Skip to main content
ACS Omega logoLink to ACS Omega
. 2024 Mar 11;9(11):12539–12552. doi: 10.1021/acsomega.3c09987

Calcium Carbonate-Based Nanoplatforms for Cancer Therapeutics: Current State of Art and Future Breakthroughs

Xiaoting Zhou †,, Qihui Wang †,, Zipeng Lei †,§, Ke Zhang , Shuxue Zhen , Huiqin Yao ‡,*, Yan Zu †,*
PMCID: PMC10955594  PMID: 38524488

Abstract

graphic file with name ao3c09987_0007.jpg

With the rapid development of nanotechnology, nanomaterials have shown immense potential for antitumor applications. Nanosized calcium carbonate (CaCO3) materials exhibit excellent biocompatibility and degradability, and have been utilized to develop platform technologies for cancer therapy. These materials can be engineered to carry anticancer drugs and functional groups that specifically target cancer cells and tissues, thereby enhancing therapeutic efficacy. Additionally, their physicochemical properties can be tailored to enable stimuli-responsive therapy and precision drug delivery. This Review consolidates recent literatures focusing on the synthesis, physicochemical properties, and multimodal antitumor therapies of CaCO3-based nanoplatforms (CBN). We also explore the current challenges and potential breakthroughs in the development of CBN for antitumor applications, providing a valuable reference for researchers in the field.

1. Introduction

Cancer is a prevalent disease threatening human health, and its treatment involves a variety of treatment modalities. With the continuous development of nanotechnology, nanoparticles (NPs) are increasingly being employed in cancer treatment, providing innovative strategies for drug transport, improving treatment efficiency, and alleviating side effects.1,2 Recently, the application of inorganic nanoparticles in drug delivery has become increasingly prominent.3,4 Nanoparticle drug delivery systems are particles between 1–500 nm in size that can deliver therapeutic drugs to specific targets in the body, thereby improving efficacy and reducing side effects. Among a variety of inorganic biominerals, due to their good biocompatibility and their superiority as an effective transport system for various types of drugs, calcium-based nanomaterials offer an exceptional range of advantages.5,6 With the advancement of nanotechnology, Ca2+ overload-induced apoptosis can be modulated by functional nanomaterials, leading to the development of a new cancer therapeutic strategy (termed Ca2+ interference therapy).7 Ca2+ overload-regulated tumor therapy does not demand any external stimuli such as light, heat and ultrasound, which is favorable for a widespread biological applications and clinical translation.8,9 Hence, there has been a growing focus on tumor therapy mediated by Ca2+ overload. In addition, calcium-based nanomaterials are generally inexpensive due to their ease of production and low-cost raw materials, and have relatively high biosafety due to biocompatible components.10

Five prevalent types of nanomaterials based on calcium include: calcium phosphate (CaP), calcium carbonate (CaCO3), calcium peroxide (CaO2), calcium hydride (CaH2), and calcium sulfide (CaS).9 Of these materials, calcium carbonate (CaCO3) is the most commonly utilized in calcium-based nanotechnology.11 Recently, CaCO3-based nanoplatforms (CBN) as drug delivery systems (DDSs) in cancer treatment have received considerable attention because of their low cost, biosafety, biodegradability, and pH sensitivity.1214 These DDSs enable the sustained and direct release of the targeted therapeutic agent. In particular, for protein and gene delivery and ultrasound (US) image-guided therapy, CBN have great potential.12,15 Another aspect of their potential use is the functionalization of CBN, which is mainly related to providing active targeting, increasing stability, and improving the ability to load the drug. In addition, the robust buffer function of CBN makes them especially competitive in alleviating the acidic tumor microenvironment (TME), which is one of the factors that most CBN may be involved in modulating tumor immunity.16 Most importantly, since the degradation products consist only of calcium (controlled by kidneys and accumulated in bones)17 and carbon dioxide (CO2, excreted by the lungs),18 the side effects of using CBN to treat tumors can be negligible.

In this Review, we summarize the development of CBN for cancer therapy. We focus on the synthesis, physicochemical properties (drug loading capabilities, responsive release, and their effects on the TME) of CBN and CaCO3-based multimodal antitumor therapies (Scheme 1). We also discuss the current problems and possible future breakthroughs in the use of CBN in antitumor applications. This Review can help researchers quickly understand the antitumor mechanism and research status of CBN, and it points out the direction for their future research.

Scheme 1. Schematic Illustration of the Synthesis, Physicochemical Properties and Multimodal Antitumor Therapies of CaCO3-Based Nanoplatforms (CBN).

Scheme 1

2. Synthesis of CaCO3-Based Nanoplatforms

2.1. Typical Methods to Prepare CaCO3 NPs

At present, common approaches for the preparation of CaCO3 NPs mainly include precipitation method, gas diffusion method, flame synthesis method, cockle shell decomposition method, biomineralization method, etc. (Table 1).10,11,19 Among them, CaCO3-based drug delivery systems typically use the solution precipitation method, microemulsion method and gas diffusion method (Figure 1).14,20,21 We will describe these synthesis methods in detail below.

Table 1. Comparison of the Main Methods of CaCO3-Based Nanoplatforms Synthesis.

Typical methods Principle Pros and cons ref
Precipitation method Calcium and carbonate ions react in water, precipitating as solid CaCO3 Pros: Easy, affordable, scalable, and adjustable particle size (10,19)
Cons: Potential particle inconsistency, limited crystal control
Gas diffusion method Gaseous CO2 reacts with dissolved Ca2+ in water to precipitate CaCO3 NPs Pros: Precise particle shape control, high purity of NPs (26,30)
Cons: Complex setup, slower production
Flame synthesis method Calcium-rich precursors combust to form CaCO3 NPs in the Flame Synthesis Method Pros: Fast NPs production, good for mass production (31)
Cons: Safety risks at high temperatures, variable particle sizes
Cockle shell decomposition Method CaCO3 NPs are obtained from Cockle shells mainly by mechanical grinding Pros: Natural source use, eco-friendly, organic incorporation (32)
Cons: Organic residue may affect purity, potentially slower process
Biomineralization method Biological entities guide CaCO3 NPs crystallization in calcium-rich solutions Pros: Green method, structured crystals, high purity and biocompatibility (33)
Cons: Slow reaction rate, precise biological control requirement

Figure 1.

Figure 1

Synthesis of CaCO3 NPs. Schematic illustration of the typical methods for preparing CaCO3 NPs used in cancer therapy: gas diffusion method, solution precipitation method, and microemulsion method.

The most well-known method for the production of CaCO3 NPs is the solution precipitation method, utilizing the reaction between Ca2+ and CO32– in aqueous solution. This method enables the mass production of CaCO3 NPs without the use of surfactants, thereby reducing the cost of production. Due to the mild conditions of preparation, many biologically active substances can be loaded into CaCO3 NPs in the process of precipitation, including small molecule drugs, genes, and proteins. For example, Vidallon et al. utilized rapid precipitation technology combined with biocompatible polymer additives such as bovine serum albumin (BSA) and polydopamine (PDA) to create CaCO3 hybrid particles.22 By using BSA and PDA, hybrid particles could have different sizes and polymorphic compositions through different reaction or mixing times. The physicochemical properties of the resulting CaCO3 hybrid particles could be adjusted by controlling other manufacturing process parameters, including PDA polymerization time, salt addition sequence, and salt-polymer additive pairing. These findings are important for the development of hybrid particle systems with customized properties for specific applications such as contrast-enhanced US and photoacoustic imaging, drug transport, and cancer treatment.

As an extension of the precipitation method, the microemulsion method is widely used in the preparation of CaCO3 NPs and gene encapsulation. The microemulsion method includes water-in-oil (W/O) reverse microemulsion method and double microemulsion method.12,23 The reverse microemulsion uses W/O microemulsion droplets as a nanoreactor.24 First, “calcium microemulsion” and “carbonate microemulsion” are prepared by adding a Ca2+ or CO32– aqueous phase to the organic phase, respectively. Then, the “calcium microemulsion” is mixed with the “carbonate microemulsion” to form CaCO3 NPs. Finally, the CaCO3 NPs are separated by a centrifuge. Khan et al. prepared pH-sensitive lipid coated cisplatin (CDDP)/oleanolic acid (OA) CaCO3 NPs (CDDP/OA-LCC NPs) using a microemulsion method.21 In vitro release experimental results showed that more drugs were released from CDDP/OA-LCC NPs under acidic pH than under alkaline pH, making them an ideal carrier for pH-sensitive chemotherapeutic drugs. In the double microemulsion method, a W/O “calcium microemulsion” is obtained by the inverse microemulsion method, and then a large amount of water phase composed of CO32– is mixed with the “calcium microemulsion”, resulting in the formation of a W/O/W double emulsion. In the W/O/W double emulsion, CaCO3 NPs are formed through the reaction of Ca2+ and CO32–.12

Amorphous calcium carbonate (ACC) filled with small drug molecules is mainly produced by the gas diffusion method.25 As shown in Figure 1, CaCl2 is dissolved in ethanol, and the solution is transferred to a glass vial. Then, two bottles of CaCl2 and ammonium bicarbonate are placed in the desiccator. The ammonium bicarbonate can produce CO2 and NH3. Then CO2 and NH3 can form CO32– and NH4+ when they dissolve in an ethanol solution. Under an alkaline NH4+ environment, CO32– reacts with Ca2+ to generate ACC. Compared with other synthesis methods producing CaCO3 with good crystallization, the advantage of the gas diffusion method is that the obtained CaCO3 NPs can load more pharmaceuticals.26 It is worth mentioning that the size, shape, and phase of CaCO3 NPs produced by these methods can be tuned by changing synthesis factors such as pH, reaction temperature, ionic concentrations and surfactants.2729

2.2. Surface Modification of CaCO3 NPs for Cancer Therapy

Like other biomedical nanomaterials, proper surface modification can make CaCO3 nanomaterials play a better role under physiological conditions, such as preventing agglomeration, increasing stability, targeting, biocompatibility, and so on.34 In the antitumor research, many materials are used to modify the surface of CaCO3 nanomaterials, such as polyethylene glycol (PEG), liposomes, folic acid (FA), lipid, hyaluronic acid (HA), polyethylenimine (PEI) and so on.3538 Macromolecules such as nuclei acids including siRNA,17,39 cancer targeting agents which include peptides and antibodies,40,41 and polymers for example PEG, PEI and HA, have been used in cancer therapy. Recently, Wu et al. have constructed G/A@CaCO3 NPs composed of glucose oxidase (GOX) and 2D antimonene quantum dots (AQDs) encapsulated in CaCO3, which were coated using a constitutive lipid bilayer and additionally PEG to enhance physiological stability.37 In another study, Wang et al. reported that mesoporous silica NPs (MSNs) modified with CaCO3 and a lipid bilayer membrane (MSNs@CaCO3@liposomes) could enable continuous drug release on TME and enhance biocompatibility. The CaCO3 membrane played a guiding role by closing the MSN pore to allow pH-triggered release of the drug to enter into the cancer cell. In addition, a lipid bilayer encapsulated by MSNs@CaCO3 enhanced cellular uptake and biocompatibility.

3. Physicochemical Properties of CaCO3-Based Nanoplatforms

3.1. Diverse Drug Loading Capabilities

The therapeutic effect of antitumor drugs usually relies on high-dose and high-frequency administration, which can lead to serious side effects and toxicity for the organism.42,43 To address this problem, drug carriers that deliver adequate amounts of drug to the site of action and maintain therapeutic levels over the course of treatment are often used.44,45 In addition, drug encapsulation strategies have the potential to improve the delivery of compounds with poorly soluble, poorly stable, and high cytotoxicity under physiological conditions. Nanocarriers have several potential advantages in drug delivery:46,47 (i) biocompatibility without significant toxicity or immune response; (ii) biodegradability in biological environments; (iii) responsiveness to low pH; (iv) easily available at low cost; and (v) stable biochemical properties that do not affect the biological activity of the payload. Due to cost-effectiveness, biosafety, biodegradability, and pH sensitivity,48,49 CaCO3 NPs can carry a variety of drugs that have killing effects on tumors, such as targeted drugs, nontargeted drugs, and genes.50,51 They have good drug loading capacity and can maintain good drug stability.

3.1.1. Loading Targeted Drugs/Genes

CaCO3 NPs can load both hydrophobic and hydrophilic molecules (such as chemotherapy drugs, nucleic acids and other functional molecules), making them suitable carriers for chemotherapy, gene therapy, photodynamic therapy (PDT), etc.15 For example, Feng et al. developed a simple, rapid, and controllable industrial method to synthesize high-drug-loaded vaterite hollow calcium carbonate (VHC).52 In the industrial preparation of CaCO3, VHC can be prepared by using l-leucine. The different reaction conditions and the possible mechanism of VHC formation were studied in detail. The VHC exhibited good biocompatibility and had a high drug loading capacity (39.68%) for doxorubicin (DOX) due to the presence of hollow nanocavities. Meanwhile, DOX@VHC exhibited good pH responsiveness for continuous release of DOX. The cytostatic rate of DOX@VHC was 91.73%, indicating that this drug could significantly inhibit tumor cells and reduce damage to normal cells. The drug-loaded VHC could provide controlled drug release and effective inhibition of tumor cells, so it has good prospects for use in cancer therapy. Gene therapy also often plays a role in tumor treatment by replacing or silencing defective genes. However, nucleic acid delivery is very difficult due to their negatively charged, large size, and prone to degradation.53 In a recent study, CaCO3 NPs could be combined with nucleic acids, which can provide a promising vector for gene therapy. Through activation of the cyclic GMP-AMP synthase-stimulator (cGAS-STING) pathway of interferon genes, cytoplasmic double-stranded DNA (dsDNA) could elicit anticancer immunity.54 In another study, Li et al. reported biomineralized growth of CaCO3 NPs to synthesize cGAS-STING agonists. The obtained DNA@CaCO3 could activate the cGAS-sting pathway in dendritic cells, making it a powerful immunostimulant.55 After intratumoral injection, DNA@CaCO3 effectively suppressed CT26 and B16-F10 tumors in mice by stimulating innate and acquired immunity to reverse the immunosuppressive TME. In addition, through biomineralization of complex CaCO3-based tumor lysates, an individualized autologous tumor vaccine with cGAS-STING activation was created, which could elicit tumor-specific immunity that not only slows down tumor growth but also synergistically suppress postoperative tumor recurrence after surgery with anti-PD-1 immunotherapy. These results demonstrate a CaCO3-based biomineralization approach for the preparation of autologous tumor vaccines in a short period of time, which has the potential for individualized immunotherapy and clinical applications.

Aparting from carrying drugs that cause direct damage to tumor cells, CaCO3 NPs can also carry drug molecules that are responsive to the external environment, thus enabling spatiotemporally selective treatment of tumors.56 For example, in order to combat castration-resistant prostate cancer (CRPC), Tan et al. constructed a TME-triggered nanoprobe (PGP/CaCO3@IR820/DTX-HA).35 The outer layer of CaCO3 effectively encapsulated the chemotherapeutic agent docetaxel (DTX) and photosensitizer IR820 on the surface of pentagonal gold prisms (PGPs), acting as a TME activator to release TME-responsive drugs. The loaded photosensitizer IR820 could exert its killing effect on tumors when activated by external (Near-Infrared Radiation) NIR-light irradiation. After hyaluronic acid modification, PGP/CaCO3@IR820/DTX-HA could synergize TME-activated photothermal therapy (PTT)/PDT/CT and tumor targeting transport. Therefore, PGP/CaCO3 @IR820/DTX-HA has great potential for clinical translation by acting as a treatment for CRPC.

3.1.2. Loading Ultrasmall NPs

In addition to carrying molecules and protein drugs, CaCO3 NPs can also load various ultrasmall NPs to achieve combination therapy. In some cases, the CaCO3 component in the nanocomposites is used as a coating material to prevent the leakage of nanodrugs and protect the activity of the material.35 Several studies have explored CaCO3 nanocomposites loaded with NPs for cancer therapy, including Fe3O4@CaCO3, CaCO3–MnSiOx, Cu2O@CaCO3, and others.5759 For example, a novel nanocatalyst consisting of CaCO3-loaded GOX and two-dimensional antimony quantum dots (AQDs) was constructed and coated by a lipid bilayer (G/A@CaCO3–PEG).37 This nanocatalyst could prolong blood circulation and remain stable under physiologically neutral conditions but decompose under a weakly acidic TME, leading to rapid drug release in the TME. After integration, GOX could effectively catalyze glucose consumption, reducing adenosine triphosphate (ATP) production and subsequently down-regulating the expression of heat shock proteins (HSPs). This effect, combined with the reversal of thermotolerance in cancer cells, enhanced the therapeutic effect of 2D AQDs-induced PTT under near-infrared (NIR) light activation. The results showed that G/A@CaCO3–PEG had high antitumor activity, with an inhibition rate of 83.92%. This strategy provides a promising approach for enhancing hyperthermia-based cancer therapy by limiting the ATP supply.

3.2. Responsive Release Capability

CaCO3 NPs can regulate pharmacokinetics by controlling drug release, thereby improving therapeutic efficacy and reducing side effects.60 Usually, CaCO3 NPs can release drugs through diffusion, carrier dissolution and recrystallization.19 Due to the controlled release of various active substances loaded, CaCO3-based nanocomposites can achieve precise cancer treatments by external stimuli, such as heat, light, and ultrasound, as well as internal stimuli, such as redox, pH, and enzymes.6164

3.2.1. pH Responsiveness

The biological systems have different pH conditions, such as tumor tissues (pH 6.5–6.8) and normal tissues (pH 7.2–7.4).65 pH is an important factor in the decomposition of CaCO3 NPs. The free protons under acidic conditions can react with CaCO3 NPs to produce CO2, which can then dissolve the CaCO3 NPs and accelerate drug release. The pH response of CaCO3 NPs decomposes and releases drugs, which is the earliest and most studied.16 Recently, Ding et al. constructed pH-sensitive CaCO3 nanocarriers loaded with tumor cell lysates (TCL) and CpG immunostimulatory oligodeoxynucleotide 1826 for triple-negative breast cancer (TNBC) immunotherapy.66 This specially designed nanovaccine CaCO3@TCL/CpG could consume lactic acid to neutralize the acidic TME, increasing the M1/M2 ratio and promoting immune cells infiltration. In addition, this nanovaccine could activate tumor dendritic cells and recruit cytotoxic T cells to eliminate tumor cells. In vitro and in vivo experiments demonstrated that this nanovaccine had high cytotoxicity in 4T1 cells and significantly inhibited tumor development in mice. In another study, CRISPR/Cas9 gene-editing nanoplatforms that respond to dual stimulation of pH/glutathione could be synergized with Ca2+-enhanced CO gas for precision cancer treatment.64 In the TME, the rapid biodegradation of the CaCO3 layer induced by pH could promote the production of H2O2 through the GOx-catalyzed oxidation of glucose, and then the generated H2O2 could further react with manganese carbonyl (MnCO) to release CO gas. At the same time, the Ca2+ overload caused by decomposition of CaCO3 disrupted the intramitochondrial calcium balance, leading to Ca2+-induced (reactive oxygen species) ROS production and activation of the mitochondrial pathway of apoptosis. Next, disulfide bonds induced by GSH were cleaved, and released Cas9/sgRNA (RNP) could ablate nuclear factor E2-related factor 2 (Nrf2) gene by interfering with ROS signaling to sensitize gas therapy. This therapeutic modality demonstrates the intelligent control function of CRISPR, ions, and gas codelivery, and provides novel ideas for precise treatment with nanomedicines.

3.2.2. Ultrasound (US)-Triggered Drug Release

US is one of the most powerful noninvasive imaging diagnostic tools available. Because of the similarity of US properties, it is difficult to distinguish between tumors and normal body tissue.20 Therefore, CaCO3 NPs have been used to construct contrast enhancement agents to improve the resolution of US imaging. In ultrasound imaging, the CaCO3-based contrast agent showed strong echo signals and excellent echo persistence in tumor tissue.67 In order to solve the low penetration efficiency of drugs in tumors, Chiang et al. constructed CaCO3-DOX silica NPs by surface modification with fluorination.68 A phospholipid layer (ADSFL NPs) was used to coat the surface of fluorinated CaCO3-DOX silica NPs to attenuate aggregation and enhance the biocompatibility. Compared with the uncoated group, the outer shell of ADSFL NPs could generate interfacial nanobubbles induced by the superhydrophobic layer, which reduced the chance of DOX leakage into water by 7-fold and induced cavitation upon ultrasonic treatment. Furthermore, DOX release from ADSFL NPs could be triggered by the US treatment, which was further enhanced 1.6 times under acidic conditions. In vivo experimental results displayed that US could induce the cavitation activity of ADSFL NPs and cause vessel rupture, with a significant increase in the DOX fluorescence intensity at tumor sites.

3.2.3. Phototriggered Drug Release

Some CBN can also accelerate drug release under the stimulation of external light sources. A common strategy is that CaCO3 nanocomposites are loaded with materials that can produce heat under light irradiation.69 In a recent study, Wang et al. constructed CaCO3-DOX@silica-indocyanine green nanospheres with a high degree of homogeneity and monodispersity, proposing a light-triggered strategy that could be used to treat drug-resistant tumors.70 Under NIR laser irradiation, the nanospheres exhibited photothermal and photodynamic effects by converting laser energy to localized heat and ROS via linked indocyanine green molecules. Furthermore, light can induce drug release from nanospheres, giving the nanospheres chemotherapeutic properties. Eventually, this combination therapy realized the treatment of drug-resistant breast cancer cells. This potentially promising platform for improved multimodal cancer therapy could be provided by these CaCO3-based phototriggered materials.

3.3. Effects on Tumor Microenvironment

Besides drug loading, CBN also plays several important roles in TME modulation, such as providing CO2,58 adjusting pH,71 and serving as a Ca2+ supercarrier,72 which induces immunogenic cell death (ICD) and autophagy thereby activating immunotherapy.

3.3.1. Acid Neutralization

CaCO3, as a biocompatible biomineral, can react with protons in the TME, thereby neutralizing tumor acidity and then enhancing the efficacy of tumor therapy. For instance, Wang et al. designed pH-responsive nanomedicines prepared by coating CaCO3 NPs with 4PI–Zn coordination organic polymers to reprogram the immunosuppressive metabolic TME, thereby enhancing the killing therapeutic effect of radiotherapy (Figure 2).25 The obtained 4PI–Zn@CaCO3 NPs (referred to as acidity-ido 1-modulated nanoparticles, AIM NPs) exhibited excellent pH-dependent dissociation and 4PI release behaviors, showing a role in reversing acidity-induced radiation resistance, respectively. In another study, Zhang et al. synthesized a novel embolic agent CaCO3-coated alginate microspheres (CaCO3-ALG MSs).73 As CaCO3 reacted with protons, this CaCO3-ALG MSs could neutralize the pH of the tumor after CaCO3 decomposition without affecting the whole morphology of the microspheres. Then transcatheter arterial embolization (TAE) treatment was performed using CaCO3-ALG MSs in an orthotropic rat hepatocellular carcinoma model. 18F-fluorodeoxyglucose microposition emission tomography/CT imaging was performed after TAE and intra-arterial administration of CaCO3-ALG MSs was found to significantly improve treatment efficacy. Further studies also confirmed that after treatment of TAE with CaCO3-ALG MS, the reversal of from immunosuppression to nonimmunosuppression of TME occurred. This study not only proposes a novel calcium-based embolic agent for improved TAE therapy in hepatocellular carcinoma but also demonstrates a clinically relevant method to treat tumors through neutralizing tumor acidity. It is important to note that that when CaCO3 nanocomposites are used to change the TME from weakly acidic to neutral, the efficacy of the drug or NPs carried by these nanocomposites is expected to remain unaffected within this neutral setting.

Figure 2.

Figure 2

Acid neutralization by CBN. (a) Scheme illustrating the utilization of AIM NPs to reverse the immunosuppressive metabolic TME for reinforced radiation therapy. Upon tumor accumulation, these intravenously injected AIM NPs can react with protons to neutralize tumor acidity and instantly release the 4PI molecule to inhibit IDO1-mediated tryptophan (Trp) metabolism. (b) Hydrodynamic size distribution profiles of AIM NPs incubated in buffer solutions at different pH values. (c) Time-dependent release profiles of 4PI from AIM NPs incubated in buffer solutions at different pH values. (d) In vitro acidity neutralization profiles of CaCO3, 4PI-Zn, and AIM NPs. Figure reproduced with permission from Wiley-VCH GmbH,25 Copyright 2021.

3.3.2. Calcium Overload

Calcium ions (Ca2+) play an important role in the regulation of many cellular functions. If intracellular Ca2+ levels exceed the amount tolerated by the cell (called Ca2+ overload), oxidative stress, mitochondrial damage and subsequently cell death can occur.74 Thus, Ca2+ overload has been intensively developed as a novel cancer treatment strategy due to its high potency and desirable safety. CBN as Ca2+ overload regulators have acid-responsive properties, which means they can increase intracellular Ca2+ levels and induce cell apoptosis and death by releasing large amounts of Ca2+ in response to the TME.7 However, due to the insufficient presence of Ca2+ at the tumor site, the entry of calcium into the tumor is inefficient, leading to limited therapeutic effects.75 In order to fully utilize the Ca2+ in CaCO3, bioactive substances such as curcumin (CUR) and capsaicin, exogenous physical stimuli such as ultrasound (US), and ROS can be employed to enhance Ca2+ overload.7678 For instance, Zheng et al. used the in situ PDA as a template in a sealing container to prepare a multichannel Ca2+ nanoregulator (CaNMCUR+CDDP) conjugated CaCO3 NPs combined with CUR to induce mitochondrial dysfunction by Ca2+ overload (Figure 3a-c).72 After the whole body was administered, PEG modified CaNMCUR+CDDP efficiently accumulated in tumor sites, entered cancer cells, and induced the mitochondria damage, which significantly enhanced mitochondrial targeting tumor suppression through the outbreak of Ca2+ release, CUR-triggered Ca2+ efflux inhibition, and chemotherapy cisplatin (CDDP). The CUR of fluorescent imaging combined with the PDA of the light sound imaging was conducive to the visualization of nanomodulators. The simple and practical design of this multichannel Ca2+ nanoregulator will help the development of multimode biological imaging guidance in the future.

Figure 3.

Figure 3

Calcium overload cancer therapy based on CBN. (a) Schematic representation of the functional pattern of multifunctional Ca2+ nanomodulator PEGCaNMCUR+CDDP. (b) Mitochondrial Ca2+ concentrations and (c) cell viabilities of MCF-7 cells after treatments with various Ca2+ nanomodulators. Figure reproduced with permission from Wiley-VCH GmbH,72 Copyright 2021. (d) Schematic illustration of M@CaCO3@KAE NP-mediated apoptosis. (e) The calcium contents of A549 cells. (f) Calcium regulated protein expression levels of A549 cells, I: Control; II: CaCO3 NPs; III: KAE; IV: CaCO3@KAE NPs; V: M@CaCO3@KAE NPs. (g) TEM images of mitochondria from A549 cells treated with M@CaCO3@KAE NPs. Figure reproduced with permission from Elsevier BV,79 Copyright 2021.

Furthermore, to enhance Ca2+ overload, promoting Ca2+ influx is also an effective strategy. Yamanol-3-O-Puchantoside (KAE) is a biologically safe flavonoid with excellent anticancer capabilities, capable of disrupting calcium homeostasis and effectively promoting internal calcium flow. Inspired by these concepts, Li et al. constructed KAE-loaded CaCO3 NPs and combined with cell membrane (M) for synergistic tumor therapy (Figure 3d-g).79 In this treatment platform (M@CaCO3@KAE), the membrane coating ensured the targeted delivery of CaCO3@KAE. Upon arrival at the tumor site, CaCO3@KAE accumulated in the TME, releasing KAE and Ca2+. KAE effectively disrupted the Ca2+ balance, while the levels of Ca2+ were significantly increased, and Ca2+ overload was magnified by the mediation of KAE. As a result, mitochondrial dysfunction occurred, leading to cell death in cancer cells. Through these combined effects, in vitro experiments demonstrated that M@CaCO3@KAE achieved tumor suppression. This study presents an alternative nanoplatform acting as a “calcium bomb” to ensure the targeting and effectiveness of tumor therapy.

3.3.3. Generation of CO2 Bubbles

CO2 is generally regarded as a nontoxic, chemically inert substance that is highly soluble in blood and tissues, making it readily absorbed in cases of gas embolism.80 Clinically, CO2 has been widely used as an insufflation gas in laparoscopic surgery for diagnosis and treatment of intra-abdominal and gynecological diseases.81,82 The extracellular pH of tumors is known to be lower than that of normal tissues, making them potential targets for therapy. Carbonic acid, exciting metal extraction, and absorption can be substituted and confirmed by a carbon regime. The generated CO2 can promote drug release, aid in US imaging, rupture cell membranes, and act as a drug carrier, among other functions. The CO2 produced by the decomposition of CaCO3 can further promote the release of drugs within the CaCO3. Leveraging this unique property, a CaCO3-based nanosystem was proposed that can generate CO2 gas and promote the release of encapsulated DOX at the tumor site.67

Due to the acoustic properties of CO2 bubbles, they can serve as a good ultrasound contrast agent, providing a novel diagnostic method for tumor treatment.83 For instance, to implement photoacoustic (PA), ultrasound, and thermal imaging guided PTT, Wan et al. prepared biomimetic pH-responsive nanoprobes using tumor cell membrane-modified polydopamine (PDA)-CaCO3 NPs (CPCaNPs).71 Once the CPCaNPs targeted and penetrated deep into the acidic TME, the CO2 bubbles generated by the decomposition of CPCaNPs significantly enhanced the US signal. Additionally, the PDA of CPCaNPs could generate PA signals and perform effective PA therapy in primary tumors. Furthermore, when these nanoprobes were combined with an immune checkpoint pathway blockade, tumor recurrence and migration were effectively inhibited. However, the endocytosis of lysosomes, can lead to the drug being phagocytized and cleared before reaching the designated location, reducing drug delivery efficiency.84 To address this issue, catalase-driven nanomotors were constructed,39 with high cellular uptake and lysosomal escape. This system provided a specific mechanism for preventing the proliferation of tumor cells and inducing cell death. The nanomotors were based on the self-determination technology of the CaCO3, PEI, and catalase coat layer. The effect of the PEI proton sponge and the CO2 produced by the CaCO3 breakthrough helped control the nanometer from the lysosome to the cytoplasm, achieving prolonged retention. Additionally, it was demonstrated that nanomotors could strongly influence the properties of tubulin through paclitaxel and siRNA, leading to programmed cell death in 4T1 tumor cells. Therefore, this system can be used as a safe and vital system of combining drugs for the active treatment of breast cancer. In another work, Janus CaCO3 micromotors (JCPMs) driven by NIR light/gas were demonstrated to overcome multiple physiological barriers for efficient drug delivery.85 The JCPMs had thermophoretic movements driven by NIR light. They featured precisely controlled start/stop switches, speeds, and motion trajectories in physiological media and autonomous propulsion driven by CO2 bubbles in acidic environments (Figure 4). Under the conditions caused by the axis, the JCPMs showed multilevel gas and drug release in cells. This strategy could ensure the active substance to practice the therapeutic effects selectively on tumors and avoid side effects. The micromotor cascade-driven model designed in this article to traverse biological barriers will improve the delivery efficiency of tumor treatment.

Figure 4.

Figure 4

Generation of CO2 bubbles from CBN. Schematic illustration of the synthesis of the Janus CaCO3 micromotors (JCPMs) and the light/gas cascade-propelled JCPMs to overcome sequential and multistaged biological barriers for precise doxorubicin delivery and efficient tumor cells’ killing. Figure reproduced with permission from Elsevier BV,85 Copyright 2021.

4. Multimodal Cancer Therapy

It is often challenging to completely eradicate tumors using a single method, and employing multiple modes of therapy can effectively destroy the tumor while reducing side effects.86,87 CBN can be used for multimodal cancer therapy (Table 2), including chemodynamic therapy (CDT), RT, PTT, PDT, sonodynamic therapy (SDT), chemotherapy, immunotherapy, starvation therapy, and gas therapy.14,51,71 Due to their excellent drug-loading capability, CBN are well-suited for multimode cancer treatment. External stimulation can activate cell stress reactions (CSR), which counteract the antitumor effect. To address this issue, Liu et al. proposed a cell stress-responsive strategy and developed GOX loaded Cu3BiS3 nanosheets (CBSG NSS) wrapped in CaCO3 (CBSG@ CaCO3) as a new type of nanoagent to achieve effective combination treatment of starvation therapy and CDT (Figure 5a).88 The CBSG@CaCO3 not only induces external stimuli, such as energy consumption and oxidation stimulation, but also disrupts the CSR mechanism. After the nanoagent enters cancer cells, the outer layer of CaCO3 breaks down and releases CBSG NS, which possesses intrinsic photothermal properties, accelerating external irritation under NIR-II laser irradiation. Meanwhile, CaCO3 can block CSR by suppressing the expression of P27 and NRF2, thereby destroying the adaptive survivability of cancer cells. Moreover, a multimodal Ca2+ nanoregulator has been developed for the first time, which combines PTT with a mitochondrial Ca2+ overload strategy to inhibit tumor development.89 The synergistic Ca2+ nanoregulator SA/Cur@CaCO3–ICG(SCCI), was prepared by cross-linking sodium alginate (SA) onto the surface of CaCO3 NPs coated with conductive adhesive. SCCI has been shown to enhance photostability, produce a large amount of Ca2+ at low pH, decrease mitochondrial membrane potential, and downregulate ATP production. This SCCI has demonstrated good targeting, biocompatibility, and antitumor effects, significantly inhibiting tumor cell proliferation and producing a direct killing effect. This therapeutic strategy, based on ion interference and PTT, holds great therapeutic potential and provides a new perspective for antitumor therapy.

Table 2. Summary of Typical CaCO3-Based Nanoplatforms (CBN) for Multimodal Cancer Therapy.

Nanoformulation Cancer type Strategy ref
CaCO3@CoP 4T1 breast cancer cells The synergy of CDT and calcium overload to maximize oxidative damage and enhance cancer therapy (92)
CaCO3/Cu/Pt (oxali Platin) 4T1 breast cancer cells Neutralize TME; oxaliplatin is used for chemotherapy; Cu2+ is used for CDT; the oxaliplatin and produced ROS could also induce the ICD of cancer cells (93)
CDDP/OA-LCC Hepatocellular carcinoma Co-delivery of CDDP and OA via a pH-sensitive CaCO3 drug delivery system for enhanced tumor efficacy and reduced adverse effects (24)
CPCa 4T1 breast cancer cells Available for tumor targeting and imaging-guided PTT with synergistic immunotherapy (71)
CaCO3/PDA-SA Melanoma Enhanced melanoma treatment by the synergistic effect of PTT and Ca2+ interference therapy (20)
LST-IR820-Ca Triple negative breast cancer Synergistic PTT/PDT therapy and exploitation of LST to reduce solid stress of the tumor through depletion of collagen I (94)
CuS@Axitinib-SiO2@2-Deoxy-d-glucose(2-DG)-CaCO3-RGD 4T1 breast cancer cells Achieve severe tumor starvation and further enhance tumor treatment with the aid of PTT, CDT, and TME improvement (95)
CaCO3-GEM-Triapine Pancreatic cancer Inhibition of cancer cell proliferation, migration and resistance to GEM using 2D PANC-1/GEM cells and 3D tumor spheroids enhanced the therapeutic efficacy of GEM-based chemotherapy. (96)
G/A@CaCO3-PEG Pancreatic cancer Increase heat-based tumor therapy by limiting ATP availability (37)
IrCOOH-CaCO3@PEG 4T1 breast cancer cells The combination of calcium overload and Ir complex irradiation with the generation of ROS leads to tumor death (78)
CaNPs@Gel-MS Hepatocellular carcinoma Reverses lactate-induced chemotherapy resistance during TACE treatment (97)
CaCO3@TCL/CpG Triple negative breast cancer Neutralize TME and promote immunotherapy (66)
O2-FeCOF@CaCO3@FA 4T1 breast cancer cells Establishment of a mitochondrial pathway that inhibits tumor growth and synergizes with PDA using dual-activated MPTP channels (98)
PGP/CaCO3@IR820/DTX-HA Castration-resistant prostate cancer TME-activated synergistic PTT/PDT/chemical therapy strategy (35)
IL-12 mRNA@cRGD-CM-CaCO3 Glioblastoma multiforme Cross the blood-brain barrier and cause necrotic prolapse-induced immune responses and IL-12 mRNA transfection by ultrasound irradiation (62)
CaCO3@Cur@QTX125@HA Colorectal cancer Specific inhibitory effects on colorectal cancer growth (78,99)

Figure 5.

Figure 5

Multimodal cancer therapy based on CBN. (a) The illustration of the synthesis of degradable CBSG@CaCO3 NPs and intracellular therapeutic mechanisms of improving drug-free cancer therapy by interdicting the cellular stress response (CSR) or not. Figure reproduced with permission from Wiley-VCH GmbH,88 Copyright 2023. (b) Schematic illustration of Cu/CaCO3@Ce6 NPs-dominated advanced cancer therapy via multiple ROS amplification. (c) Schematic illustration of the synthesis of PEG-decorated Cu/CaCO3@Ce6 NPs. (d) Relative intracellular ATP levels of 4T1 with different treatments. (e) Tumor volume changes of various groups with different treatments. Figure reproduced with permission from Wiley-VCH Gmb,90 Copyright 2022.

The regulation of ROS in tumor therapy has been affected by the inherent characteristics of the TME, such as GSH excessive expression, hypoxia, and limited H2O2 efficiency. In order to utilize ROS to promote tumor treatment, Zhao et al. have established intelligent copper-dropped CaCO3-loading acoustic agents Ce6 NPs (Cu/CaCO3@Ce6, CCC NPs), and realized the combination treatment of calcium overload enhanced CDT and SDT (Figure 5b-e).90 In the TME with weak acidity and high GSH, CCC NPs released Ca2+, Cu2+ and Ce6. The release of Cu2+ could not only consume GSH and convert it to Cu+ through oxidation reactions but also provide hydroxyl radicals (·OH) that could generate CDT by a Fenton-like reaction. Under ultrasound treatment, the oxidation inside the cell could be greatly enlarged by the outbreak of singlet oxygen from SDT. In addition, Ca2+ internal flow exacerbated the destruction of mitochondria, which further accelerated the oxidation level. The simple and feasible design of the nanoregulator based on CaCO3 will be further developed as examples of ROS-regulated cancer treatment.

Additionally, pyroptosis offers new prospects for cancer immunotherapy. Novel pyroptosis inducers based on CBN have already been developed for cancer immunotherapy. Zheng et al. designed biodegradable Ca2+ nanomodulators (CaNMs) as Pyroptosis inducers for cancer immunotherapy via mitochondrial Ca2+ overload.91 The obtained CaNMs can decompose and release Ca2+ and curcumin at low pH, causing a sudden surge of mitochondrial Ca2+ ions and ultimately leading to Pyroptosis. This work confirmed for the first time the occurrence of Pyroptosis triggered by mitochondrial Ca2+ overload and also demonstrated a strong immune response generated by CaNMs, as well as significant inhibition of tumor proliferation and lung metastasis. This work will provide new inspiration for tumor immunotherapy based on Ca2+ nanomodulators.

5. Conclusions, Challenges and Future Breakthroughs

CBN is a type of novel nanomaterial and has been widely used in the biomedical field. In recent years, the application of CBN in antitumor research has been widely investigated. However, there are still some problems with their actual antitumor applications mainly in the following aspects.

5.1. Design and Fabrication

Numerous approaches exist for synthesizing CBN, yet their efficacy can be influenced by various elements, including the temperature and duration of the reaction, the presence of surfactants, the type of equipment used, and the specific process parameters, which can restrict their potential applications in cancer therapy.100,101 If these reaction conditions are not properly controlled, then the morphology and stability of NPs will be affected. For example, the most used gas diffusion method to prepare CBN still has some shortcomings. Due to the slow reaction speed of the gas diffusion method, and the need for fine control of the reaction conditions, its production efficiency is low and the yield is relatively limited.102 When the gas diffusion method is used to prepare CBN, the particle size distribution is often uneven, which leads to an unstable quality of the product. This method requires the use of many organic solvents in the preparation process, which not only increases the cost but also causes pollution to the environment. Thus, in the future, it is still necessary to design accurate methods to control the size, composition, and surface modification to greenly synthesize large quantities of CBN.

5.2. Therapeutic Mechanism

The therapeutic effect of CBN on a tumor can be affected by many factors. For example, the particle size, surface properties, concentration, and other factors of CBN will affect its antitumor effect. The process and mechanism of the antitumor effect of CBN still need to be explored. It is necessary to study the pharmacokinetics and pharmacodynamics of CBN to understand its transport processes in vivo. For example, the transport of CBN to target organs and tissues needs to be studied. First, to break through the dynamic barrier, nanomaterials need to be stable and circulate in the bloodstream for a long time in the presence of multiple enzymes, proteins, and immune cells. Although PEGylating technology can effectively extend the cycle time to a certain extent, it still cannot effectively reduce the uptake of nanocarriers by reticuloendothelial systems.88,103 Second, CBN is depolymerized or degraded at desired action sites such as weakly acidic TME or within endosomes and lysosomes. The NPs taken up by cells are required to disintegrate and release their cargo to achieve good therapeutic effect outside or inside the cell. Thus, ideal CBN should be able to load enough biologics and release them at the target site in a sustained manner. Finally, the type and stage of the tumor will also affect the therapeutic effect of CBN. Therefore, it is necessary to further study the mechanism of action of CBN to understand how it acts on tumor cells to optimize treatment.

5.3. Biocompatibility/Biosafety

The biosafety of CBN has not been fully verified.104,105 Although some studies have shown that CBN has an effective killing effect on tumor cells, its effect on normal cells still needs further study. Although calcium is an essential element for the human body, excess calcium can lead to blood clots, hypercalcemia, and other potential dangers.106 In addition, the metabolism and excretion pathways of CBN in the body are unclear and may cause potential biological risks.107 It is still necessary to study the characteristics of absorption, distribution, biotransformation, and excretion of CBN in vivo, as well as its influencing factors and mechanism. Recent studies have assessed short-term toxicity in mice only by demonstrating immune response and organ injury after injection of CBN, which is clearly insufficient for biosafety assessment.108 For the clinical transformation of CBN, it is worth systematically studying the long-term effects of CBN using multiple models.

In summary, there are still some challenges to be overcome in CBN antitumor applications. In future studies, more attention should be paid to the biosafety, metabolic pathways, and factors affecting the therapeutic effect of CBN to provide a more reliable basis for designing CBN for tumor treatment.

Acknowledgments

The authors acknowledge financial support from National Key Research and Development Program of China (2022YFA1207600), Natural Science Foundation of Ningxia Province of China (2023AAC02039) and Key Research and Development Program of Ningxia Province of China (2023BEG02031)

Author Contributions

# (X.Z., Q.W.) These authors contributed equally to this work.

The authors declare no competing financial interest.

References

  1. Aghebati-Maleki A.; Dolati S.; Ahmadi M.; Baghbanzhadeh A.; Asadi M.; Fotouhi A.; Yousefi M.; Aghebati-Maleki L. Nanoparticles and cancer therapy: Perspectives for application of nanoparticles in the treatment of cancers. J. Cell. Physiol. 2020, 235 (3), 1962–1972. 10.1002/jcp.29126. [DOI] [PubMed] [Google Scholar]
  2. Gavas S.; Quazi S.; Karpinski T. M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16 (1), 173. 10.1186/s11671-021-03628-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Huang H.-C.; Barua S.; Sharma G.; Dey S. K.; Rege K. Inorganic nanoparticles for cancer imaging and therapy. J. Controlled Release 2011, 155 (3), 344–357. 10.1016/j.jconrel.2011.06.004. [DOI] [PubMed] [Google Scholar]
  4. Pugazhendhi A.; Edison T. N. J. I.; Karuppusamy I.; Kathirvel B. Inorganic nanoparticles: A potential cancer therapy for human welfare. Int. J. Pharmaceut. 2018, 539 (1–2), 104–111. 10.1016/j.ijpharm.2018.01.034. [DOI] [PubMed] [Google Scholar]
  5. Bai S.; Lan Y.; Fu S.; Cheng H.; Lu Z.; Liu G. Connecting Calcium-Based Nanomaterials and Cancer: From Diagnosis to Therapy. Nano-Micro Lett. 2022, 14 (1), 145. 10.1007/s40820-022-00894-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Sun Z.; Li W.; Lenzo J. C.; Holden J. A.; McCullough M. J.; O’Connor A. J.; O’Brien-Simpson N. M. The Potential of Calcium Phosphate Nanoparticles as Adjuvants and Vaccine Delivery Vehicles. Front. Mater. 2021, 8, 788373 10.3389/fmats.2021.788373. [DOI] [Google Scholar]
  7. Yao J.; Peng H.; Qiu Y.; Li S.; Xu X.; Wu A.; Yang F. Nanoplatform-mediated calcium overload for cancer therapy. J. Mater. Chem. B 2022, 10 (10), 1508–1519. 10.1039/D1TB02721B. [DOI] [PubMed] [Google Scholar]
  8. Wu X.; Han X.; Guo Y.; Liu Q.; Sun R.; Wen Z.; Dai C. Application prospect of calcium peroxide nanoparticles in biomedical field. Rev. Adv. Mater. Sci. 2023, 62 (1), 20220308 10.1515/rams-2022-0308. [DOI] [Google Scholar]
  9. Xiao Y.; Li Z.; Bianco A.; Ma B. Recent Advances in Calcium-Based Anticancer Nanomaterials Exploiting Calcium Overload to Trigger Cell Apoptosis. Adv. Funct. Mater. 2023, 33, 2209291 10.1002/adfm.202209291. [DOI] [Google Scholar]
  10. Fadia P.; Tyagi S.; Bhagat S.; Nair A.; Panchal P.; Dave H.; Dang S.; Singh S. Calcium carbonate nano- and microparticles: synthesis methods and biological applications. 3 Biotech 2021, 11 (11), 457. 10.1007/s13205-021-02995-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Dizaj S. M.; Sharifi S.; Ahmadian E.; Eftekhari A.; Adibkia K.; Lotfipour F. An update on calcium carbonate nanoparticles as cancer drug/gene delivery system. Expert Opin. Drug Del. 2019, 16 (4), 331–345. 10.1080/17425247.2019.1587408. [DOI] [PubMed] [Google Scholar]
  12. Dizaj S. M.; Barzegar-Jalali M.; Zarrintan M. H.; Adibkia K.; Lotfipour F. Calcium carbonate nanoparticles as cancer drug delivery system. Expert Opin. Drug Del. 2015, 12 (10), 1649–1660. 10.1517/17425247.2015.1049530. [DOI] [PubMed] [Google Scholar]
  13. Ibiyeye K. M.; Idris S. B.; Zuki A. B. Z. Cockle shell-derived aragonite calcium carbonate nanoparticle for targeting cancer and breast cancer stem cells. Cancer Nanotechnol. 2020, 11 (1), 10. 10.1186/s12645-020-00067-3. [DOI] [Google Scholar]
  14. Liang T. T.; Feng Z. Q.; Zhang X.; Li T. F.; Yang T. Y.; Yu L. Research progress of calcium carbonate nanomaterials in cancer therapy: challenge and opportunity. Front. Bioeng. Biotechnol. 2023, 11, 1266888 10.3389/fbioe.2023.1266888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Trushina D. B.; Borodina T. N.; Belyakov S.; Antipina M. N. Calcium carbonate vaterite particles for drug delivery: Advances and challenges. Mater. Today Adv. 2022, 14, 100214 10.1016/j.mtadv.2022.100214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Kang Y.; Xu L.; Dong J.; Huang Y.; Yuan X.; Li R.; Chen L.; Wang Z.; Ji X. Calcium-based nanotechnology for cancer therapy. Coordin. Chem. Rev. 2023, 481, 215050 10.1016/j.ccr.2023.215050. [DOI] [Google Scholar]
  17. Levingstone T. J.; Herbaj S.; Dunne N. J. Calcium Phosphate Nanoparticles for Therapeutic Applications in Bone Regeneration. Nanomaterials 2019, 9 (11), 1570. 10.3390/nano9111570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Huang H.; Zhang W.; Liu Z.; Guo H.; Zhang P. Smart responsive-calcium carbonate nanoparticles for dual-model cancer imaging and treatment. Ultrasonics 2020, 108, 106198 10.1016/j.ultras.2020.106198. [DOI] [PubMed] [Google Scholar]
  19. Zhao P.; Tian Y.; You J.; Hu X.; Liu Y. Recent advances of calcium carbonate nanoparticles for biomedical applications. Bioengineering 2022, 9 (11), 691. 10.3390/bioengineering9110691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Vidallon M. L. P.; Douek A. M.; Quek A.; McLiesh H.; Kaslin J.; Tabor R. F.; Bishop A. I.; Teo B. M. Gas-Generating, pH-Responsive Calcium Carbonate Hybrid Particles with Biomimetic Coating for Contrast-Enhanced Ultrasound Imaging. Part. Part. Syst. Charact. 2020, 37 (2), 1900471 10.1002/ppsc.201900471. [DOI] [Google Scholar]
  21. Khan M. W.; Zhao P.; Khan A.; Raza F.; Raza S. M.; Sarfraz M.; Chen Y.; Li M.; Yang T.; Ma X.; Xiang G. Synergism of cisplatin-oleanolic acid co-loaded calcium carbonate nanoparticles on hepatocellular carcinoma cells for enhanced apoptosis and reduced hepatotoxicity. Int. J. Nanomed. 2019, 14, 3753–3771. 10.2147/IJN.S196651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Vidallon M. L. P.; Yu F.; Teo B. M. Controlling the Size and Polymorphism of Calcium Carbonate Hybrid Particles Using Natural Biopolymers. Cryst. Growth Des. 2020, 20 (2), 645–652. 10.1021/acs.cgd.9b01057. [DOI] [Google Scholar]
  23. Zhou F.; Li H.; Liu Y.; Deng H.; Rong J.; Zhao J. Hyaluronan derivative decorated calcium carbonate nanoparticle as a potential platform for breast cancer synergistic therapy via blood coagulation and drug delivery. J. Drug Delivery Sci. Tec. 2023, 83, 104406 10.1016/j.jddst.2023.104406. [DOI] [Google Scholar]
  24. Khan M. W.; Zou C.; Hassan S.; Din F. U.; Razak M. Y. A.; Nawaz A.; Zeb A.; Wahab A.; Bangash S. A. Cisplatin and oleanolic acid Co-loaded pH-sensitive CaCO3 nanoparticles for synergistic chemotherapy. RSC Adv. 2022, 12 (23), 14808–14818. 10.1039/D2RA00742H. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Wang C.; Dong Z.; Hao Y.; Zhu Y.; Ni J.; Li Q.; Liu B.; Han Y.; Yang Z.; Wan J.; Yang K.; Liu Z.; Feng L. Coordination Polymer-Coated CaCO3 Reinforces Radiotherapy by Reprogramming the Immunosuppressive Metabolic Microenvironment. Adv. Mater. 2022, 34 (3), 2106520 10.1002/adma.202106520. [DOI] [PubMed] [Google Scholar]
  26. Zhao Y.; Luo Z.; Li M.; Qu Q.; Ma X.; Yu S. H.; Zhao Y. A preloaded amorphous calcium carbonate/doxorubicin@ silica nanoreactor for ph-responsive delivery of an anticancer drug. Angew. Chem., Int. Ed. 2015, 54 (3), 919–922. 10.1002/anie.201408510. [DOI] [PubMed] [Google Scholar]
  27. Achour A.; Arman A.; Islam M.; Zavarian A.; Basim Al-Zubaidi A.; Szade J. Synthesis and characterization of porous CaCO3 micro/nano-particles. Eur. Phys. J. Plus 2017, 132 (6), 267. 10.1140/epjp/i2017-11531-8. [DOI] [Google Scholar]
  28. Sun B. C.; Wang X. M.; Chen J. M.; Chu G. W.; Chen J. F.; Shao L. Synthesis of nano-CaCO3 by simultaneous absorption of CO2 and NH3 into CaCl2 solution in a rotating packed bed. Chem. Eng. J. 2011, 168 (2), 731–736. 10.1016/j.cej.2011.01.068. [DOI] [Google Scholar]
  29. Cheng J.; Sun W.; Zhang Z.; Xie M.; Zhao H.; Zeng D.; Lin X. Tetrahedral DNA–mediated biomineralization of calcium carbonate nanoparticles for pH-responsive drug delivery. J. Nanopart. Res. 2023, 25 (10), 211. 10.1007/s11051-023-05858-4. [DOI] [Google Scholar]
  30. Sepulveda F.; Butto N.; Arias J. L.; Yazdani-Pedram M.; Szewczyk P. K.; Gruszczynski A.; Stachewicz U.; Neira-Carrillo A. Effect of Porous and Nonporous Polycaprolactone Fiber Meshes on CaCO3 Crystallization Through a Gas Diffusion Method. Cryst. Growth Des. 2020, 20 (8), 5610–5625. 10.1021/acs.cgd.0c00803. [DOI] [Google Scholar]
  31. Zhang M.; Li H.; Wang C.; Wang Z.; Liu D.; Yang T.; Deng Z.; Yuan G. Performance Enhancement of the Poplar Wood Composites Biomimetic Mineralized by CaCO3. ACS Omega 2022, 7 (33), 29465–29474. 10.1021/acsomega.2c03960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Muhammad Mailafiya M.; Abubakar K.; Danmaigoro A.; Musa Chiroma S.; Bin Abdul Rahim E.; Aris Mohd Moklas M.; Abu Bakar Zakaria Z. Cockle Shell-Derived Calcium Carbonate (Aragonite) Nanoparticles: A Dynamite to Nanomedicine. Appl. Sci. 2019, 9 (14), 2897. 10.3390/app9142897. [DOI] [Google Scholar]
  33. Zheng T.; Hou D.; Leng W.; Li P.; Wei W. Preparation, characterization, and formation mechanism of different biological calcium carbonate (CaCO3) induced by Bacillus mucilaginosus and Bacillus alcalophilus. J. Nanopart. Res. 2023, 25 (9), 189. 10.1007/s11051-023-05833-z. [DOI] [Google Scholar]
  34. Memar M. Y.; Adibkia K.; Farajnia S.; Kafil H. S.; Dizaj S. M.; Ghotaslou R. Biocompatibility, cytotoxicity and antimicrobial effects of gentamicin-loaded CaCO3 as a drug delivery to osteomyelitis. J. Drug Delivery Sci. Tec. 2019, 54, 101307 10.1016/j.jddst.2019.101307. [DOI] [Google Scholar]
  35. Tan H.; Liu Y.; Hou N.; Cui S.; Liu B.; Fan S.; Yu G.; Han C.; Zheng D.; Li W.; Liu Y.; Xu B.; Wang Z.; Cui D. Tumor microenvironment pH-responsive pentagonal gold prism-based nanoplatform for multimodal imaging and combined therapy of castration-resistant prostate cancer. Acta Biomater. 2022, 141, 408–417. 10.1016/j.actbio.2022.01.012. [DOI] [PubMed] [Google Scholar]
  36. Luo W.; Li Z.; Zhang L.; Xie X. Polyethylenimine-CO2 adduct templated CaCO3 nanoparticles as anticancer drug carrier. Cancer Nanotechnol. 2023, 14 (1), 7. 10.1186/s12645-023-00156-z. [DOI] [Google Scholar]
  37. Wu J.; Cai X.; Williams G. R.; Meng Z.; Zou W.; Yao L.; Hu B.; Chen Y.; Zheng Y. 2D antimonene-integrated composite nanomedicine for augmented low-temperature photonic tumor hyperthermia by reversing cell thermoresistance. Bioact. Mater. 2022, 10, 295–305. 10.1016/j.bioactmat.2021.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Wang Y.; Zhao K.; Xie L.; Li K.; Zhang W.; Xi Z.; Wang X.; Xia M.; Xu L. Construction of calcium carbonate-liposome dual-film coated mesoporous silica as a delayed drug release system for antitumor therapy. Colloid. Surface. B 2022, 212, 112357 10.1016/j.colsurfb.2022.112357. [DOI] [PubMed] [Google Scholar]
  39. Ren J.; Hu P.; Ma E.; Zhou X.; Wang W.; Zheng S.; Wang H. Enzyme-powered nanomotors with enhanced cell uptake and lysosomal escape for combined therapy of cancer. Appl. Mater. Today 2022, 27, 101445 10.1016/j.apmt.2022.101445. [DOI] [Google Scholar]
  40. Xie B.; Zhao H.; Ding Y. F.; Wang Z.; Wang Y.; Gao C.; Wang R. Drug-free tumor therapy via spermine-responsive intracellular biomineralization. J. Controlled Release 2023, 357, 572–579. 10.1016/j.jconrel.2023.04.018. [DOI] [PubMed] [Google Scholar]
  41. Su R.; Gu J.; Sun J.; Zang J.; Zhao Y.; Zhang T.; Chen Y.; Chong G.; Yin W.; Zheng X.; Liu B.; Huang L.; Ruan S.; Dong H.; Li Y.; Li Y. CaCO3 powder-mediated biomineralization of antigen nanosponges synergize with PD-1 blockade to potentiate anti-tumor immunity. J. Nanobiotechnol. 2023, 21 (1), 120. 10.1186/s12951-023-01870-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Barnes T. A.; Amir E.; Templeton A. J.; Gomez-Garcia S.; Navarro B.; Seruga B.; Ocana A. Efficacy, safety, tolerability and price of newly approved drugs in solid tumors. Cancer Treat Rev. 2017, 56, 1–7. 10.1016/j.ctrv.2017.03.011. [DOI] [PubMed] [Google Scholar]
  43. Florea A.-M.; Büsselberg D. Cisplatin as an anti-tumor drug: cellular mechanisms of activity, drug resistance and induced side effects. Cancers 2011, 3 (1), 1351–1371. 10.3390/cancers3011351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Prakash S.; Malhotra M.; Shao W.; Tomaro-Duchesneau C.; Abbasi S. Polymeric nanohybrids and functionalized carbon nanotubes as drug delivery carriers for cancer therapy. Adv. Drug. Deliver. Rev. 2011, 63 (14–15), 1340–1351. 10.1016/j.addr.2011.06.013. [DOI] [PubMed] [Google Scholar]
  45. Lohcharoenkal W.; Wang L.; Chen Y. C.; Rojanasakul Y. Protein Nanoparticles as Drug Delivery Carriers for Cancer Therapy. Biomed Res. Int. 2014, 2014, 180549 10.1155/2014/180549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Mura S.; Nicolas J.; Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12 (11), 991–1003. 10.1038/nmat3776. [DOI] [PubMed] [Google Scholar]
  47. Perez-Herrero E.; Fernandez-Medarde A. Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur. J. Pharm. Biopharm. 2015, 93, 52–79. 10.1016/j.ejpb.2015.03.018. [DOI] [PubMed] [Google Scholar]
  48. Liu Y.; Ma X.; Zhu Y.; Lv X.; Wang P.; Feng L. pH-responsive nanomedicine co-encapsulated with Erlotinib and chlorin e6 can enable effective treatment of triple negative breast cancer via reprogramming tumor vasculature. Chem. Eng. J. 2022, 437, 135305 10.1016/j.cej.2022.135305. [DOI] [Google Scholar]
  49. Zhao X.; Wan X.; Huang T.; Yao S.; Wang S.; Ding Y.; Zhao Y.; Li Z.; Li L. Acidity-responsive nanocages as robust reactive oxygen species generators with butterfly effects for maximizing oxidative damage and enhancing cancer therapy. J. Colloid Interface Sci. 2022, 618, 270–282. 10.1016/j.jcis.2022.03.084. [DOI] [PubMed] [Google Scholar]
  50. Yin S. Y.; Hu Y.; Zheng J.; Li J.; Yang R. Tannic Acid-Assisted Biomineralization Strategy for Encapsulation and Intracellular Delivery of Protein Drugs. ACS Appl. Mater. Interfaces 2022, 14 (45), 50583–50591. 10.1021/acsami.2c15205. [DOI] [PubMed] [Google Scholar]
  51. Poudel K.; Nam K. S.; Lim J.; Ku S. K.; Hwang J.; Kim J. O.; Byeon J. H. Modified Aerotaxy for the Plug-in Manufacture of Cell-Penetrating Fenton Nanoagents for Reinforcing Chemodynamic Cancer Therapy. ACS Nano 2022, 16 (11), 19423–19438. 10.1021/acsnano.2c09136. [DOI] [PubMed] [Google Scholar]
  52. Feng Z.; Yang T.; Dong S.; Wu T.; Jin W.; Wu Z.; Wang B.; Liang T.; Cao L.; Yu L. Industrially synthesized biosafe vaterite hollow CaCO3 for controllable delivery of anticancer drugs. Mater. Today Chem. 2022, 24, 100917 10.1016/j.mtchem.2022.100917. [DOI] [Google Scholar]
  53. Nguyen J.; Szoka F. C. Nucleic acid delivery: the missing pieces of the puzzle?. Acc. Chem. Res. 2012, 45 (7), 1153–1162. 10.1021/ar3000162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Ablasser A.; Chen Z. J. cGAS in action: Expanding roles in immunity and inflammation. Science 2019, 363 (6431), eaat8657 10.1126/science.aat8657. [DOI] [PubMed] [Google Scholar]
  55. Li Q.; Dong Z.; Cao Z.; Lei H.; Wang C.; Hao Y.; Feng L.; Liu Z. A General Biomineralization Strategy to Synthesize Autologous Cancer Vaccines with cGAS-STING Activating Capacity for Postsurgical Immunotherapy. ACS Nano 2023, 17 (11), 10496–10510. 10.1021/acsnano.3c01404. [DOI] [PubMed] [Google Scholar]
  56. Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020, 10 (10), 4557–4588. 10.7150/thno.38069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Popova V.; Poletaeva Y.; Chubarov A.; Dmitrienko E. pH-Responsible Doxorubicin-Loaded Fe3O4@CaCO3 Nanocomposites for Cancer Treatment. Pharmaceuticals 2023, 15 (3), 771. 10.3390/pharmaceutics15030771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Xie C.; Zhang T.; Fu Y.; Han G.; Li X. CaCO3-MnSiOx hybrid particles to enable CO2-mediated combinational tumor therapy. Nano Res. 2022, 15 (9), 8281–8290. 10.1007/s12274-022-4471-7. [DOI] [Google Scholar]
  59. Ma C.; Zhao C.; Hong C.; Zhang L.; Chen S.; Qi Y. Temperature Platform Constructed Using a Cu2O-Cu31S16 Photothermal Conversion System for the Simple Quantitative Analysis of Tumor Markers. ACS Sustain. Chem. Eng. 2022, 10, 8517–8525. 10.1021/acssuschemeng.2c01760. [DOI] [Google Scholar]
  60. Dong Z.; Feng L.; Zhu W.; Sun X.; Gao M.; Zhao H.; Chao Y.; Liu Z. CaCO3 nanoparticles as an ultra-sensitive tumor-pH-responsive nanoplatform enabling real-time drug release monitoring and cancer combination therapy. Biomaterials 2016, 110, 60–70. 10.1016/j.biomaterials.2016.09.025. [DOI] [PubMed] [Google Scholar]
  61. Zhong W.; Wong K. H.; Xu F.; Zhao N.; Chen M. NIR-responsive polydopamine-based calcium carbonate hybrid nanoparticles delivering artesunate for cancer chemo-photothermal therapy. Acta Biomater. 2022, 145, 135–145. 10.1016/j.actbio.2022.03.051. [DOI] [PubMed] [Google Scholar]
  62. Zhao P.; Tian Y.; Lu Y.; Zhang J.; Tao A.; Xiang G.; Liu Y. Biomimetic calcium carbonate nanoparticles delivered IL-12 mRNA for targeted glioblastoma sono-immunotherapy by ultrasound-induced necroptosis. J. Nanobiotechnol. 2022, 20 (1), 525. 10.1186/s12951-022-01731-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Li X.; Gao Y.; Liu X.; Hu X.; Li Y.; Sun J.; Wang P.; Wu H.; Kim H.; Ramalingam M.; Xie S.; Wang R. Ultrasound and laser-promoted dual-gas nano-generator for combined photothermal and immune tumor therapy. Front. Bioeng. Biotechnol. 2022, 10, 1005520 10.3389/fbioe.2022.1005520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Li Y.; Pan Y.; Chen C.; Li Z.; Du S.; Luan X.; Gao Y.; Han X.; Song Y. Multistage-Responsive Gene Editing to Sensitize Ion-Interference Enhanced Carbon Monoxide Gas Therapy. Small 2022, 18 (40), 2204244 10.1002/smll.202204244. [DOI] [PubMed] [Google Scholar]
  65. Chen L. Q.; Howison C. M.; Jeffery J. J.; Robey I. F.; Kuo P. H.; Pagel M. D. Evaluations of extracellular pH within in vivo tumors using acidoCEST MRI. Magn. Reson. Med. 2014, 72 (5), 1408–1417. 10.1002/mrm.25053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Ding Y.; Yang J.; Wei H.; Wang J.; Huang S.; Yang S.; Guo Y.; Li B.; Shuai X. Construction of pH-Sensitive Nanovaccines Encapsulating Tumor Cell Lysates and Immune Adjuvants for Breast Cancer Therapy. Small 2023, 19, 2301420. 10.1002/smll.202301420. [DOI] [PubMed] [Google Scholar]
  67. Min K. H.; Min H. S.; Lee H. J.; Park D. J.; Yhee J. Y.; Kim K.; Kwon I. C.; Jeong S. Y.; Silvestre O. F.; Chen X.; Hwang Y.; Kim E.; Lee S. pH-Controlled Gas-Generating Mineralized Nanoparticles: A Theranostic Agent for Ultrasound Imaging and Therapy of Cancers. ACS Nano 2015, 9 (1), 134–145. 10.1021/nn506210a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Chiang P.-H.; Fan C.-H.; Jin Q.; Yeh C.-K. Enhancing Doxorubicin Delivery in Solid Tumor by Superhydrophobic Amorphous Calcium Carbonate–Doxorubicin Silica Nanoparticles with Focused Ultrasound. Mol. Pharmaceut. 2022, 19 (11), 3894–3905. 10.1021/acs.molpharmaceut.2c00384. [DOI] [PubMed] [Google Scholar]
  69. Kong F.; Zhang H.; Zhang X.; Liu D.; Chen D.; Zhang W.; Zhang L.; Santos H. A.; Hai M. Biodegradable Photothermal and pH Responsive Calcium Carbonate@Phospholipid@Acetalated Dextran Hybrid Platform for Advancing Biomedical Applications. Adv. Funct. Mater. 2016, 26 (34), 6158–6169. 10.1002/adfm.201602715. [DOI] [Google Scholar]
  70. Wang W.; Zhao Y.; Yan B. B.; Dong L.; Lu Y.; Yu S. H. Calcium carbonate-doxorubicin@silica-indocyanine green nanospheres with photo-triggered drug delivery enhance cell killing in drug-resistant breast cancer cells. Nano Res. 2018, 11 (6), 3385–3395. 10.1007/s12274-017-1950-3. [DOI] [Google Scholar]
  71. Wan L.; Cao Y.; Cheng C.; Tang R.; Wu N.; Zhou Y.; Xiong X.; He H.; Lin X.; Jiang Q.; Wang X.; Guo X.; Wang D.; Ran H.; Ren J.; Zhou Y.; Hu Z.; Li P. Biomimetic, pH-Responsive Nanoplatforms for Cancer Multimodal Imaging and Photothermal Immunotherapy. ACS Appl. Mater. Interfaces 2023, 15, 1784–1797. 10.1021/acsami.2c16667. [DOI] [PubMed] [Google Scholar]
  72. Zheng P.; Ding B.; Shi R.; Jiang Z.; Xu W.; Li G.; Ding J.; Chen X. A Multichannel Ca2+ Nanomodulator for Multilevel Mitochondrial Destruction-Mediated Cancer Therapy. Adv. Mater. 2021, 33 (15), 2007426 10.1002/adma.202007426. [DOI] [PubMed] [Google Scholar]
  73. Zhang A.; Xiao Z.; Liu Q.; Li P.; Xu F.; Liu J.; Tao H.; Feng L.; Song S.; Liu Z.; Huang G. CaCO3-Encapuslated Microspheres for Enhanced Transhepatic Arterial Embolization Treatment of Hepatocellular Carcinoma. Adv. Healthc. Mater. 2021, 10 (19), 2100748 10.1002/adhm.202100748. [DOI] [PubMed] [Google Scholar]
  74. Zheng P.; Ding J. Calcium ion nanomodulators for mitochondria-targeted multimodal cancer therapy. Asian J. Pharm. Sci. 2022, 17 (1), 1. 10.1016/j.ajps.2021.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Monteith G. R.; McAndrew D.; Faddy H. M.; Roberts-Thomson S. J. Calcium and cancer: targeting Ca2+ transport. Nat. Rev. Cancer 2007, 7 (7), 519–530. 10.1038/nrc2171. [DOI] [PubMed] [Google Scholar]
  76. Xu M.; Zhang J.; Mu Y.; Foda M. F.; Han H. Activation of TRPV1 by capsaicin-loaded CaCO3 nanoparticle for tumor-specific therapy. Biomaterials 2022, 284, 121520 10.1016/j.biomaterials.2022.121520. [DOI] [PubMed] [Google Scholar]
  77. Zheng P.; Ding B.; Jiang Z.; Xu W.; Li G.; Ding J.; Chen X. Ultrasound-Augmented Mitochondrial Calcium Ion Overload by Calcium Nanomodulator to Induce Immunogenic Cell Death. Nano Lett. 2021, 21 (5), 2088–2093. 10.1021/acs.nanolett.0c04778. [DOI] [PubMed] [Google Scholar]
  78. Shen J.; Liao X.; Wu W.; Feng T.; Karges J.; Lin M.; Luo H.; Chen Y.; Chao H. A pH-responsive iridium(III) two-photon photosensitizer loaded CaCO3 nanoplatform for combined Ca2+ overload and photodynamic therapy. Inorg. Chem. Front. 2022, 9 (16), 4171–4183. 10.1039/D2QI00951J. [DOI] [Google Scholar]
  79. Li Y.; Zhou S.; Song H.; Yu T.; Zheng X.; Chu Q. CaCO3 nanoparticles incorporated with KAE to enable amplified calcium overload cancer therapy. Biomaterials 2021, 277, 121080 10.1016/j.biomaterials.2021.121080. [DOI] [PubMed] [Google Scholar]
  80. Stanley I. R.; Laurence A. S.; Hill J. C. Disappearance of intraperitoneal gas following gynaecological laparoscopy. Anaesthesia 2002, 57 (1), 58–62. 10.1046/j.1365-2044.2002.02358.x. [DOI] [PubMed] [Google Scholar]
  81. Mullet C. E.; Viale J. P.; Sagnard P. E.; Miellet C. C.; Ruynat L. G.; Counioux H. C.; Motin J. P.; Boulez J. P.; Dargent D. M.; Annat G. J. Pulmonary CO2 Elimination During Surgical Procedures Using Intra- or Extraperitoneal CO2 Insufflation. Anesth. Analg. 1993, 76 (3), 622–626. 10.1213/00000539-199303000-00031. [DOI] [PubMed] [Google Scholar]
  82. Park E. Y.; Kwon J. Y.; Kim K. J. Carbon dioxide embolism during laparoscopic surgery. Yonsei Med. J. 2012, 53 (3), 459–466. 10.3349/ymj.2012.53.3.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Schutt E. G.; Klein D. H.; Mattrey R. M.; Riess J. G. Injectable microbubbles as contrast agents for diagnostic ultrasound imaging: the key role of perfluorochemicals. Angew. Chem., Int. Ed. 2003, 42 (28), 3218–3235. 10.1002/anie.200200550. [DOI] [PubMed] [Google Scholar]
  84. Kato T.; Yashiro T.; Murata Y.; Herbert D. C.; Oshikawa K.; Bando M.; Ohno S.; Sugiyama Y. Evidence that exogenous substances can be phagocytized by alveolar epithelial cells and transported into blood capillaries. Cell. Tissue Res. 2003, 311, 47–51. 10.1007/s00441-002-0647-3. [DOI] [PubMed] [Google Scholar]
  85. Zhou X.; Huang X.; Wang B.; Tan L.; Zhang Y.; Jiao Y. Light/gas cascade-propelled Janus micromotors that actively overcome sequential and multi-staged biological barriers for precise drug delivery. Chem. Eng. J. 2021, 408, 127897 10.1016/j.cej.2020.127897. [DOI] [Google Scholar]
  86. Zu Y.; Yao H.; Wang Y.; Yan L.; Gu Z.; Chen C.; Gao L.; Yin W. The age of bioinspired molybdenum-involved nanozymes: Synthesis, catalytic mechanisms, and biomedical applications. VIEW 2021, 2 (3), 20200188 10.1002/VIW.20200188. [DOI] [Google Scholar]
  87. Zu Y.; Wang Z.; Yao H.; Yan L. Oxygen-generating biocatalytic nanomaterials for tumor hypoxia relief in cancer radiotherapy. J. Mater. Chem. B 2023, 11 (14), 3071–3088. 10.1039/D2TB02751H. [DOI] [PubMed] [Google Scholar]
  88. Liu C.; Cheng S.; Zhou X.; Wang J.; Mu P.; Wang Z.; Zhang L.; Li L.; Wang C. Selective Nanoblocker of Cellular Stress Response for Improved Drug-Free Tumor Therapy. Adv. Healthc. Mater. 2023, 12 (10), 2202893 10.1002/adhm.202202893. [DOI] [PubMed] [Google Scholar]
  89. Wang C.; Li T.; Wang Z.; Li Y.; Liu Y.; Xu M.; Zhang Z.; Deng Y.; Cai L.; Zhang C.; Li C. Nano-modulators with the function of disrupting mitochondrial Ca2+ homeostasis and photothermal conversion for synergistic breast cancer therapy. J. Nanobiotechnol. 2023, 21 (1), 465. 10.1186/s12951-023-02220-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Zhao Y.; Bian Y.; Xiao X.; Liu B.; Ding B.; Cheng Z.; Ma P. a.; Lin J. Tumor Microenvironment-Responsive Cu/CaCO3-Based Nanoregulator for Mitochondrial Homeostasis Disruption-Enhanced Chemodynamic/Sonodynamic Therapy. Small 2022, 18 (38), 2204047 10.1002/smll.202204047. [DOI] [PubMed] [Google Scholar]
  91. Zheng P.; Ding B.; Zhu G.; Li C.; Lin J. Biodegradable Ca2+ nanomodulators activate pyroptosis through mitochondrial Ca2+ overload for cancer immunotherapy. Angew. Chem., Int. Ed. 2022, 61 (36), e202204904 10.1002/anie.202204904. [DOI] [PubMed] [Google Scholar]
  92. Hu H.; Ruan H.; Ruan S.; Pei L.; Jing Q.; Wu T.; Hou X.; Xu H.; Wang Y.; Feng N.; Zhang Y. Acid-responsive PEGylated branching PLGA nanoparticles integrated into dissolving microneedles enhance local treatment of arthritis. Chem. Eng. J. 2022, 431, 134196 10.1016/j.cej.2021.134196. [DOI] [Google Scholar]
  93. Tang K.; Zhang X.; Yin J.; Pan W.; Li Y.; Li N.; Tang B. A CaCO3-based synergistic immunotherapy strategy for treating primary and distal tumors. Chem. Commun. 2023, 59 (24), 3562–3565. 10.1039/D2CC07076F. [DOI] [PubMed] [Google Scholar]
  94. Chen X.; Li G.; Dai S.; Li Q.; Jiang Q.; Ou S.; Jin Z.; Wu J.; Liu L.; Huang G. Losartan-IR820-Loaded Polydopamine Nanoparticles for CO2-Generating Ultrasound Imaging-Guided Photothermal/Photodynamic Therapies of Triple-Negative Breast Cancer. ACS Appl. Nano Mater. 2023, 6 (13), 12300–12311. 10.1021/acsanm.3c01987. [DOI] [Google Scholar]
  95. Ding M.; Kong X.; Chen W.; Yan L.; Huang H.; Lv Z.; Jiang P.; Mu A.; Huang C.; Shi J. Efficient starvation therapy with three-pathway blocking in combination with PTT/CDT for TME reversal and tumor apoptosis. J. Ind. Eng. Chem. 2022, 110, 456–470. 10.1016/j.jiec.2022.03.022. [DOI] [Google Scholar]
  96. Zhao Y.; Zheng Y.; Zhu Y.; Ding K.; Zhou M.; Liu T. Co-delivery of gemcitabine and Triapine by calcium carbonate nanoparticles against chemoresistant pancreatic cancer. Int. J. Pharmaceut. 2023, 636, 122844 10.1016/j.ijpharm.2023.122844. [DOI] [PubMed] [Google Scholar]
  97. Chen M.; Guo X.; Shen L.; Ding J.; Yu J.; Chen X.; Wu F.; Tu J.; Zhao Z.; Nakajima M.; Song J.; Shu G.; Ji J. Monodisperse CaCO3-loaded gelatin microspheres for reversing lactic acid-induced chemotherapy resistance during TACE treatment. Int. J. Biol. Macromol. 2023, 231, 123160–123160. 10.1016/j.ijbiomac.2023.123160. [DOI] [PubMed] [Google Scholar]
  98. Zhou Y.; Jing S.; Liu S.; Shen X.; Cai L.; Zhu C.; Zhao Y.; Pang M. Double-activation of mitochondrial permeability transition pore opening via calcium overload and reactive oxygen species for cancer therapy. J. Nanobiotechnol. 2022, 20 (1), 188. 10.1186/s12951-022-01392-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hu S.; Xia K.; Huang X.; Zhao Y.; Zhang Q.; Huang D.; Xu W.; Chen Z.; Wang C.; Zhang Z. Multifunctional CaCO3@Cur@QTX125@HA nanoparticles for effectively inhibiting growth of colorectal cancer cells. J. Nanobiotechnol. 2023, 21 (1), 353. 10.1186/s12951-023-02104-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Boyjoo Y.; Pareek V. K.; Liu J. Synthesis of micro and nano-sized calcium carbonate particles and their applications. J. Mater. Chem. A 2014, 2 (35), 14270–14288. 10.1039/C4TA02070G. [DOI] [Google Scholar]
  101. Niu Y.-Q.; Liu J.-H.; Aymonier C.; Fermani S.; Kralj D.; Falini G.; Zhou C.-H. Calcium carbonate: Controlled synthesis, surface functionalization, and nanostructured materials. Chem. Soc. Rev. 2022, 51 (18), 7883–7943. 10.1039/D1CS00519G. [DOI] [PubMed] [Google Scholar]
  102. Wu Y.; Chang X.; Yang G.; Chen L.; Wu Q.; Gao J.; Tian R.; Mu W.; Gooding J. J.; Chen X.; Sun S. A Physiologically Responsive Nanocomposite Hydrogel for Treatment of Head and Neck Squamous Cell Carcinoma via Proteolysis-Targeting Chimeras Enhanced Immunotherapy. Adv. Mater. 2023, 35, 2210787. 10.1002/adma.202210787. [DOI] [PubMed] [Google Scholar]
  103. Kolate A.; Baradia D.; Patil S.; Vhora I.; Kore G.; Misra A. PEG — A versatile conjugating ligand for drugs and drug delivery systems. J. Controlled Release 2014, 192, 67–81. 10.1016/j.jconrel.2014.06.046. [DOI] [PubMed] [Google Scholar]
  104. Lin J.; Huang L.; Xiang R.; Ou H.; Li X.; Chen A.; Liu Z. Blood compatibility evaluations of CaCO3 particles. Biomed. Mater. 2021, 16 (5), 055010 10.1088/1748-605X/ac19bf. [DOI] [PubMed] [Google Scholar]
  105. Li G.; Liang L.; Yang J.; Zeng L.; Xie Z.; Zhong Y.; Ruan X.; Dong M.; Yang Z.; Lai G.; et al. Pulmonary hypofunction due to calcium carbonate nanomaterial exposure in occupational workers: A cross-sectional study. Nanotoxicology 2018, 12 (6), 571–585. 10.1080/17435390.2018.1465606. [DOI] [PubMed] [Google Scholar]
  106. Szydlowska K.; Tymianski M. Calcium, ischemia and excitotoxicity. Cell Calcium 2010, 47 (2), 122–129. 10.1016/j.ceca.2010.01.003. [DOI] [PubMed] [Google Scholar]
  107. Danmaigoro A.; Selvarajah G. T.; Mohd Noor M. H.; Mahmud R.; Abu Bakar M. Z. Toxicity and safety evaluation of doxorubicin-loaded cockleshell-derived calcium carbonate nanoparticle in dogs. Adv. Pharmacol. Sci. 2018, 2018, 4848602 10.1155/2018/4848602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Jaji A. Z.; Zakaria Z. A. B.; Mahmud R.; Loqman M. Y.; Hezmee M. N. M.; Abba Y.; Isa T.; Mahmood S. K. Safety assessments of subcutaneous doses of aragonite calcium carbonate nanocrystals in rats. J. Nanopart. Res. 2017, 19, 1–18. 10.1007/s11051-017-3849-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from ACS Omega are provided here courtesy of American Chemical Society

RESOURCES