Background: The development of breast cancer (BC) and how it responds to treatment have both been linked to the involvement of inflammation. Chronic inflammation is critical in carcinogenesis, leading to elevated DNA damage, impaired DNA repair machinery, cell growth, apoptosis, angiogenesis, and invasion. Studies have found several targets that selectively modulate inflammation in cancer, limit BC’s growth, and boost treatment effectiveness. Drug resistance and the absence of efficient therapeutics for metastatic and triple-negative BC contribute to the poor outlook of BC patients. Summary: To treat BC, small-molecule inhibitors, phytomedicines, and nanoparticles are conjugated to attenuate BC signaling pathways. Due to their numerous target mechanisms and strong safety records, phytomedicines and nanomedicines have received much attention in studies examining their prospects as anti-BC agents by such unfulfilled demands. Key Messages: The processes involved in the affiliation across the progression of tumors and the spread of inflammation are highlighted in this review. Furthermore, we included many drugs now undergoing clinical trials that target cancer-mediated inflammatory pathways, cutting-edge nanotechnology-derived delivery systems, and a variety of phytomedicines that presently address BC.

Recent breakthroughs in treating cancer, which is a primary cause of illness and death globally, have only had a minor impact on individual survivability. Investigators seeking to create potential anticancer medicines are increasingly interested in the factors influencing cancer performance and sustainability [1‒8]. Inflammation’s role in influencing cancer development has been underlined recently [9]. Inflammation has been linked to each stage of tumor formation, particularly onset, progression, metastasis, and relapse. Extrinsic and intrinsic tumor inflammation, induced by autoimmunity, infectious diseases, or other variables contributing to cancer and speeding up neoplastic progressions, such as obesity, smoking, and alcohol intake, invoke genetic changes or provide a favored context for tumorigenesis. Inflammation governs cancer growth and might have a negative impact on therapeutic results [10, 11]. Treatment-induced inflammation only develops after treatment but may be crucial in determining drug efficacy or resistance [12]. Cancer and immune cells communicate actively or passively via chemokine and cytokine. Inflammation’s pro-tumorigenic impacts are frequently regulated through feed-forward cycles in signaling pathways.

Nanomedicine utilizes nanosized tools, while phytomedicine utilizes phytoconstituents to treat disease and improve quality of life. The nanomedicine and phytomedicine approaches are nowadays focused because the presently available chemotherapy’s main drawbacks are tumor regrowth, resistance, and harmful effects on non-targeted cells, which might limit the use of antitumor medications and hence lower the quality of life for patients. Plant-based phytonutrients (phytomedicines) and analogs can potentially amend therapeutic utility and shorten complications in cancer patients. Several phytonutrients are physiologically active substances found in nature with high anticancer activities [13]. Nanomedicine and phytomedicine ensure effective chemotherapeutic medicines with improved efficacy and fewer side effects persist in facing the obstacles of current therapy. Cancer nanotechnology has become an innovative and potentially adjunctive therapeutic approach because of the gaps in traditional cancer treatments. It offers new possibilities for early recognition, improved therapy, prognosis, and an effective cancer diagnosis. The emerging interest in using nanotechnology for cancer control is mainly due to the therapeutic potential of certain nanostructures and the distinguishing structural characteristics of cancer. Although cancer nanotechnologies have the potential to administer dose-specific chemotherapeutics (with little toxicity), it is crucial to take into account the intricacy and kinetics of the tumor to bridge the translational bench-to-bedside divide [14, 15]. Several nanomaterials were used to discover and advance existing cancer therapies. Highlighting the determinants of inflammation in multiple stages of tumor emergence will aid in developing theranostic strategies that utilize inflammatory processes and preferentially attack the tumor-associated environments [16]. Several ways are employed to amend cancer chemotherapies’ performance while reducing their toxicities. Tumor diversity can result in poor therapeutic effects, developed resistance, and unwanted side effects.

Nonetheless, employing nanomedicines to address and modulate the tumor microenvironment is essential for improving cancer’s therapy effects [17]. Nanomedicine is primarily used as a carrier system for immunoregulatory drugs, allowing for faster and more targeted delivery of these agents to aid in mounting anticancer immune function [18]. The history of nanomedicine and phytomedicine can be traced back to ancient times [19]. In modern times, pioneers of nanomedicine, Metchnikov and Ehrlich, Nobel Prize for Medicine winners 1908, worked on nanomedicine as therapeutics and diagnostics [20]. The research on nanomedicine and phytomedicine has increased dramatically in the last 30 years of the 20th century [19]. The role of nanoparticles (NPs) in biomedicine appeared in the 1970s, and since then, around 10,000 research papers have been published [19]. In this review, we looked deeper into the role of natural products, mainly nanomedicines and phytomedicines, in cancer treatment by focusing on various inflammatory signaling pathways (Table 1, 2).

Table 1.

List of some agents targeting cancer-mediated inflammatory pathways under clinical trials and their present status

Name of drugTargetConditionPhaseNCT numberPresent status
Aspirin Cox-1/2 Gastric ulcer and colon cancer III NCT04214990 Recruiting 
NCT02467582 
Cancer-associated thrombosis in solid tumor NCT02285738 Completed 
Multiple myeloma II NCT01215344 Completed 
Celecoxib Cox-2 Primary BC III NCT02429427 Completed 
Locally advanced NSCLC NCT00046839 Completed 
Lung cancer II NCT00020878 
Stage I, II, and III of BC II NCT00201773 
Prostate cancer II NCT01220973 
Metastatic colorectal cancer II NCT00466505 
Head and neck cancer NCT00581971 
II NCT00061906 
Metastatic kidney cancer II NCT01158534 
Cervical intraepithelial neoplasia NCT00081263 
Colorectal cancer NCT00033371 
Mouth neoplasms NCT00953849 
Anaplastic glioma NCT00504660 
Multiple myeloma NCT00099047 
Liver cancer III NCT03059238 
Cytokines and chemokines 
Infliximab Chimeric TNFα antibody Pancreatic neoplasms II NCT00060502 Completed 
Lung neoplasm malignant IV NCT04036721 Recruiting 
Melanoma II NCT04305145 Recruiting 
Hepatosplenic T-cell lymphoma IV NCT01804166 Completed 
Renal cell carcinoma IV NCT02596035 Not recruiting 
Etanercept Human TNFR2-Fc fusion protein Pancreatic neoplasms II NCT00201838 Completed 
Melanoma VI NCT01053819 Completed 
Metastatic castration-resistant prostate cancer NCT03792841 Recruiting 
Leukemia II NCT00509600 Completed 
Inflammatory transcription factors 
Ruxolitinib JAK1/2 inhibitor Pancreatic cancer II NCT01423604 Completed 
Estrogen receptor (ER) + BC II NCT01594216 Completed 
Colorectal cancer NCT04303403 Recruiting 
Myeloproliferative neoplasms NCT02076191 Completed 
Acute myeloid leukemia  NCT00674479 Completed 
Hematopoietic neoplasm NCT01523171 Completed 
Bladder cancer NCT02788201 Completed 
Bortezomib NF-κB inhibitor Prostate cancer II NCT00183937  
Kidney cancer II NCT00025376 
Lung cancer II NCT00064012 
Stage IV BC II NCT00025584 
Head and neck cancer NCT00629226 
Colorectal cancer NCT00280176 
Non-small cell lung cancer II NCT01833143 
Colorectal cancer II NCT00052507 
Ovarian cancer NCT00098982 
Bladder cancer II NCT00066352 
Trabectedin TAMs cytotoxicity Prostate cancer II NCT00147212 Completed 
Ovarian cancer II NCT00113607 
Pancreatic cancer II NCT01339754 
Neoplasm metastases I/II NCT01267084 
Solitary fibrous tumors II NCT03023124 Recruiting 
Solid tumors II NCT00786838 Completed 
Soft tissue sarcoma II NCT00070109 
Liposarcoma II NCT00060944 
Brain and CNS tumors II NCT00003939 
Recurrent high-grade meningioma II NCT02234050 
Sarcoma II NCT00379145 
Metastatic liposarcoma II NCT01692496 
Name of drugTargetConditionPhaseNCT numberPresent status
Aspirin Cox-1/2 Gastric ulcer and colon cancer III NCT04214990 Recruiting 
NCT02467582 
Cancer-associated thrombosis in solid tumor NCT02285738 Completed 
Multiple myeloma II NCT01215344 Completed 
Celecoxib Cox-2 Primary BC III NCT02429427 Completed 
Locally advanced NSCLC NCT00046839 Completed 
Lung cancer II NCT00020878 
Stage I, II, and III of BC II NCT00201773 
Prostate cancer II NCT01220973 
Metastatic colorectal cancer II NCT00466505 
Head and neck cancer NCT00581971 
II NCT00061906 
Metastatic kidney cancer II NCT01158534 
Cervical intraepithelial neoplasia NCT00081263 
Colorectal cancer NCT00033371 
Mouth neoplasms NCT00953849 
Anaplastic glioma NCT00504660 
Multiple myeloma NCT00099047 
Liver cancer III NCT03059238 
Cytokines and chemokines 
Infliximab Chimeric TNFα antibody Pancreatic neoplasms II NCT00060502 Completed 
Lung neoplasm malignant IV NCT04036721 Recruiting 
Melanoma II NCT04305145 Recruiting 
Hepatosplenic T-cell lymphoma IV NCT01804166 Completed 
Renal cell carcinoma IV NCT02596035 Not recruiting 
Etanercept Human TNFR2-Fc fusion protein Pancreatic neoplasms II NCT00201838 Completed 
Melanoma VI NCT01053819 Completed 
Metastatic castration-resistant prostate cancer NCT03792841 Recruiting 
Leukemia II NCT00509600 Completed 
Inflammatory transcription factors 
Ruxolitinib JAK1/2 inhibitor Pancreatic cancer II NCT01423604 Completed 
Estrogen receptor (ER) + BC II NCT01594216 Completed 
Colorectal cancer NCT04303403 Recruiting 
Myeloproliferative neoplasms NCT02076191 Completed 
Acute myeloid leukemia  NCT00674479 Completed 
Hematopoietic neoplasm NCT01523171 Completed 
Bladder cancer NCT02788201 Completed 
Bortezomib NF-κB inhibitor Prostate cancer II NCT00183937  
Kidney cancer II NCT00025376 
Lung cancer II NCT00064012 
Stage IV BC II NCT00025584 
Head and neck cancer NCT00629226 
Colorectal cancer NCT00280176 
Non-small cell lung cancer II NCT01833143 
Colorectal cancer II NCT00052507 
Ovarian cancer NCT00098982 
Bladder cancer II NCT00066352 
Trabectedin TAMs cytotoxicity Prostate cancer II NCT00147212 Completed 
Ovarian cancer II NCT00113607 
Pancreatic cancer II NCT01339754 
Neoplasm metastases I/II NCT01267084 
Solitary fibrous tumors II NCT03023124 Recruiting 
Solid tumors II NCT00786838 Completed 
Soft tissue sarcoma II NCT00070109 
Liposarcoma II NCT00060944 
Brain and CNS tumors II NCT00003939 
Recurrent high-grade meningioma II NCT02234050 
Sarcoma II NCT00379145 
Metastatic liposarcoma II NCT01692496 
Table 2.

List of the nanomedicines in BC management by targeting specific signaling pathways

Type of nanomedicineConjugated moleculesIn vitro studyTargeting pathwaysOutcomeReference
AuNPs Constituents of Commiphora myrrha and Boswellia sacra MDA-MB-231, MCF7, and MCF-10A cell lines Cytotoxicity inducer Cytotoxicity BC cell lines [21
No damage to normal cells 
MDA-MB-231, MCF7, BT549, MCF-10A DNA damage and inducing Oxidative stress Cytotoxicity and radiation dose-dependent effects in BC cells [22
 184B5, and SUM-159 human cells 
PLGA-b-PEG polymer NPs Antisense-miR-21, miR-10b Human MDA-MB-231 cells Multitarget antagonization of native miRNA to tackle malignancy and proliferation Significant reduction in tumor growth at low doses, with a 40% reduction [23
RNA NPs based on pRNA 3-way-junction (3WJ) Anti-miR-21 RNA NPs’ potential involvement in miRNA transport in cancer therapies has been confirmed [24
PEG-PLA NPs siRNA In vivo and in vitro on SUM149, MCF-10A, and BT549 cells Cell growth is reduced, and cell apoptosis is increased when CDK1 (cyclin dependent kinase) is targeted by siRNA. Tumor development in mice has been Slowed without generating systemic damage or triggering the immune response [25
HA-chitosan NPs miR-34a and DOX In vivo and in vitro on human MDA-MB-231 cells DOX-induced apoptosis by suppressing Bcl-2. Also, miR-34a hamper cell growth via Notch-1 signaling Tumor suppression [26
pH dual-sensitive cationic unimolecular NP siRNA MDA-MB-468 TNBC cells EGFR GE11 peptide significantly elevated cellular uptake of NPs [27
OX26-PEG-Se NPs AZD2014 (vistusertib) + AZD8055, + FITC MCF7 cells JAK2/STAT3 and mTOR pathway Inhibited phosphorylation of TSC2 and small GTPase Rheb [28, 29
Iron oxide NPs (IONPs) DOX Xenograft model Wnt/β-catenin signaling Suppress GSK-3β, β-catenin assists transcription of Wnt target genes and upregulate E-cadherin and CD24 [30
Polylactic acid glycolic PD98059, a selective MAPK inhibitor Melanoma bearing mice MAPK signaling Inhibit phosphorylation of ERK [31
Acid-PD98059 
Conjugated NPs 
Ultrasmall magnetic IONP iWnt and ATF24 peptides MDA-MB-231 human BC cell Blocked Wnt/β-catenin pathway Minimized stemness of CSCs [30
Photoactive nanocarbon complex TRPV2 antibody MCF7 cells Dysregulate Wnt/β-catenin The reduced self-renewal potential of CSCs [32
Pathway via modulation of calcium influx 
HA-conjugateds Pluronic® NPs Thymoquinone MDAMB-231, MDAMB-468, 4T1 Downregulate VEGF-A through upregulation of miR-361 Attenuate angiogenesis [33
Type of nanomedicineConjugated moleculesIn vitro studyTargeting pathwaysOutcomeReference
AuNPs Constituents of Commiphora myrrha and Boswellia sacra MDA-MB-231, MCF7, and MCF-10A cell lines Cytotoxicity inducer Cytotoxicity BC cell lines [21
No damage to normal cells 
MDA-MB-231, MCF7, BT549, MCF-10A DNA damage and inducing Oxidative stress Cytotoxicity and radiation dose-dependent effects in BC cells [22
 184B5, and SUM-159 human cells 
PLGA-b-PEG polymer NPs Antisense-miR-21, miR-10b Human MDA-MB-231 cells Multitarget antagonization of native miRNA to tackle malignancy and proliferation Significant reduction in tumor growth at low doses, with a 40% reduction [23
RNA NPs based on pRNA 3-way-junction (3WJ) Anti-miR-21 RNA NPs’ potential involvement in miRNA transport in cancer therapies has been confirmed [24
PEG-PLA NPs siRNA In vivo and in vitro on SUM149, MCF-10A, and BT549 cells Cell growth is reduced, and cell apoptosis is increased when CDK1 (cyclin dependent kinase) is targeted by siRNA. Tumor development in mice has been Slowed without generating systemic damage or triggering the immune response [25
HA-chitosan NPs miR-34a and DOX In vivo and in vitro on human MDA-MB-231 cells DOX-induced apoptosis by suppressing Bcl-2. Also, miR-34a hamper cell growth via Notch-1 signaling Tumor suppression [26
pH dual-sensitive cationic unimolecular NP siRNA MDA-MB-468 TNBC cells EGFR GE11 peptide significantly elevated cellular uptake of NPs [27
OX26-PEG-Se NPs AZD2014 (vistusertib) + AZD8055, + FITC MCF7 cells JAK2/STAT3 and mTOR pathway Inhibited phosphorylation of TSC2 and small GTPase Rheb [28, 29
Iron oxide NPs (IONPs) DOX Xenograft model Wnt/β-catenin signaling Suppress GSK-3β, β-catenin assists transcription of Wnt target genes and upregulate E-cadherin and CD24 [30
Polylactic acid glycolic PD98059, a selective MAPK inhibitor Melanoma bearing mice MAPK signaling Inhibit phosphorylation of ERK [31
Acid-PD98059 
Conjugated NPs 
Ultrasmall magnetic IONP iWnt and ATF24 peptides MDA-MB-231 human BC cell Blocked Wnt/β-catenin pathway Minimized stemness of CSCs [30
Photoactive nanocarbon complex TRPV2 antibody MCF7 cells Dysregulate Wnt/β-catenin The reduced self-renewal potential of CSCs [32
Pathway via modulation of calcium influx 
HA-conjugateds Pluronic® NPs Thymoquinone MDAMB-231, MDAMB-468, 4T1 Downregulate VEGF-A through upregulation of miR-361 Attenuate angiogenesis [33

TNBC, triple-negative breast cancer.

Formerly, most cancer treatments used pharmacological drugs, surgical intervention, or radiotherapy to destroy or eradicate tumor cells surgically. Then it progressed to therapeutic strategies, with the development of medications targeting molecular mechanisms, including specific kinase inhibitors and monoclonal antibodies. These treatments dramatically refined cancer patients’ well-being and longevity. The effectiveness and safety concerns continued to restrict cancer therapies’ full potential. Nanomedicine provides a more precise targeting method for these medicines, allowing them to improve drug accumulation in tumors while minimizing systemic toxicity.

Moreover, nanomedicine was developed to solve a number of problems with conventional cancer therapy, such as the lack of cooperation of free medicines and the establishment of chemoresistance [34, 35]. Most biologically active constituents are highly soluble in water but have low absorption because they cannot cross the lipid membranes of the cells, have excessively large molecular sizes, or are poorly absorbed, resulting in a loss of bioavailability and efficacy. Such circumstances make phytomedicines unavailable in clinical settings. Some extracts are not used clinically because of these obstacles [36]. Combining natural products with nanotechnology has been a promising approach. The nano-structured systems allow the biologically active constituents to potentiate the pharmacological action by reducing the dose and side effects and improving activity. Using nano-based formulations in plant medicine research has several benefits, such as increased solubility and bioavailability, dispersion of tissue macrophages, and therapeutic properties. Nano-formulations also offer protection from toxicity, stability, and long-term administration. Emergent formulations like niosomes, liposomes, nanospheres, and phytosomes are feasible options as carrier molecules for the efficient delivery of drugs in clinical settings. Anti-inflammatory medicines have been used as a treatment strategy because of the involvement of inflammation in cancer formation and recurrence. The transport of anti-inflammatory medicaments to malignant cells is the greatest obstacle. Nanotechnology has made specialized cancer therapy both safe and effective.

However, the speed of nanomedicine development has been hindered by the complicated and varied characteristics of cancer, limited evidence on the outcome and behavior of nanomedicines in the patient’s psyche, and a lack of large-scale commercialization. A complete grasp of the tactics, developments, success, problems, and future views is required to transform the narrative from traditional cancer therapies to anti-inflammatory nanotherapeutics [37]. Tumor resistance is assisted by several signaling pathways, notably PI3K/AKT/mTOR, RAS, WNT/β-catenin, TGF-β, and notch. The adaptable inflammatory action is the body’s frontline of infection defense. NF-κB, MAPK, JAK-STAT, and PI3K/AKT are cancer’s most typically dysfunctional inflammatory mechanisms. Nanomedicine, as a platform for transporting therapeutic medicines that selectively target tumor locations, is getting popular as an attempt to alleviate major obstacles in treating cancer. Polymer and INPs, liposomes, micellar nanostructures, nanotubes, and exosomes have all been widely researched as sustainable nanostructures. Because of their toxicity, sensitivity, and quick clearance by the mononuclear phagocytes, exosomes have been identified as a promising prospective method for the bioinspired, biologically engineered, and biomimetic delivery of drugs [38].

Addressing these inflammatory pathways has been demonstrated in preclinical research to effectively suppress tumorigenesis and metastasis, suggesting that they could be useful therapeutic strategies for individuals with inflammation-related breast cancer (BC). Additional biological processes observed in inflammatory BC include suppressing the TGF-β pathway and genetic changes in proteins that regulate cell growth. More research is needed to establish its physiological activity and therapeutic roles in inflammatory BC. Therapies that target the EGFR and JAK2 pathways are likely the first and most established in terms of clinical application [39].

Attaching ligands to the NP’s surface allows them to validate and bind specifically to corresponding molecular markers on the surface of specific BC cells, making them ideal for addressing BC cells. The ligand-NP conjugation interacts with BC receptors (e.g., HER2, EGFR, VEGFR, IGF-IR), induces NP internalization via endocytosis, and transfers the linked proteins to the active areas of tumor cells via proteolytic cleavage [40]. In progressed aggressive tumors and most malignancies, enhanced amplification of systemic inflammation has been found. Radiotherapy and chemotherapy cause a high tumor-associated inflammatory process. Due to chemotherapy’s inability to identify between tumor and normal cells, emphasis has been placed recently on producing targeted medicines with greater precision for cancerous cells, greater efficacy, and reduced toxicity [41]. Small-molecule inhibitors (SMIs), organic biomolecules, composite cytokines, local irradiation, neutralizing antibodies, oncolytic viruses, Toll-like receptor agonists, and specialized pro-resolving lipid mediators are all investigated as ways to attenuate inflammatory response in cancer treatment (Fig. 1).

Fig. 1.

An overview of the mechanism of nanomedicine, phytomedicine, and SMIs blocking cancer cell survival pathways like JAK-STAT/MAPK/NF-κB/MAPK/Akt/mTOR/Wnt pathways leading to inhibition of various downstream signaling (TNFR/CD40R/EGFR/IL-6) pathways. All these factors make cancer cells die via various cell death mechanisms.

Fig. 1.

An overview of the mechanism of nanomedicine, phytomedicine, and SMIs blocking cancer cell survival pathways like JAK-STAT/MAPK/NF-κB/MAPK/Akt/mTOR/Wnt pathways leading to inhibition of various downstream signaling (TNFR/CD40R/EGFR/IL-6) pathways. All these factors make cancer cells die via various cell death mechanisms.

Close modal

Nanomedicines Targeting Inflammatory Signaling Pathways in BC

Polymer-drug conjugates, including NPs in association with certain other SMIs, phytoconstituents, and siRNA, have been the most dominant groups of targeted therapies in cancer treatment due to breakthroughs in nanotechnology. NPs are employed to incorporate active chemicals for better delivery systems in various cancer therapeutic strategies. By controlling mTOR activity, NPs could halt cellular proliferation in tumor cells [42]. Polyamidoamine, a type of nanocomposite designed for clinical use, disrupts the AKT-TSC2-mTOR signaling cascade, resulting in autophagy [43]. SiO2NP suppresses the PI3K-AKT-mTOR signaling cascade by repealing the NO-NOS system, generating an inflammatory response in tumor cells that results in apoptosis [44].

Song et al. [45] developed hyaluronic acid (HA)-coated, mannan-conjugated MnO2 NPs (Man-HA-MnO2) to modify multidrug resistance by utilizing HA’s potential to remodel anti-inflammatory, pro-tumor M2 TAMs (tumor-associated macrophages M2) to pro-inflammatory, antitumor M1 macrophages. In a breast tumor mouse model, therapy with Man-HA-MnO2 particles boosted tumor oxygenation while downregulating HIF-1α (hypoxia-inducible factor-1) and VEGF. Compared to chemotherapy alone, combining these NPs with doxorubicin (DOX) suppressed tumor development. Zuo et al. synthesized cationic lipid-aided polymeric NPs enclosed with siRNA to create a suitable siRNA delivery method for anti-breast cancer stem-cell (BCSC) therapy. The BCSCs were then effectively removed in vitro using NPs supplied with siRNA addressing the oncogene Plk1. They blocked the TGF-β type I receptor and TGF-β signaling, allowing NPs to penetrate tumor tissue more easily (Fig. 1). The tumor microenvironment (TME) was changed, and the enhanced penetration and retention impact was modified [46].

The tumor targeting with drugs/phytopharmaceuticals utilizing various nanosized carriers like liposomes, NPs, polymers, micelles, and conjugates of NPs is the main focus of nanomedicine [47]. There are two ways through which nanomedicine work. One is by covalent drug attachment using linkers to the targeted receptor, which is required to be recognized by tumor cells and is called active targeting. The other is passive targeting, where the enhanced penetration and retention effect is utilized [19]. The main factors are stability and drug release rate until the targeted tumor cell. The bioavailability of the drugs in biological tissues and fluids is quantified using various imaging techniques like magnetic resonance (MRI), gamma cameras, near-infrared (NIR) luminescence, and fluorescence and positron emission tomography (PET) are major techniques. In active targeting where the drug attachment with the targeted receptor has been traced via in vitro but very less in vivo, the progress is very slow [19]. The targeted antibody-mediated radiotherapy demonstrated less than 0.01% of the dose administered at the tumor site [48]. The pharmacological evaluation must often be conducted via dose-dependent targeting and saturation of the targeted receptor, which occurs even at low doses. In several nanomedicine technologies, biomarker estimation becomes important to estimate the efficacy and toxicity of the drugs as only very few drugs reach clinical trials and market [49]. The new advancements in novel nanomaterials have attracted increased attention toward theranostics and nano-vectors in nanomedicine in the past 10 years.

The carbon base materials consist of single-wall and multi-wall carbon nanotubes, fullerene (mainly C60), nanodiamond (ND), and graphene oxide (GO). However, all these materials are not soluble in water and can be solubilized by polyfunctionalized polyethylene glycol (PEG). Carbon cores are essentially used for scaffolds and cancer targeting; the Raman signatures have huge potential. The toxicity or safety concerns with these materials need to be addressed. Chronic use makes it an issue, although clinical investigation is a very important aspect [50, 51].

Gold nanoparticles (GNPs) have been brought to nanoscience through novel theranostic concepts based on the medium-sensitive plasmonic absorption leading to infrared light and visible light-induced collective oscillation of the electrons on the gold surface when the NP size is very tiny than the wavelength of light [52, 53]. The GNP plasmons can be useful to nanomedicine in several ways, like photo-thermal therapy [19, 52, 53]. GNPs provide versatile scaffolds for cell surface sensing using array-based “chemical nose” approaches and specific recognition. Passive cancer targeting with PEG and active cancer targeting with covalent bonds to rhTNFα (CYT-6091) have reached clinical trials [54]. The GNP is considered safer, more superior in terms of plasmonic properties than group 11 elements [19].

Despite the safety concerns, the superparamagnetic iron oxide NPs (SPIONPs) are widely used with magnets for cancer ablation by hyperthermia and MRI [55, 56]. These techniques have reached phase II for glioblastoma and prostate cancer [57]. Some other oxide NPs, like silica, encapsulate drug molecules for delivery [58]. The Quantum Dots (QD) NPs are widely applied in nanomedicine coated with ZnS/CdS for various applications, including cancer [19, 59].

The drug nano-vectors in nanomedicine have been studied extensively, including polymers, copolymers, antibodies, aptamers, proteins, and dendrimers [19, 58, 60]. Several biodegradable polymers are used in clinical studies [61]. The challenges in these polymers include poor drug loading, burst release, and poor miscibility with the drugs [62]. Dendrimers have a huge capacity to encapsulate drugs, and microbiocide Vivagel was investigated clinically [19]. The clinical investigations remain slow, although the major obstacles are reproducibility, purity, biocompatibility, and biodegradability [63].

The most successful in the nanomedicine remained are liposomes, as the anticancer drug carriers. The combination of drugs and imaging agents for diagnostics is the main example called theranostics [64]. The research on nanomedicine is expanding daily; however, there is a need for a multi-disciplinary approach between chemists, biophysicists, pharmacologists, clinicians, and biomedical engineers to produce nanoengineered drugs for the market.

Challenges in Using Nano and Phytomedicine in Modulating Signaling Pathways Involved in BC

Using NPs as drug delivery systems (DDS) is fraught with difficulties, including biodistribution, bioavailability, and potential toxicity. The use of PNPs in targeted DDS had modest results in translating preclinical methods to clinical trials for cancer treatment. Restricted ligand-receptor interaction, upregulation of surface markers on healthy and cancerous cells, the complexity of routing the therapeutics in notably minute doses to the tumor area, and NP buildup in non-targeted sites that result in nanotoxicity are some of the difficulties encountered during targeted DDS fabrication [65]. Another challenge in drug delivery is unwanted communication among biological entities and NPs, leading to unfavorable interactions and toxicity [66]. Nanocarriers’ physicochemical characteristics differ from batch to batch, and their substantial manufacture is likewise relatively challenging [67].

Nanomaterials for the delivery of drugs need to be precisely developed and evaluated to prevent nanocarrier toxicities in healthy cells. The protein crown, a biomolecule layer that surrounds NP complexes in biological contexts, oversees promoting the succeeding contacts of cells with the activated NPs. This protein crown interacts with the periventricular tissue’s surroundings, penetrates the tissue, and causes cells to internalize. By decreasing the adherence between NPs and the cell membrane, the protein corona ingestion on the NP’s surface renders the addressing capabilities of the naked NPs [68‒70]. Additionally, NPs cause protein aggregation, influencing their toxicity and targeting potential. The deposited proteins have a detrimental effect on the selectivity of NPs and help tumor cells activate mTOR, which causes cellular death [71, 72].

Clinical development will be facilitated by resolving the challenges of predictable, reproducible, and sustainable NP synthesis and NP detection and validation. Even though known pharmaceuticals have been utilized as the cargo in most approved NPs, we may anticipate that the next generation of nanomedicines will progressively include novel molecular domains and innovative therapeutics, including siRNA and mRNA genetic manipulation [15].

Nanomedicine has increased the potential for advancing BC treatment by enhancing the existing DDSs. Novel potential for upfront diagnosis and monitoring has been made possible by advancements in nanotechnology. We may better regulate the safety and usefulness of systemically delivered chemotherapeutics by adjusting the bioavailability of nanomedicines and their drug target aggregation [73]. Limited solubility, penetrability, rapid first-pass effect, and P-gp efflux are issues with the existing chemotherapeutics. Additionally, many chemotherapies can make cancer resistant. Therefore, the goal is to create an efficient therapy regimen using a nanomedicine method for BC treatment. The important discovery with the presently available therapy is that the drug, in addition to dispersing to the spot relevant to the target, also disperses to healthy tissue, resulting in significant toxicity. An alternative to the issues context of current chemotherapy treatments for BC is provided by nanomedicine. The nanomedicine techniques offer a target-specific medication employing several innovative drug delivery approaches, making them advantageous over the typical BC therapy [74]. Nanocarrier systems are being developed as a result of studies in the area of nanomedicine. These tactics involve functionalizing these nanostructures with functionalities that promote site-specific, customized drug release. Liposomes, nanogels, polymer micelles, and dendrimers are a few NP approaches that can overcome various biological obstacles in drug delivery.

NPs Conjugated Phytochemicals as Nanomedicine

The anticarcinogenic functions of phytochemicals, which control cellular processes like cell growth, cell death, migration, and invasion by attenuating various signaling pathways, are being supported by more and more investigation (Table 2). However, their diagnostic use is obstructed by poor hydrophilicity, poor absorption, destabilization, and biodegradability. Research has provided innovative formulations to increase the utilization of phytochemicals in anticancer therapy. Targeted delivery is made possible using nanostructures to extend the life span of phytochemicals in circulation and their bioavailability and durability [75]. Using nanoparticulate-based DDS in cancer treatment is promising because these naturally occurring nanosized substances have changed bioactivities and substantially reduced toxicity.

As previously mentioned, quercetin and its metabolites are essential in destroying malignant cells by focusing on inflammatory pathways. Quercetin-loaded Au nanoparticles (Qc-AuNPs) have been emphasized in reports using BC cell lines to effectively limit the epithelial-mesenchymal transition by blocking the transcription repressing agents, i.e., Snail, Slug, and Twist, and also angiogenesis and pervasiveness through the EGFR/VEGFR2-assisted pathway, and cycle arrest by impairing the EGFR/PI3K/AKT-assisted pathways [76]. AuNPs-Qu-5 has now been suggested as a promising medication delivery platform to treat BC and has been demonstrated to be more efficient in killing cancer cells than free quercetin. Resveratrol, 6-Gingerol, and other phytonutrients have also been coupled with nanomedicines to enhance their therapeutic value [75, 77].

Small-Molecule Inhibitors

SMIs are cancer therapy drugs to treat established, treatment-resistant malignancies and persistent invasive tumors. They include multitargeted and extremely targeted kinase inhibitors, DNA repair enzymes, epigenetic regulatory proteins, and proteasomes. C-MET and FLT3 inhibitors are two of the many receptor tyrosine kinase inhibitors that prevent tumor growth by concentrating on inflammatory signaling pathways. Several downstream signaling pathways, including PI3K/AKT, RAS/MAPK, FAK (focal adhesion kinase), STAT3/5, RAC/RHO, phospholipase Cg (PLC-g), SHP2, c-SRC, and CRKL, are activated when the C-MET receptor unites to its sole ligand, HGF (hepatocyte growth factor). These pathways govern viability, cell growth, invasion, and motility [78].

As crucial cytoplasmic kinases, often STKs, are activated when RTKs are activated, RTKs are a popular choice as a primary target for anticancer medications. The extracellular RTKs and cytoplasmic STKs are the targets of SMIs. Deregulated RTK activation promotes enhanced cell growth and survival, which frequently aids in cancer development. In particular, human epidermal growth factor receptor 2 (HER2), a member of the EGFR family of RTKs that is upregulated, is elevated and consequently highly expressed in some breast tumors. The phosphoinositol 3′-kinase/Akt route, the Ras/Raf/MAPK pathway, the STAT3 pathway, and the PLC-γ-CaMP pathway are only a few downstream signaling cascades stimulated by the activated tyrosine kinases [79, 80]. Chaperones, known as heat shock proteins (HSPs), aid in correctly folding and transporting proteins across the cell membrane. As they aid in the appropriate folding and transport of oncoproteins, which are crucial components of numerous signaling pathways, HSPs are linked to the growth and severity of cancer. It has been revealed that unusually higher levels of HSP27, 70, and 90 expressions are present in several malignancies, resulting in chemotherapy resistance and apoptotic inhibition. HSP90 is required for the oncogenic proteins HER2 and c-Src to function, which are essential for the development of BC. The new HSP90, SMI ganetespib, which has been demonstrated to have antitumor activities to combat several malignancies, is being researched as a potential cancer treatment [81‒83].

Fulvestrant, a specific estrogen receptor down-regulator, is an efficient endocrine therapeutic option for women with metastatic BC that is hormone-sensitive. It leads to the inhibition of BC cells. For the treatment of metastatic BC, fulvestrant has been approved. In metastatic hormone, receptor-positive BC cases that have persisted or reverted to past tamoxifen or aromatase inhibitor therapeutic processes, the coupling of fulvestrant with CDK4/6 inhibitors has demonstrated higher potency in the monotherapy process. Fulvestrant with alpelisib, an α-selective PI3K inhibitor, has been implemented for instances with PIK3CA mutant BC that has advanced on an endocrine treatment approach [84‒86].

Phytochemicals are significant chemopreventive agents that boost the production of antioxidants and the Nrf2-ARE (antioxidant-response-element) signaling pathway [87]. Phytoconstituents exhibit a variety of activities on numerous molecular targets in signaling pathways and alter the autophagy/apoptosis equilibrium, which controls cell viability. In addition to causing cell death in tumor cells, phytochemicals also cause necroptosis, which slows tumorigenesis and invasion [88‒90].

Flavonoids have antioxidant features and can alter signaling pathways, including MAPK/p38, PI3K/AKT, apoptotic processes, and Wnt, which eventually relates to their anticancer characteristics [91]. Curcumin is a polyphenol that modulates tumor cell proliferation and has received much interest in cancer therapies. It affects various cell signaling pathways, notably EGFR, erbB2 (erythroblastic oncogene-B), AKT, sonic hedgehog, Wnt/β-catenin, NF-κB, and STATs metastasis and angiogenesis [92, 93]. The primary molecules curcumin addresses to kill cancer cells are COX-2, NF-κB, TNFα, and cyclin D1. After curcumin therapies, cyclin D1 and CDK4 activation in breast and skin malignancies were greatly diminished in several in vitro investigations.

Additionally, curcumin inhibits the activity of activator protein-1 (AP-1), reducing the activation of angiogenic markers like VEGF, angiopoietin, MMP-9, and MMP-3 [94]. A flavonoid called apigenin has antiangiogenic characteristics by controlling signaling pathways, inducing apoptosis, preventing cancer cells from transforming, and arresting the cell cycle. The in vivo findings demonstrated that apigenin declined the production of Her2/neu protein and limited the PI3K/Akt/Forkhead box O-signaling cascade in BC mouse models [94‒96]. Table 3 lists small molecules and phytomedicines that aim at inflammatory pathways in BC therapy.

Table 3.

List of some naturally occurring and small synthetic molecules that target inflammatory signaling pathways in BC treatment

NameClass of compoundTargetMechanism of actionIC50References
Naturally occurring small molecules 
Andrographolide Diterpenoid Suppress PI3K/AKT pathway Prohibits activation of HIF-1α and VEGF protein, deactivates AKT, mTOR, and P70S6K 63.19 μM [97, 98
Baicalin Flavonoid Regulates PI3K/AKT, mTOR, and NF-κB Suppress hTERT and SLUG-assisted EMT 51.06 μm [99, 100
Curcumin Phyto polyphenol Inhibits VEGF, IL-8, CD-31, JAK2/STAT3, PI3K/AKT/mTOR COX-2, and NF-κB signaling Inhibits p65 and p50 genes, C-MET, survivin, Bcl-2, and cyclin D1 4.69 μM [92, 101, 102
Luteolin Flavonoid Suppress PI3K/AKT pathway Promotes caspase-3, 7, and 9, ERK, IGF, β-catenin, GSK-3β, MMP-2 and 9, iNOS, COX-2, Bcl-2, Bax, CDK2, and cyclin D and damages DNA 1.29 μg/mL [103, 104
Emodin Natural anthraquinone Suppress JAK/STAT, MAPK, PI3K/AKT, and NF-κB Suppress JAK activity and IRF4, STAT6, and C/EBPb signaling 7.22 μg/mL [105‒107
Licochalcone A Estrogenic flavonoid natural phenol, chalcone Suppress PI3K/AKT/mTOR activation Promote autophagy and apoptosis by targeting cyclins and CDKs 8.4 μM [108, 109
Quercetin Flavonoid Declines the synthesis of cytokines and inhibits the stimulation of ERK and p38 MAP kinase, and NF-κB/IκB signaling pathways It attenuates p300/HAT-mediated signaling in BC cells attenuating 7.6 μM [110, 111
Rottlerin (Rott) Polyphenol Rott increased the activation of LC3, beclin-1, and Atg12 aggregation during autophagy CSCs inhibited the Akt and mTOR phosphorylation, upregulated AMPK phosphorylation, and apoptosis 35 μM [112‒114
Cyanidin-3-glucoside (C3G) ACN found in fruits C3G exerts anti-inflammatory action and promotes miR-124 expression miR-124 dysregulated STAT3 and limits angiogenesis 150 μM [115
Delphinidin Anthocyanidin monomer Enhanced apoptosis and autophagy and suppressed mTOR, and activated the AMPK pathway In HER2+ BC cells, enhanced apoptosis 40 μM [116
Synthetic small molecules 
Everolimus Brand name-Afinitor Suppress PI3K/AKT Suppress mTOR 50 nm [117, 118
Larotrectinib Vitrakvi Regulates RAS/MAPK/ER, PI3K/AKT and PLC. Suppressed tropomyosin receptor kinase A/B/C 50 nm [119, 120
Buparlisib NVP-BKM120 Suppress PI3Kα and mTOR Decreases AKT activation, increases caspase-9 and caspase-3/7 activity, and inhibits NF-κB production 4.722 μM [121, 122
NameClass of compoundTargetMechanism of actionIC50References
Naturally occurring small molecules 
Andrographolide Diterpenoid Suppress PI3K/AKT pathway Prohibits activation of HIF-1α and VEGF protein, deactivates AKT, mTOR, and P70S6K 63.19 μM [97, 98
Baicalin Flavonoid Regulates PI3K/AKT, mTOR, and NF-κB Suppress hTERT and SLUG-assisted EMT 51.06 μm [99, 100
Curcumin Phyto polyphenol Inhibits VEGF, IL-8, CD-31, JAK2/STAT3, PI3K/AKT/mTOR COX-2, and NF-κB signaling Inhibits p65 and p50 genes, C-MET, survivin, Bcl-2, and cyclin D1 4.69 μM [92, 101, 102
Luteolin Flavonoid Suppress PI3K/AKT pathway Promotes caspase-3, 7, and 9, ERK, IGF, β-catenin, GSK-3β, MMP-2 and 9, iNOS, COX-2, Bcl-2, Bax, CDK2, and cyclin D and damages DNA 1.29 μg/mL [103, 104
Emodin Natural anthraquinone Suppress JAK/STAT, MAPK, PI3K/AKT, and NF-κB Suppress JAK activity and IRF4, STAT6, and C/EBPb signaling 7.22 μg/mL [105‒107
Licochalcone A Estrogenic flavonoid natural phenol, chalcone Suppress PI3K/AKT/mTOR activation Promote autophagy and apoptosis by targeting cyclins and CDKs 8.4 μM [108, 109
Quercetin Flavonoid Declines the synthesis of cytokines and inhibits the stimulation of ERK and p38 MAP kinase, and NF-κB/IκB signaling pathways It attenuates p300/HAT-mediated signaling in BC cells attenuating 7.6 μM [110, 111
Rottlerin (Rott) Polyphenol Rott increased the activation of LC3, beclin-1, and Atg12 aggregation during autophagy CSCs inhibited the Akt and mTOR phosphorylation, upregulated AMPK phosphorylation, and apoptosis 35 μM [112‒114
Cyanidin-3-glucoside (C3G) ACN found in fruits C3G exerts anti-inflammatory action and promotes miR-124 expression miR-124 dysregulated STAT3 and limits angiogenesis 150 μM [115
Delphinidin Anthocyanidin monomer Enhanced apoptosis and autophagy and suppressed mTOR, and activated the AMPK pathway In HER2+ BC cells, enhanced apoptosis 40 μM [116
Synthetic small molecules 
Everolimus Brand name-Afinitor Suppress PI3K/AKT Suppress mTOR 50 nm [117, 118
Larotrectinib Vitrakvi Regulates RAS/MAPK/ER, PI3K/AKT and PLC. Suppressed tropomyosin receptor kinase A/B/C 50 nm [119, 120
Buparlisib NVP-BKM120 Suppress PI3Kα and mTOR Decreases AKT activation, increases caspase-9 and caspase-3/7 activity, and inhibits NF-κB production 4.722 μM [121, 122

EMT, epithelial-mesenchymal transition.

In BC stem cells, ginsenoside F2 had anti-proliferative properties and activated the autophagosome process. In addition, ginsenoside F2 attracted GFP-tagged LC3-II to autophagosomes, stimulated the production of acidic vasculature organelles, and increased Atg-7 expression [123]. Quinacrine improved the activation of p53 and p21 and inhibited topoisomerase activation in BC cells, exhibiting anticancer effects [124]. Juglanin, typically derived from green husks, also demonstrated anticancer action. By controlling the ROS/JNK signaling system, juglanin-mediated therapy reduced G2/M phase arrest and activated apoptosis in human BC [125].

The detection, management, and mitigation of BC are several applications for nanotechnology. Numerous nanostructures were designed with the specific purpose of attacking BC that has progressed. By lowering utilization and medical costs, two key barriers to traditional cancer treatment-targeted NPs, DDS aims to lessen the negative impacts of antitumor medications [126]. Several nanostructures were utilized as delivery carriers for medications and diagnostic agents. Liposomes, polymeric vehicles including microspheres, hydrogels, polymeric NPs, dendrimers, nanowires, metallic NPs (e.g., Au, Ag, Ti), carbon nanodevices (e.g., nanotubes, NDs, graphene), inorganic particles (e.g., silica particles), and hybrid nanostructures have all been used [127]. Several nanomedicine approaches for the treatment of BC are given in Table 4.

Table 4.

Various nanomedicine approaches for BC treatment

Drug formulationsChemicals/polymersActive ingredientsCell line/receptorOutcomeReferences
Polymeric NPs of Docetaxel Pluronic F-127 DTX HER2+ cell line Increase uptake and cytotoxicity [128
SLNPs of tamoxifen TMX TMX MCF7 and MCF7-TamR Improved TAM efficacy and abolished Tam resistance by inducing death [129
Gold NPs FA, glucose, and glutamine Gold BC cell Improve tumor-specificity and radio-sensitizing ability of BSA-GNPs [130
AS1411 aptamer Aptamer-functionalized albumin, iron oxide, Au Gold, DOX MCF7 and SKBR3 human CCs Boosted cellular intake and activity [131
CNT-drug complex Hydrophilic polymer, PEG, and β-estradiol (E2) Lobaplatin ER in human Demonstrating continuous release qualities, there are no clear harmful effects [132
BC cells (HBCUs) 
Polymeric micelles Conjugated with Zileuton Zileuton BC cell line Lowered the number of CSCs in the tumor and successfully prevented metastasis [133
Trastuzumab-dendrimer-fluorine DDS Conjugated with fluorinated dendrimers Trastuzumab MCF7 cells It was effective to treat BC cells in vitro. Because there are 19F nuclei, MRI measures can be used to track the system [134
Silicon nanowire Conjugated with mebendazole (MBZ) MBZ MCF7 cells Concise biosensing was achieved through the detection of minute signaling pathways and bio-recognition events [135
Drug formulationsChemicals/polymersActive ingredientsCell line/receptorOutcomeReferences
Polymeric NPs of Docetaxel Pluronic F-127 DTX HER2+ cell line Increase uptake and cytotoxicity [128
SLNPs of tamoxifen TMX TMX MCF7 and MCF7-TamR Improved TAM efficacy and abolished Tam resistance by inducing death [129
Gold NPs FA, glucose, and glutamine Gold BC cell Improve tumor-specificity and radio-sensitizing ability of BSA-GNPs [130
AS1411 aptamer Aptamer-functionalized albumin, iron oxide, Au Gold, DOX MCF7 and SKBR3 human CCs Boosted cellular intake and activity [131
CNT-drug complex Hydrophilic polymer, PEG, and β-estradiol (E2) Lobaplatin ER in human Demonstrating continuous release qualities, there are no clear harmful effects [132
BC cells (HBCUs) 
Polymeric micelles Conjugated with Zileuton Zileuton BC cell line Lowered the number of CSCs in the tumor and successfully prevented metastasis [133
Trastuzumab-dendrimer-fluorine DDS Conjugated with fluorinated dendrimers Trastuzumab MCF7 cells It was effective to treat BC cells in vitro. Because there are 19F nuclei, MRI measures can be used to track the system [134
Silicon nanowire Conjugated with mebendazole (MBZ) MBZ MCF7 cells Concise biosensing was achieved through the detection of minute signaling pathways and bio-recognition events [135

Cancer Nanomedicine Challenges in Clinical Translation

Cancer nanotechnology has emerged as a ground-breaking and potential adjuvant therapy approach due to the shortcomings of traditional cancer treatments. It offers new opportunities for early recognition, enhanced therapy, a better prognosis, and effective cancer diagnosis. The emerging interest in using nanotechnology to control and fight cancer is primarily due to the therapeutic potential of certain nanostructures and cancer’s distinctive structural characteristics. Although cancer nanotechnologies can potentially administer dose-specific chemotherapeutics (with little toxicity), it is crucial to consider the intricacy and kinetics of the tumor to bridge the translational bench-to-bedside divide [14, 15]. Due to the lack of clinically available nanocarriers that take advantage of a broad range of nanoscale properties, the prospect of nanotechnology in medicine remains unrealized. Although therapeutic agents (chemical, physiological, or nanotechnological) addressing a portion of cancer cell subsets in vitro and even in relevant animal studies have been shown to be very efficient and cell-selective, several have collapsed during human trials. Long-term efforts to improve the regional localization selectivity of therapeutic NPs by adding targeting components to them have not shown promising outcomes. This mistake is related to using molecular targeting agents to increase identification efficiency but at the expense of far more challenging obstacles to overcome when crossing biological barriers [136].

The outcome of aggressive tumors in cancer biology, like triple-negative BC and pancreatic cancer, can be significantly improved by nanomedicine. In treating triple-negative BC, novel methods, including hemodynamic targeting, have shown potential for combating drug resistance and targeted delivery. Moreover, it is crucial to remember that the absence of professional insight in large-scale clinical-grade operations makes monetization for cancer nanomedicines difficult. Small compounds and antibodies are produced with particular expertise in modern pharmaceutical manufacturing facilities [137]. However, despite overstating preclinical outcomes, scientific studies must continue to uncover nanomaterials and pinpoint innovative approaches to nanocomposite cancer therapy. It may even learn through nature’s delivery mechanisms. It is crucial to thoroughly comprehend the therapeutic efficacy of cancer nanomodalities by looking at their physical interconnections. Translational research must take a “disease-driven” strategy instead of a “formulation-driven” one to generate clinically significant cancer nanotechnology that assists people in the long run [136].

Nano Phytomedicine as a New Therapeutic Approach

Currently, over 50% of necessary medications originate from nature. Because of its improved therapeutic effectiveness and fewer side effects relative to allopathic drugs, the usage of phytomedicine has grown over the past few years. Due to their limited hydrophilicity, lipophilicity, and improper molecular size, which leads to limited absorption, phytomedicines have demonstrated excellent in vitro activity but limited in vivo efficiency. Some of these issues can be addressed, and appropriate dosing regimens can be established with a deeper comprehension of the pharmacokinetics of phytomedicine. Using nano-based formulations in phytomedicine investigation has several benefits, such as increased solubility and bioavailability, increased therapeutic properties, protection from toxicity, increased tissue macrophage dispersion, better stability, sustained delivery, and protection from chemical and biological decay [138]. Using emerging formulations like niosomes, liposomes, nanospheres, and phytosomes is feasible. By reducing toxicity and boosting bioavailability, these DDSs could effectively lessen frequent dosing to combat non-compliance and improve therapeutic value. Therefore, incorporating NPs as DDS in the conventional medical system could aid in the fight against more severe diseases like cancer, diabetes, and asthma [36, 139].

The inflammation in cancer has a great influence on the plasticity TME. During carcinogenesis, inflammatory processes can be pro-tumorogenic/blocking antitumor immunity or anti-tumorigenic (immunosurveillance). The inflammatory process could exert signals that direct pro-tumorogenic signaling. The key role of inflammation in all stages of tumor growth calls for investigating the molecular events behind early tumorigenesis and propagation. The main goal of cancer therapy is to encourage the cancer-inhibiting inflammatory process and suppress cancer-promoting inflammation while maintaining a healthy alignment of inflammation, which is the most challenging treatment aspect. The cancer cells express the multidrug-resistant proteins to efflux out the anti-cancer drugs. The inflammation process is complex, and many reactions occur in TME. The inflammation has been proposed to alter MDR expression in cancer cells [140]. Various inflammatory mediators like chemokines, cytokines, and prostaglandins have been reported to regulate the expression of the MDR transporters in tumor cells. Such factors are crucial in the bioavailability of anticancer drugs at TME. It has been proposed that anti-inflammatory drugs such as non-steroidal anti-inflammatory drugs and glucocorticoids enhance the therapeutic outcome and maintain the balance of bioavailability of anticancer drugs [140]. It is important to uncover all the mechanisms between MDR proteins and inflammation that will be helpful in the therapeutic management of cancer subjects with malignant cancers with high death rates.

In order to improve their pharmacokinetic profile and reduce the off-target impacts, medications must be delivered via substrates that restrict drug breakdown and transformation in the circulatory and phagocytic pathways. Numerous NPs, bioflavonoids, phytocompounds, and small molecules are used in different targeting approaches, mostly inhibiting the PI3K-AKT, MAPK, and STAT3 pathways. By preventing the formation of p53 and increasing the transcription of the anti-apoptotic proteins (Bcl-2 and Bcl-xL), NF-κB and STAT3 both show good tumor-promoting processes. Most drugs block migration, penetration, and adherence to cancer cells while activating Bax proteins to cause cell death. It is necessary to research to develop low-toxicity anti-inflammatory drug combinations that can address inflammation-related signaling pathways in BCs with various phenotypic characteristics. The bioavailability problems posed by several elements, including the utilization of nanocarriers, liposomes, prosthetic polymeric micelles, phospholipid-based drug carriers, and microspheres, have recently been postulated to tackle these consequences and call for specialized investigations. Implementing any custom strategy that accents the goal of these signaling pathways to lessen toxicity and broaden the range of host defense to display profound importance for anticancer activity must be done with caution as we work to solve the challenges of conventional chemotherapies.

The authors have no conflicts of interest to declare.

No funding was received to carry out this study.

Writing original draft: Jeba Ajgar Ansari and Sakeel Ahmed; conceptualization, review and edit, and writing original draft: Jonaid Ahmad Malik; literature review: Faisal Ashraf Bhat, Afreen Khanam, Suhail Ahmad Mir, and Amr S. Abouzied; supervision and review and edit: Nafees Ahemad; and conceptualization supervision and review and edit: Sirajuheen Anwar.

Additional Information

Jeba Ajgar Ansari and Jonaid Ahmad Malik contributed equally to this work.

1.
Malik
JA
,
Jan
R
,
Ahmed
S
,
Anwar
S
.
Breast cancer drug repurposing a tool for a challenging disease
. In:
Drug repurposing - molecular aspects and therapeutic applications
.
2022 Jan
2.
Anwar
S
,
Malik
JA
,
Ahmed
S
,
Kameshwar
VA
,
Alanazi
J
,
Alamri
A
.
Can natural products targeting EMT serve as the future anticancer therapeutics
.
Molecules
.
2022
;
27
(
22
):
7668
.
3.
Anwar
S
,
Saleem
H
,
Khurshid
U
,
Ansari
SY
,
Alghamdi
S
,
Al-Khulaidi
AWA
.
Comparative phytochemical composition, oleuropein quantification, antioxidant and cytotoxic properties of Olea europaea L. leaves
.
Nat Prod Res
.
2023 Mar
37
6
1023
9
.
4.
Alafnan
A
,
Dogan
R
,
Bender
O
,
Celik
I
,
Mollica
A
,
Malik
JA
.
Beta Elemene induces cytotoxic effects in FLT3 ITD-mutated acute myeloid leukemia by modulating apoptosis
.
Eur Rev Med Pharmacol Sci
.
2023
;
27
(
8
):
3270
87
.
5.
Shinde
SS
,
Ahmed
S
,
Malik
JA
,
Hani
U
,
Khanam
A
,
Ashraf Bhat
F
.
Therapeutic delivery of tumor suppressor miRNAs for breast cancer treatment
.
J Biol Chem
.
2023
;
12
(
3
):
467
.
6.
Bender
O
,
Shoman
ME
,
Ali
TFS
,
Dogan
R
,
Celik
I
,
Mollica
A
.
Discovery of oxindole-based FLT3 inhibitors as a promising therapeutic lead for acute myeloid leukemia carrying the oncogenic ITD mutation
.
Arch Pharm
.
2023 Feb
356
2
2200407
.
7.
Malik
JA
,
Ahmed
S
,
Momin
SS
,
Shaikh
S
,
Alafnan
A
,
Alanazi
J
.
Drug repurposing : a new hope in drug discovery for prostate cancer
.
ACS Omega
.
2023
;
8
(
1
):
56
73
.
8.
Alamri
A
,
Rauf
A
,
Khalil
AA
,
Alghamdi
A
,
Alafnan
A
,
Alshammari
A
.
In silico screening of marine compounds as an emerging and promising approach against estrogen receptor alpha-positive breast cancer
.
BioMed Res Int
.
2021 Dec
2021
9734279
.
9.
Jha
NK
,
Arfin
S
,
Jha
SK
,
Kar
R
,
Dey
A
,
Gundamaraju
R
.
Re-establishing the comprehension of phytomedicine and nanomedicine in inflammation-mediated cancer signaling
.
Semin Cancer Biol
.
2022 Nov
86
Pt 2
1086
104
.
10.
Abbas
T
,
Keaton
MA
,
Dutta
A
.
Genomic instability in cancer
.
Cold Spring Harb Perspect Biol
.
2013 Mar
5
3
a012914
.
11.
Zhao
H
,
Wu
L
,
Yan
G
,
Chen
Y
,
Zhou
M
,
Wu
Y
.
Inflammation and tumor progression: signaling pathways and targeted intervention
.
Signal Transduct Target Ther
.
2021 Jul
6
1
263
46
.
12.
Greten
FR
,
Grivennikov
SI
.
Inflammation and cancer: triggers, mechanisms and consequences
.
Immunity
.
2019 Jul
51
1
27
41
.
13.
Choudhari
AS
,
Mandave
PC
,
Deshpande
M
,
Ranjekar
P
,
Prakash
O
.
Phytochemicals in cancer treatment: from preclinical studies to clinical practice
.
Front Pharmacol
.
2019
;
10
:
1614
7
.
14.
Bor
G
,
Mat Azmi
ID
,
Yaghmur
A
.
Nanomedicines for cancer therapy: current status, challenges and future prospects
.
Ther Deliv
.
2019 Jan
10
2
113
32
.
15.
Shi
J
,
Kantoff
PW
,
Wooster
R
,
Farokhzad
OC
.
Cancer nanomedicine: progress, challenges and opportunities
.
Nat Rev Cancer
.
2016 Nov
17
1
20
37
.
16.
Petros
RA
,
Desimone
JM
.
Strategies in the design of nanoparticles for therapeutic applications
.
Nat Rev Drug Discov
.
2010 Jul
9
8
615
27
.
17.
Li
J
,
Burgess
DJ
.
Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment
.
Acta Pharm Sin B
.
2020
;
10
(
11
):
2110
24
.
18.
Liu
Z
,
Jiang
W
,
Nam
J
,
Moon
JJ
,
Kim
BYS
.
Immunomodulating nanomedicine for cancer therapy
.
Nano Lett
.
2018
;
18
(
11
):
6655
9
.
19.
Astruc
D
.
Introduction to nanomedicine
.
Molecules
.
2015
21
1
E4
.
20.
Cooper
E
.
From Darwin and metchnikoff to burnet and beyond
.
Contrib Microbiol
.
2008
;
15
:
1
11
.
21.
Moses
SL
,
Edwards
VM
.
Cytotoxicity in MCF-7 and MDA-MB-231 breast cancer cells, without harming MCF-10a healthy cells
.
J Nanomed Nanotechnol
.
2016
07
02
).
22.
Swanner
J
,
Mims
J
,
Carroll
DL
,
Akman
SA
,
Furdui
CM
,
Torti
SV
.
Differential cytotoxic and radiosensitizing effects of silver nanoparticles on triple-negative breast cancer and non-triple-negative breast cells
.
Int J Nanomedicine
.
2015 Jun
10
3937
53
.
23.
Devulapally
R
,
Sekar
NM
,
Sekar
TV
,
Foygel
K
,
Massoud
TF
,
Willmann
JK
.
Polymer nanoparticles mediated codelivery of AntimiR-10b and AntimiR-21 for achieving triple negative breast cancer therapy
.
ACS Nano
.
2015 Mar
9
3
2290
302
.
24.
Shu
D
,
Li
H
,
Shu
Y
,
Xiong
G
,
Carson
WE
,
Haque
F
.
Systemic delivery of anti-miRNA for suppression of triple negative breast cancer utilizing RNA nanotechnology
.
ACS Nano
.
2015 Oct
9
10
9731
40
.
25.
Liu
Y
,
Zhu
YH
,
Mao
CQ
,
Dou
S
,
Shen
S
,
Tan
ZB
.
Triple negative breast cancer therapy with CDK1 siRNA delivered by cationic lipid assisted PEG-PLA nanoparticles
.
J Control Release
.
2014 Oct
192
114
21
.
26.
Deng
X
,
Cao
M
,
Zhang
J
,
Hu
K
,
Yin
Z
,
Zhou
Z
.
Hyaluronic acid-chitosan nanoparticles for co-delivery of MiR-34a and doxorubicin in therapy against triple negative breast cancer
.
Biomaterials
.
2014
;
35
(
14
):
4333
44
.
27.
Chen
G
,
Wang
Y
,
Xie
R
,
Gong
S
.
Tumor-targeted pH/redox dual-sensitive unimolecular nanoparticles for efficient siRNA delivery
.
J Control Release
.
2017 Aug
259
105
14
.
28.
Amani
H
,
Habibey
R
,
Shokri
F
,
Hajmiresmail
SJ
,
Akhavan
O
,
Mashaghi
A
.
Selenium nanoparticles for targeted stroke therapy through modulation of inflammatory and metabolic signaling
.
Sci Rep
.
2019 Dec
9
1
6044
.
29.
Hua
H
,
Kong
Q
,
Zhang
H
,
Wang
J
,
Luo
T
,
Jiang
Y
.
Targeting mTOR for cancer therapy
.
J Hematol Oncol
.
2019 Jul
12
1
71
19
.
30.
Miller-Kleinhenz
J
,
Guo
X
,
Qian
W
,
Zhou
H
,
Bozeman
EN
,
Zhu
L
.
Dual-targeting Wnt and uPA receptors using peptide conjugated ultra-small nanoparticle drug carriers inhibited cancer stem-cell phenotype in chemo-resistant breast cancer
.
Biomaterials
.
2018 Jan
152
47
62
.
31.
Basu
S
,
Harfouche
R
,
Soni
S
,
Chimote
G
,
Mashelkar
RA
,
Sengupta
S
.
Nanoparticle-mediated targeting of MAPK signaling predisposes tumor to chemotherapy
.
Proc Natl Acad Sci U S A
.
2009 May
106
19
7957
61
.
32.
Yu
Y
,
Yang
X
,
Reghu
S
,
Kaul
SC
,
Wadhwa
R
,
Miyako
E
.
Photothermogenetic inhibition of cancer stemness by near-infrared-light-activatable nanocomplexes
.
Nat Commun
.
2020 Aug
11
1
4117
4
.
33.
Pelon
F
,
Bourachot
B
,
Kieffer
Y
,
Magagna
I
,
Mermet-Meillon
F
,
Bonnet
I
.
Cancer-associated fibroblast heterogeneity in axillary lymph nodes drives metastases in breast cancer through complementary mechanisms
.
Nat Commun
.
2020 Jan
11
1
404
–.
34.
Falzone
L
,
Salomone
S
,
Libra
M
.
Evolution of cancer pharmacological treatments at the turn of the third millennium
.
Front Pharmacol
.
2018 Nov
9
1300
.
35.
Gao
S
,
Yang
D
,
Fang
Y
,
Lin
X
,
Jin
X
,
Wang
Q
.
Engineering nanoparticles for targeted remodeling of the tumor microenvironment to improve cancer immunotherapy
.
Theranostics
.
2019
;
9
(
1
):
126
51
.
36.
Jahangir
MA
,
Anand
C
,
Muheem
A
,
Gilani
SJ
,
Taleuzzaman
M
,
Zafar
A
.
Nano phytomedicine based delivery system for CNS disease
.
Curr Drug Metab
.
2020 May
21
9
661
73
.
37.
Praveen
TK
,
Gangadharappa
HV
,
Abu Lila
AS
,
Moin
A
,
Mehmood
K
,
Krishna
KL
.
Inflammation targeted nanomedicines: patents and applications in cancer therapy
.
Semin Cancer Biol
.
2022 Apr
86
Pt 2
645
63
.
38.
Ansari
MA
,
Thiruvengadam
M
,
Venkidasamy
B
,
Alomary
MN
,
Salawi
A
,
Chung
I-M
.
Exosome-based nanomedicine for cancer treatment by targeting inflammatory pathways: current status and future perspectives
.
Semin Cancer Biol
.
2022 Apr
86
Pt 2
678
96
.
39.
Wang
X
,
Semba
T
,
Phi
LTH
,
Chainitikun
S
,
Iwase
T
,
Lim
B
.
Targeting signaling pathways in inflammatory breast cancer
.
Cancers
.
2020
;
12
(
9
):
2479
19
.
40.
Al-Hatamleh
MAI
,
Ahmad
S
,
Boer
JC
,
Lim
J
,
Chen
X
,
Plebanski
M
.
A perspective review on the role of nanomedicine in the modulation of TNF-TNFR2 Axis in breast cancer immunotherapy
.
J Oncol
.
2019
;
2019
:
6313242
.
41.
Bedard
PL
,
Hyman
DM
,
Davids
MS
,
Siu
LL
.
Small molecules, big impact: 20 years of targeted therapy in oncology
.
Lancet
.
2020 Mar
395
10229
1078
88
.
42.
Liu
HL
,
Zhang
YL
,
Yang
N
,
Zhang
YX
,
Liu
XQ
,
Li
CG
.
A functionalized single-walled carbon nanotubeinduced autophagic cell death in human lung cells through Akt-TSC2-mTOR signaling
.
Cell Death Dis
.
2011 May
2
5
e159
.
43.
Li
C
,
Liu
H
,
Sun
Y
,
Wang
H
,
Guo
F
,
Rao
S
.
PAMAM nanoparticles promote acute lung injury by inducing autophagic cell death through the Akt-TSC2-mTOR signaling pathway
.
J Mol Cell Biol
.
2009 Oct
1
1
37
45
.
44.
Duan
J
,
Yu
Y
,
Yu
Y
,
Li
Y
,
Wang
J
,
Geng
W
.
Silica nanoparticles induce autophagy and endothelial dysfunction via the PI3K/Akt/mTOR signaling pathway
.
Int J Nanomedicine
.
2014 Nov
9
1
5131
41
.
45.
Song
M
,
Liu
T
,
Shi
C
,
Zhang
X
,
Chen
X
.
Bioconjugated manganese dioxide nanoparticles enhance chemotherapy response by priming tumor-Associated macrophages toward m1-like phenotype and attenuating tumor hypoxia
.
ACS Nano
.
2016 Jan
10
1
633
47
.
46.
He
L
,
Gu
J
,
Lim
LY
,
Yuan
ZX
,
Mo
J
.
Nanomedicine-mediated therapies to target breast cancer stem cells
.
Front Pharmacol
.
2016
7
SEP
313
3
.
47.
Farrell
D
,
Ptak
K
,
Panaro
NJ
,
Grodzinski
P
.
Nanotechnology-based cancer therapeutics--promise and challenge--lessons learned through the NCI Alliance for Nanotechnology in Cancer
.
Pharm Res
.
2011 Feb
28
2
273
8
.
48.
Jain
M
,
Venkatraman
G
,
Batra
SK
.
Optimization of radioimmunotherapy of solid tumors: biological impediments and their modulation
.
Clin Cancer Res
.
2007 Mar
13
5
1374
82
.
49.
Tan
DSW
,
Thomas
GV
,
Garrett
MD
,
Banerji
U
,
De Bono
JS
,
Kaye
SB
.
Biomarker-driven early clinical trials in oncology: a paradigm shift in drug development
.
Cancer J
.
2009 Sep
15
5
406
20
.
50.
Lim
DJ
,
Sim
M
,
Oh
L
,
Lim
K
,
Park
H
.
Carbon-based drug delivery carriers for cancer therapy
.
Arch Pharm Res
.
2014 Jan
37
1
43
52
.
51.
Bianco
A
,
Kostarelos
K
,
Prato
M
.
Applications of carbon nanotubes in drug delivery
.
Curr Opin Chem Biol
.
2005 Dec
9
6
674
9
.
52.
Dreaden
EC
,
Alkilany
AM
,
Huang
X
,
Murphy
CJ
,
El-Sayed
MA
.
The golden age: gold nanoparticles for biomedicine
.
Chem Soc Rev
.
2012 Mar
41
7
2740
79
.
53.
Daniel
MC
,
Astruc
D
.
Gold nanoparticles: assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology
.
Chem Rev
.
2004
;
104
:
293
346
.
54.
Libutti
SK
,
Paciotti
GF
,
Byrnes
AA
,
Alexander
HR
,
Gannon
WE
,
Walker
M
.
Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine
.
Clin Cancer Res
.
2010 Dec
16
24
6139
49
.
55.
Mahmoudi
M
,
Simchi
A
,
Imani
M
,
Shokrgozar
MA
,
Milani
AS
,
Häfeli
UO
.
A new approach for the in vitro identification of the cytotoxicity of superparamagnetic iron oxide nanoparticles
.
Colloids Surf B Biointerfaces
.
2010 Jan
75
1
300
9
.
56.
Gupta
AK
,
Gupta
M
.
Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications
.
Biomaterials
.
2005 Jun
26
18
3995
4021
.
57.
Jin
R
,
Lin
B
,
Li
D
.
Superparamagnetic iron oxide nanoparticles for MR imaging and therapy: design considerations and clinical applications
.
Curr Opin Pharmacol
.
2014 Oct
18
18
27
.
58.
Liong
M
,
Lu
J
,
Kovochich
M
,
Xia
T
,
Ruehm
SG
,
Nel
AE
.
Multifunctional inorganic nanoparticles for imaging, targeting, and drug delivery
.
ACS Nano
.
2008 May
2
5
889
96
.
59.
Gao
X
,
Cui
Y
,
Levenson
RM
,
Chung
LWK
,
Nie
S
.
In vivo cancer targeting and imaging with semiconductor quantum dots
.
Nat Biotechnol
.
2004 Aug
22
8
969
76
.
60.
Duncan
R
.
The dawning era of polymer therapeutics
.
Nat Rev Drug Discov
.
2003 May
2
5
347
60
.
61.
Brigger
I
,
Dubernet
C
,
Couvreur
P
.
Nanoparticles in cancer therapy and diagnosis
.
Adv Drug Deliv Rev
.
2002 Sep
54
5
631
51
.
62.
Delplace
V
,
Couvreur
P
,
Nicolas
J
.
Recent trends in the design of anticancer polymer prodrug nanocarriers
.
Polym Chem
.
2014 Mar
5
5
1529
44
.
63.
Duncan
R
,
Izzo
L
.
Dendrimer biocompatibility and toxicity
.
Adv Drug Deliv Rev
.
2005 Dec
57
15
2215
37
.
64.
Allen
TM
,
Cullis
PR
.
Liposomal drug delivery systems: from concept to clinical applications
.
Adv Drug Deliv Rev
.
2013 Jan
65
1
36
48
.
65.
Masood
F
.
Polymeric nanoparticles for targeted drug delivery system for cancer therapy
.
Mater Sci Eng C
.
2016
;
60
:
569
78
.
66.
Wang
Y
,
Santos
A
,
Evdokiou
A
,
Losic
D
.
An overview of nanotoxicity and nanomedicine research: principles, progress and implications for cancer therapy
.
J Mater Chem B
.
2015 Sep
3
36
7153
72
.
67.
Ma
S
,
Zhou
J
,
Zhang
Y
,
He
Y
,
Jiang
Q
,
Yue
D
.
Highly stable fluorinated nanocarriers with iRGD for overcoming the stability dilemma and enhancing tumor penetration in an orthotopic breast cancer
.
ACS Appl Mater Interfaces
.
2016 Oct
8
42
28468
79
.
68.
Docter
D
,
Westmeier
D
,
Markiewicz
M
,
Stolte
S
,
Knauer
SK
,
Stauber
RH
.
The nanoparticle biomolecule corona: lessons learned: challenge accepted
.
Chem Soc Rev
.
2015 Aug
44
17
6094
121
.
69.
Ke
PC
,
Lin
S
,
Parak
WJ
,
Davis
TP
,
Caruso
F
.
A decade of the protein corona
.
ACS Nano
.
2017 Dec
11
12
11773
6
.
70.
Salvati
A
,
Pitek
AS
,
Monopoli
MP
,
Prapainop
K
,
Bombelli
FB
,
Hristov
DR
.
Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface
.
Nat Nanotechnol
.
2013 Jan
8
2
137
43
.
71.
Mirshafiee
V
,
Kim
R
,
Park
S
,
Mahmoudi
M
,
Kraft
ML
.
Impact of protein pre-coating on the protein corona composition and nanoparticle cellular uptake
.
Biomaterials
.
2016 Jan
75
295
304
.
72.
Lunova
M
,
Prokhorov
A
,
Jirsa
M
,
Hof
M
,
Olżyńska
A
,
Jurkiewicz
P
.
Nanoparticle core stability and surface functionalization drive the mTOR signaling pathway in hepatocellular cell lines
.
Sci Rep
.
2017
;
7
(
1
):
16049
16
.
73.
Blanco
E
,
Hsiao
A
,
Mann
AP
,
Landry
MG
,
Meric-Bernstam
F
,
Ferrari
M
.
Nanomedicine in cancer therapy: innovative trends and prospects
.
Cancer Sci
.
2011 Jul
102
7
1247
52
.
74.
Sartaj
A
,
Baboota
S
,
Ali
J
.
Nanomedicine: a promising avenue for the development of effective therapy for breast cancer
.
Curr Cancer Drug Targets
.
2020 Mar
20
8
603
15
.
75.
Wei
QY
,
He
KM
,
Chen
JL
,
Xu
YM
,
Lau
ATY
.
Phytofabrication of nanoparticles as novel drugs for anticancer applications
.
Molecules
.
2019 Nov
24
23
4246
.
76.
Balakrishnan
S
,
Bhat
FA
,
Raja Singh
P
,
Mukherjee
S
,
Elumalai
P
,
Das
S
.
Gold nanoparticle-conjugated quercetin inhibits epithelial-mesenchymal transition, angiogenesis and invasiveness via EGFR/VEGFR-2-mediated pathway in breast cancer
.
Cell Prolif
.
2016 Dec
49
6
678
97
.
77.
Balakrishnan
S
,
Mukherjee
S
,
Das
S
,
Bhat
FA
,
Raja Singh
P
,
Patra
CR
.
Gold nanoparticles-conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 and MDA-MB-231)
.
Cell Biochem Funct
.
2017 Jun
35
4
217
31
.
78.
Organ
SL
,
Tsao
MS
.
An overview of the c-MET signaling pathway
.
Ther Adv Med Oncol
.
2011
3
1 Suppl l
S7
19
.
79.
Arora
A
,
Scholar
EM
.
Role of tyrosine kinase inhibitors in cancer therapy
.
J Pharmacol Exp Ther
.
2005 Dec
315
3
971
9
.
80.
Lavanya
V
,
Adil
M
,
Ahmed
N
,
Rishi
AK
,
Jamal
S
.
Small molecule inhibitors as emerging cancer therapeutics
.
Integr Cancer Sci Ther
.
2014
;
1
(
3
):
39
46
.
81.
Da Rocha Dias
S
,
Friedlos
F
,
Light
Y
,
Springer
C
,
Workman
P
,
Marais
R
.
Activated B-RAF is an Hsp90 client protein that is targeted by the anticancer drug 17-allylamino-17-demethoxygeldanamycin
.
Cancer Res
.
2005 Dec
65
23
10686
91
.
82.
Kamal
A
,
Thao
L
,
Sensintaffar
J
,
Zhang
L
,
Boehm
MF
,
Fritz
LC
.
A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors
.
Nature
.
2003 Sep
425
6956
407
10
.
83.
Ying
W
,
Du
Z
,
Sun
L
,
Foley
KP
,
Proia
DA
,
Blackman
RK
.
Ganetespib, a unique triazolone-containing Hsp90 inhibitor, exhibits potent antitumor activity and a superior safety profile for cancer therapy
.
Mol Cancer Ther
.
2012 Feb
11
2
475
84
.
84.
Soleja
M
,
Raj
GV
,
Unni
N
.
An evaluation of fulvestrant for the treatment of metastatic breast cancer
.
Expert Opin Pharmacother
.
2019 Oct
20
15
1819
29
.
85.
Sledge
GW
,
Toi
M
,
Neven
P
,
Sohn
J
,
Inoue
K
,
Pivot
X
.
Monarch 2: abemaciclib in combination with fulvestrant in women with HR+/HER2- advanced breast cancer who had progressed while receiving endocrine therapy
.
J Clin Oncol
.
2017 Sep
35
25
2875
84
.
86.
Sun
G
,
Rong
D
,
Li
Z
,
Sun
G
,
Wu
F
,
Li
X
.
Role of small molecule targeted compounds in cancer: progress, opportunities, and challenges
.
Front Cell Dev Biol
.
2021
;
9
:
694363
16
.
87.
Clere
N
,
Faure
S
,
Martinez
MC
,
Andriantsitohaina
R
.
Anticancer properties of flavonoids: roles in various stages of carcinogenesis
.
Cardiovasc Hematol Agents Med Chem
.
2011 Jun
9
2
62
77
.
88.
Hasima
N
,
Ozpolat
B
.
Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer
.
Cell Death Dis
.
2014 Nov
5
11
e1509
.
89.
Adams
LS
,
Phung
S
,
Yee
N
,
Seeram
NP
,
Li
L
,
Chen
S
.
Blueberry phytochemicals inhibit growth and metastatic potential of MDA-MB-231 breast cancer cells through modulation of the phosphatidylinositol 3-kinase pathway
.
Cancer Res
.
2010 May
70
9
3594
605
.
90.
Zhang
HW
,
Hu
JJ
,
Fu
RQ
,
Liu
X
,
Zhang
YH
,
Li
J
.
Flavonoids inhibit cell proliferation and induce apoptosis and autophagy through downregulation of PI3Kγ mediated PI3K/AKT/mTOR/p70S6K/ULK signaling pathway in human breast cancer cells
.
Sci Rep
.
2018 Jul
8
1
11255
13
.
91.
Koosha
S
,
Alshawsh
MA
,
Looi
CY
,
Seyedan
A
,
Mohamed
Z
.
An association map on the effect of flavonoids on the signaling pathways in colorectal cancer
.
Int J Med Sci
.
2016 Apr
13
5
374
85
.
92.
Kunnumakkara
AB
,
Bordoloi
D
,
Harsha
C
,
Banik
K
,
Gupta
SC
,
Aggarwal
BB
.
Curcumin mediates anticancer effects by modulating multiple cell signaling pathways
.
Clin Sci
.
2017 Aug
131
15
1781
99
.
93.
Li
L
,
Braiteh
FS
,
Kurzrock
R
.
Liposome-encapsulated curcumin: in vitro and in vivo effects on proliferation, apoptosis, signaling, and angiogenesis
.
Cancer
.
2005 Sep
104
6
1322
31
.
94.
George
BP
,
Chandran
R
,
Abrahamse
H
.
Role of phytochemicals in cancer chemoprevention: insights
.
Antioxidants
.
2021
;
10
(
9
):
1455
.
95.
Shukla
S
,
Bhaskaran
N
,
Babcook
MA
,
Fu
P
,
MacLennan
GT
,
Gupta
S
.
Apigenin inhibits prostate cancer progression in TRAMP mice via targeting PI3K/Akt/FoxO pathway
.
Carcinogenesis
.
2014 Feb
35
2
452
60
.
96.
Mafuvadze
B
,
Liang
Y
,
Besch-Williford
C
,
Zhang
X
,
Hyder
SM
.
Apigenin induces apoptosis and blocks growth of medroxyprogesterone acetate-dependent BT-474 xenograft tumors
.
Horm Cancer
.
2012 Aug
3
4
160
71
.
97.
Li
J
,
Zhang
C
,
Jiang
H
,
Cheng
J
.
Andrographolide inhibits hypoxia-inducible factor-1 through phosphatidylinositol 3-kinase/AKT pathway and suppresses breast cancer growth
.
Onco Targets Ther
.
2015 Feb
8
427
35
.
98.
Tohkayomatee
R
,
Reabroi
S
,
Tungmunnithum
D
,
Parichatikanond
W
,
Pinthong
D
.
Andrographolide exhibits anticancer activity against breast cancer cells (MCF-7 and MDA-MB-231 cells) through suppressing cell proliferation and inducing cell apoptosis via inactivation of ER-α receptor and PI3K/AKT/mTOR signaling
.
Molecules
.
2022 Jun
27
11
3544
.
99.
Chung
H
,
Choi
HS
,
Seo
EK
,
Kang
DH
,
Oh
ES
.
Baicalin and baicalein inhibit transforming growth factor-β1-mediated epithelial-mesenchymal transition in human breast epithelial cells
.
Biochem Biophys Res Commun
.
2015 Mar
458
3
707
13
.
100.
Yan
W
,
Ma
X
,
Zhao
X
,
Zhang
S
.
Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro
.
Drug Des Devel Ther
.
2018
;
12
:
3961
72
.
101.
Vadhan-Raj
S
,
Weber
DM
,
Wang
M
,
Giralt
SA
,
Thomas
SK
,
Alexanian
R
.
Curcumin downregulates NF-kB and related genes in patients with multiple myeloma: results of a phase I/II study
.
Blood
.
2007 Nov
110
11
1177
.
102.
Hu
S
,
Xu
Y
,
Meng
L
,
Huang
L
,
Sun
H
.
Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells
.
Exp Ther Med
.
2018 Aug
16
2
1266
72
.
103.
Tuorkey
MJ
.
Molecular targets of luteolin in cancer
.
Eur J Cancer Prev
.
2016
;
25
(
1
):
65
76
.
104.
Batra
P
,
Sharma
AK
.
Anti-cancer potential of flavonoids: recent trends and future perspectives
.
3 Biotech
.
2013 Feb
3
6
439
59
.
105.
Iwanowycz
S
,
Wang
J
,
Hodge
J
,
Wang
Y
,
Yu
F
,
Fan
D
.
Emodin inhibits breast cancer growth by blocking the tumor-promoting feedforward loop between cancer cells and macrophages
.
Mol Cancer Ther
.
2016 Aug
15
8
1931
42
.
106.
Lin
W
,
Zhong
M
,
Yin
H
,
Chen
Y
,
Cao
Q
,
Wang
C
.
Emodin induces hepatocellular carcinoma cell apoptosis through MAPK and PI3K/AKT signaling pathways in vitro and in vivo
.
Oncol Rep
.
2016 Aug
36
2
961
7
.
107.
Akkol
EK
,
Tatlı
II
,
Karatoprak
,
Ağar
OT
,
Yücel
Ç
,
Sobarzo‐sánchez
E
.
Is emodin with anticancer effects completely innocent? Two sides of the coin
.
Cancers
.
2021 Jun
13
11
2733
.
108.
Xue
L
,
Zhang
WJ
,
Fan
QX
,
Wang
LX
.
Licochalcone a inhibits PI3K/AKt/mTOR signaling pathway activation and promotes autophagy in breast cancer cells
.
Oncol Lett
.
2018 Feb
15
2
1869
73
.
109.
Arita
M
,
Koike
J
,
Yoshikawa
N
,
Kondo
M
,
Hemmi
H
.
Licochalcone A inhibits BDNF and TrkB gene expression and hypoxic growth of human tumor cell lines
.
Int J Mol Sci
.
2020 Jan
21
2
506
.
110.
Saleh
HA
,
Yousef
MH
,
Abdelnaser
A
.
The anti-in fl ammatory properties of phytochemicals and their effects on epigenetic mechanisms involved in TLR4/NF- k B-mediated in fl ammation
.
Front Immunol
.
2021
;
12
:
606069
.
111.
Song
YK
,
Yoon
JH
,
Woo
JK
,
Kang
JH
,
Lee
KR
,
Oh
SH
.
Quercetin is a flavonoid breast cancer resistance protein inhibitor with an impact on the oral pharmacokinetics of sulfasalazine in rats
.
Pharmaceutics
.
2020
;
12
(
5
):
397
.
112.
Kumar
D
,
Shankar
S
,
Srivastava
RK
.
Rottlerin-induced autophagy leads to the apoptosis in breast cancer stem cells: molecular mechanisms
.
Mol Cancer
.
2013 Dec
12
1
171
15
.
113.
Rahman
MA
,
Hannan
MA
,
Dash
R
,
Rahman
MH
,
Islam
R
,
Uddin
MJ
.
Phytochemicals as a complement to cancer chemotherapy: pharmacological modulation of the autophagy-apoptosis pathway
.
Front Pharmacol
.
2021
;
12
:
639628
0
.
114.
Song
C
,
Bae
Y
,
Jun
JJ
,
Lee
H
,
Kim
ND
,
Lee
KB
.
Identification of TG100-115 as a new and potent TRPM7 kinase inhibitor, which suppresses breast cancer cell migration and invasion
.
Biochim Biophys Acta Gen Subj
.
2017 Apr
1861
4
947
57
.
115.
Ma
X
,
Ning
S
.
Cyanidin-3-glucoside attenuates the angiogenesis of breast cancer via inhibiting STAT3/VEGF pathway
.
Phytother Res
.
2019 Jan
33
1
81
9
.
116.
Chen
J
,
Zhu
Y
,
Zhang
W
,
Peng
X
,
Zhou
J
,
Li
F
.
Delphinidin induced protective autophagy via mTOR pathway suppression and AMPK pathway activation in HER-2 positive breast cancer cells
.
BMC Cancer
.
2018 Mar
18
1
342
.
117.
Zhong
L
,
Li
Y
,
Xiong
L
,
Wang
W
,
Wu
M
,
Yuan
T
.
Small molecules in targeted cancer therapy: advances, challenges, and future perspectives
.
Signal Transduct Target Ther
.
2021
;
6
(
1
):
201
48
.
118.
Ariaans
G
,
Jalving
M
,
de Vries
EGE
,
de Jong
S
.
Anti-tumor effects of everolimus and metformin are complementary and glucose-dependent in breast cancer cells
.
BMC Cancer
.
2017 Mar
17
1
1
13
.
119.
Drilon
A
,
Laetsch
TW
,
Kummar
S
,
DuBois
SG
,
Lassen
UN
,
Demetri
GD
.
Efficacy of larotrectinib in TRK fusion–positive cancers in adults and children
.
N Engl J Med
.
2018 Feb
378
8
731
9
.
120.
Cocco
E
,
Scaltriti
M
,
Drilon
A
.
NTRK fusion-positive cancers and TRK inhibitor therapy
.
Nat Rev Clin Oncol
.
2018 Dec
15
12
731
47
.
121.
Sharma
VR
,
Sharma
AK
,
Punj
V
,
Priya
P
.
Recent nanotechnological interventions targeting PI3K/Akt/mTOR pathway: a focus on breast cancer
.
Semin Cancer Biol
.
2019 Dec
59
133
46
.
122.
Duross
AN
,
Neufeld
MJ
,
Landry
MR
,
Rosch
JG
,
Eaton
CT
,
Sahay
G
.
Micellar formulation of talazoparib and buparlisib for enhanced DNA damage in breast cancer chemoradiotherapy
.
ACS Appl Mater Interfaces
.
2019 Apr
11
13
12342
56
.
123.
Mai
TT
,
Moon
JY
,
Song
YW
,
Viet
PQ
,
Phuc
PV
,
Lee
JM
.
Ginsenoside F2 induces apoptosis accompanied by protective autophagy in breast cancer stem cells
.
Cancer Lett
.
2012 Aug
321
2
144
53
.
124.
Mohapatra
P
,
Preet
R
,
Das
D
,
Satapathy
SR
,
Choudhuri
T
,
Wyatt
MD
.
Quinacrine-mediated autophagy and apoptosis in colon cancer cells is through a p53- and p21-dependent mechanism
.
Oncol Res
.
2012
20
2–3
81
91
.
125.
Sun
C-Y
,
Zhu
Y
,
Li
X-F
,
Tang
L-P
,
Su
Z-Q
,
Wang
X-Q
.
Norcantharidin alone or in combination with crizotinib induces autophagic cell death in hepatocellular carcinoma by repressing c-Met-mTOR signaling
.
Oncotarget
.
2017 Dec
8
70
114945
55
.
126.
Hussain
Z
,
Khan
JA
,
Murtaza
S
.
Nanotechnology: an emerging therapeutic option for breast cancer
.
Crit Rev Eukaryot Gene Expr
.
2018
;
28
(
2
):
163
75
.
127.
Tong
R
,
Kohane
DS
.
New strategies in cancer nanomedicine
.
Annu Rev Pharmacol Toxicol
.
2016 Jan
56
41
57
.
128.
Karahaliloğlu
Z
,
Kilicay
E
,
Alpaslan
P
,
Hazer
B
,
Baki Denkbas
E
.
Enhanced antitumor activity of epigallocatechin gallate–conjugated dual-drug-loaded polystyrene–polysoyaoil–diethanol amine nanoparticles for breast cancer therapy
.
J Bioact Compat Polym
.
2017 Jun
33
1
38
62
.
129.
Liu
Y
,
Qiao
L
,
Zhang
S
,
Wan
G
,
Chen
B
,
Zhou
P
.
Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy
.
Acta Biomater
.
2018 Jan
66
310
24
.
130.
Yang
H
,
Liang
H
,
Xie
Y
,
Chen
Q
.
A cancer cell turn-on protein-CuSMn nanoparticle as the sensor of breast cancer cell and CH3O-PEG-phosphatide
.
Chin Chem Lett
.
2018 Oct
29
10
1528
32
.
131.
Graham-Gurysh
E
,
Kelkar
S
,
McCabe-Lankford
E
,
Kuthirummal
N
,
Brown
T
,
Kock
ND
.
Hybrid donor-acceptor polymer particles with amplified energy transfer for detection and on-demand treatment of breast cancer
.
ACS Appl Mater Interfaces
.
2018 Mar
10
9
7697
703
.
132.
Yu
S
,
Zhang
YI
,
Chen
L
,
Li
Q
,
Du
J
,
Gao
Y
.
Antitumor effects of carbon nanotube-drug complex against human breast cancer cells
.
Exp Ther Med
.
2018 Aug
16
2
1103
10
.
133.
Gener
P
,
Montero
S
,
Xandri-Monje
H
,
Díaz-Riascos
ZV
,
Rafael
D
,
Andrade
F
.
ZileutonTM loaded in polymer micelles effectively reduce breast cancer circulating tumor cells and intratumoral cancer stem cells
.
Nanomedicine
.
2020
;
24
:
102106
.
134.
Bartusik-Aebisher
D
,
Chrzanowski
G
,
Bober
Z
,
Aebisher
D
.
An analytical study of Trastuzumab-dendrimer-fluorine drug delivery system in breast cancer therapy in vitro
.
Biomed Pharmacother
.
2021 Jan
133
111053
.
135.
Shashaani
H
,
Faramarzpour
M
,
Hassanpour
M
,
Namdar
N
,
Alikhani
A
,
Abdolahad
M
.
Silicon nanowire based biosensing platform for electrochemical sensing of Mebendazole drug activity on breast cancer cells
.
Biosens Bioelectron
.
2016 Nov
85
363
70
.
136.
van der Meel
R
,
Lammers
T
,
Hennink
WE
.
Cancer nanomedicines: oversold or underappreciated
.
Expert Opin Drug Deliv
.
2017
;
14
(
1
):
1
5
.
137.
Wolfram
J
,
Ferrari
M
.
Clinical cancer nanomedicine
.
Nano Today
.
2019 Apr
25
85
98
.
138.
Gunasekaran
T
,
Haile
T
,
Nigusse
T
,
Dhanaraju
MD
.
Nanotechnology: an effective tool for enhancing bioavailability and bioactivity of phytomedicine
.
Asian Pac J Trop Biomed
.
2014 May
4
Suppl 1
S1
7
.
139.
Ansari
SH
,
Islam
F
,
Sameem
M
.
Influence of nanotechnology on herbal drugs: a Review
.
J Adv Pharm Technol Res
.
2012 Jul
3
3
142
6
.
140.
Mirzaei
SA
,
Dinmohammadi
F
,
Alizadeh
A
,
Elahian
F
.
Inflammatory pathway interactions and cancer multidrug resistance regulation
.
Life Sci
.
2019 Oct
235
116825
.