Open Access Article
Fang
Yuan†
a,
Lu
Wang†
a,
Lulu
Ning
*b,
Jianjian
Zhang
*a and
Yuan
Guo
*ac
aKey Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, P. R. China. E-mail: zhangjj@nwu.edu.cn; guoyuan@nwu.edu.cn
bShaanxi Provincial Key Laboratory of Papermaking Technology and Specialty Paper Development, College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, P. R. China. E-mail: ninglulu@sust.edu.cn
cEngineering Research Center of Western Resource Innovation Medicine Green Manufacturing of the Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710127, P. R. China
First published on 29th September 2025
Cancer therapy has made notable progress in moving towards more precise, intelligent and personalized treatment modes. However, current approaches such as chemotherapy, phototherapy, sonodynamic therapy, and immune therapy still suffer from limitations, hindering their application in clinical practice. Gasotransmitters have long been recognized as key regulators in cancer pathology, and the development of novel therapeutic agents with gas-releasing ability aiming to establish new therapeutic modes has garnered considerable attention. In this perspective, we aim to summarize the critical roles of gaseous molecules in cancer biology and their potential mechanisms for enhancing the efficacy of dominant treatment modalities. We also provide insights into recent advances in integrating gas therapy with conventional treatments to overcome current challenges and improve therapeutic outcomes, ultimately paving the way for the development of next-generation treatment paradigms.
Gas therapy (GT) has emerged as a promising therapeutic modality that utilizes bioactive gaseous molecules to regulate pathological processes and exert therapeutic effects. In recent years, several physiologically significant gases, including gasotransmitters, such as nitric oxide (NO),13 hydrogen sulfide (H2S),14 and carbon monoxide (CO),15 as well as other biocompatible gases, such as hydrogen (H2)16 and oxygen (O2),17 have attracted increasing attention as emerging agents in cancer therapy. Notably, the gasotransmitters often exhibit concentration-dependent biphasic effects, acting as a “double-edged sword” in both physiological and pathological contexts.18 For example, H2S, produced via multiple endogenous metabolic pathways, induces minimal side effects in normal cells at low concentrations.19 However, elevated levels of H2S in tumor tissues disrupt mitochondrial homeostasis and induce apoptosis, primarily through the inhibition of cytochrome c oxidase (complex IV, COX IV) in the electron transport chain (ETC). This suppression of mitochondrial bioenergetics impairs the biosynthesis of biomacromolecules and ultimately inhibits tumor proliferation.20 Similarly, both NO and CO can inhibit COX IV and other mitochondrial enzymes, further suppressing cellular energy metabolism.21 Despite considerable advances in gaseous therapeutics for tumor treatment, clinical translation remains challenging due to the need for precise regulation of the dosage and delivery strategies as improper administration may lead to severe respiratory toxicity and systemic side effects.22 Furthermore, single-agent gas therapy regimens are often insufficient to achieve complete tumor eradication.
Encouragingly, the integration of GT with other therapeutic modalities has emerged as a promising strategy, offering synergistic effects that enhance anticancer efficacy and provide a compelling direction for future development. When combined with CDT, RT, phototherapy, immunotherapy, or ferroptosis, gas molecules act as effective adjuvants. For instance: (1) O2 enhances the efficacy of PDT or SDT by alleviating tumor hypoxia;23 (2) NO reacts with ROS to generate highly reactive cytotoxic species, such as highly toxic peroxynitrite (ONOO−), thereby improving the therapeutic efficacy of PDT or SDT;24–26 (3) CO and NO can reverse multidrug resistance (MDR) via distinct mechanisms, thereby restoring chemosensitivity.27 Overall, integrating GT with existing anticancer modalities may significantly potentiate their therapeutic outcomes (Scheme 1).
In this perspective, we first outline representative biotherapeutic gases (e.g., NO, CO, and H2S), then focus on their synergistic mechanisms in tumor therapy. Our goal is to elucidate their anticancer pathways, while highlighting their unique advantages and inherent limitations. Finally, we summarize recent advances in combining GT with other treatment modalities. This perspective does not aim to provide exhaustive coverage of the field, as many recent reviews have already done so,28–31 but instead offers a focused discussion of key themes currently under research.
Recently, attention has shifted to the anti-tumor activity of NO. Several studies have demonstrated that a high local concentration of NO can suppress tumor progression by inhibiting mitochondrial enzymes and inducing DNA damage, thereby restricting the tumor growth.35 Mechanistically, this process involves the concurrent production of NO (via iNOS) and superoxide radical (O2˙−, via NADPH oxidase) in activated tumor-associated macrophages. These species react to form ONOO−, a highly potent cytotoxic molecule that kills tumor cells.36 These mechanisms can be further enhanced by immunostimulatory therapeutic strategies and can synergize with other ROS-dependent therapies to improve the therapy outcome.37
CO also exhibits pleiotropic effects on cancer progression. At low concentrations, CO promotes tumor growth through the CO/HO-1 system.41 However, at high concentrations exceeding a specific threshold, it can suppress tumor growth by inhibiting mitochondrial activity, inducing excessive ROS production and downregulating major protein expression, thereby reducing the tumor cell proliferation and survival.42,43 Additionally, CO modulates cancer metabolism by inhibiting the Warburg effect, forcing tumor cells to consume more oxygen and ultimately leading to cell death.44
Beyond their independent effects, many physiological processes require the coordinated signaling pathways between H2S and NO to be completed.49 These gasotransmitters share considerable overlapping molecular signaling pathways including PI3K/Akt and MAPK, and their interplay modulates cancer cell survival, proliferation, and the immune response.50 Deciphering these interactions, particularly the downstream signaling crosstalk among H2S, NO, and CO, may provide novel therapeutic strategies for cancer treatment.51
These pharmacological challenges have driven innovations in gas delivery systems, paralleling expanding clinical investigation. For example, a Phase 1b trial (NCT05351502) is currently evaluating a low-volume, ultra-high concentration of nitric oxide (LV-UNO) in combination with PD-1 inhibitors. The study assesses the overall response rate (ORR), duration of response (DOR), and immune-related responses in patients with tumors. Another ongoing trial (NCT05607407) explores an indirect H2S modulation strategy using methimazole in progressive glioblastoma, aiming to improve treatment outcomes and extend survival. These examples illustrate the potential advantages of integrating GT with established cancer treatment modalities. In the following sections, we discuss the mechanisms through which these gases contribute to tumor therapy, highlighting their diverse roles across different biological contexts.
Addressing the chronic inflammation offers a promising therapeutic strategy for tumor intervention. Accumulating evidence indicates that gaseous mediators are inherently involved in cellular signaling and play key roles in regulating inflammatory responses. For instance, H2S suppresses the nuclear factor κB (NF-κB) pathway and downregulates chemokines such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), thereby exerting anti-inflammatory effects.19,46 Similarly, CO exhibits therapeutic anti-inflammatory properties by inducing HO-1 production and regulating inflammatory signaling pathways, including MAPK and the NF-κB.72 Notably, inflammation not only precedes tumorigenesis but also shapes the adaptive immune response, influencing both its magnitude and specificity. This underscores inflammatory modulation as a robust therapeutic approach to restrain tumor progression.
Immune suppression represents a major obstacle for effective tumor immunotherapy. For instance, the hypoxia TME promotes the accumulation of adenosine, which inhibits effector T-cell function.74 Interestingly, recent studies have shown that H2S promotes the differentiation of central memory T cells (Tcm), a phenotype associated with enhanced antitumor responses and long-term persistence.75 Experimental evidence shows that H2S-treated T cells or those engineered to overexpress CBS (an H2S-producing enzyme) demonstrated superior tumor control in melanoma and lymphoma models. Mechanistically, this effect depends on the NAD+–Sirt1–Foxo1 axis and improved mitochondrial function.
In addition, H2S-mediated protein S-sulfhydration has emerged as a critical regulator in tumor-associated macrophages (TAMs), which typically drive immunosuppression and tumor progression. Activation of the CTH–H2S axis has been associated with the repolarization of TAMs toward the anti-tumorigenic M1 phenotype, potentially blocking their tumor-promoting activities in breast cancer.76 Moreover, both CO and H2S compromise mitochondrial integrity, resulting in the cytosolic release of mitochondrial DNA. This event enables NO and H2S to function as a gas immunoadjuvant that activates the cGAS–STING pathway, a central driver of innate immune responses.77 These findings underscore the significant role of gaseous signaling molecules in immune therapy. A deeper understanding of their interplay with adaptive immune responses could open new avenues for advancing cancer immunotherapy.
Conventional chemotherapeutic agents that inhibit metabolic enzymes or complexes are often limited by their toxicity to normal tissues. Recent studies, however, highlight the role of gaseous signaling molecules (such as H2S and NO) in cancer metabolism. Exogenous H2S enhances glucose metabolism and lactate accumulation by impairing proton export, resulting in intracellular acidification. This acidic stress disrupts homeostasis and triggers apoptotic cell death.84 NO, a byproduct of cellular metabolism, exerts context-dependent roles in tumor biology. On the one hand, NO impairs mitochondrial respiration and increases glutamine consumption in the TCA cycle, promoting tumor progression and chemotherapy resistance. Thus, inhibiting NO synthesis in stromal cells may suppress tumor-supportive effects and improve therapeutic outcomes.85 On the other hand, NO-mediated post-translational modifications,86 particularly S-nitrosation, exhibit tumor-suppressing properties in highly glycolytic or hypoxic cancer cells. These dual roles of NO underscore the importance of precision dosing and combinatorial strategies in NO-based therapies. Further studies are essential to evaluate NO donors and small molecule regulators, as targeting NO metabolism could disrupt tumor survival networks and provide novel therapeutic avenues.
NO functions as a bell-shaped effector molecule in the TME, exerting both pro- and anti-tumorigenic effects. Elevated NO levels in TME promote tumor progression and migration by upregulating caveolin-1 expression, as observed in melanoma, breast, and prostate cancer.89,90 NO also regulates angiogenesis in the TME and has been exploited as a therapeutic target. Specifically, inhibiting the expression of NOS1 can sensitize glioma tumors to radiotherapy.91 Beyond tumor initiation, stroma-derived NO has also been observed to have tumor-suppressive effects.92 These studies highlight the divergent roles of NO in the TME.
Although experimental research in this field is still in its early stages and is further complicated by the pleiotropic effects of signaling molecules on tumor progression, such mechanistic insights highlight the unique and versatile roles of gasotransmitters in modulating tumor biology and demonstrate considerable promise as therapeutic agents (Fig. 1b). However, considering the multifaceted nature of cancer progression and the limitations of single-modality treatments (Fig. 1a), increasing research efforts have shifted toward integrating GT with both conventional and emerging therapeutic strategies. In the following sections, we will discuss how gaseous signaling molecules can synergize with established treatment modalities, including CDT, PDT, PTT, SDT, and immunotherapy (Fig. 1c), to enhance therapeutic efficacy and overcome resistance mechanisms (Fig. 1d).
![]() | ||
| Fig. 2 Schematic of the design rationale of the chemiexcitation-triggered poly-prodrug for the co-activation of CO and CA-4 for combinational therapy. (a) Chemical structures of the triblock copolymers. (b) Mechanism of the H2O2-triggered co-activation of CA-4 and CO. Reproduced from ref. 93 with permission from Springer Nature, copyright 2025. (c) Reactive oxygen species (ROS) cascade nanoplatform targeting the regulation of P-glycoprotein, and the synergistic induction of ferroptosis to reverse multidrug-resistance in prostate cancer. Schematic of the mechanism of nanoplatform TK-Fc/LAE NPs with ROS cascade amplification for gas therapy/ferroptosis/chemotherapy. Reproduced from ref. 94 with permission from Wiley-VCH, copyright 2024. | ||
To overcome drug resistance in prostate cancer, Pang et al. designed a nanoplatform (TK-Fe/LAE NPs) integrating GT, ferroptosis, and chemotherapy (Fig. 2c).94 Upon activation by elevated intracellular ROS, the system disassembles to release NO, which downregulates P-glycoprotein expression, disrupts mitochondrial function and reverses multidrug resistance (MDR). Concurrently, ferrocene-mediated Fenton reactions induce ferroptosis and amplify oxidative stress, while controlled paclitaxel release ensures targeted chemotherapy. The study highlights NO's potential to overcoming drug resistance and improving therapeutic outcomes.
H2S-releasing hybrid drugs offer a promising approach to enhance therapeutic efficacy while minimizing adverse effects. For instance, NSAID–H2S conjugates exhibit synergistic pharmacological benefits by improving anti-inflammatory potency and alleviating the gastrointestinal toxicity associated with conventional NSAIDs.95 Moreover, H2S has been reported to enhance the anti-tumor effects of chemo drugs in melanoma cells.96 Matson et al. reported a dual-responsive nanoplatform (AAN-PTC–Fe2+) for glioma treatment, integrating CDT with H2S GT (Fig. 3).97 Triggered by overexpressed legumain in tumor cells, this system selectively releases H2S, which inhibits catalase activity and promotes H2O2 accumulation. Meanwhile, Fe2+ catalyzes the Fenton reaction to convert H2O2 into highly toxic hydroxyl radicals (˙OH), amplifying ROS levels and leading to enhanced tumor cell death. The co-delivery system demonstrates superior efficacy compared to the chemotherapy drug temozolomide (TMZ) and exhibits negligible cardiotoxicity, underscoring its safety profile. This strategy highlights the potential of tumor-specific H2S delivery to augment CDT and overcome the limitations of conventional chemotherapeutics.
![]() | ||
| Fig. 3 Schematic of the PHDC–Fe2+ complex chemical structure and therapeutic mechanism in the C6 glioma cell line. Reproduced from ref. 97 with permission from Wiley-VCH, copyright 2023. | ||
The integration of GT with CDT represents a potent synergistic strategy to enhance the anticancer efficacy. Gas molecules such as CO, NO, and H2S not only sensitize tumor cells to chemotherapeutic agents by disrupting mitochondrial function, reversing MDR, or inhibiting detoxifying enzymes, but also contribute to amplified oxidative stress or ferroptosis for tumor eradication. These hybrid platforms enable tumor-specific, stimulus-responsive drug release, thereby minimizing systemic toxicity while improving therapeutic precision.
Despite advances in photosensitizer (PS) design and light sources, PDT remains limited by hypoxia in late-stage solid tumors.100,101 Because PDT is oxygen-dependent, low O2 levels often limit its efficiency, and PDT itself can further exacerbate hypoxia, potentially leading to drug resistance. To address this, the most straightforward strategy is combining PDT with oxygen delivery.102,103 An alternative strategy involves alleviating hypoxia through modulation of the TME. Abnormal TME not only facilitates tumor proliferation and metastasis, but also establishes physiological barriers that impede effective penetration of therapeutics inside the tumor, posing great challenges for cancer treatment. To tackle this, Min et al. developed a hierarchical nanoplatform (denoted as T-PFRT) that can adapt to the TME via size transformation (Fig. 4a). In response to matrix metalloproteinase 2 (MMP2), T-PFRT releases the small PFRT module, which depletes stromal components and enhances O2 delivery via hemoglobin (Hb). These therapeutic effects simultaneously overcome stromal and hypoxic barriers, achieving deep tumor penetration, improved PDT performance, and enhanced overall therapeutic efficacy.104 This strategy demonstrated excellent therapeutic outcomes in the treatment of both primary and metastatic tumors.
![]() | ||
| Fig. 4 (a) Structure and composition of T-PFRT; working principles of the therapeutic components in TME normalization and photodynamic therapy for tumor inhibition and metastasis blockade. Reproduced from ref. 104 with permission from Wiley-VCH, copyright 2021. (b) Molecular structure and CO release mechanism of NanoBDP2I-S-HF. Reproduced from ref. 105 with permission from Springer Nature, copyright 2024. | ||
Simultaneous modulation of multiple parameters in the TME represents a promising therapeutic direction. Zhao et al. developed a multifunctional nanomedicine, NanoBDP2I-S-HF, which enables precise modulation of multiple gases (CO release and H2S depletion), while simultaneously enhancing PDT efficacy (Fig. 4b).105 This nanomedicine utilizes a disulfide bond to covalently conjugate a PS with the CO donor 3-hydroxyflavone (3-HF), enabling H2S-triggered bond cleavage in tumor regions. This process both reduces local H2S levels and releases 3-HF for CO-mediated therapy. Upon 660 nm laser irradiation, the PS generates cytotoxic 1O2, which subsequently oxidizes 3-HF to precisely trigger CO release. This H2S-light-1O2-responsive cascade ensures accurate gas delivery and significantly enhances PDT efficacy. Compared to single PDT treatment, NanoBDP2I-S-HF demonstrates superior therapeutic efficacy against HCT116 tumors. This multimodal-activated therapeutic strategy provides an innovative approach for improving both the precision and effectiveness of synergistic cancer treatment.
Compared with PDT, PTT typically requires higher light power due to its lower energy conversion efficiency. However, the use of high-power light sources often causes photodamage to healthy tissue and compromises its clinical feasibility. Effective tumor ablation usually demands localized temperatures exceeding 50 °C, which unavoidably harms surrounding normal tissues through heat diffusion. To address this limitation, the concept of mild PTT was proposed. This strategy focuses on suppressing heat shock protein (HSP) protein expression, which can counteract tumor thermotolerance while reducing PTT-related side effects.106
In 2023, Liu et al. constructed a co-facilitated gas-photothermal therapy nanoplatform ADT@CuSND (Fig. 5a).107 This platform enables precise and sustained delivery of H2S to tumor sites. Elevated H2S levels effectively inhibit COX IV, thereby disrupting the mitochondrial respiratory chain, inhibiting ADP conversion and downregulating HSP90, which collectively sensitize the tumor to hyperthermia. By reversing tumor thermotolerance, ADT@CuSNDs significantly enhanced mPTT efficacy, achieving effective tumor ablation with a single treatment while simultaneously minimizing damage to healthy tissues. This energy remodeling approach represents a promising paradigm for improving PTT efficacy and holds potential for the future clinical translation of tumor therapy.
![]() | ||
| Fig. 5 (a) Schematic of ADT@CuSND application in the mild photothermal therapy of a tumor. After being taken up by the cells, ADT@CuSNDs continuously released H2S into the tumor cells, which downregulated COX IV, interfered with the mitochondrial respiratory chain and blocked the energy supply in tumors. The depleted cellular ATP pool reduced the risk of the overexpression of HSP90 in response to heat stress, further reversing the thermotolerance of tumors and allowing ADT@CuSNDs to obtain enhanced mPTT. Reproduced from ref. 107 with permission from Wiley-VCH, copyright 2023. (b) Schematic of the gas/phototheranostic nanocomposite (NA1020–NO@PLX). An enhanced ICT mechanism was introduced to develop the NIR-II-peak absorbing PTA of NA1020, which was combined with thermal-sensitive NO donors to facilitate a combined low temperature PTT with gas therapy. NA1020–NO@PLX emitted the NIR-II fluorescence to guide the heat generation that simultaneously activated NO release using a laser at 1064 nm for atraumatic osteosarcoma therapy. Reproduced from ref. 109 with permission from Wiley-VCH, copyright 2023. | ||
NO has also been reported to sensitize tumor cells to PTT by inhibiting protective autophagy, thereby enhancing tumor cell death in combined treatment.108 Li et al. reported a gas/phototheranostic nanocomposite (NA1020–NO@PLX, Fig. 5b), which integrates aza-BODIPY, NA1020 with a thermal-sensitive NO donor.109 The enhanced ICT process endows the NA1020 with NIR-II-peak absorbance (1020 nm), enabling deep tissue penetration for precise imaging and PTT of deep tissue tumor. Moreover, the NA1020 exhibits a remarkable photothermal conversion efficiency, while NO release upon laser irradiation induces mitochondrial dysfunction and DNA damage, thereby augmenting the efficacy of low-temperature PTT. By combining low temperature PTT with NO delivery, significant tumor eradication in an orthotopic osteosarcoma model was observed without causing undesired tissue damage, thereby significantly minimizing the side effects commonly associated with PTT. In summary, gas/phototheranostic combination strategies have emerged as a research hotspot, underscoring the strengths of enhanced phototherapy in tumor eradication and demonstrating great potential for clinical translation.
In addition to the two classical phototherapeutic modalities, Yang et al. reported a NO-based, TME-independent phototherapeutic platform (ArgCCN, Fig. 6).110 Upon irradiation, the photogenerated holes on ArgCCN converted water into H2O2, which subsequently oxidized the arginine residues to produce NO. The burst release of NO then induced tumor cell apoptosis. Different from other therapeutic modalities, this approach does not rely on oxygen to produce ROS, nor does it exacerbate tumor hypoxia, thereby effectively overcoming the major limitations of PDT. This distinctive phototherapeutic strategy offers new perspectives for the advancement of light-based therapeutic modalities.
![]() | ||
| Fig. 6 Schematic of the microenvironment-independent NO-based phototherapeutic nanoplatform. Photogenerated hole-mediated oxidation was used to generate NO for hypoxic tumor treatment, which was achieved by poly-L-arginine-modified carbon-dot-doped g-C3N4 (ArgCCN). The holes with oxidizability were generated by laser irradiation of ArgCCN, which oxidized H2O to generate H2O2, and the H2O2 further oxidized the arginine residues to accomplish the NO production that is independent of the tumor microenvironment. Reproduced from ref. 110 with permission from Wiley-VCH, copyright 2021. | ||
A single dose of SDT is often insufficient for complete tumor eradication, as the persistent hypoxic nature of TME significantly limits its therapeutic efficacy. This highlights the potential of gas delivery to augment the therapeutic efficacy of SDT. Under this premise, Zhao et al. designed a pH/ultrasound dual-responsive biomimetic nanoplatform for combined GT and SDT, aiming to overcome the limitations of traditional PDT.116 The nanoplatform, consisting of ZIF-8 loaded with chlorin e6 (Ce6) and nitrosoglutathione (GSNO), was coated with homologous tumor cell membranes to enable active tumor targeting. In the acidic TME, ultrasound stimulation triggered the controlled release of Ce6 and NO. The released NO not only relieved hypoxia, but also synergized with ultrasound-activated Ce6 to enhance ROS and ONOO− production. This gas–sonodynamic combination resulted in effective tumor suppression with demonstrated biocompatibility, reduced phototoxicity, and improved therapeutic precision. The study provides a promising strategy for the design of multifunctional platforms in next-generation cancer therapies.
SO2 has been demonstrated to induce oxidative stress-mediated damage of biomacromolecules, making it applicable in anti-inflammatory, antibacterial, and anti-cancer therapies. In a recent study, a multicomponent polymerization strategy was developed to synthesize poly(BODIPY-sulfonamide)s (PBSAs) capable of simultaneous ROS and SO2 generation under US irradiation (Fig. 7a).117 Among them, a ROS-responsive polymer (PBSA-EG) incorporating thioketal linkers and hydrophilic chains was engineered into nanoparticles for in vivo application. Upon US activation, the system effectively released both ROS and SO2, achieving enhanced tumor suppression through synergistic sonodynamic–gas therapy. This work highlights the potential of ultrasound-triggered SO2 delivery as a powerful complement to SDT, overcoming tumor resistance and amplifying therapeutic efficacy.
![]() | ||
| Fig. 7 (a) Schematic of the preparation of PPSIs and PBSAs for the US-driven ROS and SO2 release to achieve sonodynamic and gas combination therapy. Reproduced from ref. 117 with permission from Wiley-VCH, copyright 2025. (b) Fabrication of Aza-BD@PC NPs. (c) Synergistic GT and SDT effect caused by Aza-BD@PC NPs under US irradiation for enhanced therapeutic efficacy. Reproduced from ref. 118 with permission from Wiley-VCH, copyright 2025. | ||
Glioblastoma multiforme (GBM) is the most aggressive and fatal form of glioma, characterized by rapid progression and poor prognosis. To overcome the resistance of GBM to apoptosis, Wu et al. developed a nanoplatform (Aza-BD@PC NPs, Fig. 7b), which integrates H2S-based GT with ferroptosis-augmented SDT for GBM therapy.118 Upon internalization, the nanoplatform consumed intracellular cysteine (Cys), triggering H2S release, and disrupting redox balance and metabolism, while also inducing ferroptosis. Concurrently, the released Aza-BD generated abundant 1O2 under US irradiation, enabling efficient SDT. The synergistic interplay of H2S-mediated ferroptosis and US-triggered ROS production led to significant GBM suppression (Fig. 7c), with in vivo inhibition rates up to 97.5%. This strategy offers a promising avenue to enhance SDT efficacy through ferroptosis-enabled GT in treatment-resistant tumors such as GBM.
For instance, Cheng et al. developed PEGylated Mn-doped CaS nanoparticles (MCSP) as a TME-responsive platform for gas-amplified metalloimmunotherapy against cervical cancer (Fig. 8a).120 Upon exposure to the acidic TME, MCSP rapidly releases Ca2+, Mn2+, and H2S. H2S disrupts mitochondrial oxidative phosphorylation, leading to calcium overload and pyroptosis, a pro-inflammatory cell death that enhances immunogenicity (Fig. 8b). Simultaneously, H2S-induced mitochondrial damage promotes mtDNA leakage, which synergizes with Mn2+ to robustly activate the cGAS–STING pathway, triggering dendritic cell maturation (Fig. 8c). This dual mechanism bridges innate and adaptive immunity, reducing the proportion of immunosuppressive Treg populations and promoting infiltration of CD8+ T cells and M1 macrophages. Combined with αPD-1 therapy, MCSP achieves enhanced tumor suppression, demonstrating H2S's pivotal role as a gas mediator to amplify both pyroptosis and STING-driven immune activation.
![]() | ||
| Fig. 8 (a) Mechanism of pyroptosis and activation of the cGAS–STING pathway induced by MCSP. (b) H2S-enhanced adaptive and (c) innate immune response. Reproduced from ref. 120 with permission from the American Chemical Society, copyright 2024. (d) MP@AL manufacturing process. (e) Sketch map showing the double-closed loop of MP@AL-catalyzed metabolite lactate consumption and NO replenishment. The NO produced by the reaction inhibits tumor respiration on the one hand and ensures the continuation of lactic acid depletion response and reverses the TIME; on the other hand, it induces the death of tumor immunogenic cells, which synergistically stimulates the immune response and amplifies the efficacy of immunotherapy. Reproduced from ref. 121 with permission from Wiley-VCH, copyright 2025. | ||
To overcome the limitations of conventional GT in remodeling the immunosuppressive TME, Dong et al. developed a biomimetic, pH-responsive nanosystem (MP@AL, Fig. 8d).121 This system integrates lactate metabolism regulation and NO gas immunotherapy. Lactate oxidase consumes oxygen and decomposes lactate to produce H2O2, which then oxidizes L-arginine to generate NO. NO not only disrupts mitochondrial respiration (reducing tumor oxygen consumption and enhancing lactate clearance) and reshapes the tumor immune microenvironment (TIME), but also induces ICD and activates robust antitumor immune responses. Combined with PD-L1 blockade, this dual closed-loop platform significantly inhibited tumor growth and prevented recurrence (Fig. 8e). This work illustrates that combining GT with immunometabolic modulation synergistically reverses tumor immunosuppression and enhances immunotherapy efficacy.
![]() | ||
| Fig. 9 (a) Schematic of IR&ZnPc@LNP-NO, and its response to sequential low-power and high-power US irradiation: size reduction, controlled IR and NO release, and ROS generation. (b) Illustration of the mechanism of IR&ZnPc@LNP-NO synergized with αPD-L1 for SDT-chemo-immunotherapy of deep-seated pancreatic cancer. Reproduced from ref. 122 with permission from Wiley-VCH, copyright 2025. (c) Schematic illustrating the preparation routes for the gas nanoadjuvant. (d) Schematic of the gas nanoadjuvant-based cGAS–STING pathway-dependent antitumor immune responses. Reproduced from ref. 77 with permission from Springer Nature, copyright 2023. | ||
To overcome the immunosuppressive microenvironment of triple-negative breast cancer (TNBC), Chen et al. reported a virus-mimicking gas nanoadjuvant by co-encapsulating an AIE-active photosensitizer and manganese carbonyl (MnCO) into a tetrasulfide-doped hollow mesoporous silica matrix (Fig. 9c).77 Triggered by tumor-specific GSH, the tetra-sulfide bonds enable selective drug release, enhance PDT, and generate H2S for gas-mediated immunomodulation. Upon sequential NIR laser irradiation, the AIEgen-mediated phototherapy triggers the burst of CO/Mn2+. NO gas molecules disrupt mitochondrial integrity, inducing cytosolic mtDNA leakage and activation of the cGAS–STING pathway. Mn2+ further enhances type I interferon (IFN) production, amplifying immune responses. This gas-augmented nanoplatform significantly boosts AIEgen-mediated PDT and PTT, resulting in effective tumor regression, distant tumor suppression, and prevention of metastasis and recurrence in TNBC models (Fig. 9d). The study highlights gas immunoadjuvants as powerful tools to potentiate photoimmunotherapy in poorly immunogenic tumors.
H2Se gas has recently garnered attention within the research community.59 Although it has not yet been classified as a gasotransmitter, H2Se shares many features with H2S and participates in redox-balance regulation across physiological systems. Jiang et al. constructed a Cu2−xSe@cMOF nanoplatform for combined sonodynamic/cuproptosis/gas therapy (Fig. 10a).123 Upon US stimulation, this system generated ROS and released H2Se gas, which synergistically induced oxidative stress and mitochondrial dysfunction. Concurrently, the copper core promoted cuproptosis by disrupting redox homeostasis and depleting GSH. This multifaceted strategy effectively suppressed tumor growth and metastasis. When combined with the PD-L1 immune checkpoint blockade, the platform further amplified the anti-tumor immunity. This work demonstrated how GT can be exploited to coordinate multiple therapeutic mechanisms, offering a potent strategy against primary and recurrent tumors.
![]() | ||
| Fig. 10 (a) Schematic of trimodal tumor therapy and immunotherapy using Cu2−xSe@cMOF nanoparticles. Synthesis of Cu2−xSe@cMOF and proposed dual enhancement of immune function by Cu2−xSe@cMOF for improved tumoricidal effects. Reproduced from ref. 123 with permission from the American Chemical Society, copyright 2024. (b) Schematic of the dual-targeting biomimetic nanoplatform for integrating CDT/SDT/gas therapy to boost synergistic ferroptosis against orthotopic HCC. The CO-releasing molecule CORM-401 was encapsulated into the bimetallic compound FM to form a multi-modal therapeutic nanoplatform (FM/C), which was further cloaked with a macrophage membrane inserted with the SP94-peptide to endow dual-targeting ability toward HCC. The multi-enzyme activity of FM not only promoted CDT to generate ˙OH, but also improved the SDT performance to produce 1O2 by alleviating hypoxia, which in turn activated CO release. As a result, the depletion of GSH and high level of ROS led to the accumulation of lethal lipid peroxidation (LPO) to promote ferroptosis-based tumor death in an orthotopic HCC model. Reproduced from ref. 124 with permission from Wiley-VCH, copyright 2025. | ||
To overcome the therapeutic limitations of hepatocellular carcinoma (HCC), Li et al. presented a biomimetic nanoplatform by integrating FeMoO4 and CORM-401, cloaked with a peptide-modified macrophage membrane to synergize SDT, CDT, and GT (Fig. 10b).124 The multivalent bimetallic FeMoO4 could decompose H2O2 to O2, alleviating tumor hypoxia and enhancing SDT efficiency, while its peroxidase-like activity promoted ˙OH generation via Fenton chemistry for CDT. Simultaneously, the excessive ROS triggered intracellular CO release, inducing mitochondrial dysfunction, GSH depletion, GPX4 inhibition, and lethal lipid peroxidation, thereby amplifying ferroptosis. This cascade established a robust ROS–CO–ferroptosis axis, enabling spatially and temporally controllable ferroptosis activation. The biomimetic membrane provided active HCC targeting and immune evasion, facilitating deep tumor penetration. Collectively, this gas-augmented multimodal strategy highlights the therapeutic value of CO gas as both an effector and amplifier of ferroptosis and redox-driven modalities, offering a promising approach for synergistic GT in refractory tumors such as HCC.
Given these unique features, GT is increasingly being explored in combination with established treatment modalities. A distinct advantage of this strategy is that the treatment platform can be sophisticatedly engineered to meet increasingly complex therapeutic requirements. As highlighted in this perspective, GT could be combined with several mainstream therapeutic modalities (CDT, PDT, PTT, SDT, and immunotherapy) for cancer treatment, as summarized in Table 1. In addition, biotherapeutic gases can substantially enhance therapeutic outcomes, potentially by resolving inflammation, stimulating the immune response, remodeling the tumor metabolism and bioenergetic processes, and modulating the TME.
| Gas | Structures | Therapeutic model | Trigger conditions | In vivo model | Ref. |
|---|---|---|---|---|---|
| CO | FT1@P1/P2 | CO-enhanced CDT | H2O2 | 4T1 tumor | 93 |
| NO | TK-Fc/LAE NPs | NO/ferroptosis/CDT | GSH | DU145 tumor | 94 |
| H2S | AAN-PTC–Fe2+ | H2S combination with CDT | Legumain | C6 glioma cells | 97 |
| O2 | T-PFRT | Oxygen-amplified PDT | MMP 2 | 4T1 tumor | 104 |
| CO | NanoBDP2I-S-HF | CO-amplified PDT | H2S | HCT116 tumor | 105 |
| H2S | ADT@CuSNDs | Gas-mediated sensitizing mild PTT | Hydrolysis | 4T1 tumor | 107 |
| NO | NA1020–NO@PLX | NO-mediated low temperature PTT | Light | Orthotopic osteosarcoma model | 109 |
| NO | ArgCCN | Photogenerated holes and NO gas therapy | Light | MCF-7 tumor | 110 |
| NO | GCZ@M | NO GT with SDT | pH/US | 4T1 tumor | 116 |
| SO2 | PBSA-EG | GT and SDT | US | H22 tumor-bearing mice | 117 |
| H2S | Aza-BD@PC NPs | GT and SDT | US | Glioblastoma multiforme | 118 |
| H2S | MCSP | H2S gas with immunotherapy | TME | U14 tumor | 120 |
| NO | MP@AL | Gas-immunometabolic therapeutic nanosystem | pH | Hepa1-6 tumor | 121 |
| NO | IR&ZnPc@LNP-NO | Gas-enhanced SDT-chemo-immunotherapy | US | Pancreatic cancer | 122 |
| H2S | MTHMS | Gas-enhanced PDT/PTT and immunotherapy | Light | 4T1 tumor | 77 |
| H2Se | Cu2−xSe@cMOF | GT, cuproptosis and SDT | US | 4T1 tumor | 123 |
| CO/O2 | PM-FM/C | GT, CDT and SDT | US | Hepatocellular carcinoma | 124 |
However, several key challenges must still be addressed in applying GT to cancer treatment. First, our understanding of the physicochemical and biological properties of these bioactive gases remains limited. Second, compared with conventional non-volatile drugs, gas delivery strategies are still in their infancy, particularly for in vivo applications. This creates several specific obstacles. (i) Only a few therapeutic strategies have considered the interactions and cooperative effects among gasotransmitters, even though they often act synergistically rather than independently. (ii) The heterogeneous distribution of gasotransmitters across tissues and cell types makes it difficult to determine optimal therapeutic concentrations. The intrinsic instability and variability of their derivatives further complicate accurate measurement and monitoring. (iii) Current delivery platforms, although sophisticated, raise concerns about biocompatibility. In addition, nanoplatforms that rely on endogenous triggers may lead to premature gas release, while those restricted to a single gas-release profile are often insufficient to meet the increasingly complex therapeutic demands.
To overcome these barriers and advance GT-based synergistic therapies, future research should focus on improving therapeutic efficacy, biocompatibility, and targeting efficiency, while ensuring safe and effective clinical translation. Firstly, a revolution in gas delivery mechanism is needed to diversify the development of small-molecule gas donors and expand their integration into nanoplatforms. For example, enzyme-mediated delivery could mimic endogenous gas-generating pathways, while repurposing clinically approved drugs with controllable gas-donating features or modifying natural biomolecules (1-thio-β-D-glucose) could provide safe and flexible delivery options. Secondly, dual gas donors remain rare, yet they are critical for probing gasotransmitters crosstalk, such as the cooperative roles of NO and H2S in post-translational modifications, and for designing new therapeutics. Thirdly, more precise synergy modalities are also required. Promising approaches include click-to-release chemistry (e.g., strain-promoted azide–alkyne cycloaddition and SPAAC), antibody–antigen binding for tumor targeting, covalent modification of tumor targeting peptides (e.g., RGD and NGR), and the use of pro-metabolites for metabolic labeling. Additionally, dynamic monitoring should be integrated into gas delivery systems, enabling real-time visualization of metabolic process (e.g., fluorescence-based readouts) when combined with mainstream therapies.
Taken together, gas-based therapies hold considerable promise as combination approaches that diversify treatment strategies, mitigate their side effects, and enhance therapeutic efficacy. The likelihood of clinical translation will increase as these challenges are progressively addressed. Only through a deeper understanding of the complex and sometimes paradoxical roles of gasotransmitters in cancer pathophysiology can truly novel therapeutic paradigms be realized. With continued interdisciplinary research and technological advances, GT is poised to emerge as a transformative force in next-generation cancer treatment.
Footnote |
| † These authors contributed equally to this work. |
| This journal is © The Royal Society of Chemistry 2025 |