tumor tissues before and after HBO treatment, revealing that HBO treatment led to a softening of tumor tissues. Furthermore, Wang et al.[4] observed a reduction in neovascularization, decreased vascular curvature, and increased blood perfusion at tumor tissues by HBO treatment. These experimental results suggest that HBO exerts a positive effect on the mechanical properties and vascular normalization of tumor tissues. Utilizing a 3D tumor spheroid model co-culturing cancer cells with fibroblasts, Wang et al.[4] verified that HBO treatment facilitated deep penetration of Abraxane, providing an explanation for the improved antitumor efficacy when combining HBO with commercialized nanomedicines. These investigations collectively indicate that HBO ameliorates tumor hypoxia and remodels the tumor microenvironment, mechanical properties, and vascular milieu. This leads to an elevation in the delivery efficiency of nanomedicines to tumor sites, encompassing augmented tumor accumulation, enhanced deep penetration, and cellular uptake. Consequently, these effects synergistically contribute to the efficient eradication of tumor stem cells, suppression of tumor metastasis, and heightened antineoplastic efficacy of nanomedicines. Direct suppression of CSCs and cancer metastasis: CSCs are a subpopulation of cells within tumors that possess stem cell-like properties, including self-renewal and tumorigenic capabilities. Hypoxia and the resultant hypoxia-inducible factor 1α play pivotal roles in initiating and sustaining the sternness of CSCs. These CSCs are highly resistant to conventional therapies and are often responsible for cancer recurrence and metastasis. HBO emerges as a significant modality capable of effectively counteracting the hypoxic conditions prevalent in solid tumors. Through this mechanism, HBO disrupts the maintenance and self-renewal of CSCs, consequently attenuating the stem-like properties within solid tumors. Therefore, HBO has a dual impact on CSCs. First, it diminishes the self-renewal capacity of CSCs, thereby reducing their ability to regenerate. Secondly, HBO enhances the effectiveness of nanomedicines in eliminating CSCs. This synergistic effect is attributed to HBO's improvement of nanomedicine delivery efficiency, alleviation of G0-G1 phase cell cycle arrest, and increased sensitivity to nanomedicine treatments. By decreasing tumor stemness and enhancing stem cell elimination efficiency, HBO significantly suppresses tumor metastasis.[4,6] In the orthotopic pancreatic cancer mice model, various experimental approaches, including fluorescent staining and flow cytometry analysis of sternness biomarker CD133 within tumor tissues, are employed to verify that HBO-treated groups exhibited significantly lower levels of CD133 expression compared to their untreated counterparts.[4,5,7] This finding suggests that HBO can effectively reduce the sternness of pancreatic cancer cells. Moreover, in the fibrin gel tumor spheroid assay, the authors further validated the reduction of tumor spheroid number and diameter when HBO was combined with several clinical frontline drugs such as Abraxane or Doxil,[4,5] using different tumor cell lines like Panc02 and 4T1 cancer cells. These observations indicate a decrease in tumor cell sternness due to HBO treatment. The same group further investigated both the number of intestinal metastatic nodules in Panc02 tumor models and the lung metastasis in 4T1 breast cancer model, and found that the HBO-treated groups exhibited fewer metastatic lesions compared to their untreated counterparts.[4,5] Taken together, these finding underscore the significance of HBO in modulating the aberrant microenvironment of malignant solid tumors. These findings suggest that HBO has considerable promise in clinical applications for treating matrix- and CSCs-rich solid tumors. Disrupting hypoxia-mediated immunosuppression: Tumor hypoxia is known to create an immunosuppressive microenvironment that hinders the activity of immune cells. The effectiveness of immune checkpoint blockade inhibitors against solid tumors is compromised by the hypoxia induced immunosuppressive microenvironment. HBO has been shown to reverse hypoxia-mediated immunosuppression by promoting the activation and proliferation of various immune cells. Enhanced T-cell and natural Killer cell function, along with increased production of pro-inflammatory cytokines, can lead to improved anti-tumor immunity. Furthermore, HBO's ability to promote dendritic cell maturation and antigen presentation can enhance the adaptive immune response against cancer cells. Liu et al.[8] assessed changes in immune cell infiltration at tumor tissues before and after HBO treatment and found that HBO treatment significantly increased the proportion of lymphocytes, including CD4+ and CD8+ T cells, in the tumor microenvironment, as confirmed by fluorescent staining and flow cytometry of tumor tissues. Based on these results, the authors initiated a clinical trial investigating the combination of HBO with PD-1 antibodies for hepatocellular carcinoma patients.[8] They examined a broad range of immune cell populations and found that HBO, in combination with PD-1 antibodies, resulted in a decrease in myeloid-derived suppressor cells, an increase in the M1/M2 macrophage ratio, enhanced cytotoxic T lymphocyte proportion and proliferation activity, and reduced levels of cytokines like transforming growth factor-β and interleukin-10 in the serum. In the realm of clinical practice, despite emerging as promising anticancer agents, immune checkpoint inhibitors have exhibited limited efficacy against solid tumors. This phenomenon primarily arises due to the immunosuppressive microenvironment and sparse immune cell infiltration. The investigation involving the co-administration of HBO with PD-1 antibodies for the treatment of murine pancreatic cancer underscores the significant capacity of HBO to markedly enhance immune cell, particularly T-cell, infiltration. This effect is shown to reverse the tumor's immunosuppressive microenvironment, thereby offering a conduit for immunotherapeutic interventions and presenting a novel avenue for clinically addressing solid tumors. The synergistic application of immune checkpoint inhibitor agents in conjunction with HBO holds substantial potential for yielding robust anti-tumor effects in clinical contexts.[8] HBO helps nanomedicine boost oxidative stress-induced apoptosis in tumor cells: Cancer cells are sensitive to changes in oxidative stress levels, and elevated levels of oxygen in the presence of HBO lead to an increase in reactive oxygen species (ROS) production within tumor cells. ROS plays a crucial role in inducing apoptosis (programmed cell death) in cancer cells. By elevating oxidative stress, HBO, combined with ROS-inducing nanomedicines, can boost cancer cell apoptosis. This targeted approach holds great potential in developing novel and less toxic cancer therapies. Many studies developed various nanomedicines and achieved favorable results in treating different types of tumors when combined with HBO.[4-10] For instance, HBO combined with up-conversion nanoparticles photodynamic therapy demonstrated potent therapeutic efficacy in a 4T1 tumor-bearing mouse model.9 Additionally, HBO combined with hydroxyethyl starch-coupled doxorubicin-loaded glucose oxidase nanoparticles depleted glucose within tumor cells, increased oxidative stress, and decreased cell stemness, ultimately inducing tumor cell death through the synergistic actions of chemotherapy, chemodynamic therapy, and starvation therapy.[10] These studies revealed that HBO not only enhanced oxidative stress but also increased drug penetration and accumulation while reducing tumor cell stemness, presenting a multifaceted therapeutic approach. In a holistic perspective, HBO exhibits a dual functionality within these investigations. Firstly, HBO elevates oxygen tension within solid tumors, ameliorating hypoxic conditions and thereby providing substrates for the generation of ROS. Secondly, HBO treatment remodels the tumor microenvironment, facilitating enhanced penetration and targeted accumulation of nanoparticles. Through these two aspects, HBO significantly augments the cytotoxic impact of nanoparticles utilizing principles such as photodynamic therapy or chemodynamic therapy, wherein ROS are employed for tumor cell eradication. This synergistic enhancement yields robust antineoplastic effects, thereby contributing to a substantial anti-tumor outcome. HBO modulation of tumor oxidative stress levels can guide the design of a new type of ROS-mediated tumor therapy, enabling the development of novel oxygen-dependent nanodrugs. For instance, it is possible to investigate whether HBO can enhance ROS-dependent ferroptosis. HBO therapy represents a promising approach to complement existing cancer treatments. By targeting multiple aspects of tumor progression, HBO has the potential to suppress CSCs and cancer metastasis, normalize tumor mechanics and blood vessels, disrupt hypoxia-mediated immunosuppression, and sensitize cancer cell through increased oxidative stress. The multifaceted mechanisms of HBO highlight its versatility and capacity to contribute to cancer treatments. As ongoing research continues to elucidate the full extent of HBO's benefits and optimize its integration into cancer therapy, the molecular mechanisms of HBO's effect still have significant gaps. For instance, the mechanisms underlying HBO's reduction of CAFs and ECM, and the specific subtype of CAFs affected, remain to be elucidated. The molecular basis by which HBO inhibits CSCs and the mechanism of selective CSC inhibition while sparing normal cells are still unclear. One of the current research directions is to explore the molecular mechanisms through which HBO inhibits cancer metastasis. Similarly, the precise identity of immune cells and molecules responsible for HBO- induced immune cell infiltration remains elusive. Regarding the changes in oxidative stress, a more comprehensive investigation into the mechanisms is needed, spanning from systemic oxidative stress levels in the whole organism to the impact on the mechanism of mitochondrial oxidative respiration within organelles. Nevertheless, the collective findings summarized in this study underscore the multifaceted efficacy of HBO in the realm of cancer therapy, offering promise in addressing several clinical challenges associated with solid tumors, including suboptimal chemotherapy responses, ineffective immune therapy, tumor metastasis, and relapse. Several ongoing clinical trials are currently recruiting patients, with anticipation for further preclinical investigative outcomes. In summary, HBO's multifaceted mechanisms address critical issues in the treatments of solid tumors, holding significant potential for clinical translation, thereby providing novel insights and opportunities in the landscape of cancer therapy. Finally, it is our hope that this perspective can offer a new direction for gas therapy of cancer and provide a novel approach for clinical management of cancers.
What problem does this paper attempt to address?
-
Hyperbaric oxygen regulates tumor microenvironment and boosts commercialized nanomedicine delivery for potent eradication of cancer stem-like cells
Xin Liu,Ningbing Ye,Chen Xiao,Xiaoxian Wang,Shiyou Li,Yihan Deng,Xiaoquan Yang,Zifu Li,Xiangliang Yang
DOI: https://doi.org/10.1016/j.nantod.2021.101248
IF: 17.4
2021-10-01
Nano Today
Abstract:<p>Cancer stem-like cells (CSCs) depend highly on hypoxia in solid tumors and represent an intractable challenge for marketed nanomedicines. Herein, we for the first time leveraged hyperbaric oxygen (HBO) therapy to help commercialized nanomedicines, including Doxil and Abraxane, eliminate CSCs in stroma-rich solid tumors, <em>e.g.</em>, triple negative breast cancer (TNBC) and pancreatic ductal adenocarcinoma (PDAC), for efficient cancer therapy. Mechanistically, we revealed that HBO disrupted hypoxia in solid tumors, thereby directly suppressing CSCs and cancer metastasis. More importantly, we found that HBO depleted excessive extracellular matrix, such as collagen and fibronectin, and thus normalized tumor blood vessels both structurally and functionally. As a consequence, HBO augmented the delivery of commercialized nanomedicines, but not small molecule drug, into solid tumors, in terms of tumor accumulation, deep penetration and cellular internalization, leading to efficient CSCs eradication and tumor inhibition. These results demonstrate that HBO enables commercialized nanomedicines to eliminate CSCs in stroma-rich solid tumors and suggest that the combination of HBO with commercialized nanomedicines is promising for the treatment of hypoxic solid tumors in the clinics.</p>
materials science, multidisciplinary,nanoscience & nanotechnology,chemistry
-
Hyperbaric Oxygen and Cancer: More Complex Than We Expected
Wenwu Liu,Xuejun Sun,Hengyi Tao,Kan Liu
DOI: https://doi.org/10.1007/s11523-013-0259-8
2013-01-01
Targeted Oncology
Abstract:Hyperbaric oxygen (HBO) therapy refers to the breathing ofpure oxygen while in a sealed chamber that has been pres-surized at 1.5 to 3 times normal atmospheric pressure. Todate, HBO therapy has been applied in a series of diseaseswith potential hypoxia including decompression sickness,carbonmonoxidepoisoning,gasgangrene,osteomyelitis,andso on (http://en.wikipedia.org/wiki/Hyperbaric_medicine).However, cancer is not an indication for hyperbaric oxygenin both USA and China.Recently, Moen and Stuhr reviewed the evidence on theinfluence of HBO on cancers in past 9 years [1]. Their con-clusionwasconsistentwithtwoprevioussystemicreviewsonHBO and cancer that the use of HBO in patients with malig-nanciesisconsideredsafe.Inthisreview,theysummarizedtherelationship between hypoxia and cancer and the influence ofHBO on cell survival, angiogenesis, metastasis, chemothera-py,andradiotherapy.Finally,theyreviewedtheeffectofHBOon cancer cells of different types. Although they proposedsome explanations on the effects of HBO on cancers, themechanisms are more complex than we expected.HBO, hypoxia, and angiogenesisIt has been accepted that HBO can increase the dissolvedoxygen significantly, which may rectify the hypoxia incancers. However, HBO is often used once or twice dailyfor patients, and a hypoxic environment may form betweentwo exposures. As shown in this review, hypoxia is essentialfor the progression of cancers and related to the angiogen-esis. In addition, some studies have also shown that HBOtherapy can improve the angiogenesis after ischemic injury.Our previous study also revealed that HBO pretreatment canincrease the expression of hypoxia-inducible factor and itsdownstream factor vascular endothelial growth factor [2]which is a potent pro-angiogenic factor. Thus, HBO seemsto exert promotive effect on cancer growth.HBO and metastasisMetastasis is a complex process involving local cancer cellinvasion, entry into the blood or lymph vessels, and re-penetration and colonization at a distant site [3]. Theintegrity of vascular basement membrane (VBM) iscrucial for the cancer cell invasion. Suzuki et al. foundthat HBO enhanced transendothelial permeability in ratbrains [4]. In addition, HBO preconditioning was foundto increase the expression of matrix metalloproteinase(MMP) [5] which is may break the VBM. This maypotentiate the cancer cell invasion. Of interest, HBOtherapy may reduce the insult-induced expression ofMMP [6]. Thus, whether the effect of HBO on VBMintegrity depends on the microenvironment is not clear.If so, this may partially explain the absence of influenceof HBO on the metastasis of cancers.HBO and cancer stem cellsCancer stem cells (CSCs) are cancer cells that possesscharacteristics associated with normal stem cells, specifically
-
Hyperbaric Oxygen Regulates Tumor Mechanics and Augments Abraxane and Gemcitabine Antitumor Effects Against Pancreatic Ductal Adenocarcinoma by Inhibiting Cancer-Associated Fibroblasts
Xiaoxian Wang,Ningbing Ye,Chen Xu,Chen Xiao,Zhijie Zhang,Qingyuan Deng,Shiyou Li,Jiayuan Li,Zifu Li,Xiangliang Yang
DOI: https://doi.org/10.1016/j.nantod.2022.101458
IF: 17.4
2022-01-01
Nano Today
Abstract:While the combination of Abraxane and gemcitabine (GEM) has been approved as a first-line therapy for the treatment of advanced pancreatic ductal carcinoma (PDAC), the clinical antitumor efficacy is dismal thus far. Here, we explore hyperbaric oxygen (HBO) therapy to boost antitumor efficacy of Abraxane and GEM against murine PDAC Panc02 tumors. Mechanistically, we reveal that HBO significantly constrains cancer-associated fibroblasts (CAFs) by disrupting hypoxia. As CAFs are the main producers of extracellular matrix (ECM) components, HBO effectively reduces collagen Ⅰ and fibronectin, leading to normalization of not only tumor mechanics but also blood vessels. Therefore, HBO augments drug delivery in terms of tumor accumulation, penetration, and cellular internalization. Furthermore, we demonstrate that HBO helps Abraxane and GEM eliminate cancer stem cells (CSCs) by regulating CAFs mediated CSCs niche. Consequently, HBO potentiates antitumor efficacy of Abraxane and GEM by inhibiting not only primary tumors but more importantly metastases of Panc02 tumors, with negligible side effects. Given HBO, Abraxane, and GEM are routinely applied in clinical practice, their combination is readily translatable to bedside applications.
-
Hyperbaric Oxygen Activates Enzyme‐Driven Cascade Reactions for Cooperative Cancer Therapy and Cancer Stem Cells Elimination
Yuxuan Xiong,Zhengtao Yong,Chen Xu,Qingyuan Deng,Qiang Wang,Shiyou Li,Chong Wang,Zhijie Zhang,Xiangliang Yang,Zifu Li
DOI: https://doi.org/10.1002/advs.202301278
IF: 15.1
2023-04-30
Advanced Science
Abstract:A versatile cascade bioreactor is developed to achieve an outstanding synergistic starvation‐/chemodynamic‐/chemo‐therapy with the help of hyperbaric oxygen (HBO). The amplified oxidative stress and HBO‐mediated tumor extracellular matrix degradation enable effective elimination of cancer stem cells, suppressing both orthotopic tumor and pulmonary metastasis. Tumor starvation induced by intratumor glucose depletion emerges as a promising strategy for anticancer therapy. However, its antitumor potencies are severely compromised by intrinsic tumor hypoxia, low delivery efficiencies, and undesired off‐target toxicity. Herein, a multifunctional cascade bioreactor (HCG), based on the self‐assembly of pH‐responsive hydroxyethyl starch prodrugs, copper ions, and glucose oxidase (GOD), is engineered, empowered by hyperbaric oxygen (HBO) for efficient cooperative therapy against aggressive breast cancers. Once internalized by tumor cells, HCG undergoes disassembly and releases cargoes in response to acidic tumor microenvironment. Subsequently, HBO activates GOD‐catalyzed oxidation of glucose to H2O2 and gluconic acid by ameliorating tumor hypoxia, fueling copper‐catalyzed •OH generation and pH‐responsive drug release. Meanwhile, HBO degrades dense tumor extracellular matrix, promoting tumor accumulation and penetration of HCG. Moreover, along with the consumption of glucose and the redox reaction of copper ions, the antioxidant capacity of tumor cells is markedly reduced, collectively boosting oxidative stress. As a result, the combination of HCG and HBO can not only remarkably suppress the growth of orthotopic breast tumors but also restrain pulmonary metastases by inhibiting cancer stem cells. Considering the clinical accessibility of HBO, this combined strategy holds significant translational potentials for GOD‐based therapies.
materials science, multidisciplinary,nanoscience & nanotechnology,chemistry
-
Boosting Nanomedicine Efficacy With Hyperbaric Oxygen Therapy
Xiaoxian Wang,Si Li,Xin Liu,Xian Wu,Ningbing Ye,Xiangliang Yang,Zifu Li
DOI: https://doi.org/10.1007/978-3-030-58174-9_4
2021-01-01
Abstract:Nanomedicine has been a hot topic in the field of tumor therapy in the past few decades. Because of the enhanced permeability and retention effect (EPR effect), nanomedicine can passively yet selectively accumulate at tumor tissues. As a result, it can improve drug concentration in tumor tissues and reduce drug distribution in normal tissues, thereby contributing to enhanced antitumor effect and reduced adverse effects. However, the therapeutic efficacy of anticancer nanomedicine is not satisfactory in clinical settings. Therefore, how to improve the clinical therapeutic effect of nanomedicine has become an urgent problem. The grand challenges of nanomedicine lie in how to overcome various pathophysiological barriers and simultaneously kill cancer cells effectively in hypoxic tumor microenvironment (TME). To this end, the development of novel stimuli-responsive nanomedicine has become a new research hotspot. While a great deal of progress has been made in this direction and preclinical results report many different kinds of promising multifunctional smart nanomedicine, the design of these intelligent nanomedicines is often too complicated, the requirements for the preparation processes are strict, the cost is high, and the clinical translation is difficult. Thus, it is more practical to find solutions to promote the therapeutic efficacy of commercialized nano-medicines, for example, Doxil (R), Oncaspar (R), DaunoXome (R), Abraxane (R), to name a few. Increasing attention has been paid to the combination of modern advanced medical technology and nanomedicine for the treatment of various malignancies. Recently, we found that hyperbaric oxygen (HBO) therapy could enhance Doxil (R) antitumor efficacy. Inspired by this study, we further carried out researches on the combination of HBO therapy with other nanomedicines for various cancer therapies, and revealed that HBO therapy could significantly boost antitumor efficacy of nanomedicine--mediated photodynamic therapy and photothermal therapy in different kinds of tumors, including hepatocellular carcinoma, breast cancer, and gliomas. Our results implicate that HBO therapy might be a universal strategy to boost therapeutic efficacy of nanomedicine against hypoxic solid malignancies.
-
Hyperbaric Oxygen Therapy Represses the Warburg Effect and Epithelial–Mesenchymal Transition in Hypoxic NSCLC Cells via the HIF-1α/PFKP Axis
Linling Zhang,Jingjing Ke,Shengping Min,Nan Wu,Fei Liu,Zhen Qu,Wei Li,Hongtao Wang,Zhongqing Qian,Xiaojing Wang
DOI: https://doi.org/10.3389/fonc.2021.691762
IF: 4.7
2021-07-21
Frontiers in Oncology
Abstract:Background Tumor cells initiate hypoxia-induced mechanisms to fuel cell proliferation, invasion, and metastasis, largely mediated by low O 2 -responsive Hypoxia-Inducible Factor 1 Alpha (HIF-1α). Therefore, hyperbaric oxygen therapy (HBO) is now being studied in cancer patients, but its impact upon non-small-cell lung cancer (NSCLC) cell metabolism remains uncharacterized. Methods We employed the NSCLC cell lines A549 and H1299 for in vitro studies. Glucose uptake, pyruvate, lactate, and adenosine triphosphate (ATP) assays were used to assess aerobic glycolysis (Warburg effect). A quantitative glycolytic flux model was used to analyze the flux contributions of HIF-1α-induced glucose metabolism genes. We used a Lewis lung carcinoma (LLC) murine model to measure lung tumorigenesis in C57BL/6J mice. Results HBO suppressed hypoxia-induced HIF-1α expression and downstream HIF-1α signaling in NSCLC cells. One HIF-1α-induced glucose metabolism gene—Phosphofructokinase, Platelet (PFKP)—most profoundly enhanced glycolytic flux under both low- and high-glucose conditions. HBO suppressed hypoxia-induced PFKP transactivation and gene expression via HIF-1α downregulation. HBO’s suppression of the Warburg effect, suppression of hyperproliferation, and suppression of epithelial-to-mesenchymal transition (EMT) in hypoxic NSCLC cell lines is mediated by the HIF-1α/PFKP axis. In vivo , HBO therapy inhibited murine LLC lung tumor growth in a Pfkp-dependent manner. Conclusions HBO’s repression of the Warburg effect, repression of hyperproliferation, and repression of EMT in hypoxic NSCLC cells is dependent upon HIF-1α downregulation. HIF-1α’s target gene PFKP functions as a central mediator of HBO’s effects in hypoxic NSCLC cells and may represent a metabolic vulnerability in NSCLC tumors.
oncology
-
Extremely Effective Chemoradiotherapy by Inducing Immunogenic Cell Death and Radio-Triggered Drug Release under Hypoxia Alleviation
Chunqi Zhu,Xiaomen Guo,Lihua Luo,Zhe Wu,Zhenyu Luo,Mengshi Jiang,Junlei Zhang,Bing Qin,Yingying Shi,Yan Lou,Yunqing Qiu,Jian You
DOI: https://doi.org/10.1021/acsami.9b16837
IF: 9.5
2019-01-01
ACS Applied Materials & Interfaces
Abstract:Local hypoxia in solid malignancies often results in resistance to radiotherapy (RT) and chemotherapy (CT), which may be one of the main reasons for their failure in clinical application. Especially, oxygen is an essential element for enhancing DNA damage caused by ionizing radiation in radiotherapy. Here, two biomimetic oxygen delivery systems were designed by encapsulating hemoglobin (Hb) alone into a liposome (Hb-Lipo) or co-encapsulating Hb and doxorubicin (DOX) into a liposome (DOX-Hb-Lipo). Our data indicated that both Hb-Lipo and DOX-Hb-Lipo could effectively alleviate hypoxia in tumors. We demonstrated that RT plus tumor-targeting delivery of oxygen mediated by Hb-Lipo could significantly overcome the tolerance of hypoxic cancer cells to RT, showing significantly enhanced cancer-cell killing and tumor growth inhibition ability, mainly attributing to hypoxia alleviation and increased reactive oxygen species production under RT in cancer cells. Furthermore, a melanoma model that was quite insensitive to both RT and CT was used to test the efficacy of chemoradiotherapy combined with hypoxia alleviation. RT plus Hb-Lipo only caused a limited increase in antitumor activity. However, extremely strong tumor inhibition could be obtained by RT combined with DOX-Hb-Lipo-mediated CT, attributed to radio-triggered DOX release and enhanced immunogenic cell death induced by RT under an oxygen supplement. Our study provided a valuable reference for overcoming hypoxia-induced radioresistance and a useful therapeutic strategy for cancers that are extremely insensitive to chemo- or radiotherapy.
-
Hyperbaric Oxygen Boosts PD‐1 Antibody Delivery and T Cell Infiltration for Augmented Immune Responses Against Solid Tumors
Xin Liu,Ningbing Ye,Sha Liu,Jiankun Guan,Qingyuan Deng,Zhijie Zhang,Chen Xiao,Ze‐yang Ding,Bi‐xiang Zhang,Xiao‐ping Chen,Zifu Li,Xiangliang Yang
DOI: https://doi.org/10.1002/advs.202100233
IF: 15.1
2021-06-03
Advanced Science
Abstract:<p>Aberrant mechanical properties and immunosuppression are the two key factors that limit the antitumor efficacy of T cell immune checkpoint blockade inhibitors, e.g., programmed cell death-1 antibody (PD-1 Ab), against solid tumors in the clinic. This study leverages hyperbaric oxygen (HBO) for the first time to address these two issues and reports the PD-1-Ab-mediated immune responses against various stroma-rich solid malignancies. The results demonstrate that HBO promoted PD-1 Ab delivery and T cells infiltration into tumor parenchyma by depleting the extracellular matrix's main components, such as collagen and fibronectin. Furthermore, HBO disrupts hypoxia-mediated immunosuppression and helps PD-1 Ab trigger robust cytotoxic T lymphocytes and long-lasting immunological memory to inhibit tumor relapses. Such enhanced immune responses are effective in solid tumors from rodents and the cancer cells from hepatocellular carcinoma patients. The results illustrate that HBO bolsters antitumor efficacy of PD-1 Ab, and the HBO–PD-1 Ab combination is a promising stroma-rich solid tumors' treatment in the clinic.</p>
materials science, multidisciplinary,nanoscience & nanotechnology,chemistry
-
Evaluation of use of hyperbaric oxygen in suppression of hyper-proliferation in hypoxic NSCLC cells
Jiun-Rung Chen,Haseeb Khan,Yigiter Umarzai,Abbaas Rajput,Tokalp Mamund
DOI: https://doi.org/10.14715/cmb/2023.69.14.21
2023-12-20
Abstract:Hyperbaric oxygen therapy (HBO) is being researched as a potential adjuvant treatment for solid malignancies, such as NSCLC. It can reduce tumour hypoxia and has been found to slow tumour growth, stop dedifferentiation, and reduce apoptosis resistance in hypoxic NSCLC cells. Though HBO has shown promise in treating various cancers, more study is required to determine its precise mechanism of action in NSCLC. Analyze the effect of hyperbaric oxygen on the growth of hypoxic non-small cell lung cancer cells. We used the NSCLC cell lines A549 and H1299 to analyze aerobic glycolysis in vitro. Warburg effect testing included glucose absorption, lactate, adenosine triphosphate (ATP), and pyruvate measurements. Using a quantitative glycolytic flow model, we also analyzed the effect of HIF-1-induced genes on the flux of glucose metabolism. Lewis lung carcinoma (LLC) animal models in C57BL/6J mice were used to examine the development of lung tumours. The effects of pcDNA and HIF1A on glucose uptake, lactate production, pyruvate, and ATP levels were studied in A549 and H1299 NSCLC cells. While A549's glucose absorption increased over time, H1299's was dramatically decreased by HBO treatment. The pyruvate levels were more significant in H1299, particularly in hypoxia, and were lowered by HBO. In A549, the lactate content was more effective. After HBO treatment, glucose absorption was reduced while intracellular ATP levels were maintained. Overexpression of HIF-1a was able to counteract the effect of HBO on glycolytic gene expression. PFKP is a possible therapeutic target because HBO reduces the Warburg effect in NSCLC cells by downregulating HIF-1.
-
Hyperbaric Oxygen Potentiates Doxil Antitumor Efficacy by Promoting Tumor Penetration and Sensitizing Cancer Cells
Xian Wu,Yanhong Zhu,Wei Huang,Jingqiu Li,Bixiang Zhang,Zifu Li,Xiangliang Yang
DOI: https://doi.org/10.1002/advs.201700859
IF: 15.1
2018-01-01
Advanced Science
Abstract:Hypoxia is a fundamental hallmark of solid tumors and helps contribute to chemotherapy resistance. Hyperbaric oxygen (HBO) therapy can overcome tumor hypoxia and promote chemotherapy antitumor efficacy; however, the simultaneous administration of some conventional chemotherapies, including doxorubicin (DOX), with HBO is considered an absolute contraindication. Here, DOX-loaded liposome (Doxil) is coadministered with HBO to assess the safety and efficacy of this combination treatment. By overcoming tumor hypoxia, HBO not only improves Doxil tumor penetration by decreasing the collagen deposition but also sensitizes tumor cells to Doxil. As a result, the combination treatment synergistically inhibits H22 tumor growth, with a tumor inhibition rate of 91.5%. The combination of HBO with Doxil shows neither extra side effects nor promotion of tumor metastasis. These results collectively reveal that the combination of HBO with Doxil is an effective and safe treatment modality. As both HBO and Doxil are routinely used, their combination could quickly translate to clinical trials for patients with hypoxic solid tumors.
-
Hemoglobin-based Oxygen Carriers Combined with Anticancer Drugs May Enhance Sensitivity of Radiotherapy and Chemotherapy to Solid Tumors.
Wei Wu,Qian Yang,Tao Li,Pu Zhang,Ronghua Zhou,Chengmin Yang
DOI: https://doi.org/10.1080/10731190903043218
2009-01-01
Artificial Cells Blood Substitutes and Biotechnology
Abstract:The aberrant vascular architecture in solid tumors is the key limiting factor known to ameliorate hypoxia and increase circulating anticancer drugs, thus resulting in resistance to radiotherapy and chemotherapy in tumor treatment. Previous experiments have reported hemoglobin-based oxygen carriers (HBOCs) that are effective to improve tumor oxygenation, thereby serving as potential agents target-oriented to the hypoxic tumor. Herein, we draw the hypothesis that HBOCs combined with an anticancer drug may increase oxygen bioavailability and anticancer drug retention in solid tumors and in turn contribute to enhanced sensitivity of radiotherapy and chemotherapy. This novel drug will bring a new breakthrough in the field of the development of anticancer drugs and reveal the alternative clinical use of HBOCs in tumor treatment.
-
Hyperbaric Oxygen Boosts Antitumor Efficacy of Copper-Diethyldithiocarbamate Nanoparticles Against Pancreatic Ductal Adenocarcinoma by Regulating Cancer Stem Cell Metabolism
Chen Xiao,Jiayuan Li,Ao Hua,Xing Wang,Shiyou Li,Zheng Li,Chen Xu,Zhijie Zhang,Xiangliang Yang,Zifu Li
DOI: https://doi.org/10.34133/research.0335
IF: 11
2024-01-01
Research
Abstract:Cuproptosis-based cancer nanomedicine has received widespread attention recently. However, cuproptosis nanomedicine against pancreatic ductal adenocarcinoma (PDAC) is severely limited by cancer stem cells (CSCs), which reside in the hypoxic stroma and adopt glycolysis metabolism accordingly to resist cuproptosis-induced mitochondria damage. Here, we leverage hyperbaric oxygen (HBO) to regulate CSC metabolism by overcoming tumor hypoxia and to augment CSC elimination efficacy of polydopamine and hydroxyethyl starch stabilized copper-diethyldithiocarbamate nanoparticles (CuET@PH NPs). Mechanistically, while HBO and CuET@PH NPs inhibit glycolysis and oxidative phosphorylation, respectively, the combination of HBO and CuET@PH NPs potently suppresses energy metabolism of CSCs, thereby achieving robust tumor inhibition of PDAC and elongating mice survival importantly. This study reveals novel insights into the effects of cuproptosis nanomedicine on PDAC CSC metabolism and suggests that the combination of HBO with cuproptosis nanomedicine holds significant clinical translation potential for PDAC patients.
-
Hyperbaric Oxygen‐Facilitated Cancer Treatment: A Minireview
Zi-Heng Li,Xinping Zhang,Fu-Gen Wu
DOI: https://doi.org/10.1002/anbr.202300162
2024-01-01
Advanced NanoBiomed Research
Abstract:Hypoxia in malignant tumors is a major factor in inducing the failure of clinical cancer treatment. Although several strategies have been developed to relieve hypoxia, most are still in the preclinical research phase. Therefore, hyperbaric oxygen (HBO), an approved adjuvant therapy for alleviating hypoxia clinically, is an excellent choice for enhancing the efficacy of cancer treatment that is impeded by tumor hypoxia. In this minireview, recent advances in HBO‐facilitated cancer treatment, including clinical applications and nanomedicine‐mediated cancer therapy are introduced. At the end of this minireview, the potential challenges faced by HBO therapy before clinical use are discussed. It is hoped that this review will provide a reference for future clinical research on the application of HBO in cancer treatment.
-
Synergistic Inhibitory Effect of Hyperbaric Oxygen Combined with Sorafenib on Hepatoma Cells.
Hai-Shan Peng,Ming-Bin Liao,Mei-Yin Zhang,Yin Xie,Li Xu,Yao-Jun Zhang,X F Steven Zheng,Hui-Yun Wang,Yi-Fei Chen
DOI: https://doi.org/10.1371/journal.pone.0100814
IF: 3.7
2014-01-01
PLoS ONE
Abstract:OBJECTIVES:Hypoxia is a common phenomenon in solid tumors, associated with chemotherapy and radiotherapy resistance, recurrence and metastasis. Hyperbaric oxygen (HBO) therapy can increase tissue oxygen pressure and content to prevent the resistance, recurrence and metastasis of cancer. Presently, Sorafenib is a first-line drug, targeted for hepatocellular carcinoma (HCC) but effective in only a small portion of patients and can induce hypoxia. The purpose of this study is to investigate the effect of HBO in combination with sorafenib on hepatoma cells.METHODS:Hepatoma cell lines (BEL-7402 and SK-Hep1) were treated with HBO at 2 atmosphere absolute pressure for 80 min per day or combined with sorafenib or cisplatin. At different time points, cells were tested for cell growth, colony formation, apoptosis, cell cycle and migration. Finally, miRNA from the hepatoma cells was detected by microRNA array and validated by qRT-PCR.RESULTS:Although HBO, sorafenib or cisplatin alone could inhibit growth of hepatoma cells, HBO combined with sorafenib or cisplatin resulted in much greater synergistic growth inhibition (cell proliferation and colony formation) in hepatoma cells. Similarly, the synergistic effect of HBO and sorafenib on induction of apoptosis was also observed in hepatoma cells. HBO induced G1 arrest in SK-Hep1 not in BEL-7402 cells, but enhanced cell cycle arrest induced by sorafenib in BEL-7402 treated cells. However, HBO had no obvious effect on the migration of hepatoma cells, and microRNA array analysis showed that hepatoma cells with HBO treatment had significantly different microRNA expression profiles from those with blank control.CONCLUSIONS:We show for the first time that HBO combined with sorafenib results in synergistic growth inhibition and apoptosis in hepatoma cells, suggesting a potential application of HBO combined with sorafenib in HCC patients. Additionally, we also show that HBO significantly altered microRNA expression in hepatoma cells.
-
Effects of Hyperbaric Oxygen Combined with Chemotherapy on the Proliferation of Colorectal Carcinoma in Vivo
XIA Yang,WANG Ai-guo,ZHANG Wei,HUANG Ping,HUANG Pei-lin,ZHANG Li-da
DOI: https://doi.org/10.3969/j.issn.1673-6273.2006.12.009
2006-01-01
Abstract:Objective: To study the effects of hyperbaric oxygen (HBO) or combined with chemotherapy (5-FU or paclitaxcl)on the proliferation of colon adenocarcinoma cell CT26 transplanted in mice. Methods: The cell cycle of CT26 under hyperbaric oxygen was examined with flow cytometry; BALB/c mouse tumor models were establishedand and the effects of 0.2 Mpa HBO or combined with chemotherapy (5-FU or paclitaxel) on the growth of tumors were observed. The tumor inhibitory ratio was calculated. Results: The volume inhibitory ratios or weight inhibitory ratios in different groups were as follows: HBO group(22.39%,25.77%);5-FU group(42.38%,43.61%);5-FU+HBO group(72.10%,71.47%); Paclitaxel group(26.31% ,23.04%); Paclitaxel+HBO group (33.24,30.96%). The tumor inhibitory ratio in 5-FU+HBO group was higher than that in 5-FU group or HBO group,but there was no difference between Paclitaxel+HBO group and Paclitaxel group or HBO group. Conclusion: These results show that HBO can improve the effect of cell cycle spedfic antitumor drugs, HBO itself may inhibit the growth of transplanted mouse tumor. HBO may induce cancer cells into cell cyde and accumulate in S phase in vitro; and inhibit the growth of epidermic tumors in vivo.
-
Hyperbaric oxygenation for tumour sensitisation to radiotherapy
Michael H Bennett,John Feldmeier,Robert Smee,Christopher Milross
DOI: https://doi.org/10.1002/14651858.CD005007.pub3
2012-04-18
Abstract:Background: Cancer is a common disease and radiotherapy is one well-established treatment for some solid tumours. Hyperbaric oxygenation therapy (HBOT) may improve the ability of radiotherapy to kill hypoxic cancer cells, so the administration of radiotherapy while breathing hyperbaric oxygen may result in a reduction in mortality and recurrence. Objectives: To assess the benefits and harms of radiotherapy while breathing HBO. Search methods: In March 2011 we searched The Cochrane Central Register of Controlled Trials (CENTRAL), (The Cochrane Library, Issue 3), MEDLINE, EMBASE, DORCTHIM and reference lists of articles. Selection criteria: Randomised and quasi-randomised studies comparing the outcome of malignant tumours following radiation therapy while breathing HBO versus air. Data collection and analysis: Three review authors independently evaluated the quality of the relevant trials and extracted the data from the included trials. Main results: Nineteen trials contributed to this review (2286 patients: 1103 allocated to HBOT and 1153 to control). With HBOT, there was a reduction in mortality for head and neck cancers at both one year and five years after therapy (risk ratio (RR) 0.83, P = 0.03, number needed to treat (NNT) = 11; and RR 0.82, P = 0.03, NNT = 5 respectively), as well as improved local tumour control at three months (RR with HBOT 0.58, P = 0.006, NNT = 7). The effect of HBOT varied with different fractionation schemes. Local tumour recurrence was less likely with HBOT at one year (head and neck: RR 0.66, P < 0.0001, NNT = 5), two years (uterine cervix: RR 0.60, P = 0.04, NNT = 5) and five years (head and neck: (RR 0.77, P = 0.01, NNT = 6). Any advantage is achieved at the cost of some adverse effects. There was a significant increase in the rate of both severe radiation tissue injury (RR 2.35, P < 0.0001, (number needed to harm (NNH) = 8) and the chance of seizures during therapy (RR 6.76, P = 0.03, NNH = 22) with HBOT. Authors' conclusions: There is some evidence that HBOT improves local tumour control and mortality for cancers of the head and neck, and local tumour recurrence in cancers of the head and neck, and uterine cervix. These benefits may only occur with unusual fractionation schemes. HBOT is associated with significant adverse effects including oxygen toxic seizures and severe tissue radiation injury. The methodological and reporting inadequacies of the studies included demand a cautious interpretation. More research is needed for head and neck cancer, but is probably not justified for bladder cancer. There is little evidence available concerning malignancies at other anatomical sites on which to base a recommendation.
-
Hyperbaric oxygen therapy increases the effect of 5-fluorouracil chemotherapy on experimental colorectal cancer in mice
Vanessa Foresto Machado,José Joaquim Ribeiro da Rocha,Rogério Serafim Parra,Marley Ribeiro Feitosa,Caio Abner Leite,Stefânia Bovo Minto,Sérgio Britto Garcia,Thiago Mattar Cunha,Omar Feres
DOI: https://doi.org/10.4103/2045-9912.385944
2024-09-01
Abstract:Tumor hypoxia may compromise the results of chemotherapy for treating colorectal cancer because it stimulates angiogenesis and the release of tumor growth factors. Hyperbaric oxygen (HBO) supplementation may potentiate the effects of chemotherapy in such cases. This study aimed to assess the effect of HBO therapy combined with chemotherapy on the treatment of colorectal cancer in mice. C57BL6 mice were submitted to the intrarectal instillation of N-methyl-N-nitrosoguanidine (MNNG) and treated with 5-fluorouracil (5FU) and/or HBO therapy. The MNNG group presented the highest dysplastic crypt rate. The 5FU + HBO group presented the highest rate of apoptotic cells per dysplastic crypt. The 5FU group presented the highest expression of hypoxia-inducible factor-1 alpha and CD44. HBO therapy increased the effect of 5FU on the treatment of the experimental colorectal neoplasia in mice.
-
Effects of Hyperbaric Oxygen Therapy on Tumor Growth in Murine Model of PC-3 Prostate Cancer Cell Line.
Hao Tang,Yinghao Sun,Chuanliang Xu,Tie Zhou,Xu Gao,Linhui Wang
DOI: https://doi.org/10.1016/j.urology.2008.04.057
IF: 2.633
2009-01-01
Urology
Abstract:OBJECTIVES To test the hypothesis that hyperbaric oxygen (HBO) has no effect on tumor growth in a murine model of indolent in vivo prostate cancer. HBO means breathing pure (100%) oxygen under increased atmospheric pressure. METHODS Human prostate PC-3 cells were injected into 40 severe combined-immunodeficient mice. They were randomized to undergo 20 sessions of either HBO or normobaric air in standardized conditions and observed for 4 weeks before histologic assessment of any palpable tumors that had developed. The analysis of the developed PC-3 tumors included tumor volume, microvessel density, apoptosis-associated markers (ie, p53, p27), and the proliferative index (Ki-67). RESULTS The exposure to HBO at 2 arm for 20 treatment sessions, which comprised a daily 90-minute session, 5 d/wk, had no effect on the prostate cancer volume (P > .05). No differences were observed in tumor microvessel density, proliferative index, or apoptosis markets compared with the non-HBO group (P > .05). CONCLUSIONS HBO did not have a tumor stimulatory effect on prostate cancer and could potentially be used safely in conjunction with other therapeutic modalities. UROLOGY 73: 205-208, 2009. (C) 2009 Elsevier Inc.
-
High Intensity Focused Ultrasound-Responsive and Ultrastable Cerasomal Perfluorocarbon Nanodroplets for Alleviating Tumor Multidrug Resistance and Epithelial–Mesenchymal Transition
Xiaotu Ma,Meinan Yao,Jiyun Shi,Xiaoda Li,Yu Gao,Qi Luo,Rui Hou,Xiaolong Liang,Fan Wang
DOI: https://doi.org/10.1021/acsnano.0c07287
IF: 17.1
2020-11-11
ACS Nano
Abstract:Hypoxia is a hostile hallmark of most solid tumors, which often leads to multidrug resistance (MDR) and causes the failure of chemotherapy. Hypoxia also promotes epithelial–mesenchymal transition (EMT), leading to acceleration of tumor metastasis. Many chemotherapeutic drugs can further exacerbate hypoxia and thus promote metastasis. Therefore, relieving hypoxia is necessary for chemotherapy to inhibit both MDR and EMT. Herein, highly stable cerasomal perfluorocarbon nanodroplets with an atomic layer of polyorganosiloxane surface and pH-sensitive tumor-targeting peptide (D-vPCs-O<sub>2</sub>) were fabricated to co-deliver oxygen and therapeutic drug, doxorubicin. High-intensity focused ultrasound (HIFU) was utilized to trigger the co-release of doxorubicin and oxygen and simultaneously enhance ultrasound imaging, therefore achieving imaging-guided drug delivery. Mild-temperature HIFU (M-HIFU) not only triggered oxygen release from nanodroplets but also slightly elevated tumor temperature to accelerate tumor blood flow. The oxygen release and temperature elevation jointly relieved tumor hypoxia and alleviated MDR, which greatly enhanced the drug therapeutic efficacy as compared to clinically used doxorubicin and Doxil. Overall side effects were also largely reduced owing to the ultrastable drug loading of cerasome. The improvement of insufficient chemotherapy and the relief of tumor hypoxia corporately down-regulated TGF-β1, leading to the alleviation of EMT, and therefore significantly inhibited tumor metastasis. When "D-vPCs-O<sub>2</sub> + M-HIFU" was utilized as a neoadjuvant chemotherapy, nanodroplets down-regulated heat shock proteins, reducing tumor relapse after the high-temperature HIFU (H-HIFU)-mediated hyperthermia ablation. The chemo-hyperthermia therapy totally eradicated tumors without any relapse or metastasis, providing a promising way to treat the triple-negative breast cancer, which is highly malignant, easily metastatic, and lacks effective treatments.The Supporting Information is available free of charge at <a class="ext-link" href="/doi/10.1021/acsnano.0c07287?goto=supporting-info">https://pubs.acs.org/doi/10.1021/acsnano.0c07287</a>.Supplementary methods, drug loading capability, hydrodynamic diameters, <i>in vitro</i> biocompatibility, blood biochemistry and blood routine examination, histopathology examination, <i>in vitro</i> expression level of HSP-27 and HSP-90 (<a class="ext-link" href="/doi/suppl/10.1021/acsnano.0c07287/suppl_file/nn0c07287_si_001.pdf">PDF</a>)This article has not yet been cited by other publications.
materials science, multidisciplinary,chemistry, physical,nanoscience & nanotechnology
-
Hyperbaric oxygen augments chemodynamic effect induced by probiotic-derived selenium nanoparticles to enhance cancer immune checkpoint blockade therapy
Puze Li,Yuzhu Yao,Xue Chen,Cheng Song,Meichan Wu,Jinzhao Huang,Dongdong Wang,Xuyu Li,Ban Luo,Xiangliang Yang,Jun Hu
DOI: https://doi.org/10.1016/j.cej.2023.145738
IF: 15.1
2023-09-10
Chemical Engineering Journal
Abstract:Chemodynamic therapy (CDT) was a promising approach to enhance the immunogenicity of tumor cells for potentiating immune checkpoint blockade (ICB) therapy, but usually suffered from hypoxia and immunosuppressive tumor microenvironment. Herein, we developed a hyperbaric oxygen (HBO)-strengthened CDT-nanoadjuvants platform (Se@OMV-GOx-HA) by modifying probiotic-derived selenium (Se) nanoparticles with glucose oxidase (GOx) and hyaluronic acid (HA) to boost αPD-L1-medaited ICB therapy against breast cancer. In the presence of H 2 O 2 and glutathione (GSH), Se@OMV acted as nanoenzyme to mediate the electron transfer from GSH to O 2 giving O 2 – , while GOx catalyzed the production of H 2 O 2 . Synergistically, HBO effectively improved tumor hypoxia to foster GOx-catalyzed oxidation of glucose to H 2 O 2 , then provided the essential fuels including H 2 O 2 and O 2 for Se-mediated O 2 – generation, causing strong immunogenic cell death (ICD) effect to enhance the immunogenicity of tumor cells. Under co-stimulation by Se@OMV acting as immunoadjuvants, maturation of dendritic cells and priming of cytotoxic T cells were largely promoted. Assisted by HBO, infiltration of cytotoxic T cells in mice tumor was significantly improved, which not only generated long-term antitumor immune memory, but largely boosted αPD-L1-medaited ICB therapy. Given the clinical available of HBO, this nanosystem has great clinical translation potential for cancer CDT/immunotherapy.
engineering, chemical, environmental