Cancer is among the most devastating diseases triggered by genetic mutation or due to complex intercellular, intracellular, and/or non-cellular cross-talks in the body [1, 2]. It is characterized by an abnormal growth of cells having overwhelming potential of invasion and metastasis to other tissues/organs of the body. In contrast to benign tumors that restrict to the primary sites, cancers are characterized by massive potential to spread to other parts of the body. Over 100 different types of cancer have been diagnosed in human, among them, prostate cancer (PC) is the second most aggressive and prevalent type of cancer in men with highest mortality rates after the lung cancer [1, 2]. According to the recent Cancer Statistics, percentage of population diagnosed with PC at advanced stages has alarmingly increased from 3.9–8.2% over the past decade. Owing to its enormous ability of tumorigenesis, metastasis (local or distant), multi-drug resistance (MDR), and poor disease-free survival (DFS) and overall survival (OS) rates, PC is the second leading cause of cancer-related mortalities in men (more than 370,000 deaths globally) [1, 2].
A wide variety of FDA-approved chemotherapeutics are being clinically prescribed for treatment of PC, alone or in conjunction with other viable treatments such as radiotherapy, surgery, hormonal replacement therapy, bone marrow transplant, immunotherapy, and/or personalized medicines [3, 4]. The conventional therapies produce reasonable clinical response in PC patients; however, their therapeutic significance is inadequate due to various drug-related factors such as low aqueous solubility, undesirable pharmacokinetics, narrow therapeutic index, non-specific biodistribution, and off-target adverse events [3–5]. In addition, several biological factors including, genetic mutation, altered epigenetics, enhanced drug efflux, activation of specific signaling pathways, and MDR also contribute to failure of existing conventional therapies and poor DFS and OS rates in PC patients [6, 7]. MDR is not solely related to ability of cancer cells to survive against the existing chemotherapeutics, but it is also associated with TME which play a crucial pro-oncogenic role in promoting the progression of cancer, metastasis to other body tissues, and chemoresistance.
TME consists of extracellular matrix (ECM), stromal cells (e.g., MSCs, fibroblasts, adipocytes, vasculature, etc.), red blood cells, pericytes, various immune players (e.g., macrophages, natural killer cells, dendritic cells, and lymphocytes), and cancer cells (Fig. 1B). Among various immune players, tumor-associated macrophages (TAMs) are the most abundant cells within TME that play a crucial role in the development and progression of TME via the dysregulation of various cytokines, chemokines, growth factors, angiogenesis, and immune functioning [6, 7]. Within the TME, TAMs can be polarized into two different phenotypes such as M1 (anti-inflammatory and anti-tumor) and M2 (inflammatory and pro-tumor) which are unique and distinct in functioning. M2-subtype of TAMs are of crucial concern because they promote tumor growth, metastasis, invasion, TME remodelling, and immunosuppression [6, 7]. Therefore, while designing a newer anti-neoplastic therapy, M2-subtype of TAMs should be considered as an important chemo-target in addition to cancer cells for tumbling the cancer-related fatalities.
With the advent of versatile nanotechnology-based nanocarriers such as polymeric nanoparticles, liposomes, niosomes, micelles, dendrimers, solid lipid nanoparticles, nanostructured lipid carriers, and inorganic nanoparticles, majority of challenges associated with conventional chemotherapeutics have been reasonably addressed [8–11]. However, it is worth mentioning that despite relative success of nanotechnology-based nanocarriers for treatment of PC in in vitro and in vivo models, nano-therapies are still below the expectation to provide rational therapeutic outcomes along with adequate pharmacokinetics, pharmacodynamics, and safety in humans [8–11].
To counter these challenges, various specialized strategies have been adapted in the design of nanocarriers including the passive and active targeting approaches. The passive targeting is achieved through Enhanced Permeation and Retention (EPR) effect which enables preferential diffusion of systematically available nanocarriers into TME due to poor lymphatic drainage and leaky vasculature in the tumor tissues [12, 13]. Notably, size of nanocarriers play a crucial role to attain an adequate EPR effect, and optimum size range for passive distribution of nanocarriers to TME is < 200 nm [12, 13]. On the other hand, nanocarriers have been uniquely functionalized with various targeting ligands (i.e., targeting peptides, aptamers, antibodies, and small molecules) for selective targeting of tumor cells having distinct biology (e.g., overexpressed CD44, folic acid, P-glycoprotein, transferrin receptors, etc.) [14–17]. Plethora of studies have reported that targetable NPs (passive and active) have shown better results in terms of precise site-specific targeting via recognizing specific biomarkers overexpressed on cancer cells, improving pharmacokinetics, and mitigating off-target effects compared to non-targetable NPs [14–17]. Undoubtedly, passive and active targeting strategies have shown obvious improvement in anticancer response of NPs in in vitro and in vivo models; however, literature survey of past two decades indicated that, on average, < 1% of injected NPs (targeted and non-targeted) had shown uptake into TME of solid malignant tumors [18], and these findings were really disappointment in human models.
To deal with this, cleverly designed nanocarriers are an obvious need of the current time to maximize the biodistribution of chemotherapeutics into solid malignant tumors. Dual-targeting strategy is a smart approach to resolve majority of issues related to nanocarriers [19, 20]. Dual-targeting strategy defines as anchoring of two targeting ligands on NPs corona to recognize and competitively bind to two different receptors overexpressed on cancer cells, or conjugation of one targeting ligand on NPs corona to recognize and competitively bind to two different receptors or two different target cells. Dual-targeting strategy could provide better selectivity, efficient internalization via multiple endocytosis mechanisms, inhibition of MDR, and treatment of multiple types of cancers concurrently [19, 20].
Herein, we aimed to construct polymeric nanospheres (PNSPs) as a double-edge sword for dual action against PC cells and M2-subtype of TAMs residing in the TME (Fig. 1). These PNSPs were made up of chitosan (CS) matrix encapsulated with two chemotherapeutic agents, methotrexate (MTX) and zoledronic acid (ZA) for synergistic action against PC. The ZA is a highly potent third-generation bisphosphonate with proven anticancer efficacy against PC via induction of apoptosis, inhibition of tumor invasion, downregulation of angiogenesis, and regression of tumor proliferation [21]. Likewise, MTX is a well-recognized antineoplastic agent with potent anti-oncogenic efficacy against a wide variety of cancers [22]. Owing to its folic acid-like structure, MTX has also been used as a targeting ligand for folate receptors that are heavily expressed on different types of cancer, including the PC [23]. To install dual targeting feature, engineered MTX/ZA co-loaded PNSPs were functionalized with hyaluronic acid (HA) as a targeting ligand. HA is a natural carbohydrate polymer consisting of D-glucuronic acid and N-acetyl-d-glucosamine units and it exhibits great specificity towards integral membrane glycoprotein known as cluster differentiation-44 (CD44) [24]. CD44 is a cell surface receptor heavily expressed on many types of cells, including the PC cells and TAMs, and hence has been chosen as a biological target for dual action against both PC cells and TAMs. The developed HA-decorated nanospheres (HA-PNSPs) were extensively optimized and characterized for particle size, polydispersity index (PDI), zeta potential, encapsulation efficiency (%EE), loading capacity (%LC), morphology, colloidal stability, crystallographic structure, molecular docking with CD44 receptors, in vitro release kinetics, cell uptake efficiency, cytotoxicity (IC50) against PC cells and TAMs, specific cell internalization via CD44-mediated endocytosis, and anti-metastatic potential.