Long term PCa organoids recapitulate the original tumor
A functional PCa biobank was established by collecting prostate tumor tissue pieces from prostatectomy samples (∼1 cm3 tissue) with informed written consent from the patients. We received 4-5 biopsies (Table S1) from each prostate and whenever possible, pieces were cut from both cancer and benign lesions, based on pathologist’s evaluation. Each piece was further divided into four parts that were separately processed for organoid derivation, histological analysis, genomic analysis, and patient-derived xenograft (PDX) modeling. Pathologist performed a Gleason scoring analysis for all the 36 histology samples (Table S1). PCa organoid cultures were established as described in experimental procedures and passaged every 14-20 days with a consistent passaging ratio of 1:3 either by gentle pipetting or trypsinization after dissolving the MatrigelTM blobs. After a couple of passages, aliquots of organoid lines were stored in liquid nitrogen from where they could be thawn for further expansion and assays.
Initially, ~5mm3 of fresh PCa tissue was processed into single-cell suspension (see methods) and seeded into four MatrigelTM blobs and grown for a minimum of 7 days as organoid cultures and imaged using phase contrast microcopy. The organoids generated from all patients posed heterogeneous morphologies, ranging from irregularly shaped solid cell clusters to spherical structures with a hollow lumen surrounded by columnar epithelial cells. Organoid forming capacity was measured in terms of number of organoids formed per each sample and individual organoids were classified into two main morphological groups, hollow organoids with clearly visible central lumen and solid organoids containing no or only small lumina. The total number of organoids in the different samples varied between 150 and 500 and in most of the samples there was slightly more solid than hollow organoids (Figure 1A). However, in some samples (e.g. PT-5, -6, -16, -20, -21, -23, -24, -29 and -34) more than two thirds of the organoids were solid cell clusters. Interestingly, when patient samples were stratified into two groups based on low (< 7) and high (> 7) post-operative Gleason scoring, it was observed that while hollow organoid numbers were similar in both groups, the number of solid organoids was significantly higher in the high Gleason group (Figure 1B). Upon prolonged culture also the initially hollow organoids started to develop multiple lumina as the clusters grew to form large glandular rosettes (Figure 1C). The size of the organoids continued to increase (Figure 1D, E) while the number of organoids declined during the 2-month culture period (Figure 1D, F). In some cases, highly proliferative prostate fibroblasts appeared to outcompete epithelial organoids in the culture and migrated to the bottom of the plate. In such cases, the rapidly expanding fibroblasts exhausted the nutrients leading to acidification of the culture medium. Therefore, to ensure the epithelial status of organoid cultures, epithelial cells were enriched by FACS-sorting based on the expression of an epithelial cell adhesion molecule (EpCAM, Figure 1G). The EpCAM-negative cells contained stromal cell populations of the prostate[22]. EpCAM-positive epithelial cells grown in Matrigel readily formed organoids and could be propagated in culture on long-term basis (Figure 1H). In contrast, prostate stromal cells did not form organoids but displayed an elongated fibroblastoid morphology and eventually ceased proliferation upon prolonged culture (Figure 1H).
PCa organoids recapitulates the morphological and histological features of their parental tissue
To correlate the organoid culture phenotypes with the original tumor tissue, we selected three organoid lines each representing a PCa sample classified into different grade and Gleason score category (O-12: G3a, GS3+4; O-11: G3a, GS4+4; O-9: G3b, GS3+4). The organoids were grown in Matrigel for approximately 21 days, fixed and stained for ERG, PTEN, cytokeratin 8 (CK8) and CK5 and imaged by immunofluorescence (Figure 2A). For AR-staining, organoids were processed for immunohistochemistry as the AR-antibody did not work well for immunofluorescence (Figure 2A). Immunohistochemistry was performed on the matching original tissues using the same marker proteins. The morphological features of the original patient tissue and its matching organoids showed similarity in biomarker expression and in their histological features. For example, PTEN loss is a common scenario in 40% of the PCa patients and was seen in one (PT-11) of the three patient tumors and replicated in O-11 organoid sample. Another common event is overexpression of ERG, often due to fusion with AR-responsive TMPRSS2 gene[23]. Normal prostate epithelium expresses negligible levels of ERG. ERG overexpression was observed in two of the three selected patient tumors and again was replicated in the respective organoid samples (O-9 and O-11). Incidentally, both PT-9 and PT-11 patients carried higher grade tumors with high Gleason score (Table S1). O-12, that derived from a tumor with a lower Gleason score expressed normal PTEN levels and low ERG levels in both in the original tumor lesions and in organoid cultures (Figure 2A). Disappearance of CK5-expressing basal cells is one of the reoccurring features in developing PCa lesions. In line with this, we observed depletion of CK5 positive cells in the PT-11a and corresponding O-11a representing high Gleason PCa (Figure 2A). CK5 positive cells were typically observed at the periphery of cell clusters, but in some cases, CK5 positive cells were also inside the organoids (Figure 2A). Reportedly, basal epithelial cell population is important for the growth and propagation of organoids[24]. Organoids derived from high GS (O-5, GS:4+5) and low GS (O-12, GS:3+4) showed distinct morphological features. Many low GS (O-12) organoids showed normal prostate gland-like polarized epithelial cells surrounding a lumen, whereas higher GS (O-5) organoids appeared to form more solid, tumor-like cysts (Figure 2B). High CK5 expression was observed at the outer surfaces of normal or prostatic intraepithelial neoplasia (PIN) organoids (O-12; Figure 2C). In contrast, O-5 organoids derived from a high Gleason score PCa sample PT-5, showed a more irregular and overall reduced CK5 staining.
To study the morphology and growth of PCa organoids in an in vivo setting, we performed subcutaneous transplantation of long term expanded benign hyperplastic (BH; GS3+4) O-12 and high Gleason (GS4+5) O-5 organoids in immunocompromised SCID mice. OX-12 organoid xenografts formed well-organized glandular structures with distinct layers of basal and luminal epithelium as indicated by CK5 and CK8 staining, respectively (Figure 2E). Curiously, while OX-5 organoid xenografts formed gland-like structures they displayed frequent unpolarized tumor outgrowths (Figure 2E). Overall, OX-5 organoid transplants exhibited faster growth than OX-12 transplants (728 mm3+/-386 mm3 vs. 474 mm3 +/-142 mm3 at 12 weeks, respectively) (Figure 2D). Hence, these data demonstrate that the long-term expansion of PCa organoids in vitro and in vivo preserves the growth and structural features of their tumor-of-origin. Collectively, this analysis revealed that PCa organoids express similar biological markers with their corresponding patient-derived tissue of origin, even after long-term expansion in vitro.
In vivo models are much relied in cancer drug discovery. The subcutaneous xenograft models based on established PCa cell lines are widely used in the development of anti-cancer drugs, but currently the success rate of this approach stands at ~5% to find drugs that undergo further clinical approval[25] PCa cell lines are typically adapted to in vitro culture conditions on 2D plastic surfaces over several decades and this might limit their translational relevance. Our data indicates that PDO models established from patient tumors have high resemblance to their tumor-of-origin and could thus serve as accurate models for drug development and precision medicine. To determine whether our organoid models would faithfully recapitulate the PCa phenotype, we placed selected PDOs (O- 5, -11, -12) surgically into the prostate of ICR-SCID mice. The mice were in constant observation until surgery recovery and twice weekly thereafter. Since PCa is a slow progressing cancer, the mice were observed for 90 days after which their prostates were examined in detail. This analysis revealed a remarkable resemblance between the orthotopic organoid xenografts (OOX) and their parental tumor tissue accurately reiterating the morphological traits (Figure 3A). When evaluating PCa biomarkers such as AR, PTEN, CK5 and CK8, we found that the OOX-tumors’ expression profiles resembled the corresponding original tumor tissues (Figure 3B). Importantly, O-11 and OX-11 both displayed PTEN loss seen in the original tumor, but only the OOX-11, transplanted to into mouse prostate recapitulated the histological traits of the parental tumor with cribriform and fused glands along with PTEN loss (Figure 2A, 3A-B). Similarly, OOX-12 transplants derived from low Gleason score adenocarcinoma, showed PTEN and AR expression and formed hyperproliferative glands resembling adenocarcinoma lesions in the matching original tumor. PTEN was not expressed in O-11 organoids or OOX-11 transplants, in agreement with the corresponding original tumor tissue in the respective patients (Table S2, Figures 2A and 3B). Overall, these results demonstrate that PCa organoids recapitulated the histological characteristics and differential marker expression of the original tumor tissues even after long-term culture of the organoids in vitro.
PCa organoids preserves molecular heterogeneity as in its original patient tumors.
Intratumor heterogeneity is a prominent feature of human cancers. Next, we wanted to analyze the genetic properties of the PCa organoids in more detail. The PCa-derived organoids with different Gleason scores were expanded long-term (~6 months) in culture, with a consistent passaging ratio of 1:3-1:4. The growth rate of the different organoid lines was found to vary with passage frequencies between 1-3 weeks (Figure 4A). In some cases, the reduced organoid growth rate was accompanied with an outgrowth of fibroblast-like cells (O-1, -16 and -32). To alleviate this issue and to focus on the PCa cancer cell population, few organoid lines (O-5, -9, -11, -12, -16, -24, -32 and -36) were FACS-sorted to enrich epithelial populations and subsequent genomic and other downstream analysis was performed with sorted epithelial populations of selected organoid lines (Figure 1G, H).
To determine whether the PCa organoid lines retain the original patient tumors’ mutational landscape, we performed whole exome sequencing (WES) analysis of six organoid lines (O-5a, O-5e, O-9, O-11a, O-11c and O-12) derived from four patients (PT-5, PT-9, PT-11 and PT-12). In addition to tumor organoids, matched organoid lines derived from phenotypically normal area of O-5 (5e) and O-11 (11c) were included in the analysis and cultured for more than 2 months. Organoids were sorted for CD49fhi and EpCAMlow population (basal epithelial cells enriched) and used as an internal control to compare with matching tumor lesions (O5a and O-11a). We analyzed missense variants, stop-gain variants, frameshift and inframe_indels mutations. The global variant profiles of organoid lines and their corresponding original tumor tissue were highly similar with ~95% of the mutant variants retained in almost all the samples (Figure 4B). Though we unfortunately could not get access to the patient’s germline mutational landscape, we filtered for variants present in the Catalogue of Somatic Mutations in Cancer (COSMIC) and found that the majority of all the cancer-related somatic variants present in the patient’s original tissue were retained in the corresponding PCa organoid lines (Figure 4C). We examined the mutational landscape for known driver gene mutations such as PTEN, AR, ERG, TMPRSS2, PIK3CA, SPOP and TP53 that are common in PCa and found several such mutations in our sample set (Figure 4C). Curiously, benign (PT-5e, PT-11c) and tumor (PT-5a, PT-11a) lesions and the corresponding organoids (O-5a,e and O-11a,c) all carried the same tolerated homogeneous or heterogeneous mutations in the PI3KR1 and FOXA1 genes. These types of mutations are generally considered to be associated with low risk whereas deleterious heterogeneous mutations, such as TMPRSS2, is associated with high risk of developing PCa. The observed TMPRSS2 mutations in patients were similarly found in both benign and tumor lesions as well as corresponding organoids (Figure 4C). The PT-9/O-9 pair exhibited deleterious mutation in AR and BRCA1 genes as well as a tolerated heterogeneous mutation in PIK3CA gene. Given that the mutational analysis was consistent in the sorted basal epithelial population suggest that basal stem cell-like population represents the original patient tumor tissue in terms of genetic information. However, we did observe a loss of some key mutations in organoid cultures that were present in the primary tissue. Deleterious heterogeneous mutations of SPOP detected in both PT-11a and PT-11c and PIK3CA detected in PT-11c could not be found in respective organoid cultures suggesting that PCa cells carrying these mutations were eliminated during the long-term in vitro culture.
Next, we studied how well the overall transcriptomic profiles of the primary tumor/organoid pairs were preserved. To this end, we performed genome-wide transcriptomic RNA-sequencing analysis in three selected pairs, PT/O-9, PT/O-11 and PT/O-12). Principal component analysis (PCA) indicated that the transcriptomic profiles of the organoid cultures were highly similar to their corresponding tumor samples (Figure 4D). Collectively, our genomic and transcriptomic profiling of the PCa organoids authenticate the preservation of the tumor heterogeneity including potential PCa driver mutations.
Prostate organoids show distinct drug sensitivity profiles.
Given the preservation of histological, biochemical, and genetic properties of original tumors in the in vitro organoid cultures we performed a proof-of-concept drug-sensitivity testing in four of the organoid lines (O-5, O-9, O-11 and O-12). This analysis was done to evaluate the capacity of organoid model as a platform to assess potential drug sensitivity and resistance in specific patient tumors. Moreover, when combined with functional drug sensitivity testing, the genomic characterization might give further insight into the potential mechanisms underlying drug resistance. As a primary screening step, the therapeutic response was evaluated by determining the total number and average size of organoids with different doses of enzalutamide or docetaxel, two drugs that are in regular clinical use for PCa treatment. Enzalutamide is a non-steroidal anti-androgenic drug that blocks testosterone binding to androgen receptor, and it is primarily used as androgen deprivation therapy (ADT) in the PCa patients. Docetaxel is a microtubule stabilizing cytotoxic drug typically used to treat metastatic PCa in patients with ADT-resistant tumors.
First, the organoids were grown for 3 weeks without drugs with bi-weekly medium changes followed by a 7-day treatment with a dilution series of either enzalutamide or docetaxel after which organoid number and sizes were determined. Drug dilutions were determined as reported in NCI-DTP portal[26] and by their half-maximal growth inhibitory concentration (GC50) and sensitivity was represented by measuring the area and number of organoids. Except for the O-12 organoid line, the other organoid lines showed significant resistance to enzalutamide as organoid growth was observed even at the highest concentration of enzalutamide (Figure 5A, C and E). O-9 organoids expressed high levels of ERG (Figure 2A) and were shown to contain an AR-mutation that might contribute to enzalutamide resistance (Figure 4C). O-11 organoids were PTEN-negative and expressed high levels of AR both of which could contribute to ADT-resistance[27] (Figure 3B). O-11, as well as O-5 organoids were found to express high levels of ERG that has also been associated with ADT-resistance[28] (Figure 2A). The exome-sequencing analysis revealed several putative candidate mutations in O-5 or O-11 organoid lines that could contribute to their enzalutamide-resistance. Docetaxel-treatment was effective in enzalutamide-resistant organoid samples, except for O-11 that showed some resistance at intermediate concentrations (Figure 5B, D and F). Docetaxel-resistance mechanisms are not well understood but upregulation of multidrug transporters has been suggested as the primary mechanism[29]. Of note, mutations in the DNA-repair machinery, associated with genomic instability in PCa[30], were observed in O-9 organoids (Figure 4C). Taken together, these data demonstrate the potential of the in vitro PCa organoid model as a drug screening platform that can be utilized to study patient specific drug responses.
PTEN depletion induces the growth in PCa organoids
A key property of any model system for cancer cell biological studies is their amenability for robust genetic manipulation. Loss of PTEN is one of the key genomic alterations in PCa that contributes to PCa growth and aggressiveness[9]. We selected three PTEN-positive organoid lines (O-16, O-24 and O-36) and generated isogenic PTEN-deleted variants by using previously described lentiviral constructs expressing Cas9 and PTEN-targeting gRNA[31]. Control population was transduced with the same Cas9-expressing lentiviral vector lacking a gRNA sequence. Positively transduced cells were selected using puromycin selection and 10 days post selection puromycin-resistant organoids were harvested and seeded into Matrigel blobs and grown without puromycin. Loss of PTEN expression was confirmed by immunofluorescence staining of fixed organoids (Figure 6D). To investigate whether loss of PTEN had any effect on organoid growth the different organoid variants were grown for 3 weeks followed by counting the number and overall area of the organoids in culture. While the number of organoids formed by PTEN-KO variants was not significantly increased, PTEN-KO organoids were much bigger than their respective controls (Figure 6B-C). Importantly, while most control vector transduced O-36 organoids had hollow morphology and showed distinct basal and luminal cell populations, O-36 PTEN-KO variants grew as large solid clusters with mixed basal and luminal identity based on CK5/CK8-expression (Figure 6D). Most of the control O-16 and O-24 organoids were already solid clusters but their size increased significantly upon PTEN depletion (Figure 6A).
Integrin α6 regulates PCa organoid growth.
Epithelial cells reside on a laminin-rich basement membrane (BM) that is rich of signals regulating epithelial differentiation and polarity[32]. The integrin family of ECM receptors are the primary cell surface receptors that connect prostate epithelial cells to the BM. In normal prostate epithelium, integrin α6 (CD49f) pairs with β4 to form hemidesmosomes that anchor cells to the BM by binding to laminin. Multiple studies have reported that CD49fhigh population in the prostate, as well as many other epithelial organs such as the breast, contains cells with robust organoid formation and tumor initiation capacity[33–35]. High α6-integrin expression is thus considered to mark prostate stem cells and possibly cancer stem cell pool[36]. Counterintuitively, α6β4-integrins are downregulated upon PCa progression[31] However, loss of hemidesmosome organization is due to loss of β4-subunit expression in advanced PCa, while residual α6-subunit expression persists[31, 37]. α6-subunit can also pair with β1 to form integrin α6β1 that has been implicated in the regulation and maintenance of PCa stem cells and attributes to cell survival, resistance to chemotherapy and metastasis[36]. To investigate the role of α6-integrin in PCa organoid cultures, we dissociated organoids into single cells and sorted them by FACS into four different populations using Epithelial Cell Adhesion Molecule (EpCAM, epithelial cell specific marker) and integrin α6 (CD49f) (Figure 7A). CD49flow/EpCAMhigh cells represent luminal epithelial population (LP) and double-positive CD49fhigh/EpCAMhigh cells consist of basal epithelial cells (BP)[21]. EpCAMlow/CD49flow cells presumably represent stromal cell populations (SP) whereas the identity of EpCAMlow/CD49fhigh population is unclear. It is possible that EpCAMlow/CD49fhigh represent a population enriched for cancer stem cell-like cells (PCSC) in the prostate as described for breast cancer organoids[38]. LP formed tiny and partially hollow organoids that could not be efficiently propagated long-term (Figure 7B). In contrast, both BP and PCSC populations efficiently formed organoids that showed mostly the solid phenotype, and which could be cultured on long-term basis (Figure 7B). BP organoid cultures yielded also luminal epithelial cells presumably via basal-to-luminal differentiation, as described previously[21, 39].
In addition to integrin α6 (CD49f), integrin α2 (CD49b) is also expressed in prostate epithelial cells, and high integrin α2 levels have been suggested to depict PCa stem cells[36, 40, 41]. We analyzed the expression of α2- and α6-integrins in selected PCa organoids. Immunofluorescence experiments showed that α6-integrin expression is confined to basal surfaces in both hollow and solid organoids (Figure 7C). In benign O-24 organoids, these basally localized cells also expressed high levels of CK5, a basal epithelial cell marker (Figure 7D). In solid organoids, CK5 was not as strictly restricted to basal side as α6-integrins. α2-integrin expression was also seen in basal cells in benign organoids, but it was not clearly restricted to basal surfaces in solid O-24 organoids and was only gradually decreasing towards the center of cell clusters (Figure 7C).
To test whether high levels of α6-integrins, α2-integrins or both depict prostate stem cells that possess organoid formation capability, we sorted O-24 PCa organoids based on CD49f (α6-integrin) and CD49b (α2-integrin) expression (Figure 7E). The double negative α6low/α2low population seeded onto Matrigel blobs hardly grew at all and remained as individual cells and eventually lost viability (Figure 7F). α6low/α2high cells exhibited elongated fibroblastoid morphology (Figure 7F). Regardless of the α2-integrin expression levels, cell populations expressing high levels of α6-integrin efficiently formed organoids (Figure 7F). We observed that α6hi/α2hi population formed mostly solid organoids while α6high/α2low population formed both solid and hollow organoids (Figure 7F).
To get insight into the different cell populations within prostate organoids, we performed single cell RNA sequencing (scRNA-seq) on O-36 (GS: 3+4) organoid sample. Four samples from these parent organoids were prepared: The second passage of the original patient-derived O-36 organoid (sample-1), FACS-sorted EpCAM-positive epithelial cell population derived from O-36 (sample-2), EpCAM-negative stromal population derived from the O-36 organoid (sample-3) and reaggregated epithelial and stromal O-36 populations in a co-culture setup (sample-4). After two weeks in 3D Matrigel blob culture, single cell suspensions were prepared for scRNA-seq and approximately 10,000 cells per sample were loaded into a 10X Genomics single-cell platform and analyzed with Cell Ranger software. Altogether, 40595 cells were included into the analysis that annotated 18 different clusters (Figure 8A). Interestingly, it was observed that the epithelial clusters in single and co-culture setups were relatively similar whereas the stromal populations differed significantly when cultured alone or in the co-culture settings (Figure 8B, D). Moreover, similarity in transcriptomic profiles between the original organoids and the reconstituted coculture setup was noted (Figure 8D). Cell type identification in the different clusters was determined by examining differentially expressed genes (DEGs) indicating stromal (PECAM, FN1, ACTA2) epithelial (CDH1, EPCAM, CD24) and basal (TP63, KRT5, KRT14) epithelial origin (Figure 8B, C).
As discussed above, both high α6- and α2-integrin expressing cells have been reported to carry self-renewal potency. Such stem cell-like cell populations are considered particularly critical for PCa pathogenesis and thus they represent a key target for PCa therapies [36, 42]. To assess if the α6high and α2high cells represent distinct cell populations we focused on defining ITGA2- and ITGA6-positive populations in the single cell analysis dataset. ITGA6 was highly expressed in a single epithelial cluster 9 while high ITGA2 expression depicted two clusters (Figure 8C). Comparative analysis of double negative (DN), single positive ITGA6+ and ITGA2+ and double positive (DP) ITGA6+/ITGA2+ cells revealed similarity between ITGA6+ and DP populations (Figure 8E). Curiously, the DN population expressed high levels of prostate stem cell antigen (PSCA) associated with prostate cancer progression[43, 44]. ITGA2+ cells expressed mesenchymal genes such as FN1 and VIM but also some epithelial genes although they were clearly distinct from the ITGA6+ and DP populations (Figure 8E).
Integrin α6 is required for PCa organoid growth whereas integrin α2 is dispensable.
To confirm the functional role of α6- and α2-integrins in the self-renewal capacity of PCa organoid we again implemented lentivirus-mediated CRISPR/Cas9-KO technology (Figure 9A). The knockout experiments were conducted using two different organoid lines (O-16 and O-36). The control virus expressed no specific gRNA whereas α6KO construct expressed a gRNA construct targeting the third exon-3 of ITGA6 and α2KO construct a gRNA targeting exon 5 in the ITGA2 gene[31, 45]. Transduction of the organoids was performed as described above for PTEN-KO and immunofluorescence staining confirmed efficient depletion of α6- and α2-integrins (Figure 9A). It was observed that while the number of α6KO organoids was only modestly reduced, the organoids had a dramatic growth defect when observed after 3 weeks in culture (Figure 9B, C). We could not establish long-term cultures of α6KO organoids as they eventually ceased to grow and became extinct. Any organoid colonies recovered from these cultures were found to express α6-integrins (data not shown). This α6-integrin expression might be because of in-frame editing preserving the open reading frame of ITGA6 mRNA or recovery of α6-expression due to incomplete editing in some cells. In contrast, depletion of α2-integrin expression had no significant effect on the number, growth or survival of PCa organoids. α2KO organoids formed at similar numbers and grew at rates comparable to controls (Figure 9A-C). Finally, the proliferation rates of the different organoid variants were also determined using an independent method based on Ki67 staining. In agreement with the growth analysis, Ki67 staining assay showed that α6KO organoids contained very few proliferative cells (<10%), whereas more than 30% of the cells in α2KO and control organoids showed Ki67 positivity (Figure 9D). Such levels have been considered clinically relevant depicting fast-growing prostate tumors[46]. Collectively, these results indicate that α6-integrin expression is critical for organoid formation and growth and therefore α6-, and not α2-integrins, play a key role in prostate organoid self-renewal and potentially also in the maintenance of PCa stem cell pool.