Next Article in Journal
Epigenetic Regulation Mediated by Sphingolipids in Cancer
Next Article in Special Issue
GSK2801 Reverses Paclitaxel Resistance in Anaplastic Thyroid Cancer Cell Lines through MYCN Downregulation
Previous Article in Journal
Convenient and Sensitive Measurement of Lactosylceramide Synthase Activity Using Deuterated Glucosylceramide and Mass Spectrometry
Previous Article in Special Issue
Canonical Wnt Pathway Is Involved in Chemoresistance and Cell Cycle Arrest Induction in Colon Cancer Cell Line Spheroids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer

Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5293; https://doi.org/10.3390/ijms24065293
Submission received: 2 February 2023 / Revised: 6 March 2023 / Accepted: 8 March 2023 / Published: 10 March 2023

Abstract

:
Prostate cancer (PC) is the third most frequently diagnosed cancer worldwide and the second most frequent in men. Several risk factors can contribute to the development of PC, and those include age, family history, and specific genetic mutations. So far, drug testing in PC, as well as in cancer research in general, has been performed on 2D cell cultures. This is mainly because of the vast benefits these models provide, including simplicity and cost effectiveness. However, it is now known that these models are exposed to much higher stiffness; lose physiological extracellular matrix on artificial plastic surfaces; and show changes in differentiation, polarization, and cell–cell communication. This leads to the loss of crucial cellular signaling pathways and changes in cell responses to stimuli when compared to in vivo conditions. Here, we emphasize the importance of a diverse collection of 3D PC models and their benefits over 2D models in drug discovery and screening from the studies done so far, outlining their benefits and limitations. We highlight the differences between the diverse types of 3D models, with the focus on tumor–stroma interactions, cell populations, and extracellular matrix composition, and we summarize various standard and novel therapies tested on 3D models of PC for the purpose of raising awareness of the possibilities for a personalized approach in PC therapy.

1. Prostate Cancer

Prostate cancer (PC) is the third most frequently diagnosed cancer worldwide, and the second most frequent in men, with 1.4 million cases diagnosed in 2020. In men, it is the most diagnosed cancer in 112 countries, including North and South America, Australia, and most European and African countries. The mortality differs between the high- and low-HDI (human development index), with high-HDI countries showing higher mortality rates (37.5 per 100,000) compared to low-HDI countries (5.9 per 100,000) [1]. The 5-year survival of the early-stage disease is as high as 95%, but after the PC metastasizes, the 5-year survival drops to 30% [2]. PC most often forms metastases in the bone (84%), distant lymph nodes (10.6%), liver (10.2%), and thorax (9.1%) [3].
Risk factors for developing PC include age, family history, and specific genetic mutations. Hereditary prostate cancer (HPC) is defined with three specific parameters: (a) PC in three successive generations, (b) at least two cases of PC in the family at an age of onset <55 years, and (c) three or more first-degree relatives with PC. It is still unclear if HPC differs from the sporadic disease, but men with HPC show earlier disease onset [4]. Germline mutations may affect the development and aggressiveness of PC. A recent systematic review identified germline mutations in genes involved in homologous recombination (BRCA1/2, ATM, CHEK2, NBN), mismatch repair (MLH1, MLH2, MSH6), embryonic development and regeneration (HOXB13), and regulation of the cell cycle (ATM) [5,6], while a prospective screening program associated pathogenic variants of MSH2 and MSH6 with higher PCR incidence [7]. It has also been demonstrated that men of African descent show higher PC incidence and mortality than men of other ancestry, and reports show significant differences in genomes and transcriptomes between PC of African vs. European ancestry [8,9].
There are also several environmental risk factors that contribute to the disease, such as smoking and excess body weight. Smoking has been associated with an increased relative risk of PC [10]. A recent meta-analysis investigated the role of modifiable risk factors in lower-income countries and found an association of higher fat intake with increased PC risk and of higher vegetable intake and tea consumption with lower PC risk, while alcohol, smoking, red meat intake, and high body mass index showed a trend towards increased risk [11].

2. Diagnosis and Therapy of PC

Diagnosis of PC is usually done by combining the measurement of prostate-specific antigen (PSA) blood levels and digital rectal examination, followed by multi-parametric magnetic resonance imaging and biopsy if necessary [12]. However, PSA values may be affected by other factors, such as age, body mass index, prostate volume, and genetic predisposition [13]. It has been suggested that the Prostate Health Index (Phi), which is obtained by comparing relative concentrations of total PSA, free PSA, and [-2]proPSA, provides a better prediction of aggressive PC than total and free PSA [14]. Biopsy can stratify the disease into low- (T1/2, Gleason score ≤ 6, PSA ≤ 10), intermediate-, or high-risk disease. Intermediate-risk patients are then staged for metastases using MRI or PET-CT and bone scan, while high-risk patients are staged using CT and bone scan [12]. For low-risk PC, the recommended treatment option is active surveillance using PSA [15], but curative options, such as radical prostatectomy (RP), external beam radiotherapy (RT), and low-dose-brachytherapy, can be used [12]. Intermediate-risk patients have the same options as the low-risk, with the addition of neoadjuvant and concurrent androgen deprivation therapy (ADT), while the high-risk have the option of neoadjuvant ADT and/or docetaxel-based therapy or RP with pelvic lymphadenectomy [12]. If the disease progresses during ADT, it is considered castration-resistant prostate cancer (CRPC). Metastatic PC can be hormone-naïve or castration-resistant. In the case of hormone-naïve disease, ADT is combined with chemotherapy, and for the metastatic CRPC (mCRPC), chemotherapy can be combined with a second androgen receptor (AR) inhibitor [12]. BRCA-deficient mCRPC can be treated with PARP inhibitors [16,17], while mCRPC with alterations in the PI3K/AKT signaling pathway, mostly due to deletion or mutation of PTEN [18], can be treated with AKT inhibitor Ipataserib [16]. Patients with mCRPC with disease progression after treatment with enzalutamide or abiraterone could be treated with Olaparib, which has demonstrated longer progression-free survival [19]. Other novel treatment options include the PSMA-targeted radio ligand therapy (RLT), immunotherapy, or cell-based immunotherapy, but these approaches still have limited success and are currently being tested [16,20].
The emergence of CRPC following ADT is still the major problem in the therapy of PC. ADT is based on the inhibition of the androgen receptor (AR), which is the major driver of PC and, therefore, the most important drug target. AR drives metabolic reprogramming in PC compared to healthy prostate, and this reprogramming is even more pronounced in response to chemotherapy [21,22]. Clinical studies of second-generation antiandrogens, such as enzalutamide, abiraterone, apalutamide, and darolutamide, have demonstrated effectiveness in prolonging survival time and decreasing PSA levels [23,24,25,26].
Mechanisms leading to PC progression and ADR resistance are usually associated with amplifications, mutations, altered splicing, or epigenetic reprogramming in the AR gene [27,28,29]. Another potential driver is the increase in androgen biosynthesis, which can occur through the mutation in the HSD3B1 gene [30]. Mutations in other genes may also contribute to the risk, and a panel of four genetic mutations (MSH2, CDK12, TP53, and RB1) has been proposed as a predictor of risk for early progression [31]. Complex chromosome rearrangements, or chromoplexy, which include many cancer-associated genes, have been associated with prostate cancerogenesis [32]. Therefore, many drugs are being repurposed for new application in treating CRPC [33]. New drugs and combinations of drugs designed to bypass this resistance are being tested in vitro and in vivo, such as selective AR degraders [34], AR-GR (glucocorticoid receptor) dual antagonists [35], proteolysis-targeting chimeras (PROTACs) [36], or targeted delivery of radioligands [37].
Drug testing has been traditionally performed on 2D cell cultures, where cells are grown in a monolayer attached to the surface of the dish. This model is convenient and easy to maintain, and still largely used in many laboratories worldwide. However, this model does not represent the 3D architecture of the tumor found in vivo, nor its complexity regarding the many cell types found within the tumor mass or surrounding it. The role of the tumor microenvironment has been identified as critical in facilitating tumor growth, especially the cancer-associated fibroblast population [38]. Therefore, the use of advanced 3D models is coming into focus as more biologically relevant for the purpose of identification of novel factors contributing to PC tumorigenesis and resistance, but also for drug testing.

3. Three-Dimensional Prostate Cancer Models in Drug Discovery

3.1. Spheroids vs. Organoids vs. Tumoroids

Spheroid and organoid cultures are 3D cultures composed of multiple cells with distinct and overlapping purposes. They can be useful in 3D cell research, but differ in cellular sources and protocol for establishment [39]. Spheroids are spherical clusters of broad-ranging cells, usually generated from established cell lines. They are generally cultured as free-floating aggregates, with no need for a scaffolding environment to form 3D cultures. They are considered as structures of low complexity in mirroring tumor organization, but are very popular, as they emulate properties of solid tumors in several aspects [40]. Nevertheless, they are not able to self-assemble or regenerate, and thus are not as advanced as organoids. Even though cell–cell and cell–ECM interactions are present in the spheroids, when they are larger than 500 μm, they represent non-vascularized or poorly vascularized tumors [40]. They consist of proliferating cells in the outer layer, with quiescent cells in the middle, and hypoxic and necrotic cells in the inner layer, and the metabolites are distributed in a gradient through the structure. The term prostasphere stands for self-associated PC cell lines in suspension that grow as unattached spheroids, therefore, meaning that the terms prostaspheres and PC spheroids are being used interchangeably [41,42].
Organoids are complex clusters of organ-specific cells, either stem cells or progenitor cells. Together with a given scaffolding extracellular matrix, or collagen, they can self-assemble. Histologically and genetically, they resemble the original tumor from which they were derived, both in structure and function [39,43]. They can be cultured from a very small amount of tissue and are easy for genetic manipulations [44,45]. Organoids can be maintained in long-term culture and can be cryopreserved [39].
The difference between organoids and tumoroids is that organoids lack the full complement of cells and factors found in a patient’s tumor, while tumoroids retain the full architecture of the tumor microenvironment (TME) and extracellular matrix (ECM). The cells included in the tumoroid include multiple support cells, such as cancer-associated fibroblasts (CAFs), endothelial cells, and pericytes; immune cells, such as lymphocytes, neutrophils, dendritic cells (DCs), and monocytes; and less-prevalent cells, such as myeloid-derived suppressor cells (MDSCs), mesenchymal stromal cells (MSCs), and platelets [46]. This is due to the fact that tumoroids are grown directly from fresh patient tumor tissue. Maintaining the TME and ECM is of great importance, especially in drug testing studies, because it provides the true response to conventional chemotherapeutic and targeted therapies [47].
Spheroids, in general, and prostate spheroids, as well, show upregulation of stemness markers, such as CD44, GLI1, ABCG2, and BMI1 [48]. Generation of spheroids can be used as a step in the process of enrichment of cancer stem cells (CSCs) from a cell population. Spheroids are formed, then disassociated and sorted for specific markers of stemness (e.g., CD44, CD133), thus obtaining a CSC-rich population of cells [49], and this can be applied to primary PC cultures and clinical samples, as well [50,51].

3.2. Methods for Growing 3D PC Models

3.2.1. Suspension Cell Cultures

There are several methods to develop 3D models in PC (Figure 1). The first and most popular is the self-assembly of PC cells in non-adherent culture conditions, which limits attachment of the cells on surfaces, for instance, by using agarose coating or non-adherent plastic dishes. This method is simple, low-cost, and offers a consistent yield, and it is suitable for multicellular spheroids [52]. However, some limitations lie in the difficulty controlling the spheroid size and the lack of extracellular matrix (ECM) surrogates. The uniformity of the spheroids can be achieved by generating microwells on the surface of the agarose layer, which enables generation of spheroids of a specific size [53], or by seeding a defined number of cells in each well of the round-bottomed specialized plates. In addition, this model represents problems with drug testing since it is not suitable for migration/invasion, and no cell viability assay has been developed so far [54,55,56,57,58,59,60,61]. Therefore, this model is most often used for the spheroid formation assay, where the number and size of spheroids is compared between non-treated and treated cells. A large-scale approach for this model includes the use of bioreactors with the rotating wall vessels, where a large number of suspension spheroids can be cultivated [62]. Spheroids can also be generated with microgravity, and PC cells exposed to microgravity separate into two populations, the adherent cells and the spheroids in the suspension above the adherent cells [63,64].

3.2.2. Hanging Drop

The hanging drop method is the second approach, very similar in its characteristics to the suspension culture. Cancer cells are seeded and incubated in hanging drops until they form rounded structures characterized by stable cell–cell contacts [65]. The drops can be formed by hanging the cell suspension from the lid of a petri dish, or by using specialized plates. Some advantages of this approach are the small starting numbers of cells and media volumes, uniform spheroid size that can be adjusted as necessary by modifying the number of cells during seeding, and possibility for the co-culture of different cell types [66]. However, drawbacks of this method are the difficult medium exchange, which limits drug addition; the lack of extracellular matrix surrogates; and no possibility for migration/invasion or cell viability assays [66].

3.2.3. Organ-on-a-Chip Technology

An organ-on-a-chip is a more complex approach that enables PC cells to recreate in vitro the architecture of in vivo tumor mass, which is based on microfluidic devices [67]. Microchip manufacturing methods used for this approach contain continuously perfused chambers inhabited by living cells arranged to simulate tissue- and organ-level architecture [67]. This device produces levels of tissue and organ functionality not possible with conventional 2D or 3D culture systems. Within this system, it is possible to incorporate various cell types equally distributed within the chip, and they can be kept still during media exchange. Organ-on-a-chip enables high-resolution, real-time imaging and in vitro analysis of biochemical, genetic, and metabolic activities of living cells in a functional tissue and organ context, as well as the study of tissue development, organ physiology, and disease etiology [67]. Spheroids generated in this manner are uniformly sized, and their formation is fast, with constant perfusion and uniform distribution of oxygen and nutrients. However, a drawback of this approach would be the necessity of specialized equipment and expensiveness [68].

3.2.4. Gel-Embedding

Some additional models called gel-embedding models include extracellular matrix-like gels, such as highly hydrophilic polymers with a soft tissue-like stiffness designed to mimic the extracellular protein network. Such gels include Matrigel, alginate, and collagen. Advantages of this approach are the formation of contacts between cells and the artificial extracellular matrix, as well as the possibility to perform migration/invasion assays. This model is also known as the liquid-overlay method, as the cells are first embedded in the matrix, and then the pellet is overlaid with the culture media. However, some disadvantages are the undefined composition of natural gels and the structural modification over time [69,70,71,72,73,74], as well as their impact on the lower penetration of the drugs to the destination cells, leading to lower drug efficacy [75].

3.2.5. Prefabricated Scaffolds

Prefabricated scaffolds can be considered a replacement for the ECM. They can be made of natural (i.e., collagen) or synthetic (i.e., polycaprolactone) polymers, and they create a porous environment for the physical support and growth of spheroids. Even though they ensure high tissue mimicry and maximum resemblance to the in vivo conditions, with the possibility of the use of a wide variety of materials and properties, they are much more complicated and expensive than those for gel production, and there is a risk of possible variability between scaffolds [76,77,78]. Furthermore, as in the case of Matrigel, scaffold composition may lead to spatially divergent treatment effects [75].

3.2.6. Patient-Derived Explants

The most advanced model is the ex vivo 3D culture of freshly excised PC specimens, called patient-derived explants (PDEs). This approach developed as an alternative to the use of immortalized PC cell lines to test the efficacy of new drugs in vitro or in vivo. This is basically the cultivation of tissue pieces or slices on sponge scaffolds. The advantages of this approach, besides low cost, are high tissue mimicry and direct assessment of patients’ therapeutic responses on an individual sample, which is useful for development of personalized medicine strategies. Of course, there is a limitation in the sense of reliance on fresh tissue and specialized equipment, and expertise, as well [79,80,81,82,83].

3.3. Drug Discovery and Screening

Drug discovery and screening is, in 3D cultures, most often limited to monitoring the spheroid-forming capabilities of cells exposed to treatment compared to solvent control. The size, shape, and number of spheroids are measured and compared, providing information about the effectiveness of the used substance in this setting. Many groups use proliferation and viability assays to evaluate the fitness of their spheroids, although IC50 can be an imperfect index for evaluation of spheroid viability, and the response to the same drug can differ based on the type of spheroid model (floating vs. matrix-embedded) and on the size/uniformity of spheroids [84]. Some studies go beyond this and perform sectioning and immunofluorescent staining of specific targets of interest, measure gene/protein expression in the 3D models, and even measure some metabolic parameters of performed RNA sequencing. An overview of the recent studies on PC cell lines involving spheroid/organoid/tumoroid models using different therapeutics is presented in Table 1.
Cell-based assays are still the main tool for testing the efficacy of a new compound in drug discovery. While comparing 2D and 3D cell models, it has been shown that there are some remarkable differences between the two [85]. Cellular responses to drug treatments in 3D models are for sure more similar to in vivo responses when compared to 2D models. For instance, 3D cell models are more resistant to the anticancer treatment than 2D models, which has been demonstrated for several different cancers and combinations of drugs used in these types of cancer [86,87,88,89,90]. It has been shown that drugs were often highly active in 2D models, while less active and gradually lost their activity in 3D spheroids/organoids. This would imply that certain geno- and phenotypical changes induced by 3D spheroids/organoids formation are responsible for increased drug resistance due to the signals from dynamic cellular interactions between neighboring cells and ECM input into the cellular decision-making process [87,91]. Increased drug resistance is probably due to the limited diffusion through the spheroid/organoid, which leads to the drug concentration gradient across a single spheroid/organoid and hypoxia, which has been shown to lead to the activation of genes involved in cell survival and drug sensitivity [75]. Moreover, stromal cells also have been shown to be involved in the drug resistance, and this chemoresistance is observed in vivo, as well [87,92]. Several clinical trials failed upon reaching advanced stages of drug testing due to the fact that sometimes resistance mechanisms are not active in 2D cell culture, but are seen in 3D cell culture models, as well as in vivo in a xenograft model [85].
Differences in physical and physiological properties of 2D and 3D models affect their response to the drug treatment. For instance, 2D cells are stretched out on a flat substrate, while 3D cells on a natural or synthetic scaffold material maintain normal morphology and multiple contacts with the surrounding cells, and these differences in the morphological spread contribute to the differences in the drug response between the two. Moreover, the difference in the expression and the spatial organization of surface receptors in these two models also affects the response to drugs since the levels of receptors and the binding efficiency of a drug to these receptors is different due to the difference in the structure, localization, and spatial organization of these receptors on cell surfaces [93,94]. In addition, there is a difference in cancer gene expression levels because in 2D cell culture, some genes are differently expressed, which can lead to the different response and, thus, affect the effectiveness of a drug [93,95]. Moreover, we need to take into consideration that cells in 2D and 3D are also in different stages of the cell cycle, meaning that 2D cells are mainly proliferative, while 3D cells are usually a mixture of cells in proliferation, quiescence, and even apoptosis/necrosis, having proliferating cells on the outer region and quiescent cells in the middle region due to the lack of nutrients and gas exchange, while the center of the 3D structure often contains dead cells [96]. Further, active cell proliferation is sometimes required for drugs to be effective, so only the outer layer of the 3D structure will be affected, while the quiescent cells of the middle layer may respond poorly [97].
Drug accessibility to cells and local pH is also important for the difference in drug response. In the 2D monolayer, drugs diffuse to cells equally [98]. In the 3D model, diffusion of the drugs is dependent on the distance of the cells from the surface and the local pH [99]. Both hypoxia and lower pH contribute to the drug resistance, as there is no efficient transport system to remove waste from the central region of the spheroid. Additionally, lower pH can reduce the efficiency of the uptake of the drug, resulting in increased resistance [100]. The use of patient-derived primary tumor cells for the generation of 3D models proved to be promising in evaluating cellular responses to antiproliferative cytotoxic and targeted agents, as well as in assessing the chemosensitivity and signaling pathway activity in cancer cells. This was demonstrated in some of the most common cancers, including lung, breast, and prostate cancers [98]. These findings paved the way for patient-derived 3D models in the development of personalized medicine, as the same model can be examined in vitro and in vivo for the analysis of various signaling pathways and evaluation of chemosensitivity [101,102,103].
Table 1. Recent examples of the use of 3D in vitro models for testing of various compounds and therapies.
Table 1. Recent examples of the use of 3D in vitro models for testing of various compounds and therapies.
Type of TreatmentName of the Compound/TreatmentType of 3D Model UsedReference
chemotherapeuticdocetaxelspheroids in U-bottom plates and Matrigel-embedded[104]
chemotherapeuticdocetaxel on gold nanoparticlesspheroids in low-attachment plates[105]
chemotherapeuticbortezomibspheroids in agarose-coated plates[106]
chemotherapeuticdocetaxel on microparticlesspheroids in low-attachment plates[107]
natural compoundBrachydin Cspheroids in agarose-coated dishes[108]
natural compoundBrachydin Aspheroids in agarose-coated dishes[109]
natural compoundgreen tea extractspheroids in hanging drop[110]
natural compoundperillilaldehydespheroids in poly-HEMA-coated plates[111]
natural compoundpristimerinspheroids in poly-HEMA-coated plates[112]
natural compoundcurcuminspheroids in low-attachment plates[113]
natural compoundgallic acidspheroids in hanging drops[114]
natural compoundprocyanidin B2 3,3″-di-O-gallatespheroids in low-attachment plates[115]
natural compoundrosmarinic acidspheroids in hanging drops[116]
statinsimvastatinspheroids in hanging drop (plates)[117]
statinrosuvastatinspheroids in agarose-coated plates (liquid overlay)[118]
ADTdarolutamidespheroids in low-attachment plates[119]
radionuclideradium-233spheroids in low-attachment plates[120]
radionuclide225Ac on liposomes/antibodyspheroids in low-attachment plates[121]
radionuclide64CuCl2spheroids in low-attachment plates[122]
hormone17β-estradiol or testosteronespheroids in agarose-coated wells (1 spheroid/well)[123]
antibody TNB-585 (anti-PSMA antibody)spheroids in low-attachment round-bottom plates[124]
antibody-drug conjugateantibody-drug conjugate U3-1402patient-derived xenograft organoids [125]
antibody-drug conjugateantibody-drug conjugates VH1-HLE-AF680spheroids in methocellulose + Matrigel hanging drop plates[126]
ligand-radionuclide conjugatePSMA-targeting ligand labeled with 212Pbspheroids in agarose-coated plates[127]
immunotoxinanti-PSMA immunotoxin hD7-1(VL-VH)-PE40spheroids in agarose-coated plates[128]
oncolytic virusPIV5 oncolytic virusspheroids in low-attachment plates[129]
ultrasoundfocused ultrasoundspheroids in low-attachment plates[130]
microgravitymicrogravityspheroids in microgravity or agarose-coated dishes[63]
CHK1 inhibitorMU380spheroids in low-attachment plates[131]
DNMT inhibitorCM-272spheroids in U-bottom plates[132]
kinase inhibitorponatinib, sunitinib, sorafeniborganoids[133]
kinase inhibitorDovitinib, BGJ398, or PD166866spheroids in agarose-coated plates[134]
HDAC inhibitorJazz90, Jazz167spheroids in Matrigel[135]
mPGES-1 inhibitorKH176mspheroids in Matrigel, low-attachment plates[136]
TRPM8 antagonistTRPM8 antagonistspheroids in ECM[137]
NUAK antagonistWZ4003 and HTH-02-006spheroids in low-attachment plates[138]
PKC agonistHMI-1a3spheroids in agarose-coated U-bottom plates[139]
Cyclodextrin nanospongeGSH-NSsspheroids in hanging drops[140]
cytotoxic metalIr(III)–Cu(II) Compounds on liposomesspheroids in hanging drops[141]
cytotoxic metalIrIII complex conjugated to coumarinspheroids in low-attachment plates[142]
glycoproteinfetuin-Aspheroids in low-attachment plates[143]
peptideGV1001 peptidespheroids in low-attachment plates[144]
small moleculeATPγS and ATPspheroids in spheroid culture plates[145]

4. Standard and Novel Therapies Used in 3D Models of PC

Results obtained so far from different 3D assays and approaches used in PC early-stage drug discovery encompass radiotherapy, hormone therapy, chemotherapy, targeted therapies, and novel and experimental therapies [146]. Many combined therapies, which include different approaches, are also being tested, and the most recent studies are summarized in Table 2.

4.1. Radiotherapy

Radiotherapy is recommended for localized or locally advanced PC [147]. The report of Camus et al. on the viability of 3D multicellular PC spheroids after treatment with Surface Enhanced Raman Spectroscopy (SERS) showed that this novel method for measuring intracellular redox potential and pH in 3D live cultures can actually represent a potential new platform for in vitro preclinical characterization of tumor models [148]. Radiation can be applied on 3D cultures in combination with potential sensitizers, for example, AMPK activator AICAR [149], or cytotoxic metals [150]. Apart from the classic irradiation, there is some interest in targeted delivery of radionuclides to the PC by using different targeting molecules, and this approach is also being tested in vitro on 3D cultures. Different radionuclides and isotopes, such as 233Ra, 225Ac, 212Pb, or 64CuCl2, have been tested on spheroid models, either directly or using different carriers, and showed a good effect on spheroid models [120,121,122,127]. 233Ra pre-treatment of HAp surface has shown a drastic effect on the survival of PC cells and spheroid outgrowth [120]. Spheroid cultures can be used to assess the effectiveness of delivery and to test different carriers, as was demonstrated by Salerno et al.: 225Ac α-particles were delivered by either tumor-responsive liposomes or antibodies, and they have shown that small spheroids (80–100 μm) were more effectively inhibited by the radiolabeled antibodies, large-size spheroids (300 μm) were more responsive to liposome carriers, while the combination of both carriers was the most effective for intermediate-size spheroids (200 μm) [121].

4.2. Hormone Therapy

Treatments that reduce androgen production by the testicles are the most commonly used hormone therapies for PC. ADT can be performed surgically or chemically. The surgical option is orchiectomy, a procedure to remove one or both testicles, which can reduce the level of testosterone in the blood by 90% to 95% [151]. The remaining 5% is produced by the adrenal gland, so luteinizing hormone-releasing hormone (LHRH), also known as gonadotropin-releasing hormone (GnRH), agonists are used (goserelin, triptorelin, histrelin). They produce an initial surge in luteinizing hormone (LH) and testosterone levels, but constant exposure to LHRH desensitizes the pituitary cells and, therefore, suppresses testosterone levels [152]. LHRH/GnRH antagonists, such as degarelix, abarelix, and relugolix, can also be used, as they inhibit downstream LH signaling and achieve chemical castration within 2–3 days [152]. Antiandrogen therapies, treatments that block the action of androgens in the body, are not considered typical ADT and are often used concurrently with ADT or when ADT stops working. Such treatments include androgen receptor blockers (also called androgen receptor antagonists), which compete with androgens for binding to the androgen receptor, or androgen synthesis blockers, which prevent the production of androgens. Some examples of the androgen receptor blockers include the first-generation drugs flutamide, bicalutamide, and nilutamide, and the second-generation drugs enzalutamide, apalutamide, and darolutamide [153]. Some of the approved androgen synthesis inhibitors are abiraterone, ketoconazole, and aminoglutethimide. Abiraterone is also approved in combination with prednisone to treat metastatic PC, both castration-sensitive and castration-resistant [154].
Advanced CRPC represents a major clinical problem since the gold standard, AR targeting, is not as effective in the long run as previously thought [155,156]. Therefore, a co-culture 3D model of PC and CAF has been developed, and it was demonstrated that the stromal cells reduced the sensitivity of PC cells to androgens and other drugs without altering AR levels. This has demonstrated that this kind of PC and CAF combined 3D model is necessary to understand how CAF can influence the drug response of PC cells to current therapies. Therefore, this type of 3D co-culture can be a valuable in vitro drug-testing tool [157].

4.3. Chemotherapy

Chemotherapy is used in advanced PC, CRPC, or mCRPC. Several standard drugs are used as chemotherapeutic agents for these advanced stages of PC. Standard chemotherapy begins with docetaxel combined with prednisone [158]. However, there are some recent advances in this kind of treatment. Addition of hormonal therapy to docetaxel in those with newly diagnosed PC, or the use of cabazitaxel to treat mCRPC, showed significant reduction in tumor growth and spread. Some new approaches, such as using drug-encapsulating polymersomes, which contain docetaxel and present folic acid on the surface, or micellar delivery systems for paclitaxel, have demonstrated higher cytotoxicity than using the drug in free form [159,160]. The advantage of such delivery systems is their ability to penetrate into the center of the spheroid/tumor mass, bypassing the issue of reduced drug permeability and uptake [146]. Tumor cells can be sensitized to chemotherapy by using various compounds, e.g., MU380 can sensitize docetaxel-resistant PC to gemcitabine; MF-15 can re-sensitize enzalutamide-resistant cells to anzalutamide; and paxilline can reverse the resistance to docetaxel, palitaxel, doxorubicin, and cisplatin. [117,131,161]. Some compounds can also act synergistically, such as resveratrol with docetaxel [162] or JQ1 with docetaxel [104].

4.4. Targeted Therapies

Targeted therapies are designed to stop only the growth of the cells with a specific mutation, in this way sparing the healthy cells from damage. This kind of treatment is different from chemotherapy, which damages healthy cells, along with the cancer cells [163]. There are several agents used for this kind of treatment. ANTI-ANGIOGENIC AGENTS, such as Aflibercept, Bevacizumab, and Thalidomide/Lenalidomide, target angiogenesis, which is an important process for the growth, progression, and metastasis of solid tumors [164,165,166]. TYROSINE KINASE INHIBITORS (TKIs) inhibit tyrosine kinases alone or in combination with other targets, such as the angiogenesis factors mentioned before. Tyrosine kinases are mediators of intracellular signaling pathways that control cell growth, migration, and invasion. Some examples in this group include Dasatinib, Cabozantinib, and Sunitinib [133,134,167]. ENDOTHELIN RECEPTOR ANTAGONISTS are Atrasentan and Zibotentan. Endothelins are small proteins implicated in tumor growth and metastasis [168]. CLUSTERIN INHIBITORS (Custirsen) inhibit clusterin, a cytoprotective chaperone whose transcription is promoted by the androgen receptor and heat shock Factor-1, a key mediator of carcinogenesis [169]. Other agents worth mentioning are: B-cell Lymphoma 2 (Bcl-2) inhibitors, Insulin-Like Growth Factor (IGF) inhibitors, inhibitors of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, and immunotherapeutic agents. The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth and cell cycle progression and integrates signals from growth factors and is aberrantly activated and frequently mutated in PC [170,171,172]. HDAC inhibitors have also demonstrated potential in the 3D model [135]. Targeted therapies are being tested with currently known chemotherapeutics or natural compounds in an effort to either increase the sensitivity of PC to known drugs or to re-sensitize resistant tumors (recent publications are summarized in Table 2). Some molecular targets have been tested in a 3D in vitro setting, and have been shown to affect the sensitivity of PC spheroids to treatment, for example, CD133 suppression increases sensitivity to paclitaxel [173], CDH1 loss sensitizes PC to DNA-damaging agents [174], and midkine downregulation sensitizes cells to quercetin [175]. In summary, targeted therapies demonstrate high tumor cell specificity and efficacy, while providing acceptable toxicity and side effects [164]. However, targeted therapy of mCRPC is still not showing satisfactory outcomes, and there was no difference in survival when docetaxel or prednisone were used, with or without targeted therapy [164].

4.5. Novel and Experimental Therapies

Many natural compounds are being tested for their effect on PC, and 3D spheroid models are used as either simple spheroid formation assays to assess the ability of the cells to form spheroids after treatment, or more complex studies that include measuring viability, apoptosis, metabolic parameters, and gene and protein expression. Curcumin is one of the most investigated natural compounds in this context, and it has demonstrated anti-tumor effects on many tumor types, including prostate [113,176,177]. However, the search for new natural inhibitors continues, as many are being tested on PC spheroids, such as flavokawain A [178], brachydin A [109], green tea extract [110], rosmarinic acid [116], or grape seed extract [115].
3D cultures are being used to develop novel carrier systems with the possibility to penetrate deeper into the structure of the tumor and deliver specific compounds, such as radionuclides, miRNA, and drugs. Several carriers have been tested that have shown increased permeability, even in 3D cultures [107,121,126,141,142,179,180].
MicroRNA (miRNA) molecules can be used to target specific genes of interest; however, their application in vivo remains controversial, as precise delivery to target cells is required to avoid off-target effects. The combining of specific miRNA with standard therapy is showing great potential in vitro, including on 3D cultures. siCD133 combined with paclitaxel shows a synergistic effect in vitro and inhibits spheroid formation [173]. siMRP1 combined with doxorubicin and loaded onto silicon nanoparticles shows increased retention and selectivity to the tumor in 2D and 3D conditions [181]. siEphA2 combined with a small-molecule HDAC inhibitor loaded into lipid nanoparticles induces cytotoxicity in 3D spheroids comparable to that observed in the 2D monolayer [167]. siMK in combination with quercetin results in reduced spheroid size compared to either treatment alone [175].
Table 2. Recent examples of the use of 3D in vitro models for testing of combinations of therapies.
Table 2. Recent examples of the use of 3D in vitro models for testing of combinations of therapies.
Type of TreatmentName of the Compound/TreatmentType of 3D Model UsedReference
chemotherapeutic + natural compoundlactic acid, arctigenin, docetaxelspheroids in low-attachment plates[182]
chemotherapeutic + natural compoundcurcumin, cisplatin, paclitaxel, docetaxel spheroids in Matrigel[177]
chemotherapeutic + BET inhibitorJQ1, docetaxelspheroids in Matrigel[104]
chemotherapeutic + PARP inhibitorolaparib and carboplatinPDX-derived organoids for drug sensitivity testing[183]
chemotherapeutic + radiotherapycarboplatin and radiotherapyorganoids[174]
chemotherapeutic + siRNAsiMRP1 + doxorubicinspheroids in low-attachment plates[181]
chemotherapeutic + siRNAsiCD133 + paclitaxelspheroids in Matrigel[173]
chemotherapeutic + TRAIL inhibitortaxanes + TRAILspheroids in low-attachment plates, monocultures or mixed with fibroblasts[184]
chemotherapeutic + uricosuricprobenecid, doxorubicin, cisplatinspheroids in low-attachment plates[185]
chemotherapeutic + antioxidantresveratrol + docetaxelspheroids in low-attachment plates[162]
chemotherapeutic + hypoxia-activated prodrugdocetaxel, TH-302spheroids in low-attachment plates[100]
chemotherapeutic + antibioticciprofloxacin, doxorubicin [186]
chemotherapeutic + NAMPT inhibitorFK866 + doxorubicinspheroids in bioreactor[187]
chemotherapeutic + ion channel inhibitorPaxilline + docetaxel, paclitaxel, doxorubicin, and cisplatinspheroids in low-attachment plates[161]
ADT + anti-inflammatory drug + AKR1C inhibitorMF-15, indomethacin, enzalutamidespheroids in low-attachment plates[117]
ADT + cytokineIL-23, enzolutamide, darolutamidespheroids in low-attachment plates[188]
ADT + small peptidesmall peptide Rh-2025u, enzalutamidespheroids in Matrigel[189]
ADT + small peptideEnzalutamide or Bicalutamide, recombinant NRG1 peptideorganoids, xenograft[190]
natural compound + MEK inhibitorcurcumin, PD98059spheroids in low-attachment plates[176]
natural compound + NEDD8 inhibitorflavokawain A, MLN4924spheroids in low-attachment plates[178]
acyl-CoA synthetase inhibitor + contrasting agent5-aminolevulinic acid, triacsin Cspheroids in low-attachment plates[191]
cytotoxic metal + radiation[Cu(TPZ)2]-liposomes and gamma-radiationspheroids in agarose-coated plates[150]
hyperthermia + radiationhyperthermia + electron radiationspheroids in low-attachment plates[192]
NDRG1 inhibitor + iron chelatorthiosemicarbazones, Dp44mT, DpCspheroids in collagen hydrogel (liquid overlay)[193]
OGT inhibitor + CDK inhibitorOSMI-2 + AT7519spheroids in Matrigel[194]
kinase inhibitor + siRNAsiEphA2, JIB-04 in lipid nanoparticlesspheroids in poly-HEMA-coated plates[167]
AMPK activator + radiationAICAR + radiationspheroids in agar-coated plates (liquid overlay)[149]
statin + anticonvulsantvalproic acid, simvastatinspheroids in low-attachment plates, multiple generations[195]
antioxidant + siRNAsiMK + quercetinspheroids in agarose-coated plates (liquid overlay)[175]
Finally, an important feature to investigate during PC development and metastasis is the spread of PC to the bone [196]. PC most often metastasizes to the bone, and this is the primary cause of PC-related morbidity and mortality [197]. Certain translational 3D models have been developed to mimic the complex structure of the cancer metastasis, and some valuable recent examples include breast and PC [98]. A biomimetic bone microenvironment was designed to mimic the mesenchymal-to-epithelial transition (MET) of PC using highly metastatic and non-metastatic PC cell lines [169,170,198]. Bioactive factors from osteogenic induction of human mesenchymal stem cells (MSCs) were added to the porous 3D scaffold of different compositions (e.g., PLGA, nano-hydroxyapatite (nHA)/PLGA [168], and nanohydroxyapatite/collagen mixed scaffolds [171]). Such structures mimicked the interaction between the PC and bone microenvironments and allowed for the study of novel therapeutic approaches. In addition, genetically engineered mouse models (GEMMs) that mimic PC bone metastasis can be used as an efficient model for studying the advanced stages of PC in vivo [199,200]. Until now, the improvements in the treatment of PC have mainly been made in early-stage localized disease. However, the distinction between indolent and aggressive tumors and the lack of efficient therapies of advanced PC still represent a major problem in PC management. GEMM can offer the possibility to generate new models that accurately reflect human disease and to implement this knowledge in drug discovery and screening [201]. So far, this model has been used mainly for the investigation of PC tumor initiation and progression, with little or no focus on PC metastasis to the bone. Nevertheless, one of the studies established a prostate-derived tumor line that showed frequent metastasis to the bone and growth in an immunocompetent host [202]. In this way, a useful model was generated to study the mechanisms of bone metastasis, as well as the tumor immune response. Sadly, the publications on this topic are still sparse, and there is room for expansion in this area.

5. Conclusions and Future Directions

Cancer studies are mainly relying on in vitro models, and continuous improvement of these models is crucial for the further development of cancer research. Therefore, an upgrade to the 3D model architecture is the future of PC research in order to better understand the complex mechanisms influencing PC development and progression. This is essential for more comprehensive drug discovery and screening. Novel therapies are much needed, especially for CRPC and mCRPC. Research into new 3D models, which are able to closely reflect the tumor microenvironment, have shown impressive progress in the last decade, and many options have been developed for 3D model establishment and maintenance. It is of great importance to mimic the natural growth of cancer more closely, and in this sense, it is necessary to engineer separate cancer masses and biomimetic stromal compartments containing appropriate cell populations (e.g., fibroblasts, endothelial cells, immune cells, and other ECM components). Heterotypic spheroids, organoids, and tumoroids are models of increasing complexity that take into account the contribution of ECM and supporting cell populations. Further attention should be given to the primary cancer cell lines and PDX models because this could lead to the development of personalized drug-screening platforms. The future of 3D PC research lies not only in the investigation of tumor growth, but also in studying invasion, migration, cancer stem cells plasticity, and cancer cell dormancy, which can model the interaction between cancer and stromal cells more accurately.

Author Contributions

Conceptualization: T.P.; writing—original draft preparation: T.P. and M.S.; writing—review and editing: T.P. and M.S.; funding: M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a donation of the dm-drogerie markt d.o.o., raised by the “Movember” campaign.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study did not report any data.

Acknowledgments

We would like to thank everyone who contributed to the “Movember” fundraiser action.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the conceptualization and writing of this manuscript.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Christenson, M.; Song, C.-S.; Liu, Y.-G.; Chatterjee, B. Precision Targets for Intercepting the Lethal Progression of Prostate Cancer: Potential Avenues for Personalized Therapy. Cancers 2022, 14, 892. [Google Scholar] [CrossRef] [PubMed]
  3. Gandaglia, G.; Abdollah, F.; Schiffmann, J.; Trudeau, V.; Shariat, S.F.; Kim, S.P.; Perrotte, P.; Montorsi, F.; Briganti, A.; Trinh, Q.-D.; et al. Distribution of Metastatic Sites in Patients with Prostate Cancer: A Population-Based Analysis. Prostate 2014, 74, 210–216. [Google Scholar] [CrossRef] [PubMed]
  4. Raghallaigh, H.N.; Bott, S.R. The Role of Family History and Germline Genetics in Prostate Cancer Disease Profile and Screening. In Urologic Cancers; Barber, N., Ali, A., Eds.; Exon Publications: Brisbane, Australia, 2022; ISBN 978-0-645-33205-6. [Google Scholar]
  5. Marino, F.; Totaro, A.; Gandi, C.; Bientinesi, R.; Moretto, S.; Gavi, F.; Pierconti, F.; Iacovelli, R.; Bassi, P.; Sacco, E. Germline Mutations in Prostate Cancer: A Systematic Review of the Evidence for Personalized Medicine. Prostate Cancer Prostatic Dis. 2022. [Google Scholar] [CrossRef] [PubMed]
  6. Ni Raghallaigh, H.; Eeles, R. Genetic Predisposition to Prostate Cancer: An Update. Fam. Cancer 2022, 21, 101–114. [Google Scholar] [CrossRef]
  7. Bancroft, E.K.; Page, E.C.; Brook, M.N.; Thomas, S.; Taylor, N.; Pope, J.; McHugh, J.; Jones, A.-B.; Karlsson, Q.; Merson, S.; et al. A Prospective Prostate Cancer Screening Programme for Men with Pathogenic Variants in Mismatch Repair Genes (IMPACT): Initial Results from an International Prospective Study. Lancet. Oncol. 2021, 22, 1618–1631. [Google Scholar] [CrossRef]
  8. Yuan, J.; Kensler, K.H.; Hu, Z.; Zhang, Y.; Zhang, T.; Jiang, J.; Xu, M.; Pan, Y.; Long, M.; Montone, K.T.; et al. Integrative Comparison of the Genomic and Transcriptomic Landscape between Prostate Cancer Patients of Predominantly African or European Genetic Ancestry. PLoS Genet. 2020, 16, e1008641. [Google Scholar] [CrossRef] [Green Version]
  9. Samtal, C.; El Jaddaoui, I.; Hamdi, S.; Bouguenouch, L.; Ouldim, K.; Nejjari, C.; Ghazal, H.; Bekkari, H. Review of Prostate Cancer Genomic Studies in Africa. Front. Genet. 2022, 13, 911101. [Google Scholar] [CrossRef]
  10. Plaskon, L.A.; Penson, D.F.; Vaughan, T.L.; Stanford, J.L. Cigarette Smoking and Risk of Prostate Cancer in Middle-Aged Men. Cancer Epidemiol. Biomark. Prev. 2003, 12, 604–609. [Google Scholar]
  11. Cirne, F.; Kappel, C.; Zhou, S.; Mukherjee, S.D.; Dehghan, M.; Petropoulos, J.-A.; Leong, D.P. Modifiable Risk Factors for Prostate Cancer in Low- and Lower-Middle-Income Countries: A Systematic Review and Meta-Analysis. Prostate Cancer Prostatic Dis. 2022, 25, 453–462. [Google Scholar] [CrossRef]
  12. Parker, C.; Castro, E.; Fizazi, K.; Heidenreich, A.; Ost, P.; Procopio, G.; Tombal, B.; Gillessen, S. Prostate Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up†. Ann. Oncol. 2020, 31, 1119–1134. [Google Scholar] [CrossRef] [PubMed]
  13. Cornu, J.-N.; Cancel-Tassin, G.; Cox, D.G.; Roupret, M.; Koutlidis, N.; Bigot, P.; Valeri, A.; Ondet, V.; Gaffory, C.; Fournier, G.; et al. Impact of Body Mass Index, Age, Prostate Volume, and Genetic Polymorphisms on Prostate-Specific Antigen Levels in a Control Population. Eur. Urol. 2016, 70, 6–8. [Google Scholar] [CrossRef] [PubMed]
  14. Loeb, S.; Shin, S.S.; Broyles, D.L.; Wei, J.T.; Sanda, M.; Klee, G.; Partin, A.W.; Sokoll, L.; Chan, D.W.; Bangma, C.H.; et al. Prostate Health Index (Phi) Improves Multivariable Risk Prediction of Aggressive Prostate Cancer. BJU Int. 2017, 120, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kang, S.K.; Mali, R.D.; Prabhu, V.; Ferket, B.S.; Loeb, S. Active Surveillance Strategies for Low-Grade Prostate Cancer: Comparative Benefits and Cost-Effectiveness. Radiology 2021, 300, 594–604. [Google Scholar] [CrossRef] [PubMed]
  16. Posdzich, P.; Darr, C.; Hilser, T.; Wahl, M.; Herrmann, K.; Hadaschik, B.; Grünwald, V. Metastatic Prostate Cancer—A Review of Current Treatment Options and Promising New Approaches. Cancers 2023, 15, 461. [Google Scholar] [CrossRef] [PubMed]
  17. Tsujino, T.; Takai, T.; Hinohara, K.; Gui, F.; Tsutsumi, T.; Bai, X.; Miao, C.; Feng, C.; Gui, B.; Sztupinszki, Z.; et al. CRISPR Screens Reveal Genetic Determinants of PARP Inhibitor Sensitivity and Resistance in Prostate Cancer. Nat. Commun. 2023, 14, 252. [Google Scholar] [CrossRef]
  18. Jamaspishvili, T.; Berman, D.M.; Ross, A.E.; Scher, H.I.; De Marzo, A.M.; Squire, J.A.; Lotan, T.L. Clinical Implications of PTEN Loss in Prostate Cancer. Nat. Rev. Urol. 2018, 15, 222–234. [Google Scholar] [CrossRef]
  19. de Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef]
  20. Muralidhar, A.; Potluri, H.K.; Jaiswal, T.; McNeel, D.G. Targeted Radiation and Immune Therapies-Advances and Opportunities for the Treatment of Prostate Cancer. Pharmaceutics 2023, 15, 252. [Google Scholar] [CrossRef]
  21. Petrella, G.; Corsi, F.; Ciufolini, G.; Germini, S.; Capradossi, F.; Pelliccia, A.; Torino, F.; Ghibelli, L.; Cicero, D.O. Metabolic Reprogramming of Castration-Resistant Prostate Cancer Cells as a Response to Chemotherapy. Metabolites 2022, 13, 65. [Google Scholar] [CrossRef]
  22. Lasorsa, F.; di Meo, N.A.; Rutigliano, M.; Ferro, M.; Terracciano, D.; Tataru, O.S.; Battaglia, M.; Ditonno, P.; Lucarelli, G. Emerging Hallmarks of Metabolic Reprogramming in Prostate Cancer. Int. J. Mol. Sci. 2023, 24, 910. [Google Scholar] [CrossRef] [PubMed]
  23. Sternberg, C.N.; Fizazi, K.; Saad, F.; Shore, N.D.; De Giorgi, U.; Penson, D.F.; Ferreira, U.; Efstathiou, E.; Madziarska, K.; Kolinsky, M.P.; et al. Enzalutamide and Survival in Nonmetastatic, Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2197–2206. [Google Scholar] [CrossRef] [PubMed]
  24. Armstrong, A.J.; Szmulewitz, R.Z.; Petrylak, D.P.; Holzbeierlein, J.; Villers, A.; Azad, A.; Alcaraz, A.; Alekseev, B.; Iguchi, T.; Shore, N.D.; et al. ARCHES: A Randomized, Phase III Study of Androgen Deprivation Therapy With Enzalutamide or Placebo in Men With Metastatic Hormone-Sensitive Prostate Cancer. J. Clin. Oncol. 2019, 37, 2974–2986. [Google Scholar] [CrossRef]
  25. Smith, M.R.; Hussain, M.; Saad, F.; Fizazi, K.; Sternberg, C.N.; Crawford, E.D.; Kopyltsov, E.; Park, C.H.; Alekseev, B.; Montesa-Pino, Á.; et al. Darolutamide and Survival in Metastatic, Hormone-Sensitive Prostate Cancer. N. Engl. J. Med. 2022, 386, 1132–1142. [Google Scholar] [CrossRef]
  26. Smith, M.R.; Saad, F.; Chowdhury, S.; Oudard, S.; Hadaschik, B.A.; Graff, J.N.; Olmos, D.; Mainwaring, P.N.; Lee, J.Y.; Uemura, H.; et al. Apalutamide and Overall Survival in Prostate Cancer. Eur. Urol. 2021, 79, 150–158. [Google Scholar] [CrossRef]
  27. Kneppers, J.; Severson, T.M.; Siefert, J.C.; Schol, P.; Joosten, S.E.P.; Yu, I.P.L.; Huang, C.-C.F.; Morova, T.; Altıntaş, U.B.; Giambartolomei, C.; et al. Extensive Androgen Receptor Enhancer Heterogeneity in Primary Prostate Cancers Underlies Transcriptional Diversity and Metastatic Potential. Nat. Commun. 2022, 13, 7367. [Google Scholar] [CrossRef]
  28. Shiota, M.; Akamatsu, S.; Tsukahara, S.; Nagakawa, S.; Matsumoto, T.; Eto, M. Androgen Receptor Mutations for Precision Medicine in Prostate Cancer. Endocr. Relat. Cancer 2022, 29, R143–R155. [Google Scholar] [CrossRef] [PubMed]
  29. Mao, Y.; Yang, G.; Li, Y.; Liang, G.; Xu, W.; Hu, M. Advances in the Current Understanding of the Mechanisms Governing the Acquisition of Castration-Resistant Prostate Cancer. Cancers 2022, 14, 3744. [Google Scholar] [CrossRef]
  30. Qiu, Y. A Phosphorylation Switch Controls Androgen Biosynthesis in Prostate Cancer. J. Clin. Invest. 2023, 133, e166499. [Google Scholar] [CrossRef]
  31. Wang, Z.; Yan, X.; Tang, P.; Tang, T.; Wang, Y.; Peng, S.; Wang, S.; Lan, W.; Wang, L.; Zhang, Y.; et al. Genetic Profiling of Hormone-Sensitive and Castration-Resistant Prostate Cancers and Identification of Genetic Mutations Prone to Castration-Resistant Prostate Cancer. Prostate Cancer Prostatic Dis. 2022. [Google Scholar] [CrossRef]
  32. Baca, S.C.; Prandi, D.; Lawrence, M.S.; Mosquera, J.M.; Romanel, A.; Drier, Y.; Park, K.; Kitabayashi, N.; MacDonald, T.Y.; Ghandi, M.; et al. Punctuated Evolution of Prostate Cancer Genomes. Cell 2013, 153, 666–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Malik, J.A.; Ahmed, S.; Momin, S.S.; Shaikh, S.; Alafnan, A.; Alanazi, J.; Said Almermesh, M.H.; Anwar, S. Drug Repurposing: A New Hope in Drug Discovery for Prostate Cancer. ACS Omega 2023, 8, 56–73. [Google Scholar] [CrossRef] [PubMed]
  34. Fang, Q.; Cole, R.N.; Wang, Z. Mechanisms and Targeting of Proteosome-Dependent Androgen Receptor Degradation in Prostate Cancer. Am. J. Clin. Exp. Urol. 2022, 10, 366–376. [Google Scholar] [PubMed]
  35. Chai, X.; Hu, X.-P.; Wang, X.-Y.; Wang, H.-T.; Pang, J.-P.; Zhou, W.-F.; Liao, J.-N.; Shan, L.-H.; Xu, X.-H.; Xu, L.; et al. Computationally Guided Discovery of Novel Non-Steroidal AR-GR Dual Antagonists Demonstrating Potency against Antiandrogen Resistance. Acta Pharmacol. Sin. 2023. [Google Scholar] [CrossRef]
  36. Jia, X.; Han, X. Targeting Androgen Receptor Degradation with PROTACs from Bench to Bedside. Biomed. Pharmacother. 2023, 158, 114112. [Google Scholar] [CrossRef] [PubMed]
  37. Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021, 385, 1091–1103. [Google Scholar] [CrossRef]
  38. Owen, J.S.; Clayton, A.; Pearson, H.B. Cancer-Associated Fibroblast Heterogeneity, Activation and Function: Implications for Prostate Cancer. Biomolecules 2022, 13, 67. [Google Scholar] [CrossRef]
  39. Gunti, S.; Hoke, A.T.K.; Vu, K.P.; London, N.R. Organoid and Spheroid Tumor Models: Techniques and Applications. Cancers 2021, 13, 874. [Google Scholar] [CrossRef]
  40. Nunes, A.S.; Barros, A.S.; Costa, E.C.; Moreira, A.F.; Correia, I.J. 3D Tumor Spheroids as in Vitro Models to Mimic in Vivo Human Solid Tumors Resistance to Therapeutic Drugs. Biotechnol. Bioeng. 2019, 116, 206–226. [Google Scholar] [CrossRef] [Green Version]
  41. Garraway, I.P.; Sun, W.; Tran, C.P.; Perner, S.; Zhang, B.; Goldstein, A.S.; Hahm, S.A.; Haider, M.; Head, C.S.; Reiter, R.E.; et al. Human Prostate Sphere-Forming Cells Represent a Subset of Basal Epithelial Cells Capable of Glandular Regeneration in Vivo. Prostate 2010, 70, 491–501. [Google Scholar] [CrossRef]
  42. Chen, S.; Principessa, L.; Isaacs, J.T. Human Prostate Cancer Initiating Cells Isolated Directly from Localized Cancer Do Not Form Prostaspheres in Primary Culture. Prostate 2012, 72, 1478–1489. [Google Scholar] [CrossRef] [Green Version]
  43. Lancaster, M.A.; Knoblich, J.A. Organogenesis in a Dish: Modeling Development and Disease Using Organoid Technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
  44. Schutgens, F.; Clevers, H. Human Organoids: Tools for Understanding Biology and Treating Diseases. Annu. Rev. Pathol. 2020, 15, 211–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hendriks, D.; Artegiani, B.; Hu, H.; Chuva de Sousa Lopes, S.; Clevers, H. Establishment of Human Fetal Hepatocyte Organoids and CRISPR-Cas9-Based Gene Knockin and Knockout in Organoid Cultures from Human Liver. Nat. Protoc. 2021, 16, 182–217. [Google Scholar] [CrossRef] [PubMed]
  46. Rodrigues, J.; Heinrich, M.A.; Teixeira, L.M.; Prakash, J. 3D In Vitro Model (R)Evolution: Unveiling Tumor-Stroma Interactions. Trends Cancer 2021, 7, 249–264. [Google Scholar] [CrossRef] [PubMed]
  47. Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2019, 6, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Zhang, L.; Jiao, M.; Li, L.; Wu, D.; Wu, K.; Li, X.; Zhu, G.; Dang, Q.; Wang, X.; Hsieh, J.-T.; et al. Tumorspheres Derived from Prostate Cancer Cells Possess Chemoresistant and Cancer Stem Cell Properties. J. Cancer Res. Clin. Oncol. 2012, 138, 675–686. [Google Scholar] [CrossRef] [PubMed]
  49. Gao, W.; Wang, Y.; Yu, S.; Wang, Z.; Ma, T.; Chan, A.M.-L.; Chiu, P.K.-F.; Ng, C.-F.; Wu, D.; Chan, F.L. Endothelial Nitric Oxide Synthase (ENOS)-NO Signaling Axis Functions to Promote the Growth of Prostate Cancer Stem-like Cells. Stem Cell Res. Ther. 2022, 13, 188. [Google Scholar] [CrossRef] [PubMed]
  50. Wang, S.; Huang, S.; Zhao, X.; Zhang, Q.; Wu, M.; Sun, F.; Han, G.; Wu, D. Enrichment of Prostate Cancer Stem Cells from Primary Prostate Cancer Cultures of Biopsy Samples. Int. J. Clin. Exp. Pathol. 2014, 7, 184–193. [Google Scholar] [PubMed]
  51. Rao, W.; Zhao, S.; Yu, J.; Lu, X.; Zynger, D.L.; He, X. Enhanced Enrichment of Prostate Cancer Stem-like Cells with Miniaturized 3D Culture in Liquid Core-Hydrogel Shell Microcapsules. Biomaterials 2014, 35, 7762–7773. [Google Scholar] [CrossRef] [Green Version]
  52. Mosaad, E.; Chambers, K.; Futrega, K.; Clements, J.; Doran, M.R. Using High Throughput Microtissue Culture to Study the Difference in Prostate Cancer Cell Behavior and Drug Response in 2D and 3D Co-Cultures. BMC Cancer 2018, 18, 592. [Google Scholar] [CrossRef] [PubMed]
  53. Krishnan, M.A.; Chelvam, V. Developing ΜSpherePlatform Using a Commercial Hairbrush: An Agarose 3D Culture Platform for Deep-Tissue Imaging of Prostate Cancer. ACS Appl. Bio Mater. 2021, 4, 4254–4270. [Google Scholar] [CrossRef] [PubMed]
  54. Carlsson, L.; Lennartsson, L.; Ronquist, G.; Larsson, A.; Nilson, S.; Nilsson, O. Mode of Growth Determines Differential Expression of Prostasomes in Cultures of Prostate Cancer Cell Lines and Opens for Studies of Prostasome Gene Expression. UPS J. Med. Sci. 2006, 111, 293–301. [Google Scholar] [CrossRef] [PubMed]
  55. Takir, G.G.; Debelec-Butuner, B.; Korkmaz, K.S. 3D Cell Culture Model for Prostate Cancer Cells to Mimic Inflammatory Microenvironment. Proceedings 2018, 2, 1555. [Google Scholar] [CrossRef] [Green Version]
  56. Fontana, F.; Raimondi, M.; Marzagalli, M.; Sommariva, M.; Limonta, P.; Gagliano, N. Epithelial-To-Mesenchymal Transition Markers and CD44 Isoforms Are Differently Expressed in 2D and 3D Cell Cultures of Prostate Cancer Cells. Cells 2019, 8, 143. [Google Scholar] [CrossRef] [Green Version]
  57. Enmon, R.M.; O’Connor, K.C.; Lacks, D.J.; Schwartz, D.K.; Dotson, R.S. Dynamics of Spheroid Self-Assembly in Liquid-Overlay Culture of DU 145 Human Prostate Cancer Cells. Biotechnol. Bioeng. 2001, 72, 579–591. [Google Scholar] [CrossRef]
  58. Enmon, R.M.; O’Connor, K.C.; Song, H.; Lacks, D.J.; Schwartz, D.K. Aggregation Kinetics of Well and Poorly Differentiated Human Prostate Cancer Cells. Biotechnol. Bioeng. 2002, 80, 580–588. [Google Scholar] [CrossRef]
  59. Song, H.; O’Connor, K.C.; Lacks, D.J.; Enmon, R.M.; Jain, S.K. Monte Carlo Simulation of LNCaP Human Prostate Cancer Cell Aggregation in Liquid-Overlay Culture. Biotechnol. Prog. 2003, 19, 1742–1749. [Google Scholar] [CrossRef]
  60. Song, H.; Jain, S.K.; Enmon, R.M.; O’Connor, K.C. Restructuring Dynamics of DU 145 and LNCaP Prostate Cancer Spheroids. Vitro Cell Dev. Biol. Anim. 2004, 40, 262–267. [Google Scholar] [CrossRef]
  61. O’Connor, K.C.; Venczel, M.Z. Predicting Aggregation Kinetics of DU 145 Prostate Cancer Cells in Liquid-Overlay Culture. Biotechnol. Lett. 2005, 27, 1663–1668. [Google Scholar] [CrossRef]
  62. Phelan, M.A.; Gianforcaro, A.L.; Gerstenhaber, J.A.; Lelkes, P.I. An Air Bubble-Isolating Rotating Wall Vessel Bioreactor for Improved Spheroid/Organoid Formation. Tissue Eng. Part C Methods 2019, 25, 479–488. [Google Scholar] [CrossRef] [PubMed]
  63. Dietrichs, D.; Grimm, D.; Sahana, J.; Melnik, D.; Corydon, T.J.; Wehland, M.; Krüger, M.; Vermeesen, R.; Baselet, B.; Baatout, S.; et al. Three-Dimensional Growth of Prostate Cancer Cells Exposed to Simulated Microgravity. Front. Cell Dev. Biol. 2022, 10, 841017. [Google Scholar] [CrossRef] [PubMed]
  64. Hybel, T.E.; Dietrichs, D.; Sahana, J.; Corydon, T.J.; Nassef, M.Z.; Wehland, M.; Krüger, M.; Magnusson, N.E.; Bauer, J.; Utpatel, K.; et al. Simulated Microgravity Influences VEGF, MAPK, and PAM Signaling in Prostate Cancer Cells. Int. J. Mol. Sci. 2020, 21, 1263. [Google Scholar] [CrossRef] [Green Version]
  65. Foty, R. A Simple Hanging Drop Cell Culture Protocol for Generation of 3D Spheroids. J. Vis. Exp. 2011, 2720. [Google Scholar] [CrossRef]
  66. Eder, T.; Eder, I.E. 3D Hanging Drop Culture to Establish Prostate Cancer Organoids. Methods Mol. Biol. 2017, 1612, 167–175. [Google Scholar] [CrossRef] [PubMed]
  67. Bhatia, S.N.; Ingber, D.E. Microfluidic Organs-on-Chips. Nat. Biotechnol. 2014, 32, 760–772. [Google Scholar] [CrossRef]
  68. Hsiao, A.Y.; Torisawa, Y.; Tung, Y.-C.; Sud, S.; Taichman, R.S.; Pienta, K.J.; Takayama, S. Microfluidic System for Formation of PC-3 Prostate Cancer Co-Culture Spheroids. Biomaterials 2009, 30, 3020–3027. [Google Scholar] [CrossRef] [Green Version]
  69. Härmä, V.; Virtanen, J.; Mäkelä, R.; Happonen, A.; Mpindi, J.-P.; Knuuttila, M.; Kohonen, P.; Lötjönen, J.; Kallioniemi, O.; Nees, M. A Comprehensive Panel of Three-Dimensional Models for Studies of Prostate Cancer Growth, Invasion and Drug Responses. PloS ONE 2010, 5, e10431. [Google Scholar] [CrossRef]
  70. Dolega, M.E.; Abeille, F.; Picollet-D’hahan, N.; Gidrol, X. Controlled 3D Culture in Matrigel Microbeads to Analyze Clonal Acinar Development. Biomaterials 2015, 52, 347–357. [Google Scholar] [CrossRef]
  71. Ma, L.; Li, J.; Nie, Q.; Zhang, Q.; Liu, S.; Ge, D.; You, Z. Organoid Culture of Human Prostate Cancer Cell Lines LNCaP and C4-2B. Am. J. Clin. Exp. Urol. 2017, 5, 25–33. [Google Scholar]
  72. Haq, S.; Samuel, V.; Haxho, F.; Akasov, R.; Leko, M.; Burov, S.V.; Markvicheva, E.; Szewczuk, M.R. Sialylation Facilitates Self-Assembly of 3D Multicellular Prostaspheres by Using Cyclo-RGDfK(TPP) Peptide. Onco. Targets Ther. 2017, 10, 2427–2447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Meinert, C.; Theodoropoulos, C.; Klein, T.J.; Hutmacher, D.W.; Loessner, D. A Method for Prostate and Breast Cancer Cell Spheroid Cultures Using Gelatin Methacryloyl-Based Hydrogels. Methods Mol. Biol. 2018, 1786, 175–194. [Google Scholar] [CrossRef] [PubMed]
  74. Hainline, K.M.; Gu, F.; Handley, J.F.; Tian, Y.F.; Wu, Y.; de Wet, L.; Vander Griend, D.J.; Collier, J.H. Self-Assembling Peptide Gels for 3D Prostate Cancer Spheroid Culture. Macromol. Biosci. 2019, 19, e1800249. [Google Scholar] [CrossRef] [Green Version]
  75. Van Hemelryk, A.; Mout, L.; Erkens-Schulze, S.; French, P.J.; van Weerden, W.M.; van Royen, M.E. Modeling Prostate Cancer Treatment Responses in the Organoid Era: 3D Environment Impacts Drug Testing. Biomolecules 2021, 11, 1572. [Google Scholar] [CrossRef] [PubMed]
  76. Katti, K.S.; Molla, M.S.; Karandish, F.; Haldar, M.K.; Mallik, S.; Katti, D.R. Sequential Culture on Biomimetic Nanoclay Scaffolds Forms Three-Dimensional Tumoroids. J. Biomed. Mater. Res. A 2016, 104, 1591–1602. [Google Scholar] [CrossRef] [PubMed]
  77. Cecilia, A.; Baecker, A.; Hamann, E.; Rack, A.; van de Kamp, T.; Gruhl, F.J.; Hofmann, R.; Moosmann, J.; Hahn, S.; Kashef, J.; et al. Optimizing Structural and Mechanical Properties of Cryogel Scaffolds for Use in Prostate Cancer Cell Culturing. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 71, 465–472. [Google Scholar] [CrossRef]
  78. Bäcker, A.; Erhardt, O.; Wietbrock, L.; Schel, N.; Göppert, B.; Dirschka, M.; Abaffy, P.; Sollich, T.; Cecilia, A.; Gruhl, F.J. Silk Scaffolds Connected with Different Naturally Occurring Biomaterials for Prostate Cancer Cell Cultivation in 3D. Biopolymers 2017, 107, 70–79. [Google Scholar] [CrossRef]
  79. Centenera, M.M.; Gillis, J.L.; Hanson, A.R.; Jindal, S.; Taylor, R.A.; Risbridger, G.P.; Sutherland, P.D.; Scher, H.I.; Raj, G.V.; Knudsen, K.E.; et al. Evidence for Efficacy of New Hsp90 Inhibitors Revealed by Ex Vivo Culture of Human Prostate Tumors. Clin. Cancer Res. 2012, 18, 3562–3570. [Google Scholar] [CrossRef] [Green Version]
  80. Centenera, M.M.; Raj, G.V.; Knudsen, K.E.; Tilley, W.D.; Butler, L.M. Ex Vivo Culture of Human Prostate Tissue and Drug Development. Nat. Rev. Urol. 2013, 10, 483–487. [Google Scholar] [CrossRef]
  81. Centenera, M.M.; Hickey, T.E.; Jindal, S.; Ryan, N.K.; Ravindranathan, P.; Mohammed, H.; Robinson, J.L.; Schiewer, M.J.; Ma, S.; Kapur, P.; et al. A Patient-Derived Explant (PDE) Model of Hormone-Dependent Cancer. Mol. Oncol. 2018, 12, 1608–1622. [Google Scholar] [CrossRef] [Green Version]
  82. Shafi, A.A.; Schiewer, M.J.; de Leeuw, R.; Dylgjeri, E.; McCue, P.A.; Shah, N.; Gomella, L.G.; Lallas, C.D.; Trabulsi, E.J.; Centenera, M.M.; et al. Patient-Derived Models Reveal Impact of the Tumor Microenvironment on Therapeutic Response. Eur. Urol. Oncol 2018, 1, 325–337. [Google Scholar] [CrossRef] [PubMed]
  83. Mutuku, S.M.; Trim, P.J.; Prabhala, B.K.; Irani, S.; Bremert, K.L.; Logan, J.M.; Brooks, D.A.; Stahl, J.; Centenera, M.M.; Snel, M.F.; et al. Evaluation of Small Molecule Drug Uptake in Patient-Derived Prostate Cancer Explants by Mass Spectrometry. Sci. Rep. 2019, 9, 15008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Xu, Y.; Pachnikova, G.; Wang, H.; Wu, Y.; Przybilla, D.; Schäfer, R.; Chen, Z.; Zhu, S.; Keilholz, U. IC50: An Unsuitable Measure for Large-Sized Prostate Cancer Spheroids in Drug Sensitivity Evaluation. Bosn. J. Basic Med. Sci. 2022, 22, 580–592. [Google Scholar] [CrossRef]
  85. Edmondson, R.; Broglie, J.J.; Adcock, A.F.; Yang, L. Three-Dimensional Cell Culture Systems and Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug Dev. Technol. 2014, 12, 207–218. [Google Scholar] [CrossRef] [Green Version]
  86. Birgersdotter, A.; Sandberg, R.; Ernberg, I. Gene Expression Perturbation in Vitro--a Growing Case for Three-Dimensional (3D) Culture Systems. Semin. Cancer Biol. 2005, 15, 405–412. [Google Scholar] [CrossRef] [PubMed]
  87. Breslin, S.; O’Driscoll, L. Three-Dimensional Cell Culture: The Missing Link in Drug Discovery. Drug Discov. Today 2013, 18, 240–249. [Google Scholar] [CrossRef]
  88. Hopkins, A.L. Network Pharmacology: The next Paradigm in Drug Discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef]
  89. DiMasi, J.A.; Hansen, R.W.; Grabowski, H.G. The Price of Innovation: New Estimates of Drug Development Costs. J. Health Econ. 2003, 22, 151–185. [Google Scholar] [CrossRef] [Green Version]
  90. Karlsson, H.; Fryknäs, M.; Larsson, R.; Nygren, P. Loss of Cancer Drug Activity in Colon Cancer HCT-116 Cells during Spheroid Formation in a New 3-D Spheroid Cell Culture System. Exp. Cell Res. 2012, 318, 1577–1585. [Google Scholar] [CrossRef]
  91. Kola, I. The State of Innovation in Drug Development. Clin. Pharmacol. Ther. 2008, 83, 227–230. [Google Scholar] [CrossRef]
  92. Weaver, V.M.; Petersen, O.W.; Wang, F.; Larabell, C.A.; Briand, P.; Damsky, C.; Bissell, M.J. Reversion of the Malignant Phenotype of Human Breast Cells in Three-Dimensional Culture and in Vivo by Integrin Blocking Antibodies. J. Cell Biol. 1997, 137, 231–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Oktem, G.; Sercan, O.; Guven, U.; Uslu, R.; Uysal, A.; Goksel, G.; Ayla, S.; Bilir, A. Cancer Stem Cell Differentiation: TGFβ1 and Versican May Trigger Molecules for the Organization of Tumor Spheroids. Oncol. Rep. 2014, 32, 641–649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Banerjee, M.; Li, Z.; Gao, Y.; Lai, F.; Huang, M.; Zhang, Z.; Cai, L.; Sanabria, J.; Gao, T.; Xie, Z.; et al. Inverse Agonism at the Na/K-ATPase Receptor Reverses EMT in Prostate Cancer Cells. Prostate 2021, 81, 667–682. [Google Scholar] [CrossRef] [PubMed]
  95. Goksel, G.; Bilir, A.; Uslu, R.; Akbulut, H.; Guven, U.; Oktem, G. WNT1 Gene Expression Alters in Heterogeneous Population of Prostate Cancer Cells; Decreased Expression Pattern Observed in CD133+/CD44+ Prostate Cancer Stem Cell Spheroids. J. BUON 2014, 19, 207–214. [Google Scholar]
  96. Wen, Z.; Liao, Q.; Hu, Y.; You, L.; Zhou, L.; Zhao, Y. A Spheroid-Based 3-D Culture Model for Pancreatic Cancer Drug Testing, Using the Acid Phosphatase Assay. Braz. J. Med. Biol. Res. 2013, 46, 634–642. [Google Scholar] [CrossRef] [Green Version]
  97. Tung, Y.-C.; Hsiao, A.Y.; Allen, S.G.; Torisawa, Y.; Ho, M.; Takayama, S. High-Throughput 3D Spheroid Culture and Drug Testing Using a 384 Hanging Drop Array. Analyst 2011, 136, 473–478. [Google Scholar] [CrossRef]
  98. Costard, L.S.; Hosn, R.R.; Ramanayake, H.; O’Brien, F.J.; Curtin, C.M. Influences of the 3D Microenvironment on Cancer Cell Behaviour and Treatment Responsiveness: A Recent Update on Lung, Breast and Prostate Cancer Models. Acta Biomater. 2021, 132, 360–378. [Google Scholar] [CrossRef]
  99. Chitcholtan, K.; Sykes, P.H.; Evans, J.J. The Resistance of Intracellular Mediators to Doxorubicin and Cisplatin Are Distinct in 3D and 2D Endometrial Cancer. J. Transl. Med. 2012, 10, 38. [Google Scholar] [CrossRef] [Green Version]
  100. Jouberton, E.; Voissiere, A.; Penault-Llorca, F.; Cachin, F.; Miot-Noirault, E. Multicellular Tumor Spheroids of LNCaP-Luc Prostate Cancer Cells as in Vitro Screening Models for Cytotoxic Drugs. Am. J. Cancer Res. 2022, 12, 1116–1128. [Google Scholar]
  101. Moskovits, N.; Itzhaki, E.; Tarasenko, N.; Chausky, E.; Bareket-Samish, A.; Kaufman, A.; Meerson, R.; Stemmer, S.M. Establishing 3-Dimensional Spheroids from Patient-Derived Tumor Samples and Evaluating Their Sensitivity to Drugs. J. Vis. Exp. 2022, 64564. [Google Scholar] [CrossRef]
  102. Williams, E.S.; Rodriguez-Bravo, V.; Chippada-Venkata, U.; De Ia Iglesia-Vicente, J.; Gong, Y.; Galsky, M.; Oh, W.; Cordon-Cardo, C.; Domingo-Domenech, J. Generation of Prostate Cancer Patient Derived Xenograft Models from Circulating Tumor Cells. J. Vis. Exp. 2015, 53182. [Google Scholar] [CrossRef] [Green Version]
  103. Timofeeva, O.A.; Palechor-Ceron, N.; Li, G.; Yuan, H.; Krawczyk, E.; Zhong, X.; Liu, G.; Upadhyay, G.; Dakic, A.; Yu, S.; et al. Conditionally Reprogrammed Normal and Primary Tumor Prostate Epithelial Cells: A Novel Patient-Derived Cell Model for Studies of Human Prostate Cancer. Oncotarget 2017, 8, 22741–22758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Xu, Y.; Pachnikova, G.; Przybilla, D.; Schäfer, R.; Cui, Y.; Zhou, D.; Chen, Z.; Zhao, A.; Keilholz, U. Evaluation of JQ1 Combined With Docetaxel for the Treatment of Prostate Cancer Cells in 2D- and 3D-Culture Systems. Front. Pharmacol. 2022, 13, 839620. [Google Scholar] [CrossRef]
  105. Bromma, K.; Dos Santos, N.; Barta, I.; Alexander, A.; Beckham, W.; Krishnan, S.; Chithrani, D.B. Enhancing Nanoparticle Accumulation in Two Dimensional, Three Dimensional, and Xenograft Mouse Cancer Cell Models in the Presence of Docetaxel. Sci. Rep. 2022, 12, 13508. [Google Scholar] [CrossRef] [PubMed]
  106. Kanbur, E.; Baykal, A.T.; Yerlikaya, A. Molecular Analysis of Cell Survival and Death Pathways in the Proteasome Inhibitor Bortezomib-Resistant PC3 Prostate Cancer Cell Line. Med. Oncol. 2021, 38, 112. [Google Scholar] [CrossRef] [PubMed]
  107. Paliashvili, K.; Di Maggio, F.; Ho, H.M.K.; Sathasivam, S.; Ahmed, H.; Day, R.M. A Novel Adjuvant Drug-Device Combination Tissue Scaffold for Radical Prostatectomy. Drug Deliv. 2019, 26, 1115–1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. De Oliveira, L.C.B.; Ribeiro, D.L.; do Nascimento, J.R.; da Rocha, C.Q.; Cólus, I.M.d.S.; Serpeloni, J.M. Anticancer Activities of Brachydin C in Human Prostate Tumor Cells (DU145) Grown in 2D and 3D Models: Stimulation of Cell Death and Downregulation of Metalloproteinases in Spheroids. Chem. Biol. Drug Des. 2022, 100, 747–762. [Google Scholar] [CrossRef] [PubMed]
  109. Ribeiro, D.L.; Tuttis, K.; de Oliveira, L.C.B.; Serpeloni, J.M.; Gomes, I.N.F.; Lengert, A.v.H.; da Rocha, C.Q.; Reis, R.M.; Cólus, I.M.d.S.; Antunes, L.M.G. The Antitumoral/Antimetastatic Action of the Flavonoid Brachydin A in Metastatic Prostate Tumor Spheroids In Vitro Is Mediated by (Parthanatos) PARP-Related Cell Death. Pharmaceutics 2022, 14, 963. [Google Scholar] [CrossRef]
  110. Safari, F.; Rayat Azad, N.; Alizadeh Ezdiny, A.; Pakizehkar, S.; Khazaei Koohpar, Z.; Ranji, N. Antitumor Activities of Green Tea by Up-Regulation of MiR-181a Expression in LNCaP Cells Using 3D Cell Culture Model. Avicenna J. Med. Biotechnol. 2022, 14, 89–94. [Google Scholar] [CrossRef] [PubMed]
  111. Lin, Z.; Huang, S.; LingHu, X.; Wang, Y.; Wang, B.; Zhong, S.; Xie, S.; Xu, X.; Yu, A.; Nagai, A.; et al. Perillaldehyde Inhibits Bone Metastasis and Receptor Activator of Nuclear Factor-ΚB Ligand (RANKL) Signaling-Induced Osteoclastogenesis in Prostate Cancer Cell Lines. Bioengineered 2022, 13, 2710–2719. [Google Scholar] [CrossRef]
  112. Zuo, J.; Guo, Y.; Peng, X.; Tang, Y.; Zhang, X.; He, P.; Li, S.; Wa, Q.; Li, J.; Huang, S.; et al. Inhibitory Action of Pristimerin on Hypoxia-mediated Metastasis Involves Stem Cell Characteristics and EMT in PC-3 Prostate Cancer Cells. Oncol. Rep. 2015, 33, 1388–1394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Lee, Y.-J.; Park, K.-S.; Lee, S.-H. Curcumin Targets Both Apoptosis and Necroptosis in Acidity-Tolerant Prostate Carcinoma Cells. Biomed. Res. Int. 2021, 2021, 8859181. [Google Scholar] [CrossRef] [PubMed]
  114. Jang, Y.-G.; Ko, E.-B.; Choi, K.-C. Gallic Acid, a Phenolic Acid, Hinders the Progression of Prostate Cancer by Inhibition of Histone Deacetylase 1 and 2 Expression. J. Nutr. Biochem. 2020, 84, 108444. [Google Scholar] [CrossRef]
  115. Tyagi, A.; Kumar, S.; Raina, K.; Wempe, M.F.; Maroni, P.D.; Agarwal, R.; Agarwal, C. Differential Effect of Grape Seed Extract and Its Active Constituent Procyanidin B2 3,3″-Di-O-Gallate against Prostate Cancer Stem Cells. Mol. Carcinog. 2019, 58, 1105–1117. [Google Scholar] [CrossRef] [PubMed]
  116. Jang, Y.-G.; Hwang, K.-A.; Choi, K.-C. Rosmarinic Acid, a Component of Rosemary Tea, Induced the Cell Cycle Arrest and Apoptosis through Modulation of HDAC2 Expression in Prostate Cancer Cell Lines. Nutrients 2018, 10, 1784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kafka, M.; Mayr, F.; Temml, V.; Möller, G.; Adamski, J.; Höfer, J.; Schwaiger, S.; Heidegger, I.; Matuszczak, B.; Schuster, D.; et al. Dual Inhibitory Action of a Novel AKR1C3 Inhibitor on Both Full-Length AR and the Variant AR-V7 in Enzalutamide Resistant Metastatic Castration Resistant Prostate Cancer. Cancers 2020, 12, 2092. [Google Scholar] [CrossRef]
  118. Deezagi, A.; Safari, N. Rosuvastatin Inhibit Spheroid Formation and Epithelial-Mesenchymal Transition (EMT) in Prostate Cancer PC-3 Cell Line. Mol. Biol. Rep. 2020, 47, 8727–8737. [Google Scholar] [CrossRef]
  119. Sugawara, T.; Baumgart, S.J.; Nevedomskaya, E.; Reichert, K.; Steuber, H.; Lejeune, P.; Mumberg, D.; Haendler, B. Darolutamide Is a Potent Androgen Receptor Antagonist with Strong Efficacy in Prostate Cancer Models. Int. J. Cancer 2019, 145, 1382–1394. [Google Scholar] [CrossRef]
  120. Abramenkovs, A.; Hariri, M.; Spiegelberg, D.; Nilsson, S.; Stenerlöw, B. Ra-223 Induces Clustered DNA Damage and Inhibits Cell Survival in Several Prostate Cancer Cell Lines. Transl. Oncol. 2022, 26, 101543. [Google Scholar] [CrossRef]
  121. Salerno, D.; Howe, A.; Bhatavdekar, O.; Josefsson, A.; Pacheco-Torres, J.; Bhujwalla, Z.M.; Gabrielson, K.L.; Sofou, S. Two Diverse Carriers Are Better than One: A Case Study in α-Particle Therapy for Prostate Specific Membrane Antigen-Expressing Prostate Cancers. Bioeng. Transl. Med. 2022, 7, e10266. [Google Scholar] [CrossRef]
  122. Pinto, C.I.G.; Bucar, S.; Alves, V.; Fonseca, A.; Abrunhosa, A.J.; da Silva, C.L.; Guerreiro, J.F.; Mendes, F. Copper-64 Chloride Exhibits Therapeutic Potential in Three-Dimensional Cellular Models of Prostate Cancer. Front. Mol. Biosci. 2020, 7, 609172. [Google Scholar] [CrossRef] [PubMed]
  123. Nakamura, N.; Sloper, D.T. Altered Expression of Genes Identified in Rats with Prostatic Chronic Inflammation in a Prostate Spheroid Model Treated by Estradiol/Testosterone. J. Toxicol. Sci. 2021, 46, 515–523. [Google Scholar] [CrossRef]
  124. Dang, K.; Castello, G.; Clarke, S.C.; Li, Y.; Balasubramani, A.; Boudreau, A.; Davison, L.; Harris, K.E.; Pham, D.; Sankaran, P.; et al. Attenuating CD3 Affinity in a PSMAxCD3 Bispecific Antibody Enables Killing of Prostate Tumor Cells with Reduced Cytokine Release. J. Immunother. Cancer 2021, 9, e002488. [Google Scholar] [CrossRef] [PubMed]
  125. Gil, V.; Miranda, S.; Riisnaes, R.; Gurel, B.; D’Ambrosio, M.; Vasciaveo, A.; Crespo, M.; Ferreira, A.; Brina, D.; Troiani, M.; et al. HER3 Is an Actionable Target in Advanced Prostate Cancer. Cancer Res. 2021, 81, 6207–6218. [Google Scholar] [CrossRef] [PubMed]
  126. Nessler, I.; Khera, E.; Vance, S.; Kopp, A.; Qiu, Q.; Keating, T.A.; Abu-Yousif, A.O.; Sandal, T.; Legg, J.; Thompson, L.; et al. Increased Tumor Penetration of Single-Domain Antibody-Drug Conjugates Improves In Vivo Efficacy in Prostate Cancer Models. Cancer Res. 2020, 80, 1268–1278. [Google Scholar] [CrossRef]
  127. Stenberg, V.Y.; Larsen, R.H.; Ma, L.-W.; Peng, Q.; Juzenas, P.; Bruland, Ø.S.; Juzeniene, A. Evaluation of the PSMA-Binding Ligand 212Pb-NG001 in Multicellular Tumour Spheroid and Mouse Models of Prostate Cancer. Int. J. Mol. Sci. 2021, 22, 4815. [Google Scholar] [CrossRef]
  128. Masilamani, A.P.; Dettmer-Monaco, V.; Monaco, G.; Cathomen, T.; Kuckuck, I.; Schultze-Seemann, S.; Huber, N.; Wolf, P. An Anti-PSMA Immunotoxin Reduces Mcl-1 and Bcl2A1 and Specifically Induces in Combination with the BAD-Like BH3 Mimetic ABT-737 Apoptosis in Prostate Cancer Cells. Cancers 2020, 12, 1648. [Google Scholar] [CrossRef]
  129. Kedarinath, K.; Parks, G.D. Differential In Vitro Growth and Cell Killing of Cancer versus Benign Prostate Cells by Oncolytic Parainfluenza Virus. Pathogens 2022, 11, 493. [Google Scholar] [CrossRef]
  130. Landgraf, L.; Kozlowski, A.; Zhang, X.; Fournelle, M.; Becker, F.-J.; Tretbar, S.; Melzer, A. Focused Ultrasound Treatment of a Spheroid In Vitro Tumour Model. Cells 2022, 11, 1518. [Google Scholar] [CrossRef]
  131. Drápela, S.; Khirsariya, P.; van Weerden, W.M.; Fedr, R.; Suchánková, T.; Búzová, D.; Červený, J.; Hampl, A.; Puhr, M.; Watson, W.R.; et al. The CHK1 Inhibitor MU380 Significantly Increases the Sensitivity of Human Docetaxel-Resistant Prostate Cancer Cells to Gemcitabine through the Induction of Mitotic Catastrophe. Mol. Oncol. 2020, 14, 2487–2503. [Google Scholar] [CrossRef]
  132. Moreira-Silva, F.; Outeiro-Pinho, G.; Lobo, J.; Guimarães, R.; Gaspar, V.M.; Mano, J.F.; Agirre, X.; Pineda-Lucena, A.; Prosper, F.; Paramio, J.M.; et al. G9a Inhibition by CM-272: Developing a Novel Anti-Tumoral Strategy for Castration-Resistant Prostate Cancer Using 2D and 3D in Vitro Models. Biomed. Pharmacother. 2022, 150, 113031. [Google Scholar] [CrossRef] [PubMed]
  133. Karkampouna, S.; La Manna, F.; Benjak, A.; Kiener, M.; De Menna, M.; Zoni, E.; Grosjean, J.; Klima, I.; Garofoli, A.; Bolis, M.; et al. Patient-Derived Xenografts and Organoids Model Therapy Response in Prostate Cancer. Nat. Commun. 2021, 12, 1117. [Google Scholar] [CrossRef] [PubMed]
  134. Ko, J.; Meyer, A.N.; Haas, M.; Donoghue, D.J. Characterization of FGFR Signaling in Prostate Cancer Stem Cells and Inhibition via TKI Treatment. Oncotarget 2021, 12, 22–36. [Google Scholar] [CrossRef] [PubMed]
  135. Rana, Z.; Diermeier, S.; Walsh, F.P.; Hanif, M.; Hartinger, C.G.; Rosengren, R.J. Anti-Proliferative, Anti-Angiogenic and Safety Profiles of Novel HDAC Inhibitors for the Treatment of Metastatic Castration-Resistant Prostate Cancer. Pharmaceuticals 2021, 14, 1020. [Google Scholar] [CrossRef]
  136. Jiang, X.; Renkema, H.; Smeitink, J.; Beyrath, J. Sonlicromanol’s Active Metabolite KH176m Normalizes Prostate Cancer Stem Cell MPGES-1 Overexpression and Inhibits Cancer Spheroid Growth. PLoS ONE 2021, 16, e0254315. [Google Scholar] [CrossRef]
  137. Di Donato, M.; Ostacolo, C.; Giovannelli, P.; Di Sarno, V.; Monterrey, I.M.G.; Campiglia, P.; Migliaccio, A.; Bertamino, A.; Castoria, G. Therapeutic Potential of TRPM8 Antagonists in Prostate Cancer. Sci. Rep. 2021, 11, 23232. [Google Scholar] [CrossRef]
  138. Fu, W.; Zhao, M.T.; Driver, L.M.; Schirmer, A.U.; Yin, Q.; You, S.; Freedland, S.J.; DiGiovanni, J.; Drewry, D.H.; Macias, E. NUAK Family Kinase 2 Is a Novel Therapeutic Target for Prostate Cancer. Mol. Carcinog. 2022, 61, 334–345. [Google Scholar] [CrossRef]
  139. Jäntti, M.H.; Talman, V.; Räsänen, K.; Tarvainen, I.; Koistinen, H.; Tuominen, R.K. Anticancer Activity of the Protein Kinase C Modulator HMI-1a3 in 2D and 3D Cell Culture Models of Androgen-Responsive and Androgen-Unresponsive Prostate Cancer. FEBS Open Bio 2018, 8, 817–828. [Google Scholar] [CrossRef] [Green Version]
  140. Argenziano, M.; Foglietta, F.; Canaparo, R.; Spagnolo, R.; Della Pepa, C.; Caldera, F.; Trotta, F.; Serpe, L.; Cavalli, R. Biological Effect Evaluation of Glutathione-Responsive Cyclodextrin-Based Nanosponges: 2D and 3D Studies. Molecules 2020, 25, 2775. [Google Scholar] [CrossRef]
  141. Komarnicka, U.K.; Kozieł, S.; Pucelik, B.; Barzowska, A.; Siczek, M.; Malik, M.; Wojtala, D.; Niorettini, A.; Kyzioł, A.; Sebastian, V.; et al. Liposomal Binuclear Ir(III)-Cu(II) Coordination Compounds with Phosphino-Fluoroquinolone Conjugates for Human Prostate Carcinoma Treatment. Inorg. Chem. 2022, 61, 19261–19273. [Google Scholar] [CrossRef]
  142. Novohradsky, V.; Markova, L.; Kostrhunova, H.; Kasparkova, J.; Ruiz, J.; Marchán, V.; Brabec, V. A Cyclometalated IrIII Complex Conjugated to a Coumarin Derivative Is a Potent Photodynamic Agent against Prostate Differentiated and Tumorigenic Cancer Stem Cells. Chemistry 2021, 27, 8547–8556. [Google Scholar] [CrossRef] [PubMed]
  143. Ochieng, J.; Korolkova, O.Y.; Li, G.; Jin, R.; Chen, Z.; Matusik, R.J.; Adunyah, S.; Sakwe, A.M.; Ogunkua, O. Fetuin-A Promotes 3-Dimensional Growth in LNCaP Prostate Cancer Cells by Sequestering Extracellular Vesicles to Their Surfaces to Act as Signaling Platforms. Int. J. Mol. Sci. 2022, 23, 4031. [Google Scholar] [CrossRef] [PubMed]
  144. Kim, J.W.; Park, M.; Kim, S.; Lim, S.C.; Kim, H.S.; Kang, K.W. Anti-Metastatic Effect of GV1001 on Prostate Cancer Cells; Roles of GnRHR-Mediated Gαs-CAMP Pathway and AR-YAP1 Axis. Cell Biosci. 2021, 11, 191. [Google Scholar] [CrossRef] [PubMed]
  145. Shropshire, D.B.; Acosta, F.M.; Fang, K.; Benavides, J.; Sun, L.-Z.; Jin, V.X.; Jiang, J.X. Association of Adenosine Signaling Gene Signature with Estrogen Receptor-Positive Breast and Prostate Cancer Bone Metastasis. Front. Med. 2022, 9, 965429. [Google Scholar] [CrossRef]
  146. Fontana, F.; Raimondi, M.; Marzagalli, M.; Sommariva, M.; Gagliano, N.; Limonta, P. Three-Dimensional Cell Cultures as an In Vitro Tool for Prostate Cancer Modeling and Drug Discovery. Int. J. Mol. Sci. 2020, 21, 6806. [Google Scholar] [CrossRef] [PubMed]
  147. Palacios, D.A.; Miyake, M.; Rosser, C.J. Radiosensitization in Prostate Cancer: Mechanisms and Targets. BMC Urol. 2013, 13, 4. [Google Scholar] [CrossRef] [Green Version]
  148. Camus, V.L.; Stewart, G.D.; Nailon, W.H.; McLaren, D.B.; Campbell, C.J. Measuring the Effects of Fractionated Radiation Therapy in a 3D Prostate Cancer Model System Using SERS Nanosensors. Analyst 2016, 141, 5056–5061. [Google Scholar] [CrossRef] [Green Version]
  149. Rae, C.; Mairs, R.J. AMPK Activation by AICAR Sensitizes Prostate Cancer Cells to Radiotherapy. Oncotarget 2019, 10, 749–759. [Google Scholar] [CrossRef] [Green Version]
  150. Silva, V.L.; Ruiz, A.; Ali, A.; Pereira, S.; Seitsonen, J.; Ruokolainen, J.; Furlong, F.; Coulter, J.; Al-Jamal, W.T. Hypoxia-Targeted Cupric-Tirapazamine Liposomes Potentiate Radiotherapy in Prostate Cancer Spheroids. Int. J. Pharm. 2021, 607, 121018. [Google Scholar] [CrossRef]
  151. Rove, K.O.; Crawford, E.D. Androgen Annihilation as a New Therapeutic Paradigm in Advanced Prostate Cancer. Curr. Opin. Urol. 2013, 23, 208–213. [Google Scholar] [CrossRef]
  152. Choi, E.; Buie, J.; Camacho, J.; Sharma, P.; de Riese, W.T.W. Evolution of Androgen Deprivation Therapy (ADT) and Its New Emerging Modalities in Prostate Cancer: An Update for Practicing Urologists, Clinicians and Medical Providers. Res. Rep. Urol. 2022, 14, 87–108. [Google Scholar] [CrossRef] [PubMed]
  153. Rice, M.A.; Malhotra, S.V.; Stoyanova, T. Second-Generation Antiandrogens: From Discovery to Standard of Care in Castration Resistant Prostate Cancer. Front. Oncol. 2019, 9, 801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. James, N.D.; de Bono, J.S.; Spears, M.R.; Clarke, N.W.; Mason, M.D.; Dearnaley, D.P.; Ritchie, A.W.S.; Amos, C.L.; Gilson, C.; Jones, R.J.; et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N. Engl. J. Med. 2017, 377, 338–351. [Google Scholar] [CrossRef] [PubMed]
  155. Crawford, E.D.; Heidenreich, A.; Lawrentschuk, N.; Tombal, B.; Pompeo, A.C.L.; Mendoza-Valdes, A.; Miller, K.; Debruyne, F.M.J.; Klotz, L. Androgen-Targeted Therapy in Men with Prostate Cancer: Evolving Practice and Future Considerations. Prostate Cancer Prostatic Dis. 2019, 22, 24–38. [Google Scholar] [CrossRef] [Green Version]
  156. Maughan, B.L.; Antonarakis, E.S. Androgen Pathway Resistance in Prostate Cancer and Therapeutic Implications. Expert Opin. Pharmacother. 2015, 16, 1521–1537. [Google Scholar] [CrossRef]
  157. Eder, T.; Weber, A.; Neuwirt, H.; Grünbacher, G.; Ploner, C.; Klocker, H.; Sampson, N.; Eder, I.E. Cancer-Associated Fibroblasts Modify the Response of Prostate Cancer Cells to Androgen and Anti-Androgens in Three-Dimensional Spheroid Culture. Int. J. Mol. Sci. 2016, 17, 1458. [Google Scholar] [CrossRef] [Green Version]
  158. Thomas, T.S.; Pachynski, R.K. Treatment of Advanced Prostate Cancer. Mo Med. 2018, 115, 156–161. [Google Scholar]
  159. Karandish, F.; Haldar, M.K.; You, S.; Brooks, A.E.; Brooks, B.D.; Guo, B.; Choi, Y.; Mallik, S. Prostate-Specific Membrane Antigen Targeted Polymersomes for Delivering Mocetinostat and Docetaxel to Prostate Cancer Cell Spheroids. ACS Omega 2016, 1, 952–962. [Google Scholar] [CrossRef]
  160. Du, A.W.; Lu, H.; Stenzel, M.H. Core-Cross-Linking Accelerates Antitumor Activities of Paclitaxel-Conjugate Micelles to Prostate Multicellular Tumor Spheroids: A Comparison of 2D and 3D Models. Biomacromolecules 2015, 16, 1470–1479. [Google Scholar] [CrossRef]
  161. Ohya, S.; Kajikuri, J.; Endo, K.; Kito, H.; Matsui, M. KCa1.1 K+ Channel Inhibition Overcomes Resistance to Antiandrogens and Doxorubicin in a Human Prostate Cancer LNCaP Spheroid Model. Int. J. Mol. Sci. 2021, 22, 13553. [Google Scholar] [CrossRef]
  162. Lee, S.-H.; Lee, Y.-J. Synergistic Anticancer Activity of Resveratrol in Combination with Docetaxel in Prostate Carcinoma Cells. Nutr. Res. Pract. 2021, 15, 12–25. [Google Scholar] [CrossRef] [PubMed]
  163. Moreira-Silva, F.; Henrique, R.; Jerónimo, C. From Therapy Resistance to Targeted Therapies in Prostate Cancer. Front. Oncol. 2022, 12, 877379. [Google Scholar] [CrossRef] [PubMed]
  164. Petrioli, R.; Francini, E.; Fiaschi, A.I.; Laera, L.; Roviello, G. Targeted Therapies for Prostate Cancer. Cancer Invest. 2015, 33, 276–285. [Google Scholar] [CrossRef] [PubMed]
  165. Duque, J.L.F.; Loughlin, K.R.; Adam, R.M.; Kantoff, P.; Mazzucchi, E.; Freeman, M.R. Measurement of Plasma Levels of Vascular Endothelial Growth Factor in Prostate Cancer Patients: Relationship with Clinical Stage, Gleason Score, Prostate Volume, and Serum Prostate-Specific Antigen. Clinics 2006, 61, 401–408. [Google Scholar] [CrossRef]
  166. Duque, J.L.; Loughlin, K.R.; Adam, R.M.; Kantoff, P.W.; Zurakowski, D.; Freeman, M.R. Plasma Levels of Vascular Endothelial Growth Factor Are Increased in Patients with Metastatic Prostate Cancer. Urology 1999, 54, 523–527. [Google Scholar] [CrossRef]
  167. Oner, E.; Kotmakci, M.; Baird, A.-M.; Gray, S.G.; Debelec Butuner, B.; Bozkurt, E.; Kantarci, A.G.; Finn, S.P. Development of EphA2 SiRNA-Loaded Lipid Nanoparticles and Combination with a Small-Molecule Histone Demethylase Inhibitor in Prostate Cancer Cells and Tumor Spheroids. J. Nanobiotechnol. 2021, 19, 71. [Google Scholar] [CrossRef]
  168. Bagnato, A.; Rosanò, L. The Endothelin Axis in Cancer. Int. J. Biochem. Cell. Biol. 2008, 40, 1443–1451. [Google Scholar] [CrossRef]
  169. Chi, K.N.; Hotte, S.J.; Yu, E.Y.; Tu, D.; Eigl, B.J.; Tannock, I.; Saad, F.; North, S.; Powers, J.; Gleave, M.E.; et al. Randomized Phase II Study of Docetaxel and Prednisone with or without OGX-011 in Patients with Metastatic Castration-Resistant Prostate Cancer. J. Clin. Oncol. 2010, 28, 4247–4254. [Google Scholar] [CrossRef]
  170. Toren, P.; Zoubeidi, A. Targeting the PI3K/Akt Pathway in Prostate Cancer: Challenges and Opportunities (Review). Int. J. Oncol. 2014, 45, 1793–1801. [Google Scholar] [CrossRef] [Green Version]
  171. Jones, D.T.; Valli, A.; Haider, S.; Zhang, Q.; Smethurst, E.A.; Schug, Z.T.; Peck, B.; Aboagye, E.O.; Critchlow, S.E.; Schulze, A.; et al. 3D Growth of Cancer Cells Elicits Sensitivity to Kinase Inhibitors but Not Lipid Metabolism Modifiers. Mol. Cancer Ther. 2019, 18, 376–388. [Google Scholar] [CrossRef] [Green Version]
  172. Tee, S.S.; Suster, I.; Truong, S.; Jeong, S.; Eskandari, R.; DiGialleonardo, V.; Alvarez, J.A.; Aldeborgh, H.N.; Keshari, K.R. Targeted AKT Inhibition in Prostate Cancer Cells and Spheroids Reduces Aerobic Glycolysis and Generation of Hyperpolarized [1-13C] Lactate. Mol. Cancer Res. 2018, 16, 453–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Aghajani, M.; Mokhtarzadeh, A.; Aghebati-Maleki, L.; Mansoori, B.; Mohammadi, A.; Safaei, S.; Asadzadeh, Z.; Hajiasgharzadeh, K.; Khaze Shahgoli, V.; Baradaran, B. CD133 Suppression Increases the Sensitivity of Prostate Cancer Cells to Paclitaxel. Mol. Biol. Rep. 2020, 47, 3691–3703. [Google Scholar] [CrossRef] [PubMed]
  174. Shenoy, T.R.; Boysen, G.; Wang, M.Y.; Xu, Q.Z.; Guo, W.; Koh, F.M.; Wang, C.; Zhang, L.Z.; Wang, Y.; Gil, V.; et al. CHD1 Loss Sensitizes Prostate Cancer to DNA Damaging Therapy by Promoting Error-Prone Double-Strand Break Repair. Ann. Oncol. 2017, 28, 1495–1507. [Google Scholar] [CrossRef] [PubMed]
  175. Erdogan, S.; Turkekul, K.; Dibirdik, I.; Doganlar, O.; Doganlar, Z.B.; Bilir, A.; Oktem, G. Midkine Downregulation Increases the Efficacy of Quercetin on Prostate Cancer Stem Cell Survival and Migration through PI3K/AKT and MAPK/ERK Pathway. Biomed. Pharmacother. 2018, 107, 793–805. [Google Scholar] [CrossRef]
  176. Lee, Y.-J.; Lee, S.-H. ERK1/2-Dependent Inhibition of Glycolysis in Curcumin-Induced Cytotoxicity of Prostate Carcinoma Cells. Biomed. Res. Int. 2022, 2022, 7626405. [Google Scholar] [CrossRef]
  177. Boccellino, M.; Ambrosio, P.; Ballini, A.; De Vito, D.; Scacco, S.; Cantore, S.; Feola, A.; Di Donato, M.; Quagliuolo, L.; Sciarra, A.; et al. The Role of Curcumin in Prostate Cancer Cells and Derived Spheroids. Cancers 2022, 14, 3348. [Google Scholar] [CrossRef]
  178. Song, L.; Mino, M.; Yamak, J.; Nguyen, V.; Lopez, D.; Pham, V.; Fazelpour, A.; Le, V.; Fu, D.; Tippin, M.; et al. Flavokawain A Reduces Tumor-Initiating Properties and Stemness of Prostate Cancer. Front. Oncol. 2022, 12, 943846. [Google Scholar] [CrossRef] [PubMed]
  179. Tunki, L.; Jangid, A.K.; Pooja, D.; Bhargava, S.K.; Sistla, R.; Kulhari, H. Serotonin-Functionalized Vit-E Nanomicelles for Targeting of Irinotecan to Prostate Cancer Cells. ACS Appl. Bio Mater. 2020, 3, 5093–5102. [Google Scholar] [CrossRef]
  180. Zhang, L.; Shan, X.; Meng, X.; Gu, T.; Lu, Q.; Zhang, J.; Chen, J.; Jiang, Q.; Ning, X. The First Integrins Β3-Mediated Cellular and Nuclear Targeting Therapeutics for Prostate Cancer. Biomaterials 2019, 223, 119471. [Google Scholar] [CrossRef]
  181. Tieu, T.; Wojnilowicz, M.; Huda, P.; Thurecht, K.J.; Thissen, H.; Voelcker, N.H.; Cifuentes-Rius, A. Nanobody-Displaying Porous Silicon Nanoparticles for the Co-Delivery of SiRNA and Doxorubicin. Biomater. Sci. 2021, 9, 133–147. [Google Scholar] [CrossRef]
  182. Lee, Y.-J.; Nam, H.-S.; Cho, M.-K.; Lee, S.-H. Arctigenin Induces Necroptosis through Mitochondrial Dysfunction with CCN1 Upregulation in Prostate Cancer Cells under Lactic Acidosis. Mol. Cell Biochem. 2020, 467, 45–56. [Google Scholar] [CrossRef] [PubMed]
  183. Beshiri, M.L.; Tice, C.M.; Tran, C.; Nguyen, H.M.; Sowalsky, A.G.; Agarwal, S.; Jansson, K.H.; Yang, Q.; McGowen, K.M.; Yin, J.; et al. A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening. Clin. Cancer Res. 2018, 24, 4332–4345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Grayson, K.A.; Jyotsana, N.; Ortiz-Otero, N.; King, M.R. Overcoming TRAIL-Resistance by Sensitizing Prostate Cancer 3D Spheroids with Taxanes. PLoS ONE 2021, 16, e0246733. [Google Scholar] [CrossRef] [PubMed]
  185. Uwada, J.; Mukai, S.; Terada, N.; Nakazawa, H.; Islam, M.S.; Nagai, T.; Fujii, M.; Yamasaki, K.; Taniguchi, T.; Kamoto, T.; et al. Pleiotropic Effects of Probenecid on Three-Dimensional Cultures of Prostate Cancer Cells. Life Sci. 2021, 278, 119554. [Google Scholar] [CrossRef]
  186. Davary Avareshk, A.; Jalal, R.; Gholami, J. The Effect of Ciprofloxacin on Doxorubicin Cytotoxic Activity in the Acquired Resistance to Doxorubicin in DU145 Prostate Carcinoma Cells. Med. Oncol. 2022, 39, 194. [Google Scholar] [CrossRef]
  187. Sauer, H.; Kampmann, H.; Khosravi, F.; Sharifpanah, F.; Wartenberg, M. The Nicotinamide Phosphoribosyltransferase Antagonist FK866 Inhibits Growth of Prostate Tumour Spheroids and Increases Doxorubicin Retention without Changes in Drug Transporter and Cancer Stem Cell Protein Expression. Clin. Exp. Pharmacol. Physiol. 2021, 48, 422–434. [Google Scholar] [CrossRef]
  188. Gupta, S.; Pungsrinont, T.; Ženata, O.; Neubert, L.; Vrzal, R.; Baniahmad, A. Interleukin-23 Represses the Level of Cell Senescence Induced by the Androgen Receptor Antagonists Enzalutamide and Darolutamide in Castration-Resistant Prostate Cancer Cells. Horm. Cancer 2020, 11, 182–190. [Google Scholar] [CrossRef]
  189. Di Donato, M.; Giovannelli, P.; Barone, M.V.; Auricchio, F.; Castoria, G.; Migliaccio, A. A Small Peptide Targeting the Ligand-Induced Androgen Receptor/Filamin a Interaction Inhibits the Invasive Phenotype of Prostate Cancer Cells. Cells 2021, 11, 14. [Google Scholar] [CrossRef]
  190. Zhang, Z.; Karthaus, W.R.; Lee, Y.S.; Gao, V.R.; Wu, C.; Russo, J.W.; Liu, M.; Mota, J.M.; Abida, W.; Linton, E.; et al. Tumor Microenvironment-Derived NRG1 Promotes Antiandrogen Resistance in Prostate Cancer. Cancer Cell 2020, 38, 279–296.e9. [Google Scholar] [CrossRef]
  191. Nakayama, T.; Sano, T.; Oshimo, Y.; Kawada, C.; Kasai, M.; Yamamoto, S.; Fukuhara, H.; Inoue, K.; Ogura, S.-I. Enhanced Lipid Metabolism Induces the Sensitivity of Dormant Cancer Cells to 5-Aminolevulinic Acid-Based Photodynamic Therapy. Sci. Rep. 2021, 11, 7290. [Google Scholar] [CrossRef]
  192. Rajaee, Z.; Khoei, S.; Mahdavi, S.R.; Ebrahimi, M.; Shirvalilou, S.; Mahdavian, A. Evaluation of the Effect of Hyperthermia and Electron Radiation on Prostate Cancer Stem Cells. Radiat. Environ. Biophys. 2018, 57, 133–142. [Google Scholar] [CrossRef] [PubMed]
  193. Lim, S.C.; Jansson, P.J.; Assinder, S.J.; Maleki, S.; Richardson, D.R.; Kovacevic, Z. Unique Targeting of Androgen-Dependent and -Independent AR Signaling in Prostate Cancer to Overcome Androgen Resistance. FASEB J. 2020, 34, 11511–11528. [Google Scholar] [CrossRef]
  194. Itkonen, H.M.; Poulose, N.; Steele, R.E.; Martin, S.E.S.; Levine, Z.G.; Duveau, D.Y.; Carelli, R.; Singh, R.; Urbanucci, A.; Loda, M.; et al. Inhibition of O-GlcNAc Transferase Renders Prostate Cancer Cells Dependent on CDK9. Mol. Cancer Res. 2020, 18, 1512–1521. [Google Scholar] [CrossRef] [PubMed]
  195. Iannelli, F.; Roca, M.S.; Lombardi, R.; Ciardiello, C.; Grumetti, L.; De Rienzo, S.; Moccia, T.; Vitagliano, C.; Sorice, A.; Costantini, S.; et al. Synergistic Antitumor Interaction of Valproic Acid and Simvastatin Sensitizes Prostate Cancer to Docetaxel by Targeting CSCs Compartment via YAP Inhibition. J. Exp. Clin. Cancer Res. 2020, 39, 213. [Google Scholar] [CrossRef] [PubMed]
  196. Dozzo, A.; Chullipalliyalil, K.; McAuliffe, M.; O’Driscoll, C.M.; Ryan, K.B. Nano-Hydroxyapatite/PLGA Mixed Scaffolds as a Tool for Drug Development and to Study Metastatic Prostate Cancer in the Bone. Pharmaceutics 2023, 15, 242. [Google Scholar] [CrossRef] [PubMed]
  197. Molla, M.S.; Katti, D.R.; Katti, K.S. An in Vitro Model of Prostate Cancer Bone Metastasis for Highly Metastatic and Non-Metastatic Prostate Cancer Using Nanoclay Bone-Mimetic Scaffolds. MRS Adv. 2019, 4, 1207–1213. [Google Scholar] [CrossRef]
  198. Paindelli, C.; Navone, N.; Logothetis, C.J.; Friedl, P.; Dondossola, E. Engineered Bone for Probing Organotypic Growth and Therapy Response of Prostate Cancer Tumoroids in Vitro. Biomaterials 2019, 197, 296–304. [Google Scholar] [CrossRef]
  199. Miyahira, A.K.; Sharp, A.; Ellis, L.; Jones, J.; Kaochar, S.; Larman, H.B.; Quigley, D.A.; Ye, H.; Simons, J.W.; Pienta, K.J.; et al. Prostate Cancer Research: The next Generation; Report from the 2019 Coffey-Holden Prostate Cancer Academy Meeting. Prostate 2020, 80, 113–132. [Google Scholar] [CrossRef]
  200. Ittmann, M.; Huang, J.; Radaelli, E.; Martin, P.; Signoretti, S.; Sullivan, R.; Simons, B.W.; Ward, J.M.; Robinson, B.D.; Chu, G.C.; et al. Animal Models of Human Prostate Cancer: The Consensus Report of the New York Meeting of the Mouse Models of Human Cancers Consortium Prostate Pathology Committee. Cancer Res. 2013, 73, 2718–2736. [Google Scholar] [CrossRef] [Green Version]
  201. Parisotto, M.; Metzger, D. Genetically Engineered Mouse Models of Prostate Cancer. Mol. Oncol. 2013, 7, 190–205. [Google Scholar] [CrossRef]
  202. Simons, B.W.; Kothari, V.; Benzon, B.; Ghabili, K.; Hughes, R.; Zarif, J.C.; Ross, A.E.; Hurley, P.J.; Schaeffer, E.M. A Mouse Model of Prostate Cancer Bone Metastasis in a Syngeneic Immunocompetent Host. Oncotarget 2019, 10, 6845–6854. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Outline of different methods for generating 3D models. If the starting material is cell lines, they can be grown as single cultures or co-cultures to generate spheroids or reprogrammed to IPSC and then differentiated in vitro using matrices and scaffolds to generate organoids. If patient material is used, it is usually dissociated into single cells before plating onto scaffolds, matrices, or chips to generate organoids or tumoroids. Patient material can also be implanted into animal models as xenografts. The prepared 3D cultures can then be used for a variety of applications, as presented in this schematic. Created in Biorender.com.
Figure 1. Outline of different methods for generating 3D models. If the starting material is cell lines, they can be grown as single cultures or co-cultures to generate spheroids or reprogrammed to IPSC and then differentiated in vitro using matrices and scaffolds to generate organoids. If patient material is used, it is usually dissociated into single cells before plating onto scaffolds, matrices, or chips to generate organoids or tumoroids. Patient material can also be implanted into animal models as xenografts. The prepared 3D cultures can then be used for a variety of applications, as presented in this schematic. Created in Biorender.com.
Ijms 24 05293 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Petrić, T.; Sabol, M. Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer. Int. J. Mol. Sci. 2023, 24, 5293. https://doi.org/10.3390/ijms24065293

AMA Style

Petrić T, Sabol M. Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer. International Journal of Molecular Sciences. 2023; 24(6):5293. https://doi.org/10.3390/ijms24065293

Chicago/Turabian Style

Petrić, Tina, and Maja Sabol. 2023. "Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer" International Journal of Molecular Sciences 24, no. 6: 5293. https://doi.org/10.3390/ijms24065293

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop