Fact-checked by Grok 2 weeks ago

PC3

PC-3, commonly abbreviated as PC3, is a widely used human cell line derived from a of a grade IV prostatic in a 62-year-old male. Established in 1979, it exhibits epithelial morphology, adherent growth, and a near-triploid with a modal chromosome number of 62, lacking a normal . Unlike androgen-dependent models, PC3 is androgen-independent, does not express the (AR) or (PSA), and demonstrates aggressive tumorigenic potential, forming tumors in 100% of nude mice within 21 days. PC3 cells are particularly notable for their neuroendocrine differentiation, expressing markers such as chromogranin A (CgA) and neuron-specific enolase (NSE), with focal staining for cytokeratin 8 (CK8) and positive expression of the marker CD44. These features distinguish PC3 from other common lines like , which is androgen-responsive and lacks neuroendocrine characteristics, positioning PC3 as a model for prostatic neuroendocrine (SCNC)—a rare, aggressive subtype comprising less than 1% of cancers that is unresponsive to hormonal . The line's ability to grow in standard media like F-12K supplemented with 10% at 37°C under 5% CO₂ supports its routine use in studies, , and metastasis models, particularly for bone-specific investigations. In , PC3 has become a cornerstone for elucidating mechanisms of castration-resistant progression, , and neuroendocrine transdifferentiation, with applications in for novel therapeutics targeting advanced disease stages. Its commercial availability through repositories like the American Type Culture Collection (ATCC) ensures reproducibility across global studies, contributing to over thousands of publications on biology and .

Origin and Establishment

Derivation from Patient Tissue

The PC3 cell line was derived from a bone metastasis of a grade IV prostatic adenocarcinoma obtained from a 62-year-old Caucasian male patient. The primary tumor was undifferentiated, and the metastatic tissue was sourced in 1979. The isolation process involved explanting the tumor tissue and culturing it to establish an immortalized epithelial cell line, conducted by M.E. Kaighn and colleagues at the Naval Biosciences Laboratory in Oakland, California. The resulting PC3 cells were maintained in continuous culture, demonstrating epithelial characteristics confirmed through positive keratin staining. Human origin of the PC3 cell line was verified through karyotypic , revealing a near-triploid profile with a modal number of 62 and the absence of a . Q- and C-banding techniques identified across passages, with approximately 20 marker chromosomes present in each cell.

Initial Characterization and Validation

Following its isolation, the PC3 cell line underwent initial characterization to confirm its stability and relevance as a model for human prostatic . In a seminal , researchers established PC3 as an adherent epithelial culture exhibiting a of approximately 30 hours and the ability to reach high population densities of up to 2 × 10^5 cells per square centimeter. A key validation of PC3's malignant potential involved assessing its tumorigenicity . Subcutaneous injection of PC3 cells into athymic nude mice resulted in rapid tumor formation, with revealing undifferentiated malignant cells resembling the original from which the line was derived, thus confirming its aggressive phenotype. Early phenotypic assessments also demonstrated PC3's androgen unresponsiveness, a critical feature distinguishing it from hormone-dependent prostate models. Growth assays showed no stimulation of proliferation in response to dihydrotestosterone (DHT), even at concentrations up to 10 nM, underscoring its suitability for studying advanced, castration-resistant disease. Cytogenetic analysis further validated PC3's cancerous origin by revealing a hypotriploid aneuploid karyotype with a modal chromosome number of 62 and at least 10 marker chromosomes, with observations of modal shifts (e.g., from 62 to 55 across passages), clearly differentiating it from normal prostate epithelial cells.

Biological Characteristics

Morphology and Growth Behavior

PC-3 cells display an epithelial-like morphology characterized by adherent, polygonal shapes that form cohesive monolayers in standard two-dimensional culture, lacking the glandular structures observed in more differentiated prostate epithelial cells. Under phase-contrast microscopy, these cells appear irregular and flattened with prominent nucleoli and microvilli on their surface, reflecting their origin from a poorly differentiated adenocarcinoma. This morphology supports their use in modeling aggressive prostate cancer phenotypes in vitro. In terms of growth properties, PC-3 cells exhibit a of 24-36 hours when maintained in medium supplemented with 10% (FBS), L-glutamine, and antibiotics at 37°C in a 5% CO₂ atmosphere. Although primarily anchorage-dependent for expansion, they retain the for anchorage-independent , forming colonies in soft assays that mimic tumorigenic potential. Subculturing is typically performed at 70-80% using trypsin-EDTA to prevent overgrowth, with seeding densities adjusted to 1:3 to 1:6 ratios for optimal proliferation. PC-3 cells tolerate serum-free conditions for short durations, such as 24-72 hours, allowing their use in experiments investigating independence or exosome without significant cell death.

Molecular Profile and Androgen Independence

The PC3 cell line exhibits a distinct molecular profile characterized by the absence of (AR) expression and (PSA) production, hallmarks of its hormone-refractory phenotype. This lack of AR has been confirmed through analysis, , and quantitative (qRT-PCR) on both cultured cells and xenograft tumors, showing no detectable AR protein or mRNA levels. Similarly, PSA expression is undetectable by the same methods, distinguishing PC3 from androgen-dependent models like cells. Genomically, PC3 harbors key alterations that underpin its aggressive behavior, including a truncating in TP53 (exon 5: c.414delC, p.K139Rfs*31) leading to loss of p53 protein expression, and complete homozygous deletion of PTEN. Additionally, PC3 displays relative amplification of the (approximately 2.3-fold copy number increase), which contributes to dysregulated . These genetic changes align PC3 with advanced, metastatic cancers, particularly those progressing to castration-resistant states. At the proteomic level, PC3 expresses neuroendocrine markers such as chromogranin A and neuron-specific enolase (NSE), as evidenced by and qRT-PCR, conferring a resemblance to neuroendocrine (SCNC) of the . This neuroendocrine differentiation, combined with focal cytokeratin 8 staining and expression, further supports PC3's utility as a model for aggressive, AR-negative subtypes. PC3 demonstrates , with cell proliferation remaining unaffected in androgen-deprived conditions (e.g., media supplemented with charcoal-stripped ), unlike AR-positive lines that exhibit growth inhibition upon androgen withdrawal. This independence stems from reliance on alternative signaling pathways, notably the hyperactivated PI3K/AKT axis due to PTEN loss, which sustains growth, survival, and stem-like properties in the absence of AR signaling. Inhibition of PI3K/AKT in PC3 induces , underscoring its central role in maintaining the line's viability.

Derived Variants

Paclitaxel-Resistant PC3-PR Cells

The paclitaxel-resistant PC3-PR cell line was established from the parental androgen-independent cells through chronic, stepwise exposure to escalating concentrations of , beginning at 5 nM and progressively increasing until the cells could tolerate and be maintained in up to 500 nM of the drug. This selection process yielded a subline with substantially enhanced resistance, demonstrating an IC50 of 2577.3 nM for compared to 8.6 nM in the parental PC3 cells, representing over 300-fold resistance. Key mechanisms underlying this resistance in PC3-PR cells involve upregulation of the ABCB1 (also known as MDR1) gene, which encodes an that actively transports out of the cell, reducing its intracellular accumulation and cytotoxic effects; this overexpression is driven in part by the ETS1. These molecular changes collectively impair paclitaxel's primary while preserving cell viability. Phenotypically, PC3-PR cells exhibit a slower proliferation rate than the parental PC3 line, indicative of metabolic adaptations to persistent drug stress, yet they display heightened migratory and invasive potential, facilitating aggressive tumor-like behaviors. The subline retains the androgen-independent profile of its progenitor, enabling its use in modeling advanced, hormone-refractory disease states without influence.

Holoclone-Enriched Subpopulations

Holoclone-enriched subpopulations within the PC3 cell line were first identified through single-cell cloning techniques, revealing a subset of cells capable of forming small, tightly packed colonies indicative of high proliferative capacity and self-renewal potential. These holoclones, characterized by their compact, round morphology distinct from the more loosely adherent parental PC3 cells, represent a stem-like fraction enriched for tumor-initiating properties. Isolation is achieved via limiting dilution cloning, where PC3 cells are plated at clonal densities (e.g., 1 cell per well) in 96-well plates to derive monoclonal populations, allowing selection of holoclone-forming cells based on colony appearance after 2 weeks of culture. Such holoclones comprise approximately 10% of the total PC3 population, highlighting the intrinsic heterogeneity within this androgen-independent model. These subpopulations demonstrate sustained self-renewal, as evidenced by their ability to generate secondary holoclones upon replating, and exhibit superior sphere-forming efficiency under non-adherent conditions, underscoring their stem-like behavior. Unlike paraclones or meroclones, which show limited and , holoclones maintain long-term propagability while preserving the aggressive growth traits of the parental line. Molecular profiling of PC3 holoclones reveals elevated expression of FAM65B, a associated with tumor , which contributes to their enhanced self-renewal features. These cells also display increased tumorigenicity in xenograft models, forming tumors at lower cell inocula (e.g., 1,000-10,000 cells) compared to bulk PC3 populations, with tumors exhibiting accelerated growth and vascularization. This enrichment for tumor-initiating cells positions holoclone subpopulations as a valuable tool for studying stemness without relying on surface marker-based sorting.

Research Applications

Modeling Metastasis and Invasion

The PC3 cell line has been extensively employed in in vitro assays to investigate the invasive potential of cells, particularly through -coated transwell invasion assays that simulate barriers. In these setups, PC3 cells demonstrate high invasiveness, migrating through Matrigel layers at rates significantly higher than less aggressive prostate cell lines, reflecting their propensity. This invasiveness is partly attributed to the secretion of matrix metalloproteinases (MMPs), including MMP-2 and MMP-9, which degrade and other matrix components to facilitate penetration; gelatin zymography confirms elevated MMP-2 and MMP-9 activity in PC3-conditioned media compared to non-invasive controls. Such assays have been instrumental in elucidating how molecular alterations in PC3, such as upregulated protease expression, enable tissue . In vivo, PC3 cells are commonly implanted orthotopically into the of immunocompromised mice, such as nude or NOD-SCID strains, to model metastatic progression. Following implantation, PC3 tumors develop locally and reliably metastasize to regional lymph nodes, with variable spread to distant sites including . This model recapitulates castration-resistant dynamics, as PC3's androgen independence allows tumor growth and spread even in androgen-deprived environments, mimicking advanced human disease. metastases in these models often exhibit osteolytic characteristics, with PC3 cells inducing activation and , providing insights into skeletal complications of . A pivotal discovery from PC3-based studies is the critical role of the /SDF-1 () axis in directing cells to niches. PC3 cells express high levels of the receptor, which binds SDF-1 produced by stromal cells, promoting and to the endosteal surface; neutralization of this pathway significantly reduces bone homing and metastatic burden in orthotopic models. This mechanism underscores how signaling governs organ-specific , with PC3 experiments showing that overexpression enhances bone colonization while inhibitors like AMD3100 block it, highlighting therapeutic potential.

Drug Resistance and Therapeutic Screening

The PC-3 cell line has been extensively utilized in assays to evaluate the efficacy of anti-cancer agents, particularly and (HDAC) inhibitors, in models of hormone-refractory . These screens assess compound libraries for cytotoxic potential, identifying leads that disrupt cell proliferation in androgen-independent contexts. For instance, , a prototypical , demonstrates potent activity against parental PC-3 cells, with an IC50 value of approximately 30 nM in growth inhibition assays following 24-hour exposure. Similarly, HDAC inhibitors such as suberoylanilide hydroxamic acid (SAHA) have been tested in PC-3 cells, revealing dose-dependent inhibition of proliferation through induction of and arrest. Studies employing PC-3 cells have provided critical insights into mechanisms of , notably epithelial-to-mesenchymal transition ()-mediated resistance to , a standard for advanced . -resistant PC-3 sublines exhibit upregulated markers, including (VIM) and zinc finger E-box binding homeobox 1 (ZEB1), alongside stem-like features such as expression, which correlate with reduced drug sensitivity and increased relapse risk. Reversion of via ZEB1 knockdown restores responsiveness, underscoring as a targetable driver of resistance. The nuclear factor-kappa B () pathway further contributes to this resistance in PC-3 cells, where its constitutive activation promotes survival signaling; indirect inhibition of via complex blockade enhances -induced . PC-3 cells have facilitated validation of therapeutic targets, including the 3-kinase (PI3K) pathway, which is hyperactive in androgen-independent tumors. Treatment with the PI3K inhibitor LY294002 significantly reduces cell viability in PC-3 lines by blocking Akt and downstream signals. This approach highlights PI3K as a viable target for sensitizing resistant cells, with combination strategies amplifying effects against hormone-refractory disease. The paclitaxel-resistant PC-3-PR variant, derived from parental lines, further exemplifies these applications by modeling acquired resistance in screening platforms.

Significance and Limitations

Contributions to Prostate Cancer Understanding

The PC3 cell line has significantly advanced the understanding of androgen-independent growth mechanisms in , particularly through 1990s studies that elucidated the role of interleukin-6 (IL-6) in . Research demonstrated that PC3 cells produce and respond to IL-6, promoting in an androgen-deprived , which mimics the progression to castration-resistant (CRPC). This autocrine loop was shown to activate downstream pathways like /signal transducer and activator of transcription (JAK/STAT), contributing to tumor survival and aggressiveness independent of signaling. These findings highlighted IL-6 as a key mediator in the transition from hormone-sensitive to refractory disease, influencing subsequent therapeutic targeting strategies. PC3 has provided critical evidence for neuroendocrine differentiation in aggressive cancers, establishing its characteristics as a model for small cell neuroendocrine carcinoma (SCNC). Studies confirmed that PC3 cells lack and expression while exhibiting markers such as chromogranin A and neuron-specific (NSE), akin to clinical SCNC specimens. This linkage has informed the recognition that SCNC represents 10-20% of advanced CRPC cases, often emerging post-androgen deprivation therapy and associated with poor . By recapitulating these features, PC3 has facilitated insights into lineage plasticity and treatment resistance in neuroendocrine variants. Through overexpression models in PC3, has been identified as a key prognostic in progression. Experiments overexpressing in PC3 cells revealed increased invasiveness and a potential role in , correlating with aggressive disease phenotypes observed in patient samples. Findings in patient samples have established 's cytoplasmic and nuclear localization as indicative of higher Gleason scores and biochemical recurrence risk, positioning it as a valuable marker for stratifying high-risk cases. Such models underscored 's anti-apoptotic function via protein family mechanisms, guiding its evaluation in clinical panels. As of 2025, PC3 continues to be utilized in preclinical studies exploring therapeutics, such as rosmarinic acid nanocomplexes and high-intensity models, underscoring its enduring relevance in modeling metastatic and treatment-resistant .

Model Constraints and Complementary Approaches

The PC-3 cell line exhibits significant constraints as a model for , primarily due to its lack of (AR) expression and (PSA) production, which limits its utility in studying hormone-dependent aspects of the disease. Derived from a , PC-3 over-represents advanced castration-resistant (CRPC), capturing only a subset of aggressive, androgen-independent phenotypes while failing to reflect the AR-positive and PSA-expressing characteristics prevalent in most primary tumors and earlier-stage CRPC cases. Long-term culture of PC-3 cells introduces additional challenges through genetic and phenotypic drift, where adaptations to conditions alter genomic stability, profiles, and functional properties, potentially reducing reproducibility across studies. Furthermore, PC-3 provides incomplete modeling of primary tumors, as its metastatic origin and osteolytic lesion induction do not accurately mimic the osteoblastic metastases typical of human disease. Xenograft engraftment with PC-3 also shows variable take rates, typically ranging from 50% to 70% in orthotopic models, influenced by implantation site and host factors, which complicates consistent preclinical assessments. To address these limitations, researchers often integrate PC-3 with complementary models for more comprehensive investigations. Pairing PC-3 with AR-positive cell lines like enables parallel studies of androgen-dependent and -independent mechanisms, providing a broader spectrum of disease progression. Similarly, patient-derived xenografts (PDXs) offer enhanced fidelity to human tumor heterogeneity, including AR signaling and features, and are recommended alongside PC-3 for validating therapeutic responses in advanced CRPC.

References

  1. [1]
    PC-3 - CRL-1435 - ATCC
    PC-3 is a cell line initiated from a bone metastasis of a grade IV prostatic adenocarcinoma from a 62-year-old, White, male.Missing: definition - - | Show results with:definition - -
  2. [2]
    PC3 is a cell line characteristic of prostatic small cell carcinoma
    PC3 cells do not express AR and PSA, their proliferation is independent of androgen, and they are characteristic of SCNC, unlike LNCaP cells.
  3. [3]
    PC3 Is a Cell Line Characteristic of Prostatic Small Cell Carcinoma
    Mar 22, 2011 · The purpose of this study was to compare the important and relevant features of two most commonly used PC cell lines, LNCaP and PC3, with prostatic ...<|control11|><|separator|>
  4. [4]
    Establishment and characterization of a human prostatic carcinoma ...
    Abstract. The establishment, characterization, and tumorigenicity of a new epithelial cell line (PC-3) from a human prostatic adenocarcinoma metastatic to bone ...
  5. [5]
    Cellosaurus cell line PC-3 (CVCL_0035)
    PC3 is a cell line characteristic of prostatic small cell carcinoma. ... Carbon ion irradiation of the human prostate cancer cell line PC3: a whole genome ...
  6. [6]
    Current mouse and cell models in prostate cancer research - PMC
    The PC3 cell line was originally derived from a bone metastasis of human prostatic adenocarcinoma origin (Kaighn, et al. 1979). Intravenous injection of PC3 ...Missing: derivation | Show results with:derivation
  7. [7]
    Chromosomal Analysis of Human Prostatic Adenocarcinoma Cell ...
    Karotypic analysis of PC-3 by Q- and C-banding showed the cells to be aneuploid at all culture passage levels. The modal chromosome number shifted from 62 to 55 ...Missing: PC3 | Show results with:PC3
  8. [8]
    PC-3 cell line - AcceGen
    PC-3 is a human prostate adenocarcinoma cell line originally derived from a bone metastasis in a 62-year-old Caucasian male with advanced prostate cancer.Missing: modal | Show results with:modal
  9. [9]
    Proteomic Analysis of Microvesicles Released by the Human ... - NIH
    PC-3 cells were grown in serum-free cell culture medium for 3 days and ... Searches were performed with a tolerance on mass measurement of 0.3 Da. The ...
  10. [10]
    Serum deprivation initiates adaptation and survival to oxidative ...
    Jul 27, 2020 · Serum-containing cells in each prostate cancer cell line (PC3, DU145 and LNCaP) were vulnerable to H2O2 exposure and displayed significant cell ...
  11. [11]
    A novel spontaneous model of epithelial-mesenchymal transition ...
    Jan 17, 2017 · ... mesenchymal transition (EMT). Herein, we report the development of a ... PC-3 and DU145 cell lines, for evidence of spontaneous EMT ...
  12. [12]
    Epithelial-mesenchymal transition can suppress major attributes of ...
    Apr 16, 2012 · Knockdown of EMT transcription factors in mesenchymal-like PC-3/S ... PC-3/S cells were cultured at 70% confluence, at which time they ...
  13. [13]
    Genomic analyses of the metastasis-derived prostate cancer cell ...
    Here, we report on short-read whole-genome sequencing and genomic analyses of LNCaP, VCaP, and PC3 cells stably transduced with WT AR (PC3-AR).Missing: derivation tissue origin
  14. [14]
    [PDF] On the correlation of CTCF binding and c-myc amplification ... - bioRxiv
    Sep 29, 2014 · The cell line PC3 owns the highest relative copy number, apparent by the relative amplification of C-MYC by 2.3, more than the double amount of ...
  15. [15]
    The role of PTEN/Akt/PI3K signaling in the maintenance and ... - PNAS
    Jan 6, 2009 · Our data strongly suggest that the PTEN/PI3K/Akt pathways are critical for prostate cancer stem-like cell maintenance and that targeting PI3K signaling may be ...Results And Discussion · Gene Expression Profiles Of... · Pten Loss Leads To Increased...
  16. [16]
    Kinase Pathway Promotes Autocrine Fas-Induced Death of ...
    In this study, we examined the role of Fas in mediating apoptosis induced by inhibition of the PI3K pathway in LNCaP and PC3 prostate cancer cells through ...
  17. [17]
  18. [18]
    PC3 Human Prostate Carcinoma Cell Holoclones Contain Self ...
    Mar 13, 2008 · We provide direct experimental evidence that tumor cell holoclones contain stem-like cells that can initiate serially transplantable tumors.
  19. [19]
    PC3 prostate tumor-initiating cells with molecular profile FAM65B ...
    Dec 29, 2010 · PC3 tumors are sustained by a small number of tumor-initiating cells with stem-like characteristics, including strong self-renewal and pro-angiogenic ...
  20. [20]
    Targeting MMP-9, uPAR and Cathepsin B Inhibits Invasion ... - NIH
    Down regulation of MMP-9, uPAR and CB inhibited matrigel invasion, in vitro angiogenesis and wound healing migration ability of PC3 and DU145 prostate cancer ...Matrigel Invasion Assay · Figure 2. Mtt Assay And... · Apoptotic Pathway
  21. [21]
    Inhibition of CD147 expression reduces tumor cell invasion in ...
    Gelatin zymography assay indicated that secretions of MMP-2 and MMP-9 in PC-3 cells transfected by CD147.1 and CD147.3 were lower than those in PC-3 cells trans ...
  22. [22]
    RNA interference mediated suppression of TRPV6 inhibits the ...
    May 20, 2021 · Furthermore, the growth and invasion of PC3 cell xenografts in nude mice and the expression of the invasive enzymes, MMP2, MMP9, and cathepsin B ...3. Trpv6 Sirna Transfection · 8. Cell Migration Assay · 9. Cell Invasion Assay
  23. [23]
    Bone Metastasis of Prostate Cancer Can Be Therapeutically ...
    Orthotopically implanted PC3 cells have been observed to invade locally and metastasize readily to adjacent lymph nodes (21–23). Consistent with data from the ...
  24. [24]
    An Orthotopic Murine Model of Human Prostate Cancer Metastasis
    Sep 18, 2013 · In this paper we provide a novel murine model of human PCa metastasis. In this model, the human PCa cell lines PC3-M is orthotopically implanted ...
  25. [25]
    IN VIVO MODELS OF PROSTATE CANCER METASTASIS TO BONE
    36 : Surgical orthotopic implantation allows high lung and lymph node metastatic expression of human prostate carcinoma cell line PC-3 in nude mice.
  26. [26]
    Use of the Stromal Cell-derived Factor-1/CXCR4 Pathway in ...
    SDF-1 enhances binding of prostate cancer cells to human osteosarcoma cell lines and human bone marrow endothelium. 3′-O-Acetyl-2′,7′-bis(carboxyethyl)-4-5 ...
  27. [27]
    Homing of Cancer Cells to the Bone - PMC - NIH
    The SDF-1/CXCR4 pathway has also been shown to play a significant role in metastases of prostate cancer to the bone. Several prostate cancer cells lines PC3, ...
  28. [28]
    IL-6/IL-6R as a potential key signaling pathway in prostate cancer ...
    IL-6 acts as a paracrine and autocrine growth stimulator in benign and tumor prostate cells. The levels of IL-6 and respective receptors are increased during ...Missing: paper | Show results with:paper
  29. [29]
    Case Report: Systemic Treatment and Serial Genomic Sequencing ...
    Sep 26, 2021 · Small cell carcinoma constitutes 0.5–2% of prostate cancer cases, whereas 10–20% of CRPC cases; however, the current prevalence of SCC/NEPC ...
  30. [30]
    PC3 is a cell line characteristic of prostatic small cell carcinoma
    Mar 22, 2011 · Prostatic small cell neuroendocrine carcinoma (SCNC) is a variant form of prostate cancer (PC). In contrast to adenocarcinoma, the tumor cells ...
  31. [31]
    SURVIVIN IS A POTENTIAL MEDIATOR OF PROSTATE CANCER ...
    This study suggests that Survivin may be a potentially important prognostic marker and promising therapeutic target in metastatic prostate cancer.
  32. [32]
    Survivin expression is associated with features of biologically ...
    Feb 3, 2004 · We found that survivin was overexpressed in the cell cytoplasm of approximately one-third of normal prostatic cells from prostate carcinoma ...
  33. [33]
    Enhanced tumor suppression in vitro and in vivo by co-expression of ...
    Aug 13, 2010 · ... survivin and enhanced p53 gene expression in human PC3 prostate cancer cells ... survivin protein expression might serve as a prognostic marker ...
  34. [34]
    Unique Patterns of Molecular Profiling between Human Prostate ...
    This study showed that LNCaP and PC-3 cells represent two distinct prostate cancer cell lineages. LNCaP cells retain many prostate cell specific properties.
  35. [35]
    Major changes of cell function and toxicant sensitivity in cultured ...
    Oct 8, 2018 · Genomic drift affects the functional properties of cell lines, and the reproducibility of data from in vitro studies.
  36. [36]
    Animal models of bone metastatic prostate cancer - PMC
    Considering that human prostate cancer bone metastases are generally osteoblastic, the PC3 cell line does not fully reflect the nature of human prostate cancer.
  37. [37]
    The bone microenvironment promotes tumor growth and tissue ...
    Oct 16, 2018 · Bone tissue increases the take rate in the prostate cancer cell lines LnCap and Pc3 and is associated with reduced early tumor growth. A large ...
  38. [38]
    Application of Prostate Cancer Models for Preclinical Study - NIH
    Jan 20, 2019 · The IC50 of paclitaxel against the PC3PR cells is 2577.3 nM and that against the parental PC-3 cells is 8.6 nM. MSK1 knockdown by using miR-148a ...