Abstract
Intractable human diseases such as cancers, are context dependent, unique to both the individual patient and to the specific tumor microenvironment. However, conventional cancer treatments are often nonspecific, targeting global similarities rather than unique drivers. This limits treatment efficacy across heterogeneous patient populations and even at different tumor locations within the same patient. Ultimately, this poor efficacy can lead to poor clinical outcomes and the development of treatment-resistant relapse. To prevent this and improve outcomes, it is necessary to be selective when choosing a patient’s optimal adjuvant treatment. In this review, we posit the use of personalized, tumor-specific disease models (TSM) as tools to achieve this remarkable feat. First, using ovarian cancer as a model disease, we outline the heterogeneity and complexity of both the cellular and extracellular components in the tumor microenvironment. Then we examine the advantages and disadvantages of contemporary cancer models and the rationale for personalized TSM. We discuss how to generate TSM through careful and detailed analysis of patient biopsies with contemporary analysis techniques and utilizing the resultant data to construct precision 3D models in vitro. Finally, we provide clinically relevant applications of these versatile personalized cancer models to highlight their potential impact. These models have utility towards a myriad of fundamental cancer biology and translational studies. Importantly, these approaches can be extended to other carcinomas, facilitating the discovery of new therapeutics that more effectively target the unique aspects of each individual patient’s TME.
Keywords: biomaterial, tumor microenvironment, mechanics, mechanobiology, extracellular matrix, cancers, ovarian cancers, personalized, residual disease, chemoresistance, predict relapse, cancer stem-like cells, immune cells
1. INTRODUCTION
Precision medicine, wherein a treatment is tailored to a patient’s specific tumor characteristics, has become widely popular in the last 10 years [1–3]. In precision medicine, therapies are specifically optimized based on molecular profile, cancer type, stage, and biomarkers for each patient. This process currently involves obtaining the genetic profile of the patient, which is then used to anticipate patient specific drug metabolism, response, and toxicities. Given the high molecular heterogeneity in many cancer types, the concept of personalized or precision medicine is intuitive. This is even more so, when other factors that contribute to clinical outcomes, such as ethnicity, gender, age, socioeconomic status, geographic location, and disability are considered. Precision treatments are particularly effective in increasing the treatment efficacy of cancers with targetable mutations in a subset of patients [4–6]. However, many cancers lack targetable genetic mutations, thereby making precision treatment difficult [7]. In addition, the efficacy of cancer cell-targeting therapies are often compromised by the non-cancerous components of the tumor microenvironment (TME) [8–10]. This includes tumor supporting cells and tumor secreted extracellular matrices (ECM) which form complex multifaceted interactions with cancer cells that act to modulate chemoresistance [11–14]. Unfortunately, high level cell-cell and cell-ECM interactions are often omitted in contemporaneous model systems leading to underrepresented cellular, molecular, and ECM heterogeneity and substantially impacting their clinical translation potential [15–17].
As a result of this oversight, we propose the thorough analysis of the genetic, molecular, cellular and ECM composition of patient biopsies, performed concomitantly with pathologic diagnosis, to guide construction of in vitro personalized tumor-specific models (TSM). Generating 3D microscale TSM based on this comprehensive analysis of each patient’s tumor would be followed by high throughput drug screening assays using previously developed precision models. This process would have numerous applications, ranging from personalized drug screening to identification of novel biomarkers. If successfully implemented, these applications have the potential to change the landscape of precision medicine and decrease mortality rates in many cancers. Here we present this approach through the lens of high grade serous ovarian cancer (HGSOC), which is highly heterogeneous and notorious for development of chemoresistance, relapse, and poor outcomes. Despite the focus on HGSOC in this review, we postulate that these same methods can be extended to different epithelial cancer types with their own unique genomic, molecular, and microenvironmental characteristics [18–20] (Fig. 1). Using HGSOC as a model disease allows us to highlight patient-specific differences in the TME, which may not be ubiquitous in all other epithelial tumors, but serves as a starting point from which pragmatic parallels can be drawn to other tumors.
Figure 1: Distinct and Heterogenous Microenvironments are Present in Cancers.

Human diseases, including cancers, are complex heterogeneous ecosystems. Although cancers can be categorized based on histological subtype, there are patient-specific individual variations in the tumor microenvironments (TME) constituents, which are demonstrated as varying cellular and ECM features in the schematic above. Biomaterials-based technologies enable the personalized models of patient-specific tumors, based on the analysis of the distinct TME components identified and characterized, as shown in the schematic. In this review, we describe new paradigms of personalized therapies to improve cure rates, that can be discovered and validated with the patient-specific biomaterials-based tumor models.
In the first section of this review, we start by introducing ovarian cancer and its various sources of heterogeneity, including genetic, molecular, cellular, and ECM differences between and within patients. We then briefly outline how these features interact to confer heterogenous responses to treatment. Subsequently, we review current models that recapitulate each aspect of heterogeneity before describing our proposed TSM model system and potential key applications.
2. Defining Heterogeneity in the Ovarian Cancer Microenvironment
2.1. Characteristics of High Grade Serous Ovarian Cancer
Ovarian cancer is the most lethal of all gynecological malignancies and 5th leading cause of cancer deaths among women worldwide [21,22]. As the most common type of ovarian cancer, HGSOC accounts for over 60% of ovarian cancer deaths. Patients with HGSOC are often diagnosed at advanced stages when tumors have metastasized to peritoneal organs. They receive standard therapy that combines cytoreductive surgery and platinum-taxane chemotherapy. More than half of the patients become increasingly resistant to the conventional chemotherapies within 6 months of treatment, which eventually leads to tumor relapse and death [23,24].
Characterizing these tumors reveals genomic profiles with frequent copy number variations (CNV) but also a notable deficit in mutation burden. There are only a handful of genetic mutations common between patients and they are not uniformly expressed across all HGSOC which complicates the use of targeted therapies [25–27]. Given that the majority of women with ovarian cancer have recurrent tumors and no common somatic mutations to stratify them, accelerated screening of novel or synergistic therapies are critically needed. On the other hand, large-scale HGSOC datasets have been used to stratify ovarian cancer patients into molecular subtypes based on gene expression [27,28]. This stratification has prognostic and therapeutic relevance [29], however, no clinical use has been validated [30].
Another major contributor to ovarian cancer malignancy is the unique metastatic pattern that differs from those of most other epithelial malignant diseases. Most frequently HGSOC disseminates via the transcoelomic route, with about 70% of patients developing peritoneal metastases [31,32]. Specifically, metastatic multicellular tumor spheroids disseminate with the accumulated ascites (tumor fluid) in the peritoneal cavity and spread to the surrounding abdominal organs (colon, intestine, liver, pancreas, lungs) [32,33]. They are also transported by peritoneal fluid flow to adhere to the mesothelium that covers the peritoneal surfaces where they initiate metastatic colonies [33,34]. This unique spreading pathway creates a microenvironment that enriches cancer stem-like cell (CSC) populations and treatment-resistant cancer cells [12]. Given the different characteristics of the microenvironments along various stages of metastasis, the development of in vitro 3D TSM that resemble each HGSOC-specific TME (Fig. 1) is critical for identifying therapeutics that can effectively target drug-resistant cancer cell populations. In order to recreate these microenvironments in TSM, it is necessary to better understand both the cellular and acellular components, and the role that each plays in disease progression and chemoresistance. These components and their complex interactions are described below.
2.2. Cellular heterogeneity in the TME
Cellular heterogeneity can be defined on two levels. It can refer to cell level metabolic, genetic, epigenetic, and phenotypic differences between cancer cells, or it can refer to the higher level, heterogeneous composition of all cell types in the tumor microenvironment (i.e. a tumor with high immune infiltration with high stromal content) [28,35–38]. Both definitions of cellular heterogeneity can influence disease progression and chemoresistance stressing the need to understand both levels of heterogeneity to create personalized models for precision medicine.
Cellular heterogeneity within the cancer cell population in the TME:
HGSOC is highly heterogeneous in terms of its histopathological architecture, genetic alterations, and cellular composition, which results in heterogeneous responses to chemotherapies. The histopathological architectures of HGSOC are solid masses of cells with papillary, glandular, or cribriform features, which are often accompanied by areas of extensive necrosis [39]. Some HGSOC simulate the appearance of endometrioid or transitional cell carcinoma. The morphology of these tumors is distinct, but they show similar immunoreactivity to typical HGSOC and are diagnosed accordingly [40]. HGSOC tumors share genomic features, including frequent TP53 mutations and homologous recombination deficiency. Mutations in NF1, BRCA1, BRCA2, RB1, and CDK12 are also frequent [27,41]. Most HGSOC exhibit large genetic gains and losses suggesting genomic instability. Common copy number alterations are gains in MYC and KRAS genes and losses in PTEN, RB1, and NF1 genes. Primary HGSOC can have highly heterogeneous genomic alterations, which are associated with high recurrent risk and poor prognosis [42]. Acquisition of genetic mutations over the course of disease progression is an example of the clonal evolution model of tumor heterogeneity wherein somatic mutations result in a heterogeneous mixture of different phenotypes that may respond differently to treatment [43].
In addition to genetic heterogeneity, cancer cells are also subject to epigenetic variation which influences cell phenotypes influence treatment responses [18,35,44]. Epigenetic changes are heritable modifications to gene expression that do not change the DNA, but rather alter accessibility of genes through various mechanisms including DNA methylation, histone modifications, and microRNA. These epigenetic changes occur more often than genetic mutations and happen early on in tumor progression, highlighting their role in developing heterogeneous mixtures of tumor cells and the potential impact of drugs targeting epigenetic dysregulation [44]. In ovarian cancer, an important example of epigenetic alterations and heterogeneity is the hypermethylation of BRCA1. This results in decreased BRCA1 expression which in turn promotes genomic instability in the ovarian cancer cells and leads to heterogeneous phenotypes [45]. While a detailed discussion of epigenetic modifications is beyond the scope of this review, readers are referred to more comprehensive reviews dedicated entirely to the topic [18,44,45]. Importantly, epigenetic alterations are also reported to be involved in the reprogramming of cancer cells into a rare, but clinically relevant population of cancer cells called cancer stem-like cells (CSC)[46].
CSC are a clinically relevant subpopulation that is responsible for tumor initiation, metastatic disease, recurrence, and resistance to chemo/radio-therapy [47–51]. Like normal stem cells, CSC maintain a level of pluripotency, which enables self-renewal and differentiation [49,52,53]. In addition, CSC are highly plastic cells with a heterogeneous population defined by different epigenetic and metabolic states which they pass on to their progeny. CSC progeny comprise most of the tumor mass and tend to be more responsive to chemotherapy than CSC [53]. This ultimately leads to the development of a heterogeneous tumor cell population generated from distinct CSC from the heterogeneous CSC population [35,50,51,54,55].
Self-renewing tumorigenic CSC have been identified and isolated in many cancers, including, leukemia [56], breast [57], ovarian [58], colon [59,60], prostate [61,62], brain [63–65], pancreatic [66], melanoma [67], myeloma [68], and lung [69]. Importantly, while some CSC markers are shared between different cancer types, some cancers also have unique CSC markers [70]. Even among common CSC markers, recent work by Dzobo et al. found no consistent pattern of CSC marker expression in different cancers when evaluating the expression of CSC markers including ALDH1A1, CD44, CD24, EPCAM, ICAM1, CD90, CXCR4, NES, CD133, ABCB1, and ABCG2 in colon, pancreatic, lung, and esophageal cancers [71]. This cross-cancer heterogeneity in CSC populations may indicate that CSC targeting therapies may not share efficacy across cancer types. In ovarian cancer, CSC are identified by an amalgam of biomarkers including, CD133, ALDH, CD44, CD24, and CD117, and their presence is an indicator of reduced progression-free survival and poor patient outcomes [72–84]. Ovarian CSC phenotypes are dictated in part by interactions with the other cells in the TME, which will be discussed in Section 2.3.
The resistance of CSC to treatment is attributed to overexpression of ABC transporters, enhanced ALDH activity, response to DNA damage, epithelial to mesenchymal transition (EMT), and dormancy [85,86]. Due to these resistance mechanisms, CSC are capable of surviving primary therapies and repopulating the tumor with a heterogeneous population of cancer cells [87]. In ovarian CSC, it has also been suggested that heterogeneity within the CSC population may increase the odds of the development of spontaneous escape variants [88]. Anoikis resistance is also a key feature of ovarian cancer CSC, which allows them to survive in non-adherent conditions like the malignant ascites fluid that accumulates in the peritoneal cavity in many ovarian cancer patients [51,87].
Cellular heterogeneity in the non-cancer cell composition in the TME:
Tumors are not composed merely of cancer cells that cause their uncontrolled growth. Rather, they are a heterogeneous mixture of host cells and tumor cells that interact dynamically to drive tumor progression [89,90]. Specifically, the proportion of epithelial, endothelial, lymphocyte, myeloid, and stromal cell components varies significantly between tumors [91–93]. The high degree of cellular heterogeneity both within and between patients’ tumors can affect drug response and prognosis, causing major challenges in the clinical management of many cancers [38,90]. In this section we will review the heterogeneity of cell compositions in various types of HGSOC microenvironments and discuss how these cells contribute to heterogeneity in the ECM in Section 2.3 (Fig. 2).
Figure 2: Distinct and heterogenous cellular and ECM composition and organization are present in the primary and metastatic ovarian cancers.

(Left) Production of ECM in the primary ovarian tumor gradually dysregulates over time, depositing new proteins normally not found in the ovarian germinal epithelium and tunica albuginea. The basement membrane is degraded in the primary tumor aiding in dissemination. The primary tumor is infiltrated by tumor associated macrophages, T-cells, carcinoma associated fibroblasts and endothelial cells. (Middle) The malignant ascites contains suspended single cells and cellular aggregates comprising of cancer cells, cancer stem-like cells (CSC), fibroblasts, mesothelial cells, macrophages and T-cells. ECM such as fibronectin, hyaluronan, and collagen type I are found within cellular aggregates, and in and around the fluidic ascites. (Right) Secondary omental metastases consist of colonizing cellular spheroids from the ascites. In these sites, cancers cells begin to produce new basement membrane, collagens, and hyaluronan. In all three TME, the cell and ECM subtypes are indicated with the representative schematic (not drawn to scale).
HGSOC have several unique microenvironments including the primary tumor, the malignant ascites, and secondary metastatic sites. Each of these microenvironments is accompanied by different cellular and acellular characteristics that affect tumor progression [94]. While there is overlap between the cells that are present in each microenvironment, their relative proportions vary and this can influence the signals received by the tumor cells [94]. In primary HGSOC tumors, epithelial cells have been shown to make up as much as ~68% of the population, while in a metastatic tumors, epithelial cells may only make up as few as 10% [94]. However, the immune cell composition is known to be highly variable and the stroma makes up anywhere from 10% to 60% of the tumor, indicating that a fair degree of heterogeneity exists even within the primary tumor [91–93].
CSC are enriched through dynamic interactions with cells in the surrounding TME, such as carcinoma-associated mesenchymal stem cells (CA-MSC), carcinoma-associated fibroblasts (CAF), endothelial cells (EC), and macrophages [89,90]. These interactions are exemplified by CA-MSC, which can be recruited to the primary tumor from distant locations in the body through soluble signaling or tissue resident mesenchymal stem cells (MSC) in the ovary or from the omentum. These MSC can then be converted into CA-MSC by the cancer cells [95–97]. Once converted, they have been shown to increase the number of CSC, enhance chemo-protection for CSC, and lead to tumor growth, either through paracrine signaling or indirectly by differentiation into CAF [97–105]. Importantly, CA-MSC are distinct from CAF and normal MSC, and can differentiate into several other critical components of the tumor stroma including fibroblasts, osteocytes, and adipocytes [95,105], further contributing to cellular heterogeneity. Additionally, EC in the primary tumor and secondary tumor locations can also support CSC and tumor cells [106,107]. Not only have EC been shown to induce self-renewal pathways in CSC [108], but they also protect them from cisplatin and paclitaxel treatment [109–111]. Similar to CSC recruitment of MSC to the tumor, CSC can recruit EC to the tumor through secretion of angiogenesis promoting signals, like VEGF, SDF-1. This occurs when hypoxia develops as a result of the tumor outgrowing its vasculature. Hypoxic microenvironments activate Oct-4, Sox2, Notch, VEGF, and c-MYC expression which stabilizes HIF-1α and promotes survival capacity of the CSC [70,89,112,113]. EC additionally contribute to disease progression through their role in the formation of the unique malignant ascites TME [32,114].
The malignant ascites is the buildup of fluid in the peritoneal cavity that accompanies various pathologies, including HGSOC [32]. While the exact cellular context of the ascites changes with disease progression [115], the predominant cell populations in the ascitic TME include cancer cells [116,117], CAF [116,117], leukocytes [116,117], and mesothelial cells [118]. Within the cancer cell population, ascites harbor a high proportion of treatment-resistant CSC [11,117,119]. Cells within the ascites often aggregate, to form tumor spheroids that are highly malignant, metastatic, and resistant to chemotherapy [116,120,121]. The non-tumor cells in these spheroids influence cancer cell phenotype and promote malignant characteristics. Among them, CAF are fibroblasts that are reprogrammed into a pro-tumoral phenotype. They promote EMT, tumor cell attachment to the mesothelium, and subsequent displacement of mesothelial cells [91,116,120]. This ultimately allows tumor spheroids to initiate tumor growth at secondary locations. CAF also have immunomodulatory functions, can promote angiogenesis, inhibit cancer cell apoptosis, and produce and remodel ECM [91,116,122]. CAF also have the propensity to promote stemness, chemoresistance, and tumor growth [91,97,122].
Tumor associated macrophages (TAM), another predominant cell type found in the malignant ascites as well as the primary tumor, are similar in their ability to drive components of disease progression such as angiogenesis, angiostasis, metastasis, and the function of other immune cells in the TME [123–125]. Like CAF, ovarian cancer cells also reprogram macrophages that are recruited to the tumor, into a pro-tumoral phenotype termed alternately activated M2-like macrophages [125]. These macrophages can contribute to immunosuppression by attracting regulatory T cells, inhibiting the anti-tumor response of natural killer (NK) cells and cytotoxic T cells, and inhibiting maturation of dendritic cells (DC). They can also promote drug resistance, decrease apoptosis, and promote stemness in ovarian cancer cells [125,126]. Despite the widespread influence that M2-like macrophages have on tumor progression, their prognostic effect depends on the proportion of anti-tumoral M1 macrophages, which are also present in the TME. The M1:M2 macrophage ratio in patients can vary widely, resulting in different immune responses and altered prognosis. Expectedly, higher M1:M2 ratios are associated with better outcomes for the patient [125,127].
In addition to macrophages, a host of other immune cells can be found in the ovarian cancer TME including NK cells, regulatory T cells, DC, CD8+ and CD4+ T cells, B cells, and myeloid derived suppressor cells (MDSC). Each of these cell types play a role in the complex immune response present in a tumor, with some cells like MDSC, regulatory T cells, and DC, serving tumor suppressive functions and others such as CD8+ and CD4+ T cells having antitumor effects [93,128,129].
Within the immune cell category there is remarkable heterogeneity, even at the same tumor site within the same patient [38,130–132]. The immune cell context of a tumor can change throughout tumor progression and can depend on factors like tumor location and treatment status [38,130,132]. For example, a recent study in HGSOC examined immunogenomic changes before and after neoadjuvant treatment in paired samples. Astoundingly, the authors observed that in treatment-naïve patients some areas of the same tumor could completely exclude immune cells, while different areas in the same tumor could be infiltrated [38]. Furthermore, they found that variation in immune cell genes, particularly those associated with T cells and NK cells, was responsible for the highest degree of variation between different treatment-naïve patient samples. This suggests the importance of immune heterogeneity in interpatient differences. Thorough characterization of these samples revealed that immune exclusion was associated with amplification of MYC targets and WNT signaling, which has previously been related to immune evasion [38,133]. The authors also noted increased NK cell infiltration and oligoclonal T cell expansion following treatment neoadjuvant therapy, demonstrating the plasticity of the heterogeneous immune microenvironment when perturbed [38].
Another study of immune heterogeneity in HGSOC compared the immune cell infiltration in the primary tumor and its surrounding stroma to the infiltration of corresponding omental or peritoneal metastases. Interestingly, they found variances in the number of CD45+, CD3+, CD8+ and PD-1+ cells between all primary samples and matched metastatic samples. Furthermore, immune cell infiltration in the stroma of the omentum was observed to be significantly greater than in the primary tumor stroma [130]. Post treatment analysis of the same patients/samples, revealed that an increased intratumoral CD3+ infiltration in the primary tumor was associated with platinum sensitivity indicating that high pre-therapeutic CD3+ infiltration could be an indicator that platinum therapy will be effective. Similarly, higher intratumoral CD8+ infiltration in peritoneal metastases compared to the primary tumor was also associated with platinum sensitivity [130]. These results are concordant with previous work indicating the CD3+ and CD8+ T cells are prognostic indicators for ovarian cancer [131,134,135]. Contrarily, high PD-1+ expression in peritoneal metastases is linked to poor response to platinum therapy [130]. This finding is also logical as PD-1 activation is associated with decreased anti-tumor immunity [136]. Overall, this study highlights the potential utility of immune context characterization in predicting treatment responses and disease progression. The immune cell composition in HGSOC is indisputably heterogeneous and can influence treatment response and thus needs to be taken into account in development of personalized models. That said, the immune system’s role in cancer is abundantly complex and warrants more comprehensive discussion that is beyond the scope of this review, but is actively being studied, and has been reviewed previously [128,137].
Aside from the prominent cells found directly in the malignant ascites, mesothelial cells and adipocytes can both interact with the tumor at secondary tumor sites in the peritoneal cavity and from afar through soluble signaling in the ascites to exert additional influence on disease progression [120]. Mesothelial cells, for example, can secrete factors like lysophosphatidic acid (LPA) to promote tumor cell adhesion, migration, and invasion [138,139]. They can also produce factors that inhibit drug induced apoptosis in cancer cells, following stimulation by the ascites [139,140]. Additionally, mesothelial cells are capable of transitioning into CAF via TGF-β signaling [141] to advance disease progression. Recently, mesothelial cells have even been found to induce platinum resistance in peritoneal metastasis through cell-to-cell interactions [142].
Adipocytes are prominent in the omentum, which is one of the most common sites of ovarian cancer metastasis, as well as subcutaneous tissues and the mesenteric membrane. From these locations, adipocytes can interact with tumor cells through secretion of adipokines and lipokines. Specifically, adipocytes in the omentum can attract cancer cells through IL-8 secretion and promote their proliferation through transfer of fatty acids. Importantly, adipocytes have recently been shown to confer chemoresistance to cancer cells through activation of the Akt pathway in cancer cells and secretion of a chemo-protective lipid, arachidonic acid [143]. Contrarily, adipocytes may contribute to suppression of ovarian cancer through cross-talk with ovarian cancer cells via secreted protein acidic and rich in cysteine (SPARC) [144]. These studies underscore the fact that the specific phenotype of the adipocytes and cancer cells may determine the result of adipocyte-cancer cell interactions.
As in the primary and ascites TME, ovarian cancer cells that have metastasized to secondary locations are subjected to even more diverse cellular microenvironments that can further alter the phenotype of cancer cells [145]. For example, in the omentum, adipocytes and mesothelial cells are in closer proximity to cancer cells, which is different from the TME in the ascites, where cancer cells are surrounded by mostly CAF and leukocytes. In the omentum, adipocytes are abundant with localized milky spots filled with immune cells, CAF, adipose-derived MSC, and vascular cells [146–149].
Effect of Hypoxia on Cellular Heterogeneity (cancer and non-cancer) within the TME:
When discussing cellular heterogeneity, it is also important to note the role of hypoxia in producing heterogeneous cell populations. As tumors develop, they outgrow their vasculature which leads to development of hypoxic pockets of cells [150]. These pockets of cells develop altered metabolism in order to survive the harsh conditions, which influences their proliferation, migration, and invasion [51,151]. HIF-1α plays a central role in cellular response to hypoxia and its expression is associated with poor outcomes in ovarian cancer [152]. In ovarian cancer hypoxia has been implicated in CSC enrichment through HIF-1α mediated activation of NF-κb signaling and consequent upregulation of SIRT1 [112]. Not only has hypoxia been shown to enrich for cancer stem cell phenotypes in ovarian cancer, but it also promotes CSC chemoresistance via HIF-2α mediated upregulation of BCRP, a gene encoding a transporter that can expel drugs [112,153].
Hypoxic conditions also influence the non-cancer TME cells and how they interact with the cancer cells. For example, in hypoxic environments CAF stabilize HIF-1α leading to a metabolic switch to aerobic glycolysis and corresponding lactate production. This lactate can then be used by cancer cells to promote tumor growth [150,154]. Furthermore, CAF are also known for pro-tumorigenic ECM remodeling, which may be linked to hypoxia given that fibroblasts cultured in hypoxia and associated HIF-1 have been shown to promote fibrosis and elevated ECM transcripts when fibroblasts from various organs have been exposed to hypoxic conditions [150,155]. This ultimately can influence the metastatic potential of the cancer cells [150]. Intuitively, as mentioned above, hypoxia also results in the secretion of angiogenesis promoting factors such as VEGF by tumor and stromal cells. This can lead to some new vessels temporarily restoring normoxic conditions until rapidly growing tumor cells once again exceed the reach of the new vessels. This generates further heterogeneity by creating chronic and acute hypoxic regions and development of disorganized vasculature [150].
Unsurprisingly, immune cells are not exempt from the effects of hypoxia. Tumor cells in hypoxic conditions can recruit macrophages to the tumor through secretion of chemoattractants [156]. Once they arrive, hypoxia has been shown to polarize them into an M2-like phenotype to promote tumor progression [156]. Interestingly, myeloid-derived suppressor cells (MDSC), which are normally thought to be immunosuppressive have been shown to enhance immuno-suppression in hypoxia and also to obtain an immuno-stimulatory phenotype [150]. A final example of the role of hypoxia in cellular heterogeneity stems again from the metabolic change in cancer cells brought about by HIF-1α stabilization and the switch to glycolytic dominated metabolism [51,150]. This switch ultimately creates competition for nutrients between the hypoxic cancer cells and T cells, a battle that T cells are unlikely to win. Without sufficient nutrients, the anti-tumor effects of T cells are stymied. T cells in hypoxia may even differentiate into regulatory T cells, which contribute to immunosuppression [150]. Given this evidence it is clear that as hypoxia emerges throughout a tumor, pockets of cells will also emerge with differential, often tumor supporting behavior and thereby contributes to cellular heterogeneity.
Given the effects that each cell type can have on tumor progression and chemoresistance, and the wide ranging TME present in ovarian cancer, it would be unreasonable to assume that a successful therapy for the primary tumor will be similarly effective in secondary locations such as the ascites or the omentum. Consequently, it is critical that these sources of heterogeneity be carefully considered when developing precision medicine models. The effects of this potential variability between and within patients’ tumors are compounded by the ECM in the TME. The various cell types within primary and metastatic tumor sites remodel the ECM in a context-specific manner, which in and of itself impacts drug response (Table 1).
Table 1.
Heterogeneous microenvironmental cells reprogram the extracellular matrix (ECM). The Table illustrates selected examples of the various cell types remodeling the tumor ECM through gene upregulation and increased protein and protease secretion, to promote tumor growth, angiogenesis, and metastasis.
| Type of Cell | Impact on extracellular matrix (ECM) | References |
|---|---|---|
| Cancer stem-like cells (CSC) | Overexpress several collagens, periostin, mucin 1, and tenascin-C (TN-C). Highly express decorin, lumican, biglycan, versican, aggrecan. Produce high levels of hyaluronic acid. |
[171–189,189,190] |
| Cancer-associated mesenchymal stem cells (CA-MSC) | Differentiate into CAFs and adipocytes. Remodel surrounding collagen matrix. Increase collagen production through JAK1 activation. |
[103,191–193] |
| Cancer-associated fibroblasts (CAF) | Remodel ECM to provide ideal stiffness. Create actomyosin tracks in ECM for cancer cells to follow. Secrete versican and increased amounts of collagens. Upregulate COL1A1, COL1A2, COL3A1, COL4A1, COL4A2, COL5A1, COL5A2, COL6A1, COL6A3, and collagen support genes, secreted protein acidic and rich in Cysteine (SPARC), SERPINH1, and SERPINE1. Increased expression of MMP2, MMP11, and TIMP1. |
[94,194–197] |
| Endothelial cells (EC) | Upregulation of lysyl oxidase homolog 2 (LOXL2). Secrete SPARC related modular calcium binding 2 (SMOC-2), cysteine rich with EGF like domains 2 (CRELD-2), microfibril-associated glycoprotein 2 (MAGP-2), lumican, extracellular matrix protein-1 (ECM-1). Highly express COL4A1, COL4A2, SERPINH1, and SPARC. |
[94,198,199] |
| Tumor-associated macrophages (TAM) | Secrete osteopontin, osteoactivin, collagens, fibronectins, and truncated fibronectin. Secrete matrix metalloproteases, cathepsins, lysosomal and a disintegrin and metalloproteinase (ADAM) proteases, and the urokinase-type plasminogen activator (uPA). Secrete TN-C, fibronectins (FN1). |
[200–206] |
| Adipocytes | Upregulation of tumor necrosis factor (TNF-α), osteopontin, MMP9, versican, and leptin. Secrete and process collagen type VI. Secrete endotropin. |
[207–210] |
| Mesothelial cells | Secretes collagen type I. Secrete fibronectin. |
[163,211] |
| Myeloid derived suppressor cells (MDSC) | Secrete MMPs and cathepsins. Releases TGF-β to induce LOX production. Remodel basement membrane. |
[212–214] |
| Neutrophils | Secrete MMP-9 during angiogenesis. | [215,216] |
| Dendritic cells | Release indoleamine 2,3-dioxygenase (IDO) which catalyzes tryptophan and prompts tumor angiogenesis and metastasis. | [217] |
| B cells | Generate interleukin 10 (IL-10) and immunoglobulins (IgG) which form antigen-IgG complexes to recruit immunosuppressive myeloid cells. | [218] |
2.3. ECM Heterogeneity in the TME
Cellular influence on the ECM composition:
Cells within the TME not only affect disease progression via soluble and insoluble signaling, but also exert influence through ECM remodeling (Table 1). CAF in particular, are responsible for a large portion of ECM remodeling in ovarian cancers. Fibroblasts secrete ECM proteins and proteases to develop an alternate ECM scaffolding within the ovarian cortex. However, once fibroblasts differentiate to CAF, they remodel the ECM through increased secretion of ECM proteins such as collagens, fibronectins, and tenascin-C. Secretion of proteases, and ECM modulators including matrix metalloproteinases (MMP), lysyl oxidases (LOX), and transglutaminases (TGM), which degrade and crosslink ECM components respectively is also increased [91,116,157]. Interestingly, a specific subset of CAF have been shown to produce longer collagen fibers that correlate significantly with poor clinical outcomes in head and neck, esophageal, and colorectal cancers [158]. These data support the role of a phenotypic heterogeneity within supporting tumor cells, further linking the TME to disease progression and patient outcome. While CAF are one of the most prominent remodelers of the ECM, many cells have been shown to manipulate the ECM. In ovarian cancer specifically, TAM are delineated from normal macrophages based on a set of 19 commonly upregulated ECM-related genes including LOX, LUM, COL5A1, COL5A2, COL3A1, COL1A1, COL1A2, and MMP-19 among others, clearly indicating that they contribute to ECM remodeling [159]. Cancer cells [91,159], adipocytes [160], EC [75], MSC [161], mesothelial cells [162,163], MDSC [164], neutrophils [165], and T cells [166] also aid in ECM remodeling. Furthermore, it is also well known that the ECM can influence cell phenotypes [167–170]. These bidirectional relationships highlight the urgent need for precision models that address cellular and ECM heterogeneity together, in order to more accurately reflect in vivo disease progression.
Significance of ECM in Classification and Clinical Outcomes in HGSOC:
The important relationship between cellular heterogeneity, ECM, and clinical outcome is apparent in the recently defined major subtypes (mesenchymal, immunoreactive, differentiated, proliferative and anti-mesenchymal) of HGSOC determined from RNA-seq profiling [4,27,219]. The mesenchymal subtype is represented by enhanced desmoplastic response and poor overall survival. The defining expression profile for this subtype includes increased ECM production, remodeling, cellular adhesion, signaling, and angiogenesis, which is due to a large fraction of CAF [93]. The second subtype, termed the immunoreactive subtype, is characterized by slow tumor growth, elevated cytokine and immune activation marker expression, as well as high CD3+ T cell infiltration. This subtype also features significantly better overall survival. Similarly, the differentiated subtype is associated with a higher overall survival than the mesenchymal subtype and is characterized by a low stromal response, tandem to an elevated epithelial signature and reduced growth rate. The proliferative subtype expresses proliferative and mesenchymal markers, as well as reduced immune infiltration and is associated with worse prognosis [4,27,219]. Finally, the anti-mesenchymal subtype is associated with downregulation of genes in the mesenchymal subtype. As a result, this subtype is also associated with higher overall survival [106], further cementing the relationship between the immune and mesenchymal contents of each of these subtypes and overall survival. Interestingly, recent work by Pires et al. showed that an effective T cell response resulted in lower density ECM [166]. This potentially links the improved prognosis in immunoreactive tumors compared to mesenchymal tumors to differences in ECM density brought on by distinct cell composition. This can also be expanded to the other tumor subtypes, as varying cellular and extracellular compositions might indicate responses to established chemotherapy regimens. As another example, mesenchymal and proliferative subtypes have been observed to be comparatively less responsive to treatment [220]. However, despite direct correlations of The Cancer Genome Atlas (TCGA) stratification to patient outcomes, they have yet to be utilized clinically to determine personalized treatment strategies. These RNA-seq identified HGSOC subtypes are a reminder of the cellular and extracellular complexity and heterogeneity, which necessitate personalized TSM to screen therapeutic responses. However, the different cellular and ECM compositions of the five subtypes of HGSOC, are a complex and technical challenge to recapitulate in vitro. As a result, attempts to produce personalized models of both the cellular and ECM components in ovarian cancer with the RNA-seq identified subtypes have been limited. In order to study the impact of collective gene expression of ECM and their related proteins on the treatment efficacy, new advances in our understanding of matrisome signature and structure are outlined below.
ECM Composition in the TME:
The matrisome comprises approximately 300 variations of discrete ECM proteins, each with the capacity to self-assemble into macromolecular superstructures [221]. In cancers, this milieu of macromolecules deviates heavily from the baseline of benign tissue [14,222]. Moreover, as observed in mesenchymal subtype survival statistics, ovarian cancer progression is highly correlated to matrisome dysregulation and reformation [223]. Recent advances in proteomics, imaging, single cell analysis, and bioinformatics have reinvigorated interest in how ECM structure, orientation, and composition change over tumor progression. Similar advances in 3D cell culture have demonstrated feedforward mechanisms relating to protein expression and cellular decision making [14,224]. Since a third of ovarian cancers have their highest protein expression in genes relating to ECM and adhesion [225], it is imperative that the personalized TSM are comprised of extracellular components to characterize and investigate the role of ECM in ovarian cancer outcomes.
Categorized by macromolecular organization and chemical structure, the ECM is generally divided into three subgroups, collagens, proteoglycans, and glycoproteins [170,221,226]. In the ovarian stroma collagens subgroup, collagen type I and III specifically, are thought to be the most abundant [227]. In normal ovarian tissue, collagen type I and III can be highly crosslinked, however during tumor progression collagen crosslinking becomes defective [228]. Second harmonic generation (SHG) confocal microscopy has discovered vast degradation and secretion of collagen type I fibers in HGSOC. Wherein the collagen type I of ovarian stroma – consisting of nonspecific fiber orientation – is degraded by tumor cells and replaced with a neo-reactive fiber structure [229]. This new malignant-type collagen fiber network has a higher degree of orientation, noted by a dense and ordered structure with comparative regularity over the network expanse (Fig. 2) [230]. Given that collagen type I fiber morphology and abundance are directly linked to migration and drug resistance in ovarian cancer cells respectively [185,231], the personalized tumor-specific models must incorporate this ECM component, and study the changes in its spatio-temporal morphology.
Immunohistochemistry (IHC) is commonly used to analyze ECM. However, the process of fixing, sectioning, and staining samples can damage the structure of ECM and limits the information that we can obtain from IHC analysis. Recent advances in proteomic approaches, such as peptide extraction, ultra-high pressure liquid chromatography, mass spectrometry, and bioinformatics, have provided new opportunities to obtain renewed insight into the dynamic changes in ECM during tumor progression. A comparative study by Pearce et al., has revealed a complex ECM landscape at varying HGSOC disease stage, from uninvolved, non-malignant to extensive and aggressive infiltration [92,232]. Interestingly, their findings confirm a contested theory whereby the relative abundance of collagen type I and III actually decreases in late stage and secondary tumors, likely due to a gradual dysregulation of ECM production in the TME. In addition, they identify the expression of fibronectin, cartilage oligomeric matrix protein (COMP), versican, collagen XI, collagen X, collagen I, and hyaluron to be highly correlated to disease score and the tumor stiffness. As COMP, versican, and collagen XI are normally reserved for stiff tissues such as bone or cartilage, their presence in HGSOC further highlights ECM dysregulation in the TME. Intriguingly, a decrease in key basement membrane genes relating to laminin, collagen type IV, and perlecan were also found in late stage, primary tumors. Decay of basal membrane components have previously been linked to intraperitoneal dissemination [170,228,233]. Similarly, increased expression of glycosaminoglycans (GAG) and proteoglycans such as hyaluron and versican in the metastatic tumor are associated with cancer cell migration by expanding local tissue space, increasing tumor modulus, and improving mesothelial adhesion [233–236]. Beyond migration, hyaluron is also thought to promote chemoresistance through CD44 binding, PI3K/AKT, p53, and MAPK survival pathways [237,238]. Another ECM component, tenascin-C, is found between the epithelia and the stroma of malignant tumors, with its expression levels being closely associated with tumor grade [239]. Tenascin-C also mediates αvβ3, αvβ5, α6β4 integrin interactions in concert with periostin, fibronectin, and type I and IV collagen, further shown to promote paclitaxel resistance in ovarian cancer [240,241]. The glycoprotein, fibronectin is also found to be highly expressed in metastatic tumors, and is closely correlated to tumor stage and growth [242–245]. Moreover, fibronectin is thought to play a prominent role in transcoelomic metastasis, with high concentrations found in patient ascites, associated with mesothelial adhesion and metastasis (Fig. 2) [163,246–248].
The ECM at metastatic sites is also remodeled to promote disease progression. For example, omental metastasis undergoes ECM remodeling during colonization. Fibrillar collagens are the most prevalent macromolecule, with substantial clusters of collagen type I entwined between fibroblasts, below the mesothelium surface, and within the basement membrane. Similar to primary tumors, a general increase in glycoprotein and proteoglycan production is observed in the TME of omental metastasis (Fig. 2) [232]. Interestingly, proteins that define the basement membrane are relatively replete in omental metastasis, theoretically aiding in the establishment of the tumor colony [249]. Specifically, an increased expression of laminin, collagen type IV and fibronectin are observed in the basement membrane of omental metastases [247,250–252].
Targeting the components of ECM to improve patient outcomes:
Due to the relationships between ECM, cells, and clinical outcomes, targeting critical ECM components can improve the efficacy of precision treatment [8,253–255]. The promise of targeting ECM components is exemplified by losartan, an FDA approved anti-hypertensive agent that blocks angiotensin II receptor type 1 (AT1) [256]. In ovarian cancer bearing mice, losartan treatment significantly lowered collagen and hyaluronic acid concentration in addition to decreasing the number of fibroblasts and α-SMA+ stromal cells. The decrease of ECM resulted in lowered solid stress and increased intra-tumoral chemotherapy penetration. In breast and pancreatic cancers, losartan has shown decreases in intratumoral expression of thrombospondin-1 (THBS-1) and activated TGF-β in CAF. As a result, losartan is now being evaluated in Phase II clinical trials for treatment of pancreatic cancer along with chemotherapy [257]. Given the heterogeneity of ECM components in HGSOC and the promise of precision ECM targeting treatments, it is vital to integrate different components of the ECM into personalized models.
Further, targeting ECM protein stabilizers that contribute to tumor stroma stiffness and crosslinking of collagen and elastin is also a promising lead [258,259]. For example, the mitigation of lysyl oxidases (LOX) resulted in decreased tumor burden and collagen remodeling in HGSOC associated omental metastasis [211,260]. Similarly in pancreatic cancers, the combination of LOX inhibition and gemcitabine reduced collagen fibril density, metastasis, and extended disease-free survival in mouse models [261]. In yet another example, LOX inhibition reduced collagen cross-linking and fibronectin assembly, increased drug penetration, induced apoptosis, and re-sensitized triple negative breast cancers to chemotherapy [262,263]. These successes have led to clinical trials where LOX inhibition efficacy is being investigated in myelofibrosis and pancreatic cancer patients [264]. Given the increased prevalence of collagens and elastin in mesenchymal and other ECM-rich stromal HGSOC subtypes, it is essential to target these components and their modifiers that generate, stabilize, and orient structural proteins.
2.4. Extracellular Matrix Properties within the TME
In healthy individuals both the cellular and acellular contents of the ovaries are tightly regulated by the peritoneal membrane. However, during tumor progression, components of the TME are gradually dysregulated. In primary tumors, this dysregulation (Section 2.2 and 2.3) aids in tumor cell dissemination into the peritoneal cavity, often leading to the formation of malignant ascites [265,266]. Retention of ascitic fluid in the peritoneal cavity prompts a unique mechanical environment, further augmenting the mechanical forces present in the ovarian TME, including matrix stiffness, shear stress, compression, and tensile stresses [12,267,268].
As a consequence of HGSOC associated desmoplasia, the ECM in ovarian tumors continuously stiffens [269]. The average Young’s modulus for ovarian tumors is cited at 5 kPa but contains discrete regions with moduli ranging from 16 to 35 kPa [269,270]. Elevated ECM stiffness has been shown to influence metastasis, invasion, proliferation, and chemoresistance [16,253,271–275]. Numerous studies have proven stiffer substrates enhance the metastatic phenotypes of cancer cells [276–280]. However within the field of ovarian cancer, studies have resulted in contradictory findings [269,281–284].
Ovarian cancer cells within the peritoneal ascites experience a range of compressive and shear stresses, originating from hydrostatic pressure, growth induced stress, as well as ascitic, vascular, and interstitial fluid flow respectively [12]. In the intraperitoneal space, shear stress is estimated to range between 0.14 and 11 dynes/cm2, while compressive stress is thought to range between 4.7 and 18.9 kPa [285–287]. External dynamic stimuli, including shear stress and compression, have been correlated with increased metastasis, CSC enrichment, chemoresistance, and proliferation in a variety of cancers. The effects of compression on ovarian cancer is currently thought to be contradictory and requires further attention [288–293]. Evidently, the full role that mechanical stimuli plays in ovarian tumor progression and treatment response is not well understood. This represents yet another gap in knowledge that could be filled with investigation using the patient-derived, personalized TSM.
2.5. Evidence of complex interactions within the TME
As outlined above, heterogeneity in ovarian cancer is made up of cellular, ECM, and mechanical components. Yet, rather than existing in isolated states, TME constituents are inherently coupled, cooperating together to promote events within tumorigenesis. When cells in the tumor remodel the ECM, they invariably change the mechanical properties of their surroundings. This in turn alters cellular phenotypes and consequently, disease progression and chemoresistance. Within breast tumors for instance, a tight correlation exists between immune infiltration, ECM density, and drug response [169,263]. Cohorts of tumors that respond well to treatment display muted ECM density and a higher degree of T cell infiltration [294–296]. Understanding this interconnected system has enabled researchers to predict measures – including relapse – more accurately [169,297–299]. In ovarian tumors, stiffness correlates strongly with disease stage whereby late stage tumors generally have a higher modulus [92]. The increase in stiffness and tumor stage promotes ovarian cancer cells to become increasingly pliable further promoting migration and invasive potential [300,301]. This feed forward mechanism demonstrates how stromal cells change ECM, in turn affecting cancer cell behavior, and finally how this behavior aids in disease progression. Coupling events like these highlight why comprehensive models are crucial in our understanding of ovarian cancer behavior, and why we might need to alter our approach to precision medicine.
3. MAKING PERSONALIZED TME MODELS for HGSOC
Due to the diversity in origin, architecture, and composition of HGSOC [220], conventionally available models need to be further improved to meet the needs of clinical translation [10,11,33,302,303]. In this section, we will review contemporary models used to replicate tumor cellular and acellular heterogeneity and discuss the critical next steps in the development of tumor-specific models for precision medicine applications. As evidenced throughout this review, the proposed patient-informed models contain enormous potential to improve patient care and clinical outcomes.
3.1. Models of Cellular heterogeneity
Conventional approaches of drug screening include the use of isogenic cell lines in 2D culture or propagated as xenografts, genetically engineered mouse models (GEMM), patient-derived xenografts (PDX), and patient derived organoids (PDOs) [10,302–304]. While 2D cell line cultures are easy to use and inexpensive, they have long been known to poorly represent in vivo conditions, making observed drug responses unreliable [305]. Newer 2D models might use cell lines derived from patient samples, however generation of a new cell line is prone to low success rates and fibroblast contamination. Moreover, the cell lines that are successfully established will have persevered through a strong selection pressure in 2D in vitro conditions making them a poor representation of the heterogeneous tumor cell population [19].
GEMM and PDX on the other hand are labor- and time-intensive, making it challenging for them to contribute to humanized drug screening and individualized therapy on a clinical scale [19,305,306]. Furthermore, GEMM are limited in their ability to consistently generate tumors on a reliable timeline as these tumors form with heterogeneous latency periods and growth rates [307]. PDX formed from heterogeneous populations of patient cells maintain cellular heterogeneity, however are susceptible to copy number alterations with passaging and loss of human immune cells in the tumor which may result in unrepresentative drug responses compared to the patient responses [308]. The immunocompromised nature of PDX [307] also prevents evaluation of a functioning immune system in drug response. Overall, mouse models have struggled to translate into the clinic with only about 5% of the drugs tested in mouse models performing well enough in phase III trials, to be licensed for clinical usage [308]. However, a promising newer variation of mouse model, called ‘humanized mice’ has been developed with human immune cells, allowing for evaluation of tumor interactions with the immune system [308]. Despite this improvement, the technical challenge of developing these models remains, and hinders use as high throughput screening systems.
On the other side of the spectrum, 3D in vitro co-cultures are ideal for high throughput drug screening and have played an important role in our evaluation of interactions between two or more cell types in the TME. For example, using recently developed 3D co-culture heterospheroid of CSC and CA-MSC, PDGF and Hedgehog crosstalk was found to be a key signaling mechanism, involved in increasing stemness, metastatic potential, and chemoresistance in CSC [309]. Similar heterospheroid models were utilized to show that ovarian cancer cells reprogram normal ovarian and omental MSC into pro-tumoral CA-MSC, presenting evidence that ovarian cancer cells catalyze the formation of their own pro-tumoral microenvironment [95]. Similarly, to dissect the signaling between immune cells and ovarian tumor cells, recent 3D models have featured co-cultures with immune cells. Since the adaptive and innate immune systems are both responsive and influential to the ovarian TME, they both have roles in the initiation and resolution of inflammatory response [32,33]. Ovarian cancer cells form spheroids in the ascitic fluid due in part to their interactions with macrophages, and these heterospheroids are thought to aid in transcoelomic metastasis [126]. Utilizing a hanging drop non-adherent 3D suspension model, ovarian CSC and activated macrophages can be brought in close association, simulating the physiologic environment of non-adherent malignant ascites. These 3D heterospheroids illustrate that pro-tumoral macrophages promote chemoresistant and invasive phenotypes in CSC, further leading to CSC enrichment. This model was used in the discovery that reciprocal paracrine signaling via WNT/β-catenin between macrophages and ovarian CSC promoted pro-tumoral environments including polarization of macrophages into M2-like phenotypes, and increased expression of the stem marker ALDH in CSC. This suggested that the WNT/β-catenin pathway could be a potentially effective target for new therapeutics to specifically eradicate the immuno-modulation of macrophages by CSC that contribute to recurrent disease [167]. These types of models have been made with tumor cells and endothelial cells [310], mesothelial cells [311], and adipocytes [144]. While co-cultures are advantageous in examining interactions between two or three cell types, they still do not replicate patient-specific cellular and acellular heterogeneity, and thus motivate the development of multiscale personalized TSM.
PDO have been instrumental in our ability to replicate cell-cell interactions with realistic cell compositions in vitro and have had some success in predicting drug response [19,305]. PDO are formed from either a single patient cell or a heterogeneous population of patient-derived cells grown into organoids that are embedded in basement membrane extract with an appropriate growth factor and small molecule cocktail to replicate the in vivo TME [19]. A panel of ovarian PDO have been successfully created for long term in vitro culture of all subtypes of epithelial ovarian cancers. The panel of organoids was generated with cells from primary tumors, metastatic lesions, ascites, and pleural puncture. Through in-depth analysis, they demonstrated that tumor organoids maintain histological characteristics like nuclear and cellular atypia and expression of tumor biomarkers like p53 and pax8, similar to their source samples. This study showed that passaging the organoids did not result in any genomic changes compared to the original tumors. The organoids also show hallmarks of ovarian cancers, including the significant number of copy number variations, recurrent mutations and tumor heterogeneity [36]. However, a drawback of these organoids is their cell composition, as tumor organoids averaged 88 ± 23% of cancer cell content, while the actual tumors contained only 49 ± 9% tumor cell content across all samples [19]. HGSOC organoids have also been utilized for screening compounds, while maintaining the intra- and inter-patient tumor heterogeneity and mutation status, and matching the parental tumors genetically and functionally [312–314]. Another drawback to conventional organoids is the use of basement membrane extract, such as Matrigel, for organoid culture [19]. As we discussed above, the dynamic interactions between the ECM and the cells in a tumor have a profound effect on tumor progression and clinical outcomes, which deems the use of extracts with uncontrolled ECM composition problematic. A more thorough review of these PDO models was published previously [10].
3.2. Models of ECM heterogeneity
The ECM constituents of the TME make up a rich mosaic of collagens, proteoglycans, and glycoproteins. Each macromolecule has a unique function, position, and concentration that impart chemical and physical cues to surrounding cells. Changes to the local tumor ECM impact cancer progression and individual patient response to treatment [14,16,315]. Cataloging and modeling the TME and cell-matrix interactions are vital to the long-term improvement of patient outcomes. ECM constituents, similar to TME cell heterogeneity, are disparate and unique to individuals. It is therefore important that 3D biomaterials-based personalized models are improved to better replicate a tumor’s protein rich ECM [316]. Although a plethora of 3D model systems have been utilized to study cancers, few capture the complexity of the tumor ECM, and none combine the vast biochemical and biophysical interactions present in a singular model.
The most widely utilized contemporary model for 3D ovarian carcinomas is the murine derived Matrigel, a solubilized basement membrane harvested from mouse sarcoma. However, with undefined constituents, batch to batch variability, and poor biophysical properties, it does not accurately replicate the ovarian tumor stroma [317]. Moreover, Matrigel fails to replicate the physiology of the primary tumor, as the basement membrane is lost during disease progression (section 2.3). Naturally derived protein scaffolds address some of these limitations by providing physiologically relevant chemical cues and magnifying key cell-matrix interactions. For example, the role of α5β1-integrin interactions during ovarian metastasis was discovered using fibronectin coated surfaces (polyethylene glycol [PEG] has been utilized to further regulate surface modulus) [269,318]. Studies have documented an increase in ordered collagen type I expression in ovarian carcinomas, enhancing fibrotic microenvironment at primary and secondary sites [319]. 3D collagen matrices have induced MMP matrix degradation in ovarian cancer cell lines which may support recent proteomics and SHG findings, whereby mature collagen matrices are replaced by ordered fibers [320]. Collagen type I concentrations of 3.6 and 3.4 mg/mL have been used to grow ovarian cancer cell lines [321,322]. The modulus of collagen gels generally ranges from 1 to 4 kPa, with a scaffold of 3.6 mg/mL providing a modulus of 1.7 kPa (Poisson’s ratio, 0.3) [321,323]. Notwithstanding, collagen fiber density has been shown to impact cellular behavior independent of the materials bulk stiffness [323]. This decoupling of stiffness and collagen associated adhesion motif (GFOGER) further contextualizes the complexity of the TME. Therefore, in order to create personalized TSM, it is vital to control both material stiffness and collagen concentration within collagen containing biomimetic scaffolds. Although GFOGER is the relevant binding site in the collagen triple helices, RGD cues have also been widely investigated in cancer. Shear thinning and partially crosslinked gelatin scaffolds (which have exposed RGD epitopes) achieve a higher stiffness than collagen scaffolds, and can be 3D printed to fine tune pore size and permeability [324].
Hydrogels have also been created using interpenetrating networks (IPN) to decouple physical and chemical characteristics within TME biomimetics. Thus, IPN hydrogels can be designed to contain relevant bioactive substrates while maintaining physiologically relevant mechanical signaling – including modulus, viscoelasticity, pore size, and permeability. For example, combining collagen type I and collagen type III together has shown increased invasion of transformed fallopian tube epithelial cells compared to collagen type I alone [325]. IPN hydrogels may also allow for the manipulation of discrete components in a spatio-temporal manner. A recent study shows that collagen type I morphology could be synthesized and manipulated using multiphoton excited photochemistry to align fibers within a solubilized gelatin methacrylate scaffolding [229]. Porosity can also be fine-tuned in IPN hydrogels, as with 0.125% (w/v) agarose + 1% (w/v) alginate containing 7.5% to 10% collagen type I IPN, which achieved average pore sizes of 66.6 to 62.8 μm respectively, within ovarian cancer cultures [326]. Polysaccharide hydrogels such as alginate and agarose are bioinert, and can reach the higher stiffness regimes present in ovarian tumors [12,327]. Even when exposed to long durations of physical perturbations, such as those present in studies involving mechanotransduction, agarose maintains form and structure [12]. Ovarian cancer cells, encapsulated in 3% (w/v) agarose and 0.5 mg/mL collagen type I IPN hydrogels exposed to elevated compressive stress, have shown increased invasiveness and chemoresistance [328]. The capacity to decouple physical and chemical signaling is mirrored by 4 and 8 arm PEG, with each arm capable of functional modification to include RGD or MMP motifs [282,329]. Using a PEG-MMP-RGD functionalized hydrogel system (1.5/2.0/2.5% (w/v)), ovarian cancer cells have been shown to express variable proliferative rates and spheroid formation faculty in response to increased stiffness (G’ = 241/637/1201 Pa) [282].
Self-assembling peptide-based bioinks can be programmed to assemble into well-defined structures in situ [330]. With the capability to form fine-tuned alignments, attain mechanical properties ranging from 0.6 to 205 kPa, and incorporate protein-mimetic epitopes that encourage angiogenesis and migration, they make an appealing prospect for the future of personalized tumor-specific models [331,332]. In a recently published report, ovarian TME cells were encapsulated within peptide amphiphiles/keratin (PA/K) bioinks – including HHL, RGD, and GHK epitope sequences, to enhance ECM co-assembly, cell adhesion, and proliferation respectively – specialized with organized ECM proteins [333]. The PA/K peptide hydrogels reached a stiffness of 7 kPa, and were shown to maintain stability for up to 28 days under cell culture conditions. In parallel, the self-assembling peptide RADA16-I 1% (w/v) recapitulated the effects of collagen type I models on ovarian cancer cell cultures. As RADA16-I also mimics the porosity of tumor ECM (with a pore size of 5 – 200 nm and nano fibers approximately 1000 nm in length), it can also serve as a respectable tumor model [334]. While the methods outlined above (mimicking the structure and properties of the ECM) have advanced our understanding of cancer cell-ECM interactions, there remains substantial room for improvement through the incorporation of patient and disease-specific cell and ECM compositions.
3.3. Proposed Personalized Tumor-Specific Precision Medicine Platform
Given the indisputable and coupled relationship of cellular and ECM heterogeneity with therapy response, we propose that the next step in the development of personalized and tumor models for precision medicine will need to be grounded in evidence from functional and descriptive -omics data. Specifically, we propose a thorough characterization of patient biopsies using existing and emergent technologies to determine molecular, cellular, and ECM composition. In light of innovative work from the Hynes group, leveraging proteomics has been shown to faithfully enumerate the matrisome of complex diseases [335–338]. Despite the elucidation of breast and pancreatic cancer matrisome, limited investigation has been done to define the matrisome of ovarian cancer (section 2.2). Next generation imaging and informatics modalities, including Multiplex FISH [339], multiplexed ion beam imaging – time of flight (MIBI-TOF) [340], and tissue based cyclic immunofluorescence (t-CyCIF) [341] high throughput histology will also provide a crucial spatial and quantitative snapshots of tumor heterogeneity (including protein and cellular content). The utilization of peptide extraction, ultra-high pressure liquid chromatography, mass spectrometry, mass cytometry [342], single cell sequencing [93], and bioinformatics can further inform the cellular and acellular composition.
Information gleaned from comprehensive characterization of a patient’s heterogeneous tumor composition using these tools allows for personalized re-engineering of the tumor with the same composition in vitro. However, due to the limited size of patient samples it will be important to select only the most effective characterization techniques. Generating these TSM within a high throughput culture system (like a 384-well hanging drop plate) enables drug screening to determine which tumor microenvironment compositions or more generally, which patients might respond to certain treatments or combination treatments (Fig. 4). By incorporating both cellular heterogeneity and ECM complexity into personalized disease platforms, patient-specific cell-cell and cell-ECM signaling will be more complete and we expect that treatment response predictions will be more accurate, thereby improving outcomes for cancer patients [238,333]. Furthermore, by utilizing recent advances in the fields of cellular and 3D in vitro ECM modeling to create these platforms we would pave the way for a plethora of critical downstream applications described in the following section.
Figure 4.

Proposed clinical workflow for patient-derived tumor-specific 3D models which can predict therapy response and identify the most effective yet non-toxic therapies or combinations, leading to sustained and durable responses.
4. Applications of Personalized Tumor-Specific Models
Personalized tumor-specific models have enormous potential to improve patient care and clinical outcomes. In this section, we will identify and discuss the potential high-impact applications that could be realized with these highly individualized models. TSM will be utilized in high throughput assays whereby drug response data, corresponding to individualized tumor compositions, is collected over time and analyzed in silica to determine appropriate course of action (Fig. 4). This workflow could be used for numerous critical applications:
Personalized screening for drug compounds and combinations:
Using the data obtained from characterizing patient biopsies, personalized TSM can be generated and used for screening. Based on the results of the screening, a drug or a combination of drugs will be selected based on efficacy in personalized TSM. Next, the features that are most important in determining a patient’s response to a wide variety of treatments could be discerned. This could expedite the discovery of effective adjuvant combination treatments to reduce disease burden and improve cure rates.
Identify biomarkers of chemosensitivity:
Identifying novel biomarkers of chemosensitivity is another critical function of TSM. After generating TSM from a large cohort of patients over time, patterns and tumor characteristics associated with responses to specific therapies could be identified. In the long term, complex combinations of biomarkers indicating sensitivity to certain therapies could be recognized, enabling rapid clinical treatment decisions following characterization of a patient’s tumor, potentially without the need to rebuild their microenvironments in TSM. In this situation, not only would the treatment be highly personalized, but it would decrease time to treatment, as there would be a large library of data to reference. With more precise sets of biomarkers and decreased time to treatment, clinical outcomes could drastically improve.
Predict and prevent relapses and treat minimal residual disease:
Most HGSOC relapse within the first 2 years of diagnosis, even after optimal de-bulking, chemo-, and targeted-therapies (PARP inhibitor, anti-angiogenic antibodies, etc.). As the tumor cells evolve post-therapy and develop resistance, the proposed TSM model could be applied to the evolved disease, and off-target cytotoxicity could be better avoided. Similarly, to prevent relapse, optimal cocktails of compounds that target the sources of minimal residual disease based on cataloged data indicating the key features responsible for relapse could be designed for future use. Along with TSM, the predictive models would calculate appropriate sequential treatments thereby extending remission period.
TSM could also be utilized to study quiescent tumor cells, which are conventionally difficult to study in vitro. As quiescent cancer cells that have escaped previous therapies resurface again in the TSM, their evolution can be studied and monitored to understand how different residual clonal populations take hold, and sequenced. These results can then be used to identify the druggable targets in the clonal chemoresistant populations that emerge following various primary treatments.
Predict the characteristics of future relapses:
By utilizing TSM, tumor recurrence could be modeled through serial passaging, a previously established process [343] wherein TSM would be created, dispersed after a period of culture, and then re-generated from the same cell population. This process would address the development of chemoresistance and the enhancement of CSC populations simultaneously.
5. Conclusions
In this article, we have laid out why personalized tumor-specific models (TSM) are crucial for improving precision medicine outcomes. The tumor microenvironment (TME) is made up of an amalgam of cellular and acellular components that all interact dynamically, and drive disease progression. However, it is difficult to capture the complex cellular and acellular features of the TME in conventional animal models and in vitro platforms. By omitting many of these features, biochemical and biophysical cues and epigenetic changes are lost in many contemporary models. This loss of biomarkers will lead to flawed drug screening results which will fail to select the optimal drug for a patient, thus causing a suboptimal outcome. For this reason, we propose taking elements of the TME, obtained through thorough biopsy characterization, and integrating them into personalized TSM in high throughput culture systems. In theory, this will recapitulate the patient’s heterogeneity, and thus could produce a representative chemoresistance profile. Once this process is repeated with a significant number of patients, TSM may facilitate identification of biomarkers for chemosensitivity and relapse based on the initial characterization of another patient’s tumor (without the need to generate the TSM and perform the drug screening). This would minimize lag time between diagnosis and the start of treatment, which could further improve outcomes. Importantly, although we presented this proposal through the example of ovarian cancer, we postulate that the same approaches may be useful for other types of cancers, and in general for many human diseases.
Analogous to our advances in genomic, proteomic, matrisome and other emergent platforms, we will soon be able to ‘sequence’ both the cellular and ECM constituents of each individual tumors. This will enable the recreation of biomimetic 3D TME for each patient, including appropriate cell and ECM constituents, in physiologically relevant abundance and orientation. Each patient tumor will be multiplexed on high throughput arrays to determine the best course of treatment. Additionally, by integrating various tools for sequencing, mathematical modeling, and biomaterials engineering, we will achieve the true promise of personalized medicine. The development and application of TSM is a crucial element for realizing the potential of precision medicine. The advance of personalized 3D microscale tumor models will facilitate the discovery of new therapeutics (drugs, pro-drugs, enzymes, genes, viruses, etc.) that target patient-specific features (genetic, epigenetic, molecular, and/or cellular) that are driving that patient’s disease. Ultimately, by using biomaterials to make TSM, the clinical goal of personalized cancer therapies for each patient can be met and will improve the lives of countless cancer patients across the world.
Figure 3: Selected examples of contemporary 3D platforms that can be applied towards personalized biomaterials-based patient-specific multiscale tumor models.

A) Heterospheroids in 3D suspension co-culture, that comprise of small number of ovarian cancer stem-like cells and M2-like alternatively activated macrophages, as described in [95]. B) 3D patient-derived in vitro ovarian cancer organoids grown in Matrigel, as characterized in [204]. C) In vivo patient derived xenograft (PDX) models in immune compromised animals described in [207]. D) 3D hydrogels based on a single ECM component, such as collagen type I, as described in [145]. E) 3D interpenetrating hydrogel created from multiple ECM components, for example, alginate-agarose-collagen type I as described in [223]. F) Self-assembling 3D scaffold, for example, peptide amphiphiles and keratin hydrogel, as described in [229]. (All schematics are original works based on previously published methods.)
Acknowledgments:
We would like to acknowledge Ms. Vineta Chugh for her assistance with creating Figures for this work.
Funding:
This work is supported by the American Cancer Society Research Scholar Award RSG-19-003-01-CCE (G.M.), the Office of the Assistant Secretary of Defense for Health Affairs through the Ovarian Cancer Research Program under Award No. W81XWH-16-1-0426 (G.M.), W81XWH-13-1-0134 (G.M.) and W81XWH-15-1-0221 (Y.Y.H), DOD Investigator Initiated award W81XWH-18-0346 (G.M.), Rivkin Center for Ovarian Cancer (G.M.), Michigan Ovarian Cancer Alliance (G.M.), NSF EFRI DChem (Award number 2029139), NIH/NIDCR Tissue Engineering and Regeneration Training Grant T32DE00007057 (M. E. B.) and the Rackham Research Grant (C.S.S.). Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under award number P30CA046592, NIH/NCI R03 CA216127 (Y.Y.H), Discovery To Cure Ovarian Cancer Research Grant (Y.Y.H), and Colleen’s Dream Foundation Research Grant (Y.Y.H)
Footnotes
Conflicts of Interest: The authors declare no conflict of interest.
References
- [1].Seyhan AA, Carini C, Are innovation and new technologies in precision medicine paving a new era in patients centric care?, J Transl Med. 17 (2019) 114. 10.1186/s12967-019-1864-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Gameiro GR, Sinkunas V, Liguori GR, Auler-Júnior JOC, Precision Medicine: Changing the way we think about healthcare, Clinics (Sao Paulo). 73 (2018) e723. 10.6061/clinics/2017/e723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Le Tourneau C, Borcoman E, Kamal M, Molecular profiling in precision medicine oncology, Nat Med. 25 (2019) 711–712. 10.1038/s41591-019-0442-2. [DOI] [PubMed] [Google Scholar]
- [4].Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, Traficante N, Fereday S, Hung JA, Chiew Y-E, Haviv I, Group AOCS, Gertig D, deFazio A, Bowtell DDL, Novel Molecular Subtypes of Serous and Endometrioid Ovarian Cancer Linked to Clinical Outcome, Clin Cancer Res. 14 (2008) 5198–5208. 10.1158/1078-0432.CCR-08-0196. [DOI] [PubMed] [Google Scholar]
- [5].Tomao F, Boccia SM, Sassu CM, Chirra M, Palaia I, Petrella MC, Di Donato V, Colombo N, Benedetti Panici P, First-Line Treatment with Olaparib for Early Stage BRCA-Positive Ovarian Cancer: May It Be Possible? Hypothesis Potentially Generating a Line of Research, Cancer Manag Res. 12 (2020) 5479–5489. 10.2147/CMAR.S194874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Coward JI, Middleton K, Murphy F, New perspectives on targeted therapy in ovarian cancer, Int J Womens Health. 7 (2015) 189–203. 10.2147/IJWH.S52379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Jiang Y-Z, Liu Y, Xiao Y, Hu X, Jiang L, Zuo W-J, Ma D, Ding J, Zhu X, Zou J, Verschraegen C, Stover DG, Kaklamani V, Wang Z-H, Shao Z-M, Molecular subtyping and genomic profiling expand precision medicine in refractory metastatic triple-negative breast cancer: the FUTURE trial, Cell Research. 31 (2021) 178–186. 10.1038/s41422-020-0375-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Senthebane DA, Rowe A, Thomford NE, Shipanga H, Munro D, Mazeedi MAMA, Almazyadi HAM, Kallmeyer K, Dandara C, Pepper MS, Parker MI, Dzobo K, The Role of Tumor Microenvironment in Chemoresistance: To Survive, Keep Your Enemies Closer, Int J Mol Sci. 18 (2017). 10.3390/ijms18071586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Dzobo K, Taking a Full Snapshot of Cancer Biology: Deciphering the Tumor Microenvironment for Effective Cancer Therapy in the Oncology Clinic, OMICS. 24 (2020) 175–179. 10.1089/omi.2020.0019. [DOI] [PubMed] [Google Scholar]
- [10].Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G, Integrated cancer tissue engineering models for precision medicine, PLOS ONE. 14 (2019) e0216564. 10.1371/journal.pone.0216564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Bregenzer ME, Horst EN, Mehta P, Novak CM, Repetto T, Mehta G, The Role of Cancer Stem Cells and Mechanical Forces in Ovarian Cancer Metastasis, Cancers (Basel). (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Novak C, Horst E, Mehta G, Review: Mechanotransduction in ovarian cancer: Shearing into the unknown, APL Bioengineering. 2 (2018) 031701. 10.1063/1.5024386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Hinshaw DC, Shevde LA, The Tumor Microenvironment Innately Modulates Cancer Progression, Cancer Res. 79 (2019) 4557–4566. 10.1158/0008-5472.CAN-18-3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Eble JA, Niland S, The extracellular matrix in tumor progression and metastasis, Clin Exp Metastasis. 36 (2019) 171–198. 10.1007/s10585-019-09966-1. [DOI] [PubMed] [Google Scholar]
- [15].Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C, Parker MI, Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development, OMICS: A Journal of Integrative Biology. 22 (2018) 17–34. 10.1089/omi.2017.0174. [DOI] [PubMed] [Google Scholar]
- [16].Paszek MJ, Weaver VM, The tension mounts: mechanics meets morphogenesis and malignancy, J Mammary Gland Biol Neoplasia. 9 (2004) 325–342. 10.1007/s10911-004-1404-x. [DOI] [PubMed] [Google Scholar]
- [17].Zhao Y, Cao J, Melamed A, Worley M, Gockley A, Jones D, Nia HT, Zhang Y, Stylianopoulos T, Kumar AS, Mpekris F, Datta M, Sun Y, Wu L, Gao X, Yeku O, Del Carmen MG, Spriggs DR, Jain RK, Xu L, Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma, Proc Natl Acad Sci U S A. 116 (2019) 2210–2219. 10.1073/pnas.1818357116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Bai H, Cao D, Yang J, Li M, Zhang Z, Shen K, Genetic and epigenetic heterogeneity of epithelial ovarian cancer and the clinical implications for molecular targeted therapy, J Cell Mol Med. 20 (2016) 581–593. 10.1111/jcmm.12771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Kopper O, de Witte CJ, Lõhmussaar K, Valle-Inclan JE, Hami N, Kester L, Balgobind AV, Korving J, Proost N, Begthel H, van Wijk LM, Revilla SA, Theeuwsen R, van de Ven M, van Roosmalen MJ, Ponsioen B, Ho VWH, Neel BG, Bosse T, Gaarenstroom KN, Vrieling H, Vreeswijk MPG, van Diest PJ, Witteveen PO, Jonges T, Bos JL, van Oudenaarden A, Zweemer RP, Snippert HJG, Kloosterman WP, Clevers H, An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity, Nat Med. 25 (2019) 838–849. 10.1038/s41591-019-0422-6. [DOI] [PubMed] [Google Scholar]
- [20].Patch A-M, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, Kassahn KS, Newell F, Quinn MCJ, Kazakoff S, Quek K, Wilhelm-Benartzi C, Curry E, Leong HS, Hamilton A, Mileshkin L, Au-Yeung G, Kennedy C, Hung J, Chiew Y-E, Harnett P, Friedlander M, Quinn M, Pyman J, Cordner S, O’Brien P, Leditschke J, Young G, Strachan K, Waring P, Azar W, Mitchell C, Traficante N, Hendley J, Thorne H, Shackleton M, Miller DK, Arnau GM, Tothill RW, Holloway TP, Semple T, Harliwong I, Nourse C, Nourbakhsh E, Manning S, Idrisoglu S, Bruxner TJC, Christ AN, Poudel B, Holmes O, Anderson M, Leonard C, Lonie A, Hall N, Wood S, Taylor DF, Xu Q, Fink JL, Waddell N, Drapkin R, Stronach E, Gabra H, Brown R, Jewell A, Nagaraj SH, Markham E, Wilson PJ, Ellul J, McNally O, Doyle MA, Vedururu R, Stewart C, Lengyel E, Pearson JV, Waddell N, deFazio A, Grimmond SM, Bowtell DDL, Whole–genome characterization of chemoresistant ovarian cancer, Nature. 521 (2015) 489–494. 10.1038/nature14410. [DOI] [PubMed] [Google Scholar]
- [21].Aletti GD, Gallenberg MM, Cliby WA, Jatoi A, Hartmann LC, Current management strategies for ovarian cancer, Mayo Clin Proc. 82 (2007) 751–70. 10.4065/82.6.751. [DOI] [PubMed] [Google Scholar]
- [22].Siegel RL, Miller KD, Jemal A, Cancer statistics, 2016, CA Cancer J Clin. 66 (2016) 7–30. 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
- [23].Marcus CS, Maxwell GL, Darcy KM, Hamilton CA, McGuire WP, Current approaches and challenges in managing and monitoring treatment response in ovarian cancer, J Cancer. 5 (2014) 25–30. 10.7150/jca.7810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Weberpals JI, Koti M, Squire JA, Targeting genetic and epigenetic alterations in the treatment of serous ovarian cancer, Cancer Genet. 204 (2011) 525–35. 10.1016/j.cancergen.2011.09.004. [DOI] [PubMed] [Google Scholar]
- [25].Schwartz M, Camacho-Vanegas O, Wood AM, Dashkoff M, Whitelock C, Harkins TT, Cohen CJ, Beddoe AM, Dottino P, Martignetti JA, Applying Precision Medicine to Ovarian Cancer, Int J Gynecol Cancer. 28 (2018) 479–485. 10.1097/IGC.0000000000001190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Govindarajan M, Wohlmuth C, Waas M, Bernardini MQ, Kislinger T, High-throughput approaches for precision medicine in high-grade serous ovarian cancer, J Hematol Oncol. 13 (2020) 134. 10.1186/s13045-020-00971-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Cancer N Genome Atlas Research, Integrated genomic analyses of ovarian carcinoma, Nature. 474 (2011) 609–615. 10.1038/nature10166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Zhang Y, McNerny EG, Terajima M, Raghavan M, Romanowicz G, Zhang Z, Zhang H, Kamiya N, Tantillo M, Zhu P, Scott GJ, Ray MK, Lynch M, Ma PX, Morris MD, Yamauchi M, Kohn DH, Mishina Y, Loss of BMP signaling through BMPR1A in osteoblasts leads to greater collagen cross-link maturation and material-level mechanical properties in mouse femoral trabecular compartments, Bone. 88 (2016) 74–84. 10.1016/j.bone.2016.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Verhaak RGW, Tamayo P, Yang J-Y, Hubbard D, Zhang H, Creighton CJ, Fereday S, Lawrence M, Carter SL, Mermel CH, Kostic AD, Etemadmoghadam D, Saksena G, Cibulskis K, Duraisamy S, Levanon K, Sougnez C, Tsherniak A, Gomez S, Onofrio R, Gabriel S, Chin L, Zhang N, Spellman PT, Zhang Y, Akbani R, Hoadley KA, Kahn A, Köbel M, Huntsman D, Soslow RA, Defazio A, Birrer MJ, Gray JW, Weinstein JN, Bowtell DD, Drapkin R, Mesirov JP, Getz G, Levine DA, Meyerson M, Prognostically relevant gene signatures of high-grade serous ovarian carcinoma, J Clin Invest. 123 (2013) 517–525. 10.1172/JCI65833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Konecny GE, Winterhoff B, Wang C, Gene-expression signatures in ovarian cancer: Promise and challenges for patient stratification, Gynecol Oncol. 141 (2016) 379–385. 10.1016/j.ygyno.2016.01.026. [DOI] [PubMed] [Google Scholar]
- [31].Ahmed N, Stenvers KL, Getting to know ovarian cancer ascites: opportunities for targeted therapy-based translational research, Front Oncol. 3 (2013) 256. 10.3389/fonc.2013.00256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Kipps E, Tan DSP, Kaye SB, Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research, Nat Rev Cancer. 13 (2013) 273–282. 10.1038/nrc3432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Lengyel E, Ovarian Cancer Development and Metastasis, The American Journal of Pathology. 177 (2010) 1053–1064. 10.2353/ajpath.2010.100105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Naora H, Montell DJ, Ovarian cancer metastasis: integrating insights from disparate model organisms, Nature Reviews. Cancer 5 (2005) 355–66. 10.1038/nrc1611. [DOI] [PubMed] [Google Scholar]
- [35].Roberts CM, Cardenas C, Tedja R, The Role of Intra-Tumoral Heterogeneity and Its Clinical Relevance in Epithelial Ovarian Cancer Recurrence and Metastasis, Cancers (Basel). 11 (2019). 10.3390/cancers11081083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Brooks MD, Burness ML, Wicha MS, Therapeutic Implications of Cellular Heterogeneity and Plasticity in Breast Cancer, Cell Stem Cell. 17 (2015) 260–271. 10.1016/j.stem.2015.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Yan Y, Leontovich AA, Gerdes MJ, Desai K, Dong J, Sood A, Santamaria-Pang A, Mansfield AS, Chadwick C, Zhang R, Nevala WK, Flotte TJ, Ginty F, Markovic SN, Understanding heterogeneous tumor microenvironment in metastatic melanoma, PLoS One. 14 (2019). 10.1371/journal.pone.0216485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Jiménez-Sánchez A, Cybulska P, Mager KL, Koplev S, Cast O, Couturier D-L, Memon D, Selenica P, Nikolovski I, Mazaheri Y, Bykov Y, Geyer FC, Macintyre G, Gavarró LM, Drews RM, Gill MB, Papanastasiou AD, Sosa RE, Soslow RA, Walther T, Shen R, Chi DS, Park KJ, Hollmann T, Reis-Filho JS, Markowetz F, Beltrao P, Vargas HA, Zamarin D, Brenton JD, Snyder A, Weigelt B, Sala E, Miller ML, Unraveling tumor–immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy, Nature Genetics. 52 (2020) 582–593. 10.1038/s41588-020-0630-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Kurman RJ, International Agency for Research on Cancer, World Health Organization, WHO classification of tumours of female reproductive organs, International Agency for Research on Cancer, Lyon, 2014. [Google Scholar]
- [40].Kurman RJ, Shih I-M, The Dualistic Model of Ovarian Carcinogenesis: Revisited, Revised, and Expanded, The American Journal of Pathology. 186 (2016) 733–747. 10.1016/j.ajpath.2015.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Kanchi KL, Johnson KJ, Lu C, McLellan MD, Leiserson MD, Wendl MC, Zhang Q, Koboldt DC, Xie M, Kandoth C, McMichael JF, Wyczalkowski MA, Larson DE, Schmidt HK, Miller CA, Fulton RS, Spellman PT, Mardis ER, Druley TE, Graubert TA, Goodfellow PJ, Raphael BJ, Wilson RK, Ding L, Integrated analysis of germline and somatic variants in ovarian cancer, Nat Commun. 5 (2014) 3156. 10.1038/ncomms4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Takaya H, Nakai H, Sakai K, Nishio K, Murakami K, Mandai M, Matsumura N, Intratumor heterogeneity and homologous recombination deficiency of high-grade serous ovarian cancer are associated with prognosis and molecular subtype and change in treatment course, Gynecol Oncol. 156 (2020) 415–422. 10.1016/j.ygyno.2019.11.013. [DOI] [PubMed] [Google Scholar]
- [43].Nowell PC, The clonal evolution of tumor cell populations, Science. 194 (1976) 23–28. 10.1126/science.959840. [DOI] [PubMed] [Google Scholar]
- [44].Guo M, Peng Y, Gao A, Du C, Herman JG, Epigenetic heterogeneity in cancer, Biomarker Research. 7 (2019) 23. 10.1186/s40364-019-0174-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A, Chiappinelli KB, Epigenetic therapy for ovarian cancer: promise and progress, Clinical Epigenetics. 11 (2019) 7. 10.1186/s13148-018-0602-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Muñoz P, Iliou MS, Esteller M, Epigenetic alterations involved in cancer stem cell reprogramming, Mol Oncol. 6 (2012) 620–636. 10.1016/j.molonc.2012.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Burgos-Ojeda D, Rueda BR, Buckanovich RJ, Ovarian cancer stem cell markers: Prognostic and therapeutic implications, Cancer Letters. 322 (2012) 1–7. http://dx.doi.org.proxy.lib.umich.edu/10.1016/j.canlet.2012.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Kreso A, Dick JE, Evolution of the cancer stem cell model, Cell Stem Cell. 14 (2014) 275–91. 10.1016/j.stem.2014.02.006. [DOI] [PubMed] [Google Scholar]
- [49].Magee JA, Piskounova E, Morrison SJ, Cancer Stem Cells: Impact, Heterogeneity, and Uncertainty, Cancer Cell. 21 (2012) 283–296. 10.1016/j.ccr.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Eun K, Ham SW, and Kim H, Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting, BMB Reports. 50 (2017) 117–125. 10.5483/BMBRep.2017.50.3.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Ahmed N, Escalona R, Leung D, Chan E, Kannourakis G, Tumour microenvironment and metabolic plasticity in cancer and cancer stem cells: Perspectives on metabolic and immune regulatory signatures in chemoresistant ovarian cancer stem cells, Seminars in Cancer Biology. 53 (2018) 265–281. 10.1016/j.semcancer.2018.10.002. [DOI] [PubMed] [Google Scholar]
- [52].Korkaya H, Wicha MS, Cancer stem cells: nature versus nurture, Nat Cell Biol. 12 (2010) 419–421. 10.1038/ncb0510-419. [DOI] [PubMed] [Google Scholar]
- [53].Wang T, Shigdar S, Gantier MP, Hou Y, Wang L, Li Y, Shamaileh HA, Yin W, Zhou SF, Zhao X, Duan W, Cancer stem cell targeted therapy: progress amid controversies, Oncotarget. 6 (2015) 44191–206. 10.18632/oncotarget.6176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Boesch M, Zeimet AG, Reimer D, Schmidt S, Gastl G, Parson W, Spoeck F, Hatina J, Wolf D, Sopper S, The side population of ovarian cancer cells defines a heterogeneous compartment exhibiting stem cell characteristics, Oncotarget. 5 (2014) 7027–7039. 10.18632/oncotarget.2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Batlle E, Clevers H, Cancer stem cells revisited, Nat Med. 23 (2017) 1124–1134. 10.1038/nm.4409. [DOI] [PubMed] [Google Scholar]
- [56].Bonnet D, Dick JE, Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell, Nat Med. 3 (1997) 730–737. 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
- [57].Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF, Prospective identification of tumorigenic breast cancer cells, Proc Natl Acad Sci U S A. 100 (2003) 3983–3988. 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Zhang S, Balch C, Chan MW, Lai H-C, Matei D, Schilder JM, Yan PS, Huang TH-M, Nephew KP, Identification and characterization of ovarian cancer-initiating cells from primary human tumors, Cancer Res. 68 (2008) 4311–4320. 10.1158/0008-5472.CAN-08-0364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].O’Brien CA, Pollett A, Gallinger S, Dick JE, A human colon cancer cell capable of initiating tumour growth in immunodeficient mice, Nature. 445 (2007) 106–110. 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
- [60].Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R, Identification and expansion of human colon-cancer-initiating cells, Nature. 445 (2007) 111–115. 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
- [61].Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ, Prospective identification of tumorigenic prostate cancer stem cells, Cancer Res. 65 (2005) 10946–10951. 10.1158/0008-5472.CAN-05-2018. [DOI] [PubMed] [Google Scholar]
- [62].Patrawala L, Calhoun T, Schneider-Broussard R, Li H, Bhatia B, Tang S, Reilly JG, Chandra D, Zhou J, Claypool K, Coghlan L, Tang DG, Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells, Oncogene. 25 (2006) 1696–1708. 10.1038/sj.onc.1209327. [DOI] [PubMed] [Google Scholar]
- [63].Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A, Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma, Cancer Res. 64 (2004) 7011–7021. 10.1158/0008-5472.CAN-04-1364. [DOI] [PubMed] [Google Scholar]
- [64].Singh SK, Clarke ID, Hide T, Dirks PB, Cancer stem cells in nervous system tumors, Oncogene. 23 (2004) 7267–7273. 10.1038/sj.onc.1207946. [DOI] [PubMed] [Google Scholar]
- [65].Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB, Identification of human brain tumour initiating cells, Nature. 432 (2004) 396–401. 10.1038/nature03128. [DOI] [PubMed] [Google Scholar]
- [66].Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM, Identification of pancreatic cancer stem cells, Cancer Res. 67 (2007) 1030–1037. 10.1158/0008-5472.CAN-06-2030. [DOI] [PubMed] [Google Scholar]
- [67].Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH, Identification of cells initiating human melanomas, Nature. 451 (2008) 345–349. 10.1038/nature06489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I, McNiece I, Lin L, Ambinder RF, Peacock C, Watkins DN, Huff CA, Jones RJ, Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance, Cancer Res. 68 (2008) 190–197. 10.1158/0008-5472.CAN-07-3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Eramo A, Lotti F, Sette G, Pilozzi E, Biffoni M, Di Virgilio A, Conticello C, Ruco L, Peschle C, De Maria R, Identification and expansion of the tumorigenic lung cancer stem cell population, Cell Death Differ. 15 (2008) 504–514. 10.1038/sj.cdd.4402283. [DOI] [PubMed] [Google Scholar]
- [70].Dzobo K, Senthebane DA, Ganz C, Thomford NE, Wonkam A, Dandara C, Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review, Cells. 9 (2020). 10.3390/cells9081896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Dzobo K, Ganz C, Thomford NE, Senthebane DA, Cancer Stem Cell Markers in Relation to Patient Survival Outcomes: Lessons for Integrative Diagnostics and Next-Generation Anticancer Drug Development, OMICS. 25 (2021) 81–92. 10.1089/omi.2020.0185. [DOI] [PubMed] [Google Scholar]
- [72].Silva IA, Bai S, McLean K, Yang K, Griffith K, Thomas D, Ginestier C, Johnston C, Kueck A, Reynolds RK, Wicha MS, Buckanovich RJ, Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival, Cancer Research. 71 (2011) 3991–4001. 10.1158/0008-5472.CAN-10-3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Landen CN, Goodman B, Katre AA, Steg AD, Nick AM, Stone RL, Miller LD, Mejia PV, Jennings NB, Gershenson DM, Bast RC, Coleman RL, Lopez-Berestein G, Sood AK, Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer, Mol Cancer Ther. 9 (2010) 3186–99. 10.1158/1535-7163.MCT-10-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Zhang J, Guo X, Chang DY, Rosen DG, Mercado-Uribe I, Liu J, CD133 expression associated with poor prognosis in ovarian cancer, Mod Pathol. 25 (2012) 456–64. 10.1038/modpathol.2011.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Choi Y-J, Ingram PN, Yang K, Coffman L, Iyengar M, Bai S, Thomas DG, Yoon E, Buckanovich RJ, Identifying an ovarian cancer cell hierarchy regulated by bone morphogenetic protein 2, Proc. Natl. Acad. Sci. U.S.A 112 (2015) E6882–6888. 10.1073/pnas.1507899112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Curley MD, Therrien VA, Cummings CL, Sergent PA, Koulouris CR, Friel AM, Roberts DJ, Seiden MV, Scadden DT, Rueda BR, Foster R, CD133 expression defines a tumor initiating cell population in primary human ovarian cancer, Stem Cells. 27 (2009) 2875–83. 10.1002/stem.236. [DOI] [PubMed] [Google Scholar]
- [77].Sharrow AC, Perkins B, Collector MI, Yu W, Simons BW, Jones RJ, Characterization of aldehyde dehydrogenase 1 high ovarian cancer cells: Towards targeted stem cell therapy, Gynecol Oncol. 142 (2016) 341–348. 10.1016/j.ygyno.2016.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Zhou Q, Chen A, Song H, Tao J, Yang H, Zuo M, Prognostic value of cancer stem cell marker CD133 in ovarian cancer: a meta-analysis, Int J Clin Exp Med. 8 (2015) 3080–8. [PMC free article] [PubMed] [Google Scholar]
- [79].Baba T, Convery PA, Matsumura N, Whitaker RS, Kondoh E, Perry T, Huang Z, Bentley RC, Mori S, Fujii S, Marks JR, Berchuck A, Murphy SK, Epigenetic regulation of CD133 and tumorigenicity of CD133+ ovarian cancer cells, Oncogene. 28 (2009) 209–218. 10.1038/onc.2008.374. [DOI] [PubMed] [Google Scholar]
- [80].Kryczek I, Liu S, Roh M, Vatan L, Szeliga W, Wei S, Banerjee M, Mao Y, Kotarski J, Wicha MS, Liu R, Zou W, Expression of aldehyde dehydrogenase and CD133 defines ovarian cancer stem cells, Int J Cancer. 130 (2012) 29–39. 10.1002/ijc.25967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Murphy SK, Targeting ovarian cancer-initiating cells, Anticancer Agents Med Chem. 10 (2010) 157–163. 10.2174/187152010790909272. [DOI] [PubMed] [Google Scholar]
- [82].Steg AD, Bevis KS, Katre AA, Ziebarth A, Alvarez RD, Zhang K, Conner M, Landen CN, Stem cell pathways contribute to clinical chemoresistance in ovarian cancer, Clin Cancer Res. 18 (2012) 869–881. 10.1158/1078-0432.CCR-11-2188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Charafe-Jauffret E, Ginestier C, Iovino F, Tarpin C, Diebel M, Esterni B, Houvenaeghel G, Extra JM, Bertucci F, Jacquemier J, Xerri L, Dontu G, Stassi G, Xiao Y, Barsky SH, Birnbaum D, Viens P, Wicha MS, Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer, Clin Cancer Res. 16 (2010) 45–55. 10.1158/1078-0432.CCR-09-1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Condello S, Morgan CA, Nagdas S, Cao L, Turek J, Hurley TD, Matei D, beta-Catenin-regulated ALDH1A1 is a target in ovarian cancer spheroids, Oncogene. 34 (2015) 2297–308. 10.1038/onc.2014.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Alison MR, Lin W-R, Lim SML, Nicholson LJ, Cancer stem cells: In the line of fire, Cancer Treatment Reviews. 38 (2012) 589–598. 10.1016/j.ctrv.2012.03.003. [DOI] [PubMed] [Google Scholar]
- [86].Singh A, Settleman J, EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer, Oncogene. 29 (2010) 4741–51. http://dx.doi.org.proxy.lib.umich.edu/10.1038/onc.2010.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Keyvani V, Farshchian M, Esmaeili S-A, Yari H, Moghbeli M, Nezhad S-RK, Abbaszadegan MR, Ovarian cancer stem cells and targeted therapy, Journal of Ovarian Research. 12 (2019) 120. 10.1186/s13048-019-0588-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Hatina J, Boesch M, Sopper S, Kripnerova M, Wolf D, Reimer D, Marth C, Zeimet AG, Ovarian Cancer Stem Cell Heterogeneity, in: Birbrair A (Ed.), Stem Cells Heterogeneity in Cancer, Springer International Publishing, Cham, 2019: pp. 201–221. 10.1007/978-3-030-14366-4_12. [DOI] [PubMed] [Google Scholar]
- [89].Pieterse Z, Amaya-Padilla MA, Singomat T, Binju M, Madjid BD, Yu Y, Kaur P, Ovarian cancer stem cells and their role in drug resistance, Int J Biochem Cell Biol. 106 (2019) 117–126. 10.1016/j.biocel.2018.11.012. [DOI] [PubMed] [Google Scholar]
- [90].Dzobo K, Dandara C, Broadening Drug Design and Targets to Tumor Microenvironment? Cancer-Associated Fibroblast Marker Expression in Cancers and Relevance for Survival Outcomes, OMICS. 24 (2020) 340–351. 10.1089/omi.2020.0042. [DOI] [PubMed] [Google Scholar]
- [91].Dasari S, Fang Y, Mitra AK, Cancer Associated Fibroblasts: Naughty Neighbors That Drive Ovarian Cancer Progression, Cancers (Basel). 10 (2018). 10.3390/cancers10110406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Pearce OMT, Delaine-Smith RM, Maniati E, Nichols S, Wang J, Böhm S, Rajeeve V, Ullah D, Chakravarty P, Jones RR, Montfort A, Dowe T, Gribben J, Jones JL, Kocher HM, Serody JS, Vincent BG, Connelly J, Brenton JD, Chelala C, Cutillas PR, Lockley M, Bessant C, Knight MM, Balkwill FR, Deconstruction of a Metastatic Tumor Microenvironment Reveals a Common Matrix Response in Human Cancers, Cancer Discov. 8 (2018) 304–319. 10.1158/2159-8290.CD-17-0284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Izar B, Tirosh I, Stover EH, Wakiro I, Cuoco MS, Alter I, Rodman C, Leeson R, Su M-J, Shah P, Iwanicki M, Walker SR, Kanodia A, Melms JC, Mei S, Lin J-R, Porter CBM, Slyper M, Waldman J, Jerby-Arnon L, Ashenberg O, Brinker TJ, Mills C, Rogava M, Vigneau S, Sorger PK, Garraway LA, Konstantinopoulos PA, Liu JF, Matulonis U, Johnson BE, Rozenblatt-Rosen O, Rotem A, Regev A, A single-cell landscape of high-grade serous ovarian cancer, Nat Med. 26 (2020) 1271–1279. 10.1038/s41591-020-0926-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Shih AJ, Menzin A, Whyte J, Lovecchio J, Liew A, Khalili H, Bhuiya T, Gregersen PK, Lee AT, Identification of grade and origin specific cell populations in serous epithelial ovarian cancer by single cell RNA-seq, PLoS One. 13 (2018) e0206785. 10.1371/journal.pone.0206785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Coffman LG, Pearson AT, Frisbie LG, Freeman Z, Christie E, Bowtell DD, Buckanovich RJ, Ovarian Carcinoma-Associated Mesenchymal Stem Cells Arise from Tissue-Specific Normal Stroma, Stem Cells. 37 (2019) 257–269. 10.1002/stem.2932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Verardo R, Piazza S, Klaric E, Ciani Y, Bussadori G, Marzinotto S, Mariuzzi L, Cesselli D, Beltrami AP, Mano M, Itoh M, Kawaji H, Lassmann T, Carninci P, Hayashizaki Y, Forrest ARR, Fantom Consortium, Beltrami CA, Schneider C, Specific mesothelial signature marks the heterogeneity of mesenchymal stem cells from high-grade serous ovarian cancer, Stem Cells. 32 (2014) 2998–3011. 10.1002/stem.1791. [DOI] [PubMed] [Google Scholar]
- [97].Chan T-S, Shaked Y, Tsai KK, Targeting the Interplay Between Cancer Fibroblasts, Mesenchymal Stem Cells, and Cancer Stem Cells in Desmoplastic Cancers, Frontiers in Oncology. 9 (2019). 10.3389/fonc.2019.00688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].McLean K, Gong Y, Choi Y, Deng N, Yang K, Bai S, Cabrera L, Keller E, McCauley L, Cho KR, Buckanovich RJ, Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production, J. Clin. Invest 121 (2011) 3206–3219. 10.1172/JCI45273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Lis R, Touboul C, Raynaud CM, Malek JA, Suhre K, Mirshahi M, Rafii A, Mesenchymal cell interaction with ovarian cancer cells triggers pro-metastatic properties, PLoS One. 7 (2012) e38340. 10.1371/journal.pone.0038340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Klopp AH, Gupta A, Spaeth E, Andreeff M, Marini F, Concise review: Dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth?, Stem Cells. 29 (2011) 11–19. 10.1002/stem.559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Sun T, Sun B, Ni C, Zhao X, Wang X, Qie S, Zhang D, Gu Q, Qi H, Zhao N, Pilot study on the interaction between B16 melanoma cell-line and bone-marrow derived mesenchymal stem cells, Cancer Lett. 263 (2008) 35–43. 10.1016/j.canlet.2007.12.015. [DOI] [PubMed] [Google Scholar]
- [102].Beckermann BM, Kallifatidis G, Groth A, Frommhold D, Apel A, Mattern J, Salnikov AV, Moldenhauer G, Wagner W, Diehlmann A, Saffrich R, Schubert M, Ho AD, Giese N, Büchler MW, Friess H, Büchler P, Herr I, VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma, Br J Cancer. 99 (2008) 622–631. 10.1038/sj.bjc.6604508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, Ganesan S, Glod JW, Banerjee D, Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells, Cancer Res. 68 (2008) 4331–4339. 10.1158/0008-5472.CAN-08-0943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Zhu W, Xu W, Jiang R, Qian H, Chen M, Hu J, Cao W, Han C, Chen Y, Mesenchymal stem cells derived from bone marrow favor tumor cell growth in vivo, Exp Mol Pathol. 80 (2006) 267–274. 10.1016/j.yexmp.2005.07.004. [DOI] [PubMed] [Google Scholar]
- [105].Coffman LG, Choi Y-J, McLean K, Allen BL, di Magliano MP, Buckanovich RJ, Human carcinoma-associated mesenchymal stem cells promote ovarian cancer chemotherapy resistance via a BMP4/HH signaling loop, Oncotarget. 7 (2016) 6916–6932. 10.18632/oncotarget.6870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Wang X, Zhu Q, Lin Y, Wu L, Wu X, Wang K, He Q, Xu C, Wan X, Wang X, Crosstalk between TEMs and endothelial cells modulates angiogenesis and metastasis via IGF1-IGF1R signalling in epithelial ovarian cancer, Br J Cancer. 117 (2017) 1371–1382. 10.1038/bjc.2017.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Winiarski BK, Wolanska KI, Rai S, Ahmed T, Acheson N, Gutowski NJ, Whatmore JL, Epithelial ovarian cancer-induced angiogenic phenotype of human omental microvascular endothelial cells may occur independently of VEGF signaling, Transl Oncol. 6 (2013) 703–714. 10.1593/tlo.13529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Thibault B, Castells M, Delord J-P, Couderc B, Ovarian cancer microenvironment: implications for cancer dissemination and chemoresistance acquisition, Cancer Metastasis Rev. 33 (2014) 17–39. 10.1007/s10555-013-9456-2. [DOI] [PubMed] [Google Scholar]
- [109].Prieto-Vila M, Takahashi R-U, Usuba W, Kohama I, Ochiya T, Drug Resistance Driven by Cancer Stem Cells and Their Niche, Int J Mol Sci. 18 (2017). 10.3390/ijms18122574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Varas-Godoy M, Rice G, Illanes SE, The Crosstalk between Ovarian Cancer Stem Cell Niche and the Tumor Microenvironment, Stem Cells Int. 2017 (2017) 5263974. 10.1155/2017/5263974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Xiang T, Long H, He L, Han X, Lin K, Liang Z, Zhuo W, Xie R, Zhu B, Interleukin-17 produced by tumor microenvironment promotes self-renewal of CD133+ cancer stem-like cells in ovarian cancer, Oncogene. 34 (2015) 165–176. 10.1038/onc.2013.537. [DOI] [PubMed] [Google Scholar]
- [112].Qin J, Liu Y, Lu Y, Liu M, Li M, Li J, Wu L, Hypoxia-inducible factor 1 alpha promotes cancer stem cells-like properties in human ovarian cancer cells by upregulating SIRT1 expression, Scientific Reports. 7 (2017) 10592. 10.1038/s41598-017-09244-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, Nedaeinia R, Haghjooy Javanmard S, Taherian M, Ahmadlou M, Salehi R, Sadeghi B, Manian M, The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment, Cancer Cell International. 21 (2021) 62. 10.1186/s12935-020-01719-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Nagy JA, Herzberg KT, Dvorak JM, Dvorak HF, Pathogenesis of Malignant Ascites Formation: Initiating Events That Lead to Fluid Accumulation, Cancer Res. 53 (1993) 2631–2643. [PubMed] [Google Scholar]
- [115].Kim S, Kim B, Song YS, Ascites modulates cancer cell behavior, contributing to tumor heterogeneity in ovarian cancer, Cancer Sci. 107 (2016) 1173–1178. 10.1111/cas.12987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Kan T, Wang W, Ip PP, Zhou S, Wong AS, Wang X, Yang M, Single-cell EMT-related transcriptional analysis revealed intra-cluster heterogeneity of tumor cell clusters in epithelial ovarian cancer ascites, Oncogene. 39 (2020) 4227–4240. 10.1038/s41388-020-1288-2. [DOI] [PubMed] [Google Scholar]
- [117].Gao Q, Yang Z, Xu S, Li X, Yang X, Jin P, Liu Y, Zhou X, Zhang T, Gong C, Wei X, Liu D, Sun C, Chen G, Hu J, Meng L, Zhou J, Sawada K, Fruscio R, Grunt TW, Wischhusen J, Vargas-Hernández VM, Pothuri B, Coleman RL, Heterotypic CAF-tumor spheroids promote early peritoneal metastatis of ovarian cancer, J Exp Med. 216 (2019) 688–703. 10.1084/jem.20180765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Hashimoto K, Honda K, Matsui H, Nagashima Y, Oda H, Flow Cytometric Analysis of Ovarian Cancer Ascites: Response of Mesothelial Cells and Macrophages to Cancer, Anticancer Res. 36 (2016) 3579–3584. [PubMed] [Google Scholar]
- [119].Kenda Suster N, Virant-Klun I, Presence and role of stem cells in ovarian cancer, World J Stem Cells. 11 (2019) 383–397. 10.4252/wjsc.v11.i7.383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Han Q, Huang B, Huang Z, Cai J, Gong L, Zhang Y, Jiang J, Dong W, Wang Z, Tumor cell-fibroblast heterotypic aggregates in malignant ascites of patients with ovarian cancer, Int J Mol Med. (2019). 10.3892/ijmm.2019.4361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Al Habyan S, Kalos C, Szymborski J, McCaffrey L, Multicellular detachment generates metastatic spheroids during intra-abdominal dissemination in epithelial ovarian cancer, Oncogene. 37 (2018) 5127–5135. 10.1038/s41388-018-0317-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Hussain A, Voisin V, Poon S, Karamboulas C, Bui NHB, Meens J, Dmytryshyn J, Ho VW, Tang KH, Paterson J, Clarke BA, Bernardini MQ, Bader GD, Neel BG, Ailles LE, Distinct fibroblast functional states drive clinical outcomes in ovarian cancer and are regulated by TCF21, J Exp Med. 217 (2020). 10.1084/jem.20191094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Yin M, Shen J, Yu S, Fei J, Zhu X, Zhao J, Zhai L, Sadhukhan A, Zhou J, Tumor-Associated Macrophages (TAMs): A Critical Activator In Ovarian Cancer Metastasis, Onco Targets Ther. 12 (2019) 8687–8699. 10.2147/OTT.S216355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Zhou K, Cheng T, Zhan J, Peng X, Zhang Y, Wen J, Chen X, Ying M, Targeting tumor-associated macrophages in the tumor microenvironment, Oncol Lett. 20 (2020) 234. 10.3892/ol.2020.12097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Nowak M, Klink M, The Role of Tumor-Associated Macrophages in the Progression and Chemoresistance of Ovarian Cancer, Cells. 9 (2020). 10.3390/cells9051299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Raghavan S, Mehta P, Xie Y, Lei YL, Mehta G, Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments, Journal for ImmunoTherapy of Cancer. 7 (2019) 190. 10.1186/s40425-019-0666-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Zhang M, He Y, Sun X, Li Q, Wang W, Zhao A, Di W, A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients, Journal of Ovarian Research. 7 (2014) 19. 10.1186/1757-2215-7-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Cai DL, Jin L-P, Immune Cell Population in Ovarian Tumor Microenvironment, J Cancer. 8 (2017) 2915–2923. 10.7150/jca.20314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Bellora F, Castriconi R, Dondero A, Pessino A, Nencioni A, Liggieri G, Moretta L, Mantovani A, Moretta A, Bottino C, TLR activation of tumor-associated macrophages from ovarian cancer patients triggers cytolytic activity of NK cells, Eur J Immunol. 44 (2014) 1814–1822. 10.1002/eji.201344130. [DOI] [PubMed] [Google Scholar]
- [130].Dötzer K, Schlüter F, Schoenberg MB, Bazhin AV, Edler von Koch F, Schnelzer A, Anthuber S, Grab D, Czogalla B, Burges A, Werner J, Mahner S, Mayer B, Immune Heterogeneity Between Primary Tumors and Corresponding Metastatic Lesions and Response to Platinum Therapy in Primary Ovarian Cancer, Cancers. 11 (2019) 1250. 10.3390/cancers11091250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Jiménez-Sánchez A, Memon D, Pourpe S, Veeraraghavan H, Li Y, Vargas HA, Gill MB, Park KJ, Zivanovic O, Konner J, Ricca J, Zamarin D, Walther T, Aghajanian C, Wolchok JD, Sala E, Merghoub T, Snyder A, Miller ML, Heterogeneous Tumor-Immune Microenvironments among Differentially Growing Metastases in an Ovarian Cancer Patient, Cell. 170 (2017) 927–938.e20. 10.1016/j.cell.2017.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Fridman WH, Zitvogel L, Sautès–Fridman C, Kroemer G, The immune contexture in cancer prognosis and treatment, Nat Rev Clin Oncol. 14 (2017) 717–734. 10.1038/nrclinonc.2017.101. [DOI] [PubMed] [Google Scholar]
- [133].Petralia F, Tignor N, Reva B, Koptyra M, Chowdhury S, Rykunov D, Krek A, Ma W, Zhu Y, Ji J, Calinawan A, Whiteaker JR, Colaprico A, Stathias V, Omelchenko T, Song X, Raman P, Guo Y, Brown MA, Ivey RG, Szpyt J, Guha Thakurta S, Gritsenko MA, Weitz KK, Lopez G, Kalayci S, Gümüş ZH, Yoo S, da Veiga Leprevost F, Chang H-Y, Krug K, Katsnelson L, Wang Y, Kennedy JJ, Voytovich UJ, Zhao L, Gaonkar KS, Ennis BM, Zhang B, Baubet V, Tauhid L, Lilly JV, Mason JL, Farrow B, Young N, Leary S, Moon J, Petyuk VA, Nazarian J, Adappa ND, Palmer JN, Lober RM, Rivero-Hinojosa S, Wang L-B, Wang JM, Broberg M, Chu RK, Moore RJ, Monroe ME, Zhao R, Smith RD, Zhu J, Robles AI, Mesri M, Boja E, Hiltke T, Rodriguez H, Zhang B, Schadt EE, Mani DR, Ding L, Iavarone A, Wiznerowicz M, Schürer S, Chen XS, Heath AP, Rokita JL, Nesvizhskii AI, Fenyö D, Rodland KD, Liu T, Gygi SP, Paulovich AG, Resnick AC, Storm PB, Rood BR, Wang P, Francis A, Morgan AM, Waanders AJ, Viaene AN, Buccoliero AM, Chinnaiyan AM, Leonard CA, Kline CN, Caporalini C, Kinsinger CR, Li C, Kram DE, Hanson D, Appert E, Kawaler EA, Raabe EH, Jackson EM, Greenfield JP, Stone GS, Getz G, Grant G, Teo GC, Pollack IF, Cain JE, Foster JB, Phillips JJ, Palma JE, Ketchum KA, Ruggles KV, Blumenberg L, Cornwell M, Sarmady M, Domagalski MJ, Cieślik MP, Santi M, Li MM, Ellis MJ, Wyczalkowski MA, Connors M, Scagnet M, Gupta N, Edwards NJ, Vitanza NA, Vaske OM, Becher O, McGarvey PB, Firestein R, Mueller S, Winebrake SG, Dhanasekaran SM, Cai S, Partap S, Patton T, Le T, Lorentzen TD, Liu W, Bocik WE, Integrated Proteogenomic Characterization across Major Histological Types of Pediatric Brain Cancer, Cell. 183 (2020) 1962–1985.e31. 10.1016/j.cell.2020.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Li J, Wang J, Chen R, Bai Y, Lu X, The prognostic value of tumor-infiltrating T lymphocytes in ovarian cancer, Oncotarget. 8 (2017) 15621–15631. 10.18632/oncotarget.14919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Ovarian Tumor Tissue Analysis (OTTA) Consortium, Goode EL, Block MS, Kalli KR, Vierkant RA, Chen W, Fogarty ZC, Gentry-Maharaj A, Tołoczko A, Hein A, Bouligny AL, Jensen A, Osorio A, Hartkopf A, Ryan A, Chudecka-Głaz A, Magliocco AM, Hartmann A, Jung AY, Gao B, Hernandez BY, Fridley BL, McCauley BM, Kennedy CJ, Wang C, Karpinskyj C, de Sousa CB, Tiezzi DG, Wachter DL, Herpel E, Taran FA, Modugno F, Nelson G, Lubiński J, Menkiszak J, Alsop J, Lester J, García-Donas J, Nation J, Hung J, Palacios J, Rothstein JH, Kelley JL, de Andrade JM, Robles-Díaz L, Intermaggio MP, Widschwendter M, Beckmann MW, Ruebner M, Jimenez-Linan M, Singh N, Oszurek O, Harnett PR, Rambau PF, Sinn P, Wagner P, Ghatage P, Sharma R, Edwards RP, Ness RB, Orsulic S, Brucker SY, Johnatty SE, Longacre TA, Ursula E, McGuire V, Sieh W, Natanzon Y, Li Z, Whittemore AS, deFazio Anna, Staebler A, Karlan BY, Gilks B, Bowtell DD, Høgdall E, Candido dos Reis FJ, Steed H, Campbell IG, Gronwald J, Benítez J, Koziak JM, Chang-Claude J, Moysich KB, Kelemen LE, Cook LS, Goodman MT, García MJ, Fasching PA, Kommoss S, Deen S, Kjaer SK, Menon U, Brenton JD, Pharoah PD, Chenevix-Trench G, Huntsman DG, Winham SJ, Köbel M, Ramus SJ, Dose-Response Association of CD8+ Tumor-Infiltrating Lymphocytes and Survival Time in High-Grade Serous Ovarian Cancer, JAMA Oncol. 3 (2017) e173290. 10.1001/jamaoncol.2017.3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Patsoukis N, Wang Q, Strauss L, Boussiotis VA, Revisiting the PD-1 pathway, Science Advances. 6 (2020) eabd2712. 10.1126/sciadv.abd2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Gonzalez H, Hagerling C, Werb Z, Roles of the immune system in cancer: from tumor initiation to metastatic progression, Genes Dev. 32 (2018) 1267–1284. 10.1101/gad.314617.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Carroll MJ, Fogg KC, Patel HA, Krause HB, Mancha A-S, Patankar MS, Weisman PS, Barroilhet L, Kreeger PK, Alternatively-Activated Macrophages Upregulate Mesothelial Expression of P-Selectin to Enhance Adhesion of Ovarian Cancer Cells, Cancer Res. 78 (2018) 3560–3573. 10.1158/0008-5472.CAN-17-3341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Matte I, Lane D, Laplante C, Garde Granger P, Rancourt C, Piché A, Ovarian cancer ascites enhance the migration of patient-derived peritoneal mesothelial cells via cMet pathway through HGF-dependent and -independent mechanisms, International Journal of Cancer. 137 (2015) 289–298. 10.1002/ijc.29385. [DOI] [PubMed] [Google Scholar]
- [140].Matte I, Lane D, Bachvarov D, Rancourt C, Piché A, Role of malignant ascites on human mesothelial cells and their gene expression profiles, BMC Cancer. 14 (2014) 288. 10.1186/1471-2407-14-288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Rynne-Vidal A, Au-Yeung CL, Jiménez-Heffernan JA, Pérez-Lozano ML, Cremades-Jimeno L, Bárcena C, Cristóbal-García I, Fernández-Chacón C, Yeung TL, Mok SC, Sandoval P, López-Cabrera M, Mesothelial-to-mesenchymal transition as a possible therapeutic target in peritoneal metastasis of ovarian cancer, J. Pathol 242 (2017) 140–151. 10.1002/path.4889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Yoshihara M, Kajiyama H, Yokoi A, Sugiyama M, Koya Y, Yamakita Y, Liu W, Nakamura K, Moriyama Y, Yasui H, Suzuki S, Yamamoto Y, Ricciardelli C, Nawa A, Shibata K, Kikkawa F, Ovarian cancer-associated mesothelial cells induce acquired platinum-resistance in peritoneal metastasis via the FN1/Akt signaling pathway, Int J Cancer. 146 (2020) 2268–2280. 10.1002/ijc.32854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Yang J, Zaman MM, Vlasakov I, Roy R, Huang L, Martin CR, Freedman SD, Serhan CN, Moses MA, Adipocytes promote ovarian cancer chemoresistance, Sci Rep. 9 (2019). 10.1038/s41598-019-49649-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].John B, Naczki C, Patel C, Ghoneum A, Qasem S, Salih Z, Said N, Regulation of the bi-directional cross-talk between ovarian cancer cells and adipocytes by SPARC, Oncogene. 38 (2019) 4366–4383. 10.1038/s41388-019-0728-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Heindl A, Lan C, Rodrigues DN, Koelble K, Yuan Y, Similarity and diversity of the tumor microenvironment in multiple metastases: critical implications for overall and progression-free survival of high-grade serous ovarian cancer, Oncotarget. 7 (2016) 71123–71135. 10.18632/oncotarget.12106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Motohara T, Masuda K, Morotti M, Zheng Y, El-Sahhar S, Chong KY, Wietek N, Alsaadi A, Karaminejadranjbar M, Hu Z, Artibani M, Gonzalez LS, Katabuchi H, Saya H, Ahmed AA, An evolving story of the metastatic voyage of ovarian cancer cells: cellular and molecular orchestration of the adipose-rich metastatic microenvironment, Oncogene. 38 (2019) 2885–2898. 10.1038/s41388-018-0637-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Wen Y, Guo Y, Huang Z, Cai J, Wang Z, Adipose-derived mesenchymal stem cells attenuate cisplatin-induced apoptosis in epithelial ovarian cancer cells, Molecular Medicine Reports. 16 (2017) 9587–9592. 10.3892/mmr.2017.7783. [DOI] [PubMed] [Google Scholar]
- [148].Zhang Y, Tang H, Cai J, Zhang T, Guo J, Feng D, Wang Z, Ovarian cancer-associated fibroblasts contribute to epithelial ovarian carcinoma metastasis by promoting angiogenesis, lymphangiogenesis and tumor cell invasion, Cancer Lett. 303 (2011) 47–55. 10.1016/j.canlet.2011.01.011. [DOI] [PubMed] [Google Scholar]
- [149].Ma X, The omentum, a niche for premetastatic ovarian cancer, Journal of Experimental Medicine. 217 (2020) e20192312. 10.1084/jem.20192312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Petrova V, Annicchiarico-Petruzzelli M, Melino G, Amelio I, The hypoxic tumour microenvironment, Oncogenesis. 7 (2018) 1–13. 10.1038/s41389-017-0011-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Paredes F, Williams HC, San Martin A, Metabolic adaptation in hypoxia and cancer, Cancer Letters. 502 (2021) 133–142. 10.1016/j.canlet.2020.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G, Integrated cancer tissue engineering models for precision medicine, PLoS ONE. 14 (2019) e0216564. 10.1371/journal.pone.0216564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].He M, Wu H, Jiang Q, Liu Y, Han L, Yan Y, Wei B, Liu F, Deng X, Chen H, Zhao L, Wang M, Wu X, Yao W, Zhao H, Chen J, Wei M, Hypoxia-inducible factor-2α directly promotes BCRP expression and mediates the resistance of ovarian cancer stem cells to adriamycin, Mol Oncol. 13 (2019) 403–421. 10.1002/1878-0261.12419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, De Donatis A, Lanciotti M, Serni S, Cirri P, Chiarugi P, Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay, Cancer Res. 72 (2012) 5130–5140. 10.1158/0008-5472.CAN-12-1949. [DOI] [PubMed] [Google Scholar]
- [155].Gilkes DM, Bajpai S, Chaturvedi P, Wirtz D, Semenza GL, Hypoxia-inducible Factor 1 (HIF-1) Promotes Extracellular Matrix Remodeling under Hypoxic Conditions by Inducing P4HA1, P4HA2, and PLOD2 Expression in Fibroblasts*, Journal of Biological Chemistry. 288 (2013) 10819–10829. 10.1074/jbc.M112.442939. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- [156].Henze A-T, Mazzone M, The impact of hypoxia on tumor-associated macrophages, J Clin Invest. 126 (n.d.) 3672–3679. 10.1172/JCI84427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Santi A, Kugeratski FG, Zanivan S, Cancer Associated Fibroblasts: The Architects of Stroma Remodeling, Proteomics. 18 (2018). 10.1002/pmic.201700167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Hanley CJ, Noble F, Ward M, Bullock M, Drifka C, Mellone M, Manousopoulou A, Johnston HE, Hayden A, Thirdborough S, Liu Y, Smith DM, Mellows T, Kao WJ, Garbis SD, Mirnezami A, Underwood TJ, Eliceiri KW, Thomas GJ, A subset of myofibroblastic cancer-associated fibroblasts regulate collagen fiber elongation, which is prognostic in multiple cancers, Oncotarget. 7 (2016) 6159–6174. 10.18632/oncotarget.6740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Finkernagel F, Reinartz S, Lieber S, Adhikary T, Wortmann A, Hoffmann N, Bieringer T, Nist A, Stiewe T, Jansen JM, Wagner U, Müller-Brüsselbach S, Müller R, The transcriptional signature of human ovarian carcinoma macrophages is associated with extracellular matrix reorganization, Oncotarget. 7 (2016) 75339–75352. 10.18632/oncotarget.12180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Dai L, Song K, Di W, Adipocytes: active facilitators in epithelial ovarian cancer progression?, Journal of Ovarian Research. 13 (2020) 115. 10.1186/s13048-020-00718-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [161].Ghosh D, Pena CM, Quach N, Xuan B, Lee AH, Dawson MR, Senescent mesenchymal stem cells remodel extracellular matrix driving breast cancer cells to a more-invasive phenotype, J Cell Sci. 133 (2020). 10.1242/jcs.232470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [162].Mitra AK, Ovarian Cancer Metastasis: A Unique Mechanism of Dissemination, Tumor Metastasis. (2016). 10.5772/64700. [DOI] [Google Scholar]
- [163].Kenny HA, Chiang C-Y, White EA, Schryver EM, Habis M, Romero IL, Ladanyi A, Penicka CV, George J, Matlin K, Montag A, Wroblewski K, Yamada SD, Mazar AP, Bowtell D, Lengyel E, Mesothelial cells promote early ovarian cancer metastasis through fibronectin secretion, J Clin Invest. 124 (2014) 4614–4628. 10.1172/JCI74778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Trovato R, Canè S, Petrova V, Sartoris S, Ugel S, De Sanctis F, The Engagement Between MDSCs and Metastases: Partners in Crime, Front Oncol. 10 (2020) 165. 10.3389/fonc.2020.00165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Baeriswyl V, Christofori G, The angiogenic switch in carcinogenesis, Semin Cancer Biol. 19 (2009) 329–337. 10.1016/j.semcancer.2009.05.003. [DOI] [PubMed] [Google Scholar]
- [166].Pires A, Greenshields-Watson A, Jones E, Smart K, Lauder SN, Somerville M, Milutinovic S, Kendrick H, Hindley JP, French R, Smalley MJ, Watkins WJ, Andrews R, Godkin A, Gallimore A, Immune Remodeling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection, Cancer Immunol Res. (2020). 10.1158/2326-6066.CIR-20-0070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Fogg KC, Renner CM, Christian H, Walker A, Marty-Santos L, Khan A, Olson WR, Parent C, O’Shea A, Wellik DM, Weisman PS, Kreeger PK, Ovarian Cells Have Increased Proliferation in Response to Heparin-Binding Epidermal Growth Factor as Collagen Density Increases, Tissue Eng Part A. 26 (2020) 747–758. 10.1089/ten.tea.2020.0001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Petersen OW, Rønnov-Jessen L, Weaver VM, Bissell MJ, Differentiation and cancer in the mammary gland: shedding light on an old dichotomy, Adv Cancer Res. 75 (1998) 135–161. 10.1016/s0065-230x(08)60741-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Acerbi I, Cassereau L, Dean I, Shi Q, Au A, Park C, Chen YY, Liphardt J, Hwang ES, Weaver VM, Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration, Integrative Biology. 7 (2015) 1120–1134. 10.1039/C5IB00040H. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Cho A, Howell VM, Colvin EK, The Extracellular Matrix in Epithelial Ovarian Cancer – A Piece of a Puzzle, Front Oncol. 5 (2015). 10.3389/fonc.2015.00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Kirkland SC, Type I collagen inhibits differentiation and promotes a stem cell-like phenotype in human colorectal carcinoma cells, British Journal of Cancer. 101 (2009) 320–326. 10.1038/sj.bjc.6605143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Motegi H, Kamoshima Y, Terasaka S, Kobayashi H, Houkin K, Type 1 collagen as a potential niche component for CD133-positive glioblastoma cells, Neuropathology. 34 (2014) 378–385. 10.1111/neup.12117. [DOI] [PubMed] [Google Scholar]
- [173].Lambert AW, Khuan Wong C, Ozturk S, Papageorgis P, Raghunathan R, Alekseyev Y, Gower AC, Reinhard orn M., Abdolmaleky HM, Thiagalingam S, Tumor Cell-Derived Periostin Regulates Cytokines That Maintain Breast Cancer Stem Cells, (2016). 10.1158/1541-7786.MCR-15-0079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Curry JM, Thompson KJ, Rao SG, Besmer DM, Murphy AM, Grdzelishvili VZ, Ahrens WA, McKillop IH, Sindram D, Iannitti DA, Martinie JB, Mukherjee P, The use of a novel MUC1 antibody to identify cancer stem cells and circulating MUC1 in mice and patients with pancreatic cancer, Journal of Surgical Oncology. 107 (2013) 713–722. 10.1002/jso.23316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Du WW, Fang L, Yang X, Sheng W, Yang BL, Seth A, Zhang Y, Yang BB, Yee AJ, The role of versican in modulating breast cancer cell self-renewal, Molecular Cancer Research. 11 (2013) 443–455. 10.1158/1541-7786.MCR-12-0461. [DOI] [PubMed] [Google Scholar]
- [176].Begum A, Ewachiw T, Jung C, Huang A, Norberg KJ, Marchionni L, Mcmillan R, Penchev V, Rajeshkumar NV, Maitra A, Wood L, Wang C, Wolfgang C, Dejesus-Acosta A, Laheru D, Shapiro IM, Padval M, Pachter JA, Weaver DT, Rasheed ZA, Matsui W, The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma, (2017). 10.1371/journal.pone.0180181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Nie S, Gurrea M, Zhu J, Thakolwiboon S, Heth JA, Muraszko KM, Fan X, Lubman DM, Tenascin-C: A Novel Candidate Marker for Cancer Stem Cells in Glioblastoma Identified by Tissue Microarrays, (2014). 10.1021/pr5008653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Fukunaga-Kalabis M, Martinez G, Nguyen TK, Kim D, Santiago-Walker A, Roesch A, Herlyn M, Tenascin-C promotes melanoma progression by maintaining the ABCB5-positive side population, Oncogene. 29 (2010) 6115–6124. 10.1038/onc.2010.350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Zhou W, Ke SQ, Huang Z, Flavahan W, Fang X, Paul J, Wu L, Sloan AE, McLendon RE, Li X, Rich JN, Bao S, Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth, Nature Cell Biology. 17 (2015) 170–182. 10.1038/ncb3090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Mikheev AM, Mikheeva SA, Trister AD, Tokita MJ, Emerson SN, Parada CA, Born DE, Carnemolla B, Frankel S, Kim D-H, Oxford RG, Kosai Y, Tozer-Fink KR, Manning TC, Silber JR, Rostomily RC, Periostin is a novel therapeutic target that predicts and regulates glioma malignancy, Neuro-Oncology. 17 (2015) 372–382. 10.1093/neuonc/nou161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Wang X, Liu J, Wang Z, Huang Y, Liu W, Zhu X, Cai Y, Fang X, Lin S, Yuan L, Ouyang G, Periostin Contributes to the Acquisition of Multipotent Stem Cell-Like Properties in Human Mammary Epithelial Cells and Breast Cancer Cells, PLoS ONE. 8 (2013) e72962–e72962. 10.1371/journal.pone.0072962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Stroopinsky D, Rosenblatt J, Ito K, Mills H, Yin L, Rajabi H, Vasir B, Kufe T, Luptakova K, Arnason J, Nardella C, Levine JD, Joyce R, Galinsky I, Reiter Y, Stone R, Pandolfi PP, Kufe D, Avigan D, MUC1 IS A POTENTIAL TARGET FOR THE TREATMENT OF ACUTE MYELOID LEUKEMIA STEM CELLS, (n.d.). 10.1158/0008-5472.CAN-13-0677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Engelmann K, Shen H, Finn OJ, MCF7 side population cells with characteristics of cancer stem/progenitor cells express the tumor antigen MUC1, Cancer Research. 68 (2008) 2419–2426. 10.1158/0008-5472.CAN-07-2249. [DOI] [PubMed] [Google Scholar]
- [184].Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF, Huelsken J, Interactions between cancer stem cells and their niche govern metastatic colonization, Nature. 481 (2012) 85–91. 10.1038/nature10694. [DOI] [PubMed] [Google Scholar]
- [185].Januchowski R, Świerczewska M, Sterzyńska K, Wojtowicz K, Nowicki M, Zabel M, Increased Expression of Several Collagen Genes is Associated with Drug Resistance in Ovarian Cancer Cell Lines, J Cancer. 7 (2016) 1295–1310. 10.7150/jca.15371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Okuda H, Kobayashi A, Xia B, Watabe M, Pai SK, Hirota S, Xing F, Liu W, Pandey PR, Fukuda K, Modur V, Ghosh A, Wilber A, Watabe K, Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem-like cells with macrophages and stromal cells, Cancer Research. 72 (2012) 537–547. 10.1158/0008-5472.CAN-11-1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Fang DD, Kim YJ, Lee CN, Aggarwal S, McKinnon K, Mesmer D, Norton J, Birse CE, He T, Ruben SM, Moore PA, Expansion of CD133+ colon cancer cultures retaining stem cell properties to enable cancer stem cell target discovery, British Journal of Cancer. 102 (2010) 1265–1275. 10.1038/sj.bjc.6605610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].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, Oncology Reports. 32 (2014) 641–649. 10.3892/or.2014.3252. [DOI] [PubMed] [Google Scholar]
- [189].Oskarsson T, Acharyya S, H-F Zhang X, Vanharanta S, Tavazoie SF, Morris PG, Downey RJ, Manova-Todorova K, Brogi E, Massagu J, Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs, Nature Medicine. (2011). 10.1038/nm.2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Liu B, Xu T, Xu X, Cui Y, Xing X, Biglycan promotes the chemotherapy resistance of colon cancer by activating NF-κB signal transduction, Molecular and Cellular Biochemistry. 449 (2018) 285–294. 10.1007/s11010-018-3365-1. [DOI] [PubMed] [Google Scholar]
- [191].Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, Andreeff M, Marini F, Mesenchymal Stem Cell Transition to Tumor-Associated Fibroblasts Contributes to Fibrovascular Network Expansion and Tumor Progression, PLoS ONE. 4 (2009) e4992–e4992. 10.1371/journal.pone.0004992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Lim E-J, Suh Y, Kim S, Kang S-G, Lee S-J, Force-mediated proinvasive matrix remodeling driven by tumor-associated mesenchymal stem-like cells in glioblastoma, BMB Rep. 51 (2018) 182–187. 10.5483/bmbrep.2018.51.4.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Cavarretta IT, Altanerova V, Matuskova M, Kucerova L, Culig Z, Altaner C, Adipose Tissue–derived Mesenchymal Stem Cells Expressing Prodrug-converting Enzyme Inhibit Human Prostate Tumor Growth, (2010). 10.1038/mt.2009.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Yeung T-L, Leung CS, Wong K-K, Samimi G, Thompson MS, Liu J, Zaid TM, Ghosh S, Birrer MJ, Mok SC, TGF-β modulates ovarian cancer invasion by upregulating CAF-derived versican in the tumor microenvironment, Cancer Res. 73 (2013) 5016–5028. 10.1158/0008-5472.CAN-13-0023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Li M, Wang J, Wang C, Xia L, Xu J, Xie X, Lu W, Microenvironment remodeled by tumor and stromal cells elevates fibroblast-derived COL1A1 and facilitates ovarian cancer metastasis, Exp Cell Res. 394 (2020) 112153. 10.1016/j.yexcr.2020.112153. [DOI] [PubMed] [Google Scholar]
- [196].Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, Sahai E, Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells, Nat Cell Biol. 9 (2007) 1392–1400. 10.1038/ncb1658. [DOI] [PubMed] [Google Scholar]
- [197].Hooper S, Gaggioli C, Sahai E, A chemical biology screen reveals a role for Rab21-mediated control of actomyosin contractility in fibroblast-driven cancer invasion, British Journal of Cancer. 102 (2010) 392–402. 10.1038/sj.bjc.6605469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [198].Albig AR, Roy TG, Becenti DJ, Schiemann WP, Transcriptome analysis of endothelial cell gene expression induced by growth on matrigel matrices: identification and characterization of MAGP-2 and lumican as novel regulators of angiogenesis, Angiogenesis. 10 (2007) 197–216. 10.1007/s10456-007-9075-z. [DOI] [PubMed] [Google Scholar]
- [199].Barry-Hamilton V, Spangler R, Marshall D, McCauley S, Rodriguez HM, Oyasu M, Mikels A, Vaysberg M, Ghermazien H, Wai C, Garcia CA, Velayo AC, Jorgensen B, Biermann D, Tsai D, Green J, Zaffryar-Eilot S, Holzer A, Ogg S, Thai D, Neufeld G, Van Vlasselaer P, Smith V, Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment, Nature Medicine. 16 (2010) 1009–1017. 10.1038/nm.2208. [DOI] [PubMed] [Google Scholar]
- [200].Hildenbrand R, Dilger I, Hörlin A, Stutte HJ, Urokinase and macrophages in tumour angiogenesis, British Journal of Cancer. 72 (1995) 818–823. 10.1038/bjc.1995.419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Solinas G, Schiarea S, Liguori M, Fabbri M, Pesce S, Zammataro L, Pasqualini F, Nebuloni M, Chiabrando C, Mantovani A, Allavena P, Tumor-Conditioned Macrophages Secrete Migration-Stimulating Factor: A New Marker for M2-Polarization, Influencing Tumor Cell Motility, The Journal of Immunology. 185 (2010) 642–652. 10.4049/jimmunol.1000413. [DOI] [PubMed] [Google Scholar]
- [202].Steitz AM, Steffes A, Finkernagel F, Unger A, Sommerfeld L, Jansen JM, Wagner U, Graumann J, Müller R, Reinartz S, Tumor-associated macrophages promote ovarian cancer cell migration by secreting transforming growth factor beta induced (TGFBI) and tenascin C, Cell Death & Disease. 11 (2020) 1–15. 10.1038/s41419-020-2438-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Murphy G, The ADAMs: Signalling scissors in the tumour microenvironment, Nature Reviews Cancer. 8 (2008) 929–941. 10.1038/nrc2459. [DOI] [PubMed] [Google Scholar]
- [204].Wai PY, Kuo PC, Osteopontin: regulation in tumor metastasis, (2007). 10.1007/s10555-007-9104-9. [DOI] [PubMed] [Google Scholar]
- [205].Arnold SA, Rivera LB, Miller AF, Carbon JG, Dineen SP, Xie Y, Castrillon DH, Sage EH, Puolakkainen P, Bradshaw AD, Brekken RA, Lack of host SPARC enhances vascular function and tumor spread in an orthotopic murine model of pancreatic carcinoma, DMM Disease Models and Mechanisms. 3 (2010) 57–72. 10.1242/dmm.003228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Agrawal S, Anderson P, Durbeej M, Van Rooijen N, Ivars F, Opdenakker G, Sorokin LM, Dystroglycan is selectively cleaved at the parenchymal basement membrane at sites of leukocyte extravasation in experimental autoimmune encephalomyelitis, Journal of Experimental Medicine. 203 (2006) 1007–1016. 10.1084/jem.20051342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Campo-Verde-Arbocco F, López-Laur JD, Romeo LR, Giorlando N, Bruna FA, Contador DE, López-Fontana G, Santiano FE, Sasso CV, Zyla LE, López-Fontana CM, Calvo JC, Carón RW, Creydt VP, Human renal adipose tissue induces the invasion and progression of renal cell carcinoma, 2017. www.impactjournals.com/oncotarget. [DOI] [PMC free article] [PubMed]
- [208].Ribeiro R, Monteiro C, Cunha V, Oliveira MJ, Freitas M, Fraga A, Príncipe P, Lobato C, Lobo F, Morais A, Silva V, Sanches-Magalhães J, Oliveira J, Pina F, Mota-Pinto A, Lopes C, Medeiros R, Human periprostatic adipose tissue promotes prostate cancer aggressiveness in vitro, 2012. 10.1186/1756-9966-31-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].Park J, Scherer PE, Adipocyte-derived endotrophin promotes malignant tumor progression, Journal of Clinical Investigation. 122 (2012) 4243–4256. 10.1172/JCI63930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [210].Iyengar P, Espina V, Williams TW, Lin Y, Berry D, Jelicks LA, Lee H, Temple K, Graves R, Pollard J, Chopra N, Russell RG, Sasisekharan R, Trock BJ, Lippman M, Calvert VS, Petricoin EF, Liotta L, Dadachova E, Pestell RG, Lisanti MP, Bonaldo P, Scherer PE, Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment, Journal of Clinical Investigation. 115 (2005) 1163–1176. 10.1172/JCI23424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [211].Natarajan S, Foreman KM, Soriano MI, Rossen NS, Shehade H, Fregoso DR, Eggold JT, Krishnan V, Dorigo O, Krieg AJ, Heilshorn SC, Sinha S, Fuh KC, Rankin EB, Collagen Remodeling in the Hypoxic Tumor-Mesothelial Niche Promotes Ovarian Cancer Metastasis, Cancer Res. 79 (2019) 2271–2284. 10.1158/0008-5472.CAN-18-2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [212].Pickup MW, Laklai H, Acerbi I, Owens P, Gorska AE, Chytil A, Aakre M, Weaver VM, Moses HL, Stromally derived lysyl oxidase promotes metastasis of transforming growth factor-β-deficient mouse mammary carcinomas, Cancer Research. 73 (2013) 5336–5346. 10.1158/0008-5472.CAN-13-0012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [213].Condamine T, Ramachandran I, Youn J-I, Gabrilovich DI, Regulation of Tumor Metastasis by Myeloid-Derived Suppressor Cells, Annual Review of Medicine. 66 (2015) 97–110. 10.1146/annurev-med-051013-052304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, Carbone DP, Matrisian LMM, Richmond A, Lin PC, Moses HL, Abrogation of TGFβ Signaling in Mammary Carcinomas Recruits Gr-1+CD11b+ Myeloid Cells that Promote Metastasis, Cancer Cell. 13 (2008) 23–35. 10.1016/j.ccr.2007.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Pahler JC, Tazzyman S, Erez N, Chen YY, Murdoch C, Nozawa H, Lewis CE, Hanahan D, Plasticity in tumor-promoting inflammation: Impairment of macrophage recruitment evokes a compensatory neutrophil response, Neoplasia. 10 (2008) 329–339. 10.1593/neo.07871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Nozawa H, Chiu C, Hanahan D, Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis, Proceedings of the National Academy of Sciences of the United States of America. 103 (2006) 12493–12498. 10.1073/pnas.0601807103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Mellor AL, Munn DH, IDO expression by dendritic cells: Tolerance and tryptophan catabolism, Nature Reviews Immunology. 4 (2004) 762–774. 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
- [218].De Visser KE, Korets LV, Coussens LM, De novo carcinogenesis promoted by chronic inflammation is B lymphocyte dependent, Cancer Cell. 7 (2005) 411–423. 10.1016/j.ccr.2005.04.014. [DOI] [PubMed] [Google Scholar]
- [219].Leong HS, Galletta L, Etemadmoghadam D, George J, Köbel M, Ramus SJ, Bowtell D, Efficient molecular subtype classification of high-grade serous ovarian cancer, J Pathol. 236 (2015) 272–7. 10.1002/path.4536. [DOI] [PubMed] [Google Scholar]
- [220].Lisio M-A, Fu L, Goyeneche A, Gao Z, Telleria C, High-Grade Serous Ovarian Cancer: Basic Sciences, Clinical and Therapeutic Standpoints, International Journal of Molecular Sciences. 20 (2019) 952. 10.3390/ijms20040952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].Hynes RO, Naba A, Overview of the Matrisome—An Inventory of Extracellular Matrix Constituents and Functions, Cold Spring Harb Perspect Biol. 4 (2012). 10.1101/cshperspect.a004903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Kalluri R, The biology and function of fibroblasts in cancer, Nat. Rev. Cancer 16 (2016) 582–598. 10.1038/nrc.2016.73. [DOI] [PubMed] [Google Scholar]
- [223].Zigrino P, Löffek S, Mauch C, Tumor–stroma interactions: their role in the control of tumor cell invasion, Biochimie. 87 (2005) 321–328. 10.1016/j.biochi.2004.10.025. [DOI] [PubMed] [Google Scholar]
- [224].Ahmed N, Riley C, Rice G, Quinn M, Role of integrin receptors for fibronectin, collagen and laminin in the regulation of ovarian carcinoma functions in response to a matrix microenvironment, Clin Exp Metastasis. 22 (2005) 391–402. 10.1007/s10585-005-1262-y. [DOI] [PubMed] [Google Scholar]
- [225].Chen F, Chandrashekar DS, Varambally S, Creighton CJ, Pan-cancer molecular subtypes revealed by mass-spectrometry-based proteomic characterization of more than 500 human cancers, Nature Communications. 10 (2019) 5679. 10.1038/s41467-019-13528-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Lu P, Weaver VM, Werb Z, The extracellular matrix: a dynamic niche in cancer progression, J Cell Biol. 196 (2012) 395–406. 10.1083/jcb.201102147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [227].Lind A-K, Weijdegård B, Dahm-Kähler P, Mölne J, Sundfeldt K, Brännström M, Collagens in the human ovary and their changes in the perifollicular stroma during ovulation, Acta Obstetricia et Gynecologica Scandinavica. 85 (2006) 1476–1484. 10.1080/00016340601033741. [DOI] [PubMed] [Google Scholar]
- [228].Kauppila S, Bode MK, Stenbäck F, Risteli L, Risteli J, Cross-linked telopeptides of type I and III collagens in malignant ovarian tumours in vivo, British Journal of Cancer. 81 (1999) 654–661. 10.1038/sj.bjc.6690743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Alkmin S, Brodziski R, Simon H, Hinton D, Goldsmith RH, Patankar M, Campagnola PJ, Role of Collagen Fiber Morphology on Ovarian Cancer Cell Migration Using Image-Based Models of the Extracellular Matrix, Cancers. 12 (2020) 1390. 10.3390/cancers12061390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Nadiarnykh O, LaComb RB, Brewer MA, Campagnola PJ, Alterations of the extracellular matrix in ovarian cancer studied by Second Harmonic Generation imaging microscopy, BMC Cancer. 10 (2010) 94. 10.1186/1471-2407-10-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].Flate E, Stalvey JRD, Motility of select ovarian cancer cell lines: Effect of extra-cellular matrix proteins and the involvement of PAK2, International Journal of Oncology. 45 (2014) 1401–1411. 10.3892/ijo.2014.2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Naba A, Pearce OMT, Del Rosario A, Ma D, Ding H, Rajeeve V, Cutillas PR, Balkwill FR, Hynes RO, Characterization of the Extracellular Matrix of Normal and Diseased Tissues Using Proteomics, J. Proteome Res 16 (2017) 3083–3091. 10.1021/acs.jproteome.7b00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Ricciardelli C, Rodgers RJ, Extracellular Matrix of Ovarian Tumors, Semin Reprod Med. 24 (2006) 270–282. 10.1055/s-2006-948556. [DOI] [PubMed] [Google Scholar]
- [234].Schaefer L, Schaefer RM, Proteoglycans: from structural compounds to signaling molecules, Cell Tissue Res. 339 (2009) 237. 10.1007/s00441-009-0821-y. [DOI] [PubMed] [Google Scholar]
- [235].Ween MP, Oehler MK, Ricciardelli C, Role of Versican, Hyaluronan and CD44 in Ovarian Cancer Metastasis, Int J Mol Sci. 12 (2011) 1009–1029. 10.3390/ijms12021009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Catterall JB, Jones LMH, Turner GA, Membrane protein glycosylation and CD44 content in the adhesion of human ovarian cancer cells to hyaluronan, Clin Exp Metastasis. 17 (1999) 583–591. 10.1023/A:1006756518500. [DOI] [PubMed] [Google Scholar]
- [237].Sato N, Kohi S, Hirata K, Goggins M, Role of hyaluronan in pancreatic cancer biology and therapy: Once again in the spotlight, Cancer Sci. 107 (2016) 569–575. 10.1111/cas.12913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [238].Balduit A, Agostinis C, Mangogna A, Maggi V, Zito G, Romano F, Romano A, Ceccherini R, Grassi G, Bonin S, Bonazza D, Zanconati F, Ricci G, Bulla R, The Extracellular Matrix Influences Ovarian Carcinoma Cells’ Sensitivity to Cisplatinum: A First Step towards Personalized Medicine, Cancers. 12 (2020) 1175. 10.3390/cancers12051175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Wilson KE, Langdon SP, Lessells AM, Miller WR, Expression of the extracellular matrix protein tenascin in malignant and benign ovarian tumours, British Journal of Cancer. 74 (1996) 999–1004. 10.1038/bjc.1996.480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [240].Gillan L, Matei D, Fishman DA, Gerbin CS, Karlan BY, Chang DD, Periostin secreted by epithelial ovarian carcinoma is a ligand for alpha(V)beta(3) and alpha(V)beta(5) integrins and promotes cell motility, Cancer Res. 62 (2002) 5358–5364. [PubMed] [Google Scholar]
- [241].Tumbarello DA, Temple J, Brenton JD, β3 integrin modulates transforming growth factor beta induced (TGFBI) function and paclitaxel response in ovarian cancer cells, Molecular Cancer. 11 (2012) 36. 10.1186/1476-4598-11-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [242].Franke FE, Von Georgi R, Zygmunt M, Münstedt K, Association between fibronectin expression and prognosis in ovarian carcinoma, Anticancer Res. 23 (2003) 4261–4267. [PubMed] [Google Scholar]
- [243].Villegas-Pineda JC, Toledo-Leyva A, Osorio-Trujillo JC, Hernández-Ramírez VI, Talamás-Rohana P, The translational blocking of α5 and α6 integrin subunits affects migration and invasion, and increases sensitivity to carboplatin of SKOV-3 ovarian cancer cell line, Exp Cell Res. 351 (2017) 127–134. 10.1016/j.yexcr.2017.01.010. [DOI] [PubMed] [Google Scholar]
- [244].Meenakshi Sundaram DN, Kucharski C, Parmar MB, Kc RB, Uludağ H, Polymeric Delivery of siRNA against Integrin-β1 (CD29) to Reduce Attachment and Migration of Breast Cancer Cells, Macromol Biosci. 17 (2017). 10.1002/mabi.201600430. [DOI] [PubMed] [Google Scholar]
- [245].Gopal S, Veracini L, Grall D, Butori C, Schaub S, Audebert S, Camoin L, Baudelet E, Radwanska A, Beghelli-de la Forest Divonne S, Violette SM, Weinreb PH, Rekima S, Ilie M, Sudaka A, Hofman P, Van Obberghen-Schilling E, Fibronectin-guided migration of carcinoma collectives, Nat Commun. 8 (2017) 14105. 10.1038/ncomms14105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [246].Wilhelm O, Hafter R, Coppenrath E, Pflanz M, Schmitt M, Babic R, Linke R, Gössner W, Graeff H, Fibrin-Fibronectin Compounds in Human Ovarian Tumor Ascites and Their Possible Relation to the Tumor Stroma, Cancer Res. 48 (1988) 3507–3514. [PubMed] [Google Scholar]
- [247].Kenny HA, Kaur S, Coussens LM, Lengyel E, The initial steps of ovarian cancer cell metastasis are mediated by MMP-2 cleavage of vitronectin and fibronectin, J Clin Invest. 118 (2008) 1367–1379. 10.1172/JCI33775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [248].Rieppi M, Vergani V, Gatto C, Zanetta G, Allavena P, Taraboletti G, Giavazzi R, Mesothelial cells induce the motility of human ovarian carcinoma cells, Int J Cancer. 80 (1999) 303–307. . [DOI] [PubMed] [Google Scholar]
- [249].Januchowski R, Zawierucha P, Ruciński M, Nowicki M, Zabel M, Extracellular matrix proteins expression profiling in chemoresistant variants of the A2780 ovarian cancer cell line, Biomed Res Int. 2014 (2014) 365867. 10.1155/2014/365867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [250].Kenny HA, Dogan S, Zillhardt M, Mitra A, Yamada SD, Krausz T, Lengyel E, Organotypic Models of Metastasis: A 3 Dimensional Culture Mimicking the Human Peritoneum and Omentum for the Study of the Early Steps of Ovarian Cancer Metastasis, Cancer Treat Res. 149 (2009) 335–351. 10.1007/978-0-387-98094-2_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [251].Sawada K, Mitra AK, Radjabi AR, Bhaskar V, Kistner EO, Tretiakova M, Jagadeeswaran S, Montag A, Becker A, Kenny HA, Peter ME, Ramakrishnan V, Yamada SD, Lengyel E, Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target, Cancer Res. 68 (2008) 2329–2339. 10.1158/0008-5472.CAN-07-5167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [252].Witz CA, Montoya-Rodriguez IA, Cho S, Centonze VE, Bonewald LF, Schenken RS, Composition of the extracellular matrix of the peritoneum, J Soc Gynecol Investig. 8 (2001) 299–304. 10.1016/s1071-5576(01)00122-8. [DOI] [PubMed] [Google Scholar]
- [253].Senthebane DA, Jonker T, Rowe A, Thomford NE, Munro D, Dandara C, Wonkam A, Govender D, Calder B, Soares NC, Blackburn JM, Parker MI, Dzobo K, The Role of Tumor Microenvironment in Chemoresistance: 3D Extracellular Matrices as Accomplices, Int J Mol Sci. 19 (2018). 10.3390/ijms19102861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [254].Wu T, Dai Y, Tumor microenvironment and therapeutic response, Cancer Lett. 387 (2017) 61–68. 10.1016/j.canlet.2016.01.043. [DOI] [PubMed] [Google Scholar]
- [255].Hui L, Chen Y, Tumor microenvironment: Sanctuary of the devil, Cancer Lett. 368 (2015) 7–13. 10.1016/j.canlet.2015.07.039. [DOI] [PubMed] [Google Scholar]
- [256].Zhao Y, Cao J, Melamed A, Worley M, Gockley A, Jones D, Nia HT, Zhang Y, Stylianopoulos T, Kumar AS, Mpekris F, Datta M, Sun Y, Wu L, Gao X, Yeku O, Del Carmen MG, Spriggs DR, Jain RK, Xu L, Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma, Proc Natl Acad Sci U S A. 116 (2019) 2210–2219. 10.1073/pnas.1818357116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [257].Murphy JE, Wo JY-L, Ryan DP, Jiang W, Yeap BY, Duda GD, Allen JN, Blaszkowsky LS, Ferrone C, Parikh AR, Nipp RD, Zhu AX, Goyal L, Lillemoe KD, DeLaney TF, Jain RK, Clark JW, Boucher Y, Fernandez-del Castillo C, Hong TS, Potentially curative combination of TGF-b1 inhibitor losartan and FOLFIRINOX (FFX) for locally advanced pancreatic cancer (LAPC): R0 resection rates and preliminary survival data from a prospective phase II study, Journal of Clinical Oncology. 36 (2018) 4116–4116. 10.1200/JCO.2018.36.15_suppl.4116. [DOI] [Google Scholar]
- [258].Hauge A, Rofstad EK, Antifibrotic therapy to normalize the tumor microenvironment, Journal of Translational Medicine. 18 (2020) 207. 10.1186/s12967-020-02376-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [259].Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SFT, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM, Matrix crosslinking forces tumor progression by enhancing integrin signaling, Cell. 139 (2009) 891–906. 10.1016/j.cell.2009.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [260].Cox TR, Gartland A, Erler JT, Lysyl Oxidase, a Targetable Secreted Molecule Involved in Cancer Metastasis, Cancer Res. 76 (2016) 188–92. 10.1158/0008-5472.Can-15-2306. [DOI] [PubMed] [Google Scholar]
- [261].Jiang H, Torphy RJ, Steiger K, Hongo H, Ritchie AJ, Kriegsmann M, Horst D, Umetsu SE, Joseph NM, McGregor K, Pishvaian MJ, Blais EM, Lu B, Li M, Hollingsworth M, Stashko C, Volmar K, Yeh JJ, Weaver VM, Wang ZJ, Tempero MA, Weichert W, Collisson EA, Pancreatic ductal adenocarcinoma progression is restrained by stromal matrix, J Clin Invest. 130 (2020) 4704–4709. 10.1172/jci136760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [262].Saatci O, Kaymak A, Raza U, Ersan PG, Akbulut O, Banister CE, Sikirzhytski V, Tokat UM, Aykut G, Ansari SA, Dogan HT, Dogan M, Jandaghi P, Isik A, Gundogdu F, Kosemehmetoglu K, Dizdar O, Aksoy S, Akyol A, Uner A, Buckhaults PJ, Riazalhosseini Y, Sahin O, Targeting lysyl oxidase (LOX) overcomes chemotherapy resistance in triple negative breast cancer, Nat Commun. 11 (2020) 2416. 10.1038/s41467-020-16199-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [263].Maller O, Drain AP, Barrett AS, Borgquist S, Ruffell B, Zakharevich I, Pham TT, Gruosso T, Kuasne H, Lakins JN, Acerbi I, Barnes JM, Nemkov T, Chauhan A, Gruenberg J, Nasir A, Bjarnadottir O, Werb Z, Kabos P, Chen Y-Y, Hwang ES, Park M, Coussens LM, Nelson AC, Hansen KC, Weaver VM, Tumour-associated macrophages drive stromal cell-dependent collagen crosslinking and stiffening to promote breast cancer aggression, Nature Materials. (2020) 1–12. 10.1038/s41563-020-00849-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [264].Johnston KA, Lopez KM, Lysyl oxidase in cancer inhibition and metastasis, Cancer Lett. 417 (2018) 174–181. 10.1016/j.canlet.2018.01.006. [DOI] [PubMed] [Google Scholar]
- [265].Puiffe M-L, Le Page C, Filali-Mouhim A, Zietarska M, Ouellet V, Tonin PN, Chevrette M, Provencher DM, Mes-Masson A-M, Characterization of Ovarian Cancer Ascites on Cell Invasion, Proliferation, Spheroid Formation, and Gene Expression in an In Vitro Model of Epithelial Ovarian Cancer, Neoplasia. 9 (2007) 820–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [266].Tan DS, Agarwal R, Kaye SB, Mechanisms of transcoelomic metastasis in ovarian cancer, The Lancet Oncology. 7 (2006) 925–934. 10.1016/S1470-2045(06)70939-1. [DOI] [PubMed] [Google Scholar]
- [267].Ip CKM, Li S-S, Tang MYH, Sy SKH, Ren Y, Shum HC, Wong AST, Stemness and chemoresistance in epithelial ovarian carcinoma cells under shear stress, Scientific Reports. 6 (2016) 26788. 10.1038/srep26788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [268].Mo L, Pospichalova V, Huang Z, Murphy SK, Payne S, Wang F, Kennedy M, Cianciolo GJ, Bryja V, Pizzo SV, Bachelder RE, Ascites Increases Expression/Function of Multidrug Resistance Proteins in Ovarian Cancer Cells, PLOS ONE. 10 (2015) e0131579. 10.1371/journal.pone.0131579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [269].McKenzie AJ, Hicks SR, Svec KV, Naughton H, Edmunds ZL, Howe AK, The mechanical microenvironment regulates ovarian cancer cell morphology, migration, and spheroid disaggregation, Scientific Reports. 8 (2018) 7228. 10.1038/s41598-018-25589-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [270].Ansardamavandi A, Tafazzoli-Shadpour M, Omidvar R, Nili F, An AFM-Based Nanomechanical Study of Ovarian Tissues with Pathological Conditions, Int J Nanomedicine. 15 (2020) 4333–4350. 10.2147/IJN.S254342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [271].Cassereau L, Miroshnikova YA, Ou G, Lakins J, Weaver VM, A 3D tension bioreactor platform to study the interplay between ECM stiffness and tumor phenotype, Journal of Biotechnology. 193 (2015) 66–69. 10.1016/j.jbiotec.2014.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [272].Seewaldt V, ECM stiffness paves the way for tumor cells, Nature Medicine. 20 (2014) 332–3. http://dx.doi.org.proxy.lib.umich.edu/10.1038/nm.3523. [DOI] [PubMed] [Google Scholar]
- [273].Shieh AC, Biomechanical Forces Shape the Tumor Microenvironment, Ann Biomed Eng. 39 (2011) 1379–1389. 10.1007/s10439-011-0252-2. [DOI] [PubMed] [Google Scholar]
- [274].Zustiak SP, Dadhwal S, Medina C, Steczina S, Chehreghanianzabi Y, Ashraf A, Asuri P, Three-dimensional matrix stiffness and adhesive ligands affect cancer cell response to toxins, Biotechnol. Bioeng 113 (2016) 443–452. 10.1002/bit.25709. [DOI] [PubMed] [Google Scholar]
- [275].Bordeleau F, Mason BN, Lollis EM, Mazzola M, Zanotelli MR, Somasegar S, Califano JP, Montague C, LaValley DJ, Huynh J, Mencia-Trinchant N, Negrón Abril YL, Hassane DC, Bonassar LJ, Butcher JT, Weiss RS, Reinhart-King CA, Matrix stiffening promotes a tumor vasculature phenotype, Proc Natl Acad Sci U S A. 114 (2017) 492–497. 10.1073/pnas.1613855114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [276].Pathak A, Kumar S, Biophysical regulation of tumor cell invasion: moving beyond matrix stiffness, Integr. Biol 3 (2011) 267–278. 10.1039/C0IB00095G. [DOI] [PubMed] [Google Scholar]
- [277].Schedin P, Keely PJ, Mammary Gland ECM Remodeling, Stiffness, and Mechanosignaling in Normal Development and Tumor Progression, Cold Spring Harbor Perspectives in Biology. 3 (2011). 10.1101/cshperspect.a003228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [278].Schrader J, Gordon-Walker TT, Aucott RL, Deemter M, Quaas A, Walsh S, Benten D, Forbes SJ, Wells RG, Iredale JP, Matrix Stiffness Modulates Proliferation, Chemotherapeutic Response and Dormancy in Hepatocellular Carcinoma Cells, Hepatology. 53 (2011) 1192–1205. 10.1002/hep.24108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [279].Ulrich TA, de J. Pardo EM, Kumar S, The Mechanical Rigidity of the Extracellular Matrix Regulates the Structure, Motility, and Proliferation of Glioma Cells, Cancer Res. 69 (2009) 4167–4174. 10.1158/0008-5472.CAN-08-4859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [280].Yoon A-R, Stasinopoulos I, Kim JH, Yong HM, Kilic O, Wirtz D, Bhujwalla ZM, An SS, COX-2 dependent regulation of mechanotransduction in human breast cancer cells, Cancer Biology & Therapy. 16 (2015) 430–437. 10.1080/15384047.2014.1003004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [281].Guo Z, Zhang T, Fang K, Dou J, Zhou N, Ma X, Gu N, The effects of macroporosity and stiffness of poly[(methyl vinyl ether)-alt-(maleic acid)] cross-linked egg white simulations of an aged extracellular matrix on the proliferation of ovarian cancer cells, RSC Adv. 6 (2016) 43892–43900. 10.1039/C6RA05134K. [DOI] [Google Scholar]
- [282].Loessner D, Stok KS, Lutolf MP, Hutmacher DW, Clements JA, Rizzi SC, Bioengineered 3D platform to explore cell–ECM interactions and drug resistance of epithelial ovarian cancer cells, Biomaterials. 31 (2010) 8494–8506. 10.1016/j.biomaterials.2010.07.064. [DOI] [PubMed] [Google Scholar]
- [283].McGrail DJ, Kieu QMN, Dawson MR, The malignancy of metastatic ovarian cancer cells is increased on soft matrices through a mechanosensitive Rho–ROCK pathway, J Cell Sci. 127 (2014) 2621–2626. 10.1242/jcs.144378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [284].Zhang T, Chen J, Zhang Q, Dou J, Gu N, Poly(ethylene glycol)-cross linked poly(methyl vinyl ether-co-maleic acid)hydrogels for three-dimensional human ovarian cancer cell culture, Colloids and Surfaces A: Physicochemical and Engineering Aspects. 422 (2013) 81–89. 10.1016/j.colsurfa.2013.01.030. [DOI] [Google Scholar]
- [285].Avraham-Chakim L, Elad D, Zaretsky U, Kloog Y, Jaffa A, Grisaru D, Fluid-flow induced wall shear stress and epithelial ovarian cancer peritoneal spreading, PLoS ONE. 8 (2013) e60965. 10.1371/journal.pone.0060965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [286].Jeffrey B, Udaykumar HS, Schulze KS, Flow fields generated by peristaltic reflex in isolated guinea pig ileum: impact of contraction depth and shoulders, American Journal of Physiology-Gastrointestinal and Liver Physiology. 285 (2003) G907–G918. 10.1152/ajpgi.00062.2003. [DOI] [PubMed] [Google Scholar]
- [287].Gajewski TF, Schreiber H, Fu YX, Innate and adaptive immune cells in the tumor microenvironment, Nat Immunol. 14 (2013) 1014–1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [288].Jain RK, Martin JD, Stylianopoulos T, The role of mechanical forces in tumor growth and therapy, Annu Rev Biomed Eng. 16 (2014) 321–346. 10.1146/annurev-bioeng-071813-105259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [289].Cheng G, Tse J, Jain RK, Munn LL, Micro-Environmental Mechanical Stress Controls Tumor Spheroid Size and Morphology by Suppressing Proliferation and Inducing Apoptosis in Cancer Cells, PLOS ONE. 4 (2009) e4632. 10.1371/journal.pone.0004632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [290].Frieden BR, Gatenby RA, Cancer suppression by compression, Bull Math Biol. 77 (2015) 71–82. 10.1007/s11538-014-0051-7. [DOI] [PubMed] [Google Scholar]
- [291].Olcum M, Ozcivici E, Daily application of low magnitude mechanical stimulus inhibits the growth of MDA-MB-231 breast cancer cells in vitro, Cancer Cell Int. 14 (2014) 102. 10.1186/s12935-014-0102-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [292].Tse JM, Cheng G, Tyrrell JA, Wilcox-Adelman SA, Boucher Y, Jain RK, Munn LL, Mechanical compression drives cancer cells toward invasive phenotype, PNAS. 109 (2012) 911–916. 10.1073/pnas.1118910109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [293].Demou ZN, Gene Expression Profiles in 3D Tumor Analogs Indicate Compressive Strain Differentially Enhances Metastatic Potential, Ann Biomed Eng. 38 (2010) 3509–3520. 10.1007/s10439-010-0097-0. [DOI] [PubMed] [Google Scholar]
- [294].Netti PA, Berk DA, Swartz MA, Grodzinsky AJ, Jain RK, Role of extracellular matrix assembly in interstitial transport in solid tumors, Cancer Res. 60 (2000) 2497–2503. [PubMed] [Google Scholar]
- [295].Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR, Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma, Cancer Cell. 21 (2012) 418–429. 10.1016/j.ccr.2012.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [296].Wang J-J, Siu MK-Y, Jiang Y-X, Chan DW, Cheung AN-Y, Ngan HY-S, Chan KK-L, Infiltration of T cells promotes the metastasis of ovarian cancer cells via the modulation of metastasis-related genes and PD-L1 expression, Cancer Immunol Immunother. 69 (2020) 2275–2289. 10.1007/s00262-020-02621-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [297].Joyce MH, Lu C, James ER, Hegab R, Allen SC, Suggs LJ, Brock A, Phenotypic Basis for Matrix Stiffness-Dependent Chemoresistance of Breast Cancer Cells to Doxorubicin, Front Oncol. 8 (2018). 10.3389/fonc.2018.00337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [298].Fenner J, Stacer AC, Winterroth F, Johnson TD, Luker KE, Luker GD, Macroscopic stiffness of breast tumors predicts metastasis, Sci Rep. 4 (2014) 5512. 10.1038/srep05512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [299].Cazet AS, this link will open in a new window Link to external site, Hui MN, Elsworth BL, Wu SZ, Roden D, Chan C-L, Skhinas JN, Collot R, Yang J, Harvey K, Johan MZ, Cooper C, Nair R, Herrmann D, this link will open in a new window Link to external site, McFarland A, Deng N, Ruiz-Borrego M, Rojo F, Trigo JM, Bezares S, Caballero R, Lim E, Timpson P, O’Toole S, Watkins DN, Cox TR, this link will open in a new window Link to external site, Samuel MS, this link will open in a new window Link to external site, Martín M, Swarbrick A, Targeting stromal remodeling and cancer stem cell plasticity overcomes chemoresistance in triple negative breast cancer, Nature Communications. 9 (2018) 1–18. 10.1038/s41467-018-05220-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [300].Chen M, Zeng J, Ruan W, Zhang Z, Wang Y, Xie S, Wang Z, Yang H, Examination of the relationship between viscoelastic properties and the invasion of ovarian cancer cells by atomic force microscopy, Beilstein J. Nanotechnol 11 (2020) 568–582. 10.3762/bjnano.11.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [301].Ketene AN, Schmelz EM, Roberts PC, Agah M, The effects of cancer progression on the viscoelasticity of ovarian cell cytoskeleton structures, Nanomedicine: Nanotechnology, Biology and Medicine. 8 (2012) 93–102. 10.1016/j.nano.2011.05.012. [DOI] [PubMed] [Google Scholar]
- [302].Scott CL, Becker MA, Haluska P, Samimi G, Patient-derived xenograft models to improve targeted therapy in epithelial ovarian cancer treatment, Front Oncol. 3 (2013) 295. 10.3389/fonc.2013.00295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [303].Ricci F, Broggini M, Damia G, Revisiting ovarian cancer preclinical models: implications for a better management of the disease, Cancer Treat Rev. 39 (2013) 561–568. 10.1016/j.ctrv.2013.01.005. [DOI] [PubMed] [Google Scholar]
- [304].Lengyel E, Burdette JE, Kenny HA, Matei D, Pilrose J, Haluska P, Nephew KP, Hales DB, Stack MS, Epithelial ovarian cancer experimental models, Oncogene. 33 (2014) 3619–3633. 10.1038/onc.2013.321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [305].Yang H, Sun L, Liu M, Mao Y, Patient-derived organoids: a promising model for personalized cancer treatment, Gastroenterol Rep (Oxf). 6 (2018) 243–245. 10.1093/gastro/goy040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [306].Ben-David U, Ha G, Tseng Y-Y, Greenwald NF, Oh C, Shih J, McFarland JM, Wong B, Boehm JS, Beroukhim R, Golub TR, Patient-derived xenografts undergo mouse-specific tumor evolution, Nature Genetics. 49 (2017) 1567–1575. 10.1038/ng.3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [307].Vandamme TF, Use of rodents as models of human diseases, J Pharm Bioallied Sci. 6 (2014) 2–9. 10.4103/0975-7406.124301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [308].Magnotti E, Marasco WA, The latest animal models of ovarian cancer for novel drug discovery, Expert Opin Drug Discov. 13 (2018) 249–257. 10.1080/17460441.2018.1426567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [309].Raghavan S, Snyder CS, Wang A, McLean K, Zamarin D, Buckanovich RJ, Mehta G, Carcinoma-Associated Mesenchymal Stem Cells Promote Chemoresistance in Ovarian Cancer Stem Cells via PDGF Signaling, Cancers (Basel). 12 (2020). 10.3390/cancers12082063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [310].Shoval H, Karsch-Bluman A, Brill-Karniely Y, Stern T, Zamir G, Hubert A, Benny O, Tumor cells and their crosstalk with endothelial cells in 3D spheroids, Sci Rep. 7 (2017) 10428. 10.1038/s41598-017-10699-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [311].Shishido A, Mori S, Yokoyama Y, Hamada Y, Minami K, Qian Y, Wang J, Hirose H, Wu X, Kawaguchi N, Nagumo S, Matsuura N, Yamamoto H, Mesothelial cells facilitate cancer stem-like properties in spheroids of ovarian cancer cells, Oncology Reports. 40 (2018) 2105-. [DOI] [PubMed] [Google Scholar]
- [312].Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ, Feltmate CM, Nucci MR, Swisher EM, Nguyen H, Yang C, Morizane R, Kochupurakkal BS, Do KT, Konstantinopoulos PA, Liu JF, Bonventre JV, Matulonis UA, Shapiro GI, Berkowitz RS, Crum CP, D’Andrea AD, Prediction of DNA Repair Inhibitor Response in Short Term Patient-Derived Ovarian Cancer Organoids, Cancer Discov. (2018) CD-18-0474. 10.1158/2159-8290.CD-18-0474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [313].Phan N, Hong JJ, Tofig B, Mapua M, Elashoff D, Moatamed NA, Huang J, Memarzadeh S, Damoiseaux R, Soragni A, A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids, Communications Biology. 2 (2019) 78. 10.1038/s42003-019-0305-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [314].Maru Y, Hippo Y, Current Status of Patient-Derived Ovarian Cancer Models, Cells. 8 (2019) 505. 10.3390/cells8050505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [315].Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z, Concepts of extracellular matrix remodelling in tumour progression and metastasis, Nat Commun. 11 (2020) 5120. 10.1038/s41467-020-18794-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [316].Lee HJ, Mun S, Pham DM, Kim P, Extracellular Matrix-Based Hydrogels to Tailoring Tumor Organoids, ACS Biomater. Sci. Eng (2021). 10.1021/acsbiomaterials.0c01801. [DOI] [PubMed] [Google Scholar]
- [317].Stock K, Estrada MF, Vidic S, Gjerde K, Rudisch A, Santo VE, Barbier M, Blom S, Arundkar SC, Selvam I, Osswald A, Stein Y, Gruenewald S, Brito C, van Weerden W, Rotter V, Boghaert E, Oren M, Sommergruber W, Chong Y, de Hoogt R, Graeser R, Capturing tumor complexity in vitro : Comparative analysis of 2D and 3D tumor models for drug discovery, Scientific Reports. 6 (2016) 28951. 10.1038/srep28951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [318].Mitra AK, Sawada K, Tiwari P, Mui K, Gwin K, Lengyel E, Ligand-independent activation of c-Met by fibronectin and α 5 β 1 -integrin regulates ovarian cancer invasion and metastasis, Oncogene. 30 (2011) 1566–1576. 10.1038/onc.2010.532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [319].Micek HM, Visetsouk MR, Masters KS, Kreeger PK, Engineering the extracellular matrix to model the evolving tumor microenvironment, IScience. (2020) 101742. 10.1016/j.isci.2020.101742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [320].Barbolina MV, Adley BP, Ariztia EV, Liu Y, Stack MS, Microenvironmental Regulation of Membrane Type 1 Matrix Metalloproteinase Activity in Ovarian Carcinoma Cells via Collagen-induced EGR1 Expression, J. Biol. Chem 282 (2007) 4924–4931. 10.1074/jbc.M608428200. [DOI] [PubMed] [Google Scholar]
- [321].Fleszar AJ, Walker A, Kreeger PK, Notbohm J, Substrate curvature induces fallopian tube epithelial cell invasion via cell–cell tension in a model of ovarian cortical inclusion cysts, Int Bio (Cam). 11 (2019) 342–352. 10.1093/intbio/zyz028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [322].Liu M, Zhang X, Long C, Xu H, Cheng X, Chang J, Zhang C, Zhang C, Wang X, Collagen-based three-dimensional culture microenvironment promotes epithelial to mesenchymal transition and drug resistance of human ovarian cancer in vitro, RSC Advances. 8 (2018) 8910–8919. 10.1039/C7RA13742G. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [323].Berger AJ, Linsmeier KM, Kreeger PK, Masters KS, Decoupling the effects of stiffness and fiber density on cellular behaviors via an interpenetrating network of gelatin-methacrylate and collagen, Biomaterials. 141 (2017) 125–135. 10.1016/j.biomaterials.2017.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [324].Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, Woodruff TK, Shah RN, A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice, Nature Communications. 8 (2017) 15261. 10.1038/ncomms15261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [325].Fleszar AJ, Walker A, Porubsky V, Flanigan W, James D, Campagnola PJ, Weisman PS, Kreeger PK, The Extracellular Matrix of Ovarian Cortical Inclusion Cysts Modulates Invasion of Fallopian Tube Epithelial Cells, APL Bioeng. 2 (2018). 10.1063/1.5022595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [326].Shin S, Ikram M, Subhan F, Yeong Kang H, Lim Y, Lee R, Jin S, Hun Jeong Y, Kwak J-Y, Na Y-J, Yoon S, Alginate–marine collagen–agarose composite hydrogels as matrices for biomimetic 3D cell spheroid formation, RSC Advances. 6 (2016) 46952–46965. 10.1039/C6RA01937D. [DOI] [Google Scholar]
- [327].Xu G, Yin F, Wu H, Hu X, Zheng L, Zhao J, In vitro ovarian cancer model based on three-dimensional agarose hydrogel, J Tissue Eng. 5 (2014). 10.1177/2041731413520438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [328].Novak CM, Horst EN, Lin E, Mehta G, Compressive Stimulation Enhances Ovarian Cancer Proliferation, Invasion, Chemoresistance, and Mechanotransduction via CDC42 in a 3D Bioreactor, Cancers. 12 (2020) 1521. 10.3390/cancers12061521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [329].Brooks EA, Gencoglu MF, Corbett DC, Stevens KR, Peyton SR, An omentum-inspired 3D PEG hydrogel for identifying ECM-drivers of drug resistant ovarian cancer, APL Bioengineering. 3 (2019) 026106. 10.1063/1.5091713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [330].Ferreira LP, Gaspar VM, Mano JF, Decellularized Extracellular Matrix for Bioengineering Physiomimetic 3D in Vitro Tumor Models, Trends in Biotechnology. 38 (2020) 1397–1414. 10.1016/j.tibtech.2020.04.006. [DOI] [PubMed] [Google Scholar]
- [331].Yang Z, Xu H, Zhao X, Designer Self-Assembling Peptide Hydrogels to Engineer 3D Cell Microenvironments for Cell Constructs Formation and Precise Oncology Remodeling in Ovarian Cancer, Advanced Science. 7 (2020) 1903718. 10.1002/advs.201903718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [332].Koch F, Müller M, König F, Meyer N, Gattlen J, Pieles U, Peters K, Kreikemeyer B, Mathes S, Saxer S, Mechanical characteristics of beta sheet-forming peptide hydrogels are dependent on peptide sequence, concentration and buffer composition, Royal Society Open Science. 5 (n.d.) 171562. 10.1098/rsos.171562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [333].Hedegaard CL, Redondo-Gómez C, Tan BY, Ng KW, Loessner D, Mata A, Peptide-protein coassembling matrices as a biomimetic 3D model of ovarian cancer, Science Advances. 6 (2020) eabb3298. 10.1126/sciadv.abb3298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [334].Yang Z, Zhao X, A 3D model of ovarian cancer cell lines on peptide nanofiber scaffold to explore the cell–scaffold interaction and chemotherapeutic resistance of anticancer drugs, Int J Nanomedicine. 6 (2011) 303–310. 10.2147/IJN.S15279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [335].Naba A, Clauser KR, Hoersch S, Liu H, Carr SA, Hynes RO, The Matrisome: In Silico Definition and In Vivo Characterization by Proteomics of Normal and Tumor Extracellular Matrices, Mol Cell Proteomics. 11 (2012). 10.1074/mcp.M111.014647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [336].Tian C, Öhlund D, Rickelt S, Lidström T, Huang Y, Hao L, Zhao RT, Franklin O, Bhatia SN, Tuveson DA, Hynes RO, Cancer Cell–Derived Matrisome Proteins Promote Metastasis in Pancreatic Ductal Adenocarcinoma, Cancer Res. 80 (2020) 1461–1474. 10.1158/0008-5472.CAN-19-2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [337].Hebert JD, Myers SA, Naba A, Abbruzzese G, Lamar JM, Carr SA, Hynes RO, Proteomic Profiling of the ECM of Xenograft Breast Cancer Metastases in Different Organs Reveals Distinct Metastatic Niches, Cancer Res. 80 (2020) 1475–1485. 10.1158/0008-5472.CAN-19-2961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [338].Tian C, Clauser KR, Öhlund D, Rickelt S, Huang Y, Gupta M, Mani DR, Carr SA, Tuveson DA, Hynes RO, Proteomic analyses of ECM during pancreatic ductal adenocarcinoma progression reveal different contributions by tumor and stromal cells, PNAS. 116 (2019) 19609–19618. 10.1073/pnas.1908626116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [339].Kearney L, Multiplex-FISH (M-FISH): technique, developments and applications, Cytogenet Genome Res. 114 (2006) 189–198. 10.1159/000094202. [DOI] [PubMed] [Google Scholar]
- [340].Keren L, Bosse M, Thompson S, Risom T, Vijayaragavan K, McCaffrey E, Marquez D, Angoshtari R, Greenwald NF, Fienberg H, Wang J, Kambham N, Kirkwood D, Nolan G, Montine TJ, Galli SJ, West R, Bendall SC, Angelo M, MIBI-TOF: A multiplexed imaging platform relates cellular phenotypes and tissue structure, Science Advances. 5 (2019) eaax5851. 10.1126/sciadv.aax5851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [341].Lin J-R, Izar B, Wang S, Yapp C, Mei S, Shah PM, Santagata S, Sorger PK, Highly multiplexed immunofluorescence imaging of human tissues and tumors using t-CyCIF and conventional optical microscopes, ELife. 7 (2018) e31657. 10.7554/eLife.31657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [342].Spitzer MH, Nolan GP, Mass Cytometry: Single Cells, Many Features, Cell. 165 (2016) 780–791. 10.1016/j.cell.2016.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [343].Ward Rashidi MR, Mehta P, Bregenzer M, Raghavan S, Fleck EM, Horst EN, Harissa Z, Ravikumar V, Brady S, Bild A, Rao A, Buckanovich RJ, Mehta G, Engineered 3D Model of Cancer Stem Cell Enrichment and Chemoresistance, Neoplasia. 21 (2019) 822–836. 10.1016/j.neo.2019.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
