Next Article in Journal
Development of Innate-Immune-Cell-Based Immunotherapy for Adult T-Cell Leukemia–Lymphoma
Next Article in Special Issue
Induced Pluripotent Stem Cell-Derived Fibroblasts Efficiently Engage Senescence Pathways but Show Increased Sensitivity to Stress Inducers
Previous Article in Journal
Heat Shock Protein SSA1 Enriched in Hypoxic Secretome of Candida albicans Exerts an Immunomodulatory Effect via Regulating Macrophage Function
Previous Article in Special Issue
Conditional Reprogramming Modeling of Bladder Cancer for Clinical Translation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interconversion of Cancer Cells and Induced Pluripotent Stem Cells

by
Drishty B. Sarker
1,
Yu Xue
1,
Faiza Mahmud
1,
Jonathan A. Jocelyn
1 and
Qing-Xiang Amy Sang
1,2,*
1
Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA
2
Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4380, USA
*
Author to whom correspondence should be addressed.
Cells 2024, 13(2), 125; https://doi.org/10.3390/cells13020125
Submission received: 19 December 2023 / Revised: 7 January 2024 / Accepted: 8 January 2024 / Published: 10 January 2024
(This article belongs to the Special Issue Reprogrammed Cells in Disease Modeling and Drug Discovery II)

Abstract

:
Cancer cells, especially cancer stem cells (CSCs), share many molecular features with induced pluripotent stem cells (iPSCs) that enable the derivation of induced pluripotent cancer cells by reprogramming malignant cells. Conversely, normal iPSCs can be converted into cancer stem-like cells with the help of tumor microenvironment components and genetic manipulation. These CSC models can be utilized in oncogenic initiation and progression studies, understanding drug resistance, and developing novel therapeutic strategies. This review summarizes the role of pluripotency factors in the stemness, tumorigenicity, and therapeutic resistance of cancer cells. Different methods to obtain iPSC-derived CSC models are described with an emphasis on exposure-based approaches. Culture in cancer cell-conditioned media or cocultures with cancer cells can convert normal iPSCs into cancer stem-like cells, aiding the examination of processes of oncogenesis. We further explored the potential of reprogramming cancer cells into cancer-iPSCs for mechanistic studies and cancer dependencies. The contributions of genetic, epigenetic, and tumor microenvironment factors can be evaluated using these models. Overall, integrating iPSC technology into cancer stem cell research holds significant promise for advancing our knowledge of cancer biology and accelerating the development of innovative and tailored therapeutic interventions.

Graphical Abstract

1. Introduction

Induced pluripotent stem cell (iPSC) technology has been the cornerstone of stem cell research and regenerative medicine since its emergence [1]. The long quest for a more accessible and robust pluripotent cell resource culminated in the breakthrough of pluripotency induction in somatic cells by Yamanaka and Takahashi in 2006 using a combination of four transcription factors—Oct4, Sox2, c-Myc, and Klf4—that are also referred to as “Yamanaka factors” [2]. The demonstration thereby supplanted the need for controversial embryonic stem cell (ESC) sources for generating differentiated human cells of diverse lineages [3,4]. Over the past decade and a half, iPSCs have been extensively utilized in scientific studies due to their enormous scope in cell therapy [5,6,7,8] and disease modeling [9,10] for mechanism research and drug discovery.
Cancer is a generic name for a constellation of malignancies characterized by uncontrolled growth and division [11]. At the origin of cancer is an abnormal cell that acquires either a causative mutation or causative mutations before undergoing oncogenic transformation [12]. The global cancer burden is on the rise, and addressing the high-mortality epidemic requires expedited translational research powered through an expanded availability of suitable cancer models. The promise of iPSC technology in cancer modeling has been substantiated through abundant methods for introducing genetic and epigenetic changes in cells that can drive malignant transformation [13,14,15]. Such models are advantageous in studying cancer initiation and altered pathways related to progression and metastasis [16,17,18]. Moreover, cancer dependencies can be discovered with the help of iPSC-derived cancer avatars, thereby aiding the identification and validation of therapeutic targets for anticancer drug development [19].
In addition to performing oncogenic manipulation in normal iPSCs, pluripotency induction in malignant or pre-malignant cells can generate cancer stem cell (CSC) models [20,21]. CSCs are considered tumor-initiating cells with a high self-renewal and differentiation potential, like that of iPSCs, and are involved in cancer therapeutic resistance and relapse [22]. Therefore, the intertwined aspects of pluripotency and malignancy can be explored at large in iPSC-derived cancer models for a comprehensive understanding of CSC behavior and effective cancer therapies. On the contrary, some cancer-derived induced pluripotent stem cells exhibit reduced malignancy themselves and further differentiate into benign cells of distinct lineages [23,24]. The outcome of reprogramming thus seems to be dictated by an intricate relationship between genetic and epigenetic determinants in cancer cells—a deconvolution of which may provide valuable insight into oncogenic processes and strategies to modulate them.
This review first discusses the highly similar molecular characteristics of iPSCs and cancer cells with an emphasis on pluripotency genes. The origin and biology of cancer stem cells are then reviewed as the malignant counterpart of normal pluripotent stem cells. Finally, a detailed account of the interconversion of cancer cells and iPSCs is presented to highlight the challenges and opportunities regarding the application of iPSC technology in cancer research.

2. Shared Molecular Features between iPSCs and Cancer Cells

The application of iPSC technology in cancer research benefits from a high extent of shared molecular features between iPSCs and cancer cells (Figure 1A). This section explores the parallel molecular signatures and signaling pathways commonly manifested between iPSCs and cancer cells.

2.1. The Role of the Four De Facto Pluripotency Inducers in Cancer Cells

Octamer-binding transcription factor 4 (Oct4), also known as Oct3 or POU5F1 (POU domain, class 5, transcription factor 1), is a prominent regulator of the induction and maintenance of cellular pluripotency and is capable of reprogramming neural stem cells to pluripotency individually [30]. Oct4 overexpression, although circumstantial, has been linked to tumorigenesis in gastric cancer cells [31] and lung adenocarcinoma [32]. Additionally, Oct4 has been observed to play a role in the maintenance and progression of tumors in breast cancer [33], nasopharyngeal carcinoma [34], bladder cancer [35], rectal cancer [36], brain cancer [37], and ovarian cancer [38] as well as in chemoresistance in bladder cancer [39]. Oct4 predominantly exerts its functions through the formation of complexes with other proteins, such as Sox2, Nanog, beta-catenin, and others. Given its participation in multiple pathways associated with tumorigenesis and tumor maintenance, Oct4 may emerge as a promising target for cancer treatment strategies [40].
SRY (sex-determining region Y)-box 2, also known as Sox2, is another crucial factor for pluripotency induction in human somatic cells, and its cooperativity with Oct4 is instrumental for its function [41]. Sox2 overexpression has been linked to oncogenic initiation, amplification, and maintenance in ovarian [38], lung [42], and pancreatic [43] cancers. It has been observed to play a role in late carcinogenesis in prostate [44] and pancreatic cancer [45] as well as in chemoresistance in prostate cancer [44].
Transcription factor Krüppel-like factor 4 (Klf4) interacts directly with Oct4 and Sox2 to activate Nanog, which is another transcription factor that is responsible for the maintenance of pluripotent cells in the inner cell mass of blastocysts by blocking stem cell differentiation [46,47]. Unlike the other pluripotency genes, Klf4 has been observed to possess a tumor-suppressive role in gastric [48] and colorectal [49] cancers. However, its oncogenic role is indicated in the cases of breast cancer [50] and squamous cell carcinoma of the esophagus [51]. The diverse functions exhibited by Klf4 likely stem from its distinctive structure that encompasses both transcriptional activation and repression domains. Furthermore, numerous proteins associated with tumorigenesis, such as p21, p27, p53, and Cyclin D, are downstream targets of Klf4. Its involvement in inflammation and precancerous lesions adds to its significance in tumorigenesis studies [52].
Transcription factor c-Myc enhances pluripotency to generate high-quality iPSCs along with the previously mentioned factors [53]. Overexpression of c-Myc has been tied to prostatic neoplasia [54], and the inactivation of its antagonist tumor suppressors BRCA1 and SMARCB1 has been observed to result in its overexpression in breast cancer [55] and malignant brain rhabdoid tumors [56], respectively. Conversely, c-Myc inactivation has been shown to cause the regression of liver tumors, corroborating its role as an oncogene [57].

2.2. The Role of Auxiliary Pluripotency-Related Factors in Cancer Cells

Despite being related to Klf4, which has known tumor-suppressive properties, Nanog has been linked to the promotion of tumor growth in breast cancer [58] as well as chemoresistance and regenerative capacity in prostate cancer [59]. It has also been observed to be overexpressed in hypoxia-induced aggressiveness of prostatic and pancreatic cancers [60,61].
Glis family zinc finger 1 (Glis1) is a pro-reprogramming factor that acts in collaboration with the main pluripotency inducers [62,63]. Overexpression of Glis1 has been reported in breast cancer cells where it enhances cell migration and invasion capacity together with CUX1 [64]. A similar role of the protein is observed in ovarian cancer cells as well [65].
LIN28a/b (LIN28) is an RNA-binding protein that regulates development-associated genes post-transcriptionally [66]. It has been shown to enhance reprogramming efficiency [67] and has been linked to the aggressiveness of esophageal cancer [68], the initiation and maintenance of liver cancer [69], chemoresistance in breast cancer [70], and the suppression of the p53 tumor suppressor gene [71].
The shared features between iPSCs and cancer cells are summarized in Table 1.

3. Cancer Stem Cells

Cancer stem cells (CSCs) constitute a small subset of cells within tumors, demonstrating the capacity for self-renewal, differentiation, and the initiation of tumor formation upon transplantation into an animal host [22]. They also display a higher expression of drug efflux pumps and elevated DNA repair activity [72]. These properties position CSCs as the primary contributors to resistance against chemotherapy and radiotherapy as well as the recurrence and relapse of cancer [22,72].

3.1. Theories of Cancer Stem Cell Origin

The origin of the CSC concept can be traced back to 1875 when Julius Cohnheim et al. proposed the “embryonal rest” theory, which suggests that cancerous growth may arise from residual embryonic cells persisting after development and remaining dormant until activation [73,74,75]. The initial modern identification of CSCs occurred through a CD34+/CD38− subpopulation of malignant cells being isolated from human acute myeloid leukemia (AML) [76]. This specific cell type demonstrated the ability to generate colony-forming progenitors when engrafted into SCID mice. Over time, various CSC biomarkers have been identified for different cancers, including breast, prostate, brain, stomach, liver, and others [77].
CSC genesis within tumors remains a point of contention. The “progenitor origin model” postulates that CSCs emerge from adult stem or progenitor cells undergoing carcinogenesis due to accumulated mutations [78], with some retaining stemness characteristics while others differentiate. This model complies with the “hierarchy model”, which states that tumors comprise a large number of differentiated or differentiating bulk cancer cells without proliferative capacity and a small population of CSCs that give rise to the bulk cells through division and subsequent differentiation [79]. The progenitor hypothesis identifies CSCs as the cells of cancer’s origin, but some argue that CSCs are instead “cancer-propagating” cells rather than “cancer-initiating” cells in the original tumor [12]. Alternatively, the “stochastic model” of CSC origin suggests that CSCs arise de novo from any cancer cell in the tumor under appropriate microenvironmental cues [79]. The hierarchical and stochastic models are successful in explaining the characteristic tumor architectures of disparate cancers [76,80,81]; their unification is as yet elusive in the context of CSC origin.

3.2. Cellular Plasticity in Cancer Cells

Cancer cell plasticity is a fundamental concept of the stochastic model of CSC origin and enables the acquisition of stem cell features by bulk cancer cells. In 2013, Ischenko et al. showed that the conditional expression of oncogenic KrasG12D in non-stem mouse cells resulted in the emergence of stemness features and metastatic potential. Subsequent analysis revealed c-Myc as a determining factor in the transformation of KrasG12D-expressing cells [82]. Further supporting the idea, Schwitalla et al. demonstrated a positive correlation between NF-κB signaling and the Wnt pathway that drives the dedifferentiation of colon cancer cells [83]. More recently, BIRC3 overexpression has been linked to enhanced self-renewal and stemness maintenance in glioblastoma cell lines and patient-derived glioblastoma cells [25].
In addition, microenvironmental signals also contribute to the generation of CSCs. Vermeulen et al. reported the impact of myofibroblast-secreted factors, such as the hepatocyte growth factor (HGF), in amplifying Wnt signaling activity and consequently triggering the formation of CSCs in colon cancer cells [84]. In an unexpected revelation, Landsberg et al. discovered that proinflammatory cytokine tumor necrosis factor-alpha (TNF-α) could facilitate the dedifferentiation of melanoma cells, leading to resistance against cytotoxic T cells [85].

3.3. Pluripotency-Associated Genes in Cancer Stem-like Feature Acquisition

Elucidating the role of pluripotency-associated genes in CSC formation and malignant activity is critical for obtaining desirable cancer stem-like cells in vitro. In 2010, Riggi et al. discovered that the EWS-FLI-1 fusion gene, the primary driver of Ewing sarcoma, could stimulate the expression of oncogenes SOX2, OCT4, and NANOG in human pediatric mesenchymal stem cells. Among these factors, SOX2 emerged as the pivotal element, with its lone expression capable of inducing CSC features in a primary tumor [26]. Yin et al. later demonstrated that the co-expression of OCT4 and NANOG in hepatocellular carcinoma (HCC) accelerated the epithelial–mesenchymal transition (EMT) process through the STAT3/Snail signaling pathway. The transfected HCC cells acquired CSC traits, including self-renewal, drug resistance, high tumorigenicity, and increased proliferation [86].
Recently, Liu et al. showed that biomechanical forces facilitate the interaction between TAZ and NANOG leading to the upregulation of SOX2 and OCT4, thereby enhancing CSC properties in human breast cancer [87]. Qi et al. previously identified a strong association between KLF4 and the stemness of human osteosarcoma cancer cells. Overexpression of KLF4 resulted in an increased sphere-forming potential, elevated expression of stemness genes, and heightened metastatic potential, with the p38 MAPK pathway implicated in the cell transition [88]. Kim et al. demonstrated that c-MYC could promote stemness and tumorigenicity in triple-negative breast cancer by inhibiting tumor suppressor zinc finger transcription factor 148 (ZNF148) [89]. Additionally, high c-MYC activity was observed in CD133+ colon CSCs, and the knockdown of c-MYC significantly attenuated CSC properties both in vitro and in vivo [90].

4. Deriving Cancer Stem-like Cells from iPSCs

Conversion of iPSCs into cancer stem-like cells can be achieved through the genetic manipulation of the iPSCs [14,91]. Given the lethal effect of some oncogenic mutations on iPSCs [91,92], inducible gene expression systems are widely used to obtain progenitor cells with CSC characteristics. Such systems are extremely useful for studying the stage- and tissue-specific oncogenicity of cancer-predisposing mutations in early carcinogenesis events. However, as previously discussed, the cell origin of cancer may not be identical to cancer-propagating or cancer-initiating CSCs [12]. Therefore, iPSCs with cancer-causing mutations are more useful for cancer initiation studies than CSC modeling. Yet, a handful of studies, mostly focusing on brain tumor modeling, reported cancer stem cell-like properties in engineered/edited iPSC-derived stem and progenitor cell types. Haag et al. showed that the H3.3-K27M mutation in iPSC-derived neural stem cells led to stemness maintenance and increased proliferation through the gliomagenic cells [91], while an earlier study by Koga et al. demonstrated the high self-renewal capacity of PTEN−/−; NF1−/− and TP53−/−; PDGFRAΔ8–9 iPSC-derived high-grade glioma (HGG) spheres [14]. In 2012, Friedmann-Morvinski et al. conducted a study on mouse models where they introduced a lentiviral construct carrying two shRNA sequences against NF1 and TP53 genes into astrocytes and neurons to dedifferentiate the cells into glioblastoma multiforme (GBM)-forming neural progenitor/stem-like cells [93]. Though not relating to iPSCs, this study provides strong evidence for CSC induction through the conditional manipulation of gene expression even in differentiated cells.
Exposure of normal stem cells to malignant niches has been proposed as a model for CSC origin [94,95,96]. Conforming to this hypothesis, CSC models were generated in vitro either by exposing normal iPSCs to conditioned media (CM) from cancer cells of different tissue origin [13,97,98,99,100] or by coculturing the iPSCs with the cancer cells [13] (Figure 2A). A seminal study involving various mouse cancer cell lines demonstrated that CM-dependent CSC derivation from mouse iPSCs (miPSCs) was more successful than the coculture method [13]. The CM-exposed miPSCs generally exhibited CSC-like properties by manifesting self-renewal in sphere-formation assays and forming tumors when transplanted into nude mice [13,97,98,99,100]. A noteworthy observation is that tumors derived from different CM-based CSCs exhibit varying angiogenic and metastatic potential apparently due to the difference in the expression of metastasis and angiogenesis regulators, such as IFNγ and MMPs, in corresponding CSCs [13].
CM-based CSCs can be differentiated into various stromal cells that bolster tumor growth. Subpopulations of CM CSCs have been shown to express VEGF-A [97,99] and FGF2 [99] angiogenic factors in vitro and in vivo to mediate the differentiation and maturation of CSC-derived endothelial cells through paracrine signaling [99]. Furthermore, iPSC-derived CSC-like cells have also been reported to give rise to cancer-associated fibroblasts [100], supporting tumor maintenance and CSC survival [101]. Tumor-associated myoepithelial cells can also arise from these models upon mammary fat pad transplantation [102].
The long-term tumorigenicity of these CSC models remains largely uncharacterized. However, serial transplantation of CSCs induced using pancreatic carcinoma-conditioned media into mice was shown to form more aggressive cancers with the downregulation of the CSC markers of CD133, CD24a, and EpCAM [98]. miPSCs exposed to Lewis lung carcinoma (LLC)-CM were shown to harbor a hypomethylated genome with an enhanced PI3K–Akt pathway [103] that was retained by subsequent progenies. It is important to note that the PI3K–Akt pathway has been reported as highly necessary for the viability of human iPSCs [104]. Later analysis uncovered prostaglandin E2 (PGE2) enrichment in LLC-CM and a higher conversion of miPSCs into CSCs with PGE2 as an additive in the culture media [105]. In addition, extracellular vesicles from LLC cells have been shown to render miPSCs capable of forming tumors in vivo [106]. Exposure to the E-cadherin-Fc chimera protein was also shown to promote CSC features in colon cancer cells [107].
A higher degree of plasticity distinguishes cancer stem cells from bulk cancer cells [108], and the distinction is particularly relevant in the context of the CM-dependent generation of malignant cells from iPSCs. A recent study on the U87MG glioblastoma cell line revealed that CM from bulk cancer cells and cancer stem cell-generated neuron-like glioblastoma cells and spherical glioblastoma stem cells from iPSCs, respectively. While both the induced cell types showed the MGMT, GLI2, LEF1, and β-catenin overexpression characteristic of glioblastoma, only the cancer stem-like cells expressed stem cell markers CD133 and CD44. This finding emphasizes CM compositional variation as a critical determiner of the molecular features of the induced cells [109].
An expedited in vivo approach to converting miPSCs into CSCs entails mixing cancer cells with iPSCs before injecting them into immunocompromised mice. Pancreatic cancer stem cells generated through this method showed varied morphology (mesenchymal-like vs. epithelial-like), tumorigenicity, differentiation capacity, drug sensitivity, and gene expression depending on the distinct microenvironment provided by different cancer cell-CM. However, generated CSCs usually exhibit enhanced invasion ability as well as upregulated energy production and cancer-related pathways compared with the parental iPSC lines [110].
Finally, the differentiation of iPSCs into cervical reserve cells using small molecules has been described. These reserve cells possibly give rise to cervical cancer stem cells [111].

Tumorigenicity and Tumor-Promoting Potential of Other iPSC-Derived Cells

In an in vitro pancreatic ductal adenocarcinoma (PDAC) model, iPSC-derived stellate cells were cocultured with patient-derived organoids or cancer cells to mimic tumor stroma. The stellate cells promoted the growth of some organoids and cells but not all. The observation suggests that the growth of cancer cells depends on the properties of the original tumor and is differentially influenced by a given stromal cell type [112].
A wealth of reports has described the generation, application, and overall usability of iPSC-derived mesenchymal stem cells (iMSCs) for tissue regeneration, cancer therapy, and the treatment of immune-related diseases [113,114,115,116,117]. However, the oncogenicity of iMSCs, especially iMSCs derived from mutation-carrying iPSCs, requires careful investigation. A study showed that iMSCs derived from BRCA+/− iPSCs showed elevated expressions of VEGF, PDGF, and ANGPT angiogenic factors and promoted the formation of an extended vascular network both in vitro and in vivo. The haploinsufficient mesenchymal stem cells were also characterized by higher migration ability and significantly upregulated Periostin expression compared with its BRCA+/+ counterpart. An enhanced tumorigenic and metastatic potential of BRCA+/− iMSCs was also observed when co-injected with 4T1 breast cancer cells into the mammary tissue of NOD-SCID mice [17]. Promisingly, iMSCs derived from disease-free iPSCs are less protumorigenic, while bone marrow-derived mesenchymal stem cells tend to support tumor growth and invasion by producing PGE2, IL6, and other protumor factors [118,119]. It will be worthwhile to examine the basis of the dissimilar cancer-promoting properties between iMSCs and mesenchymal stem cells from other sources [120,121,122].
Table 2 summarizes the derivation of cancer stem-like cells from iPSCs without the need for genetic manipulation.

5. Reprogramming Cancer Cells to Obtain Cancer Stem-like Cells

With the advent of iPSC technology, it has been possible to convert patient-derived somatic cells with cancer-predisposing germline mutation into iPSCs for disease modeling and therapy evaluation [123,124,125]. Beyond these goals, directed dedifferentiation of malignant cells can yield CSC-like cells (Figure 2B) for cancer research. The direct reprogramming of non-stem bulk cancer cells into CSC-like cells can occur through the transfection of either CSC-promoting protein-coding genes [26,82] or pluripotency-associated genes, which is discussed in great detail in this section. An alternative method for direct reprogramming utilizes exposure-based approaches to drive CSC formation by cancer cells (Figure 1B).
Relying on reprogramming factors to induce pluripotency, an attempt to reprogram colorectal cancer cell lines with the Yamanaka cocktail yielded cancer-iPSCs that could differentiate into three germ layer lineages. Nevertheless, the down-regulation of pluripotency genes and a distinct miRNA profile suggest incomplete or partial reprogramming toward pluripotency. Additionally, the cancer-iPSCs attained an epithelial/mesenchymal hybrid phenotype owing to dysregulated miRNA expression [126].
Cancer cell reprogramming has proved an effective tool in translational medical research on myeloproliferative disorders. iPSCs derived from malignant cells of juvenile myelomonocytic leukemia (JMML) through lentiviral OSKM expression showed higher proliferative capacity in cultures and produced myeloid cells of pathological features upon differentiation [20]. More JMML models have been established after with the help of iPSC technology to facilitate the study of disease mechanisms and potent therapeutics [127,128]. iPSCs generated from imatinib-sensitive chronic myelogenous leukemia (CML) patients showed imatinib insensitivity despite restored the expression of the BCR-ABL oncoprotein. Hematopoietic differentiation of the CML-iPSCs regained sensitivity to imatinib, though a fraction of immature cells were still resistant to kinase inhibitors like CML-iPSCs [129]. Similar results were obtained for KBM7 leukemia cell line-derived iPSCs [130]. Recently, a streamlined OSKM-based reprogramming method has been described to derive iPSCs from genetically diverse AML patients, which can model the disease reliably upon xenotransplantation [131]. AML-iPSCs have previously been shown to lose the epigenetic memory of parental cells and lack oncogenic potential. However, the hematopoietic differentiation of these cells reinstates leukemic properties [132]. Cancer-iPSCs that retain oncogenic mutations and malignant properties of source cells thus present a robust and renewable source of in vitro models for drug screening and mechanistic studies of hematological malignancies [20,133,134,135,136,137].
Toward the derivation of brain CSC models, pluripotency induction using OCT4 and JDP2 reprogramming factors conferred DAOY medulloblastoma cell lines with higher in vivo tumorigenicity [27]. The enhancement of tumor formation potency may partly be explained by mTOR activation through epigenetic rewiring due to increased Oct4 [138]. Moreover, a study on DAOY, D341, and D283 medulloblastoma cell lines characterized undifferentiated cell subpopulations in each cell line phenotypically and functionally to conclude that D283 has the highest level of CSC enrichment among the medulloblastoma cell lines [139]. Genetic alterations and the position of the cell of tumor origin within the differentiation hierarchy may account for the observed heterogeneity of stemness features in cancer cell lines [140]. Glioblastoma cell lines T731 and T653 were reprogrammed with Oct4, Sox2, and Klf4 to derive induced pluripotent cancer cells, and the use of a small molecule, PD98059, was shown to increase reprogramming efficiency [141]. Fully reprogrammed iPSCs have been generated from plexiform neurofibroma cells with mutations in the NF1 tumor suppressor gene, potentiating faithful modeling of the developmental tumor [142,143].
Melanoma cell lines have been widely studied for CSC generation. R545 murine melanoma cells were reprogrammed with Oct4, Klf4, and c-Myc pluripotency inducers to obtain non-tumorigenic cancer-iPSCs. Chimeric mice embodying R545-iPSC-derived cells did not develop tumors at five months [24]. In a later work, constitutive expression of OCT4, KLF4, and SOX2 in BRAF-mutant HT-144 human melanoma cells produced metastable iPSCs with reduced tumorigenicity and therapeutic resistance against MAPK inhibitors. These cancer-iPSCs could differentiate into neurons and fibroblast-like cells beyond melanocyte lineages due to the programming-induced resetting of the melanoma epigenetic memory [144]. Apart from pluripotency factor-based reprogramming, the transfer of R545 cell nuclei into enucleated mice oocytes led to the development of pluripotent embryonic stem cells with the potential to generate nuclear transfer chimeras. However, the chimeric mice developed multiple myeloma lesions at a very early age [29].
Reprogramming various human gastrointestinal cancer cell lines with OSKM factors produced cancer-iPSCs with lower proliferation in vitro. The differentiated cancer-iPSCs showed a decreased tumorigenic potential in vivo. These iPSCs could also undergo differentiation into cell lineages from all three germ layers and showed higher sensitivity to anticancer therapies [23]. However, the introduction of OSK into SW480 colon cancer cell lines grown on a serum-containing medium produced CSCs with high dye-efflux activity, self-renewal capacity, and sustained tumorigenicity in serial transplantation [21]. Hep3B liver cancer cells reprogrammed with four factors expressed pluripotency markers and showed stemness that declined over time [145]. The reprogramming of the PLC/PRF/5 hepatoma cell line yielded tumorigenic cells with aggressive phenotypes [146].
A study on PANC-1 human pancreatic cancer cells revealed that the CSC subpopulation with higher c-MET expression is more amenable to OSKM-based reprogramming [147]. A previous reprogramming work on the same cell line reported a regression in tumorigenic potency of reprogrammed cancer cells upon four factor transduction [148]. Concerning patient-derived pancreatic tumors, an iPSC line derived from primary PDAC cells showed pluripotency features and progressed from early to late invasive stages of PDAC upon differentiation [149]. Chiou et al. demonstrated that co-expression of Oct4 and Nanog in A549 lung adenocarcinoma cells led to an augmentation of the CD133+ subpopulation, intensifying their capabilities in EMT, drug resistance, and tumor initiation [150].
In addition to transcription factor-mediated reprogramming, various novel methods have emerged for manipulating the differentiation stage of cancer cells. In 2021, Suzuka et al. discovered that double-network hydrogels (DN gel), comprising PAMPS and PDMAAm, could swiftly reprogram brain cancer cells into CSCs. The DN gel interacts with surface proteins, leading to differential spatiotemporal activation of protein kinases and, consequently, the induction of stemness protein expression, such as GLI1. The interface between the gel and cells hosts protein complexes produced by the cells, creating an optimal niche for the maintenance of CSCs [28]. Thus, this technology allows for the isolation of CSCs or the reversion of cancer cells into a stem cell state.
Table 3 lists the findings from the studies aimed at pluripotency induction in cancer cells.

6. Discussion

Induced pluripotent cancer stem cells (iPSCs) have added significantly to the understanding of cancer research by providing a robust source of in vitro cancer models for the study of cancer initiation, progression, metastasis, and therapeutic vulnerabilities. Their striking similarities with cancer cells at molecular levels, high proliferative capacity, and tumor formation potency have stimulated a more direct use of iPSCs in the given area of research. The stem cell model of cancer propagation emphasizes the presence and active role of cancer stem cells (CSCs) in malignant tumor progression and therapy resistance. The “stemness” properties shared between CSCs and iPSCs thus fueled the characterization of the tumorigenic potential of different iPSC lines. Moreover, the CSC-like behavior of iPSCs in response to tumor microenvironment (TME) components and cancer cell reprogramming to obtain CSC-like cells present areas of growing research interest.
So far, the most feasible way for converting iPSCs into cancer stem cells is the exposure of iPSCs to conditioned media (CM) from cancer cell culture. CM possess and provide key TME components that activate cancer-related pathways and drive epigenetic changes for malignant transformation of the iPSCs [13,100]. However, it is important to note that the CM-based CSCs might be able to mimic the transcriptomic landscape of native CSCs in vivo only, limiting their usefulness in in vitro studies [98,131]. While genetic manipulation of iPSCs can sporadically generate CSCs, the significance of epigenetic changes associated with CSC behavior is typically overlooked in these methods [116,151]. Contrarily, the CM-dependent conversion of iPSCs into CSCs postulates that malignant transformation is achievable without known genetic abnormalities, including key driver mutations [98,103]. However, oncogenic mutations are involved in the development of CSC features in cancer cells as revealed through the better reprogramming efficiency of p53-null or mutant liver cancer cells compared with their wild-type counterparts [145]. SW48 colorectal cancer cells with heterozygous p53 R273H missense mutation gave rise to an increased number of CSCs, signifying the contribution of an aberrant genetic makeup toward CSC development [152]. Hence, the reconciliation of these mutually exclusive CSC-induction schemes warrants an extensive characterization of genetic and microenvironment factors that govern CSC generation from normal stem cells.
A significant limitation of the in vitro functional assessment of iPSC-derived CSCs is the lack of a foolproof method for differentiating between generated CSCs and uninduced iPSCs. Sphere-formation assays are frequently employed to determine the self-renewal capacity of iPSC-derived CSCs, but they do not distinguish the tumorigenic population from uninduced iPSCs since the latter also exhibit a self-renewal ability [153]. Marker analysis provides an alternative identification approach to cancer stem cells, though their use is often contested due to an inconsistent association with CSC identity [154,155,156]. Hence, a combination of phenotypic and functional studies is recommended for CSC identification. The undefined components of CM and the sustainability of acquired CSC characteristics by iPSC-derived CSCs also present some gaps in knowledge of the CSC derivation process. An intrinsic limitation of these iPSC-derived cancer models is that only early stages of carcinogenesis can be studied with these platforms. The insufficiency regarding late-stage tumor studies may be overcome with additional TME components in three-dimensional tumor models or animal experiments.
Reprogramming cancer cells has been presented as a viable tool for generating CSC models for cancer pathway and biomarker studies and anticancer drug screening. It also offers a potent therapeutic approach due to the loss of tumorigenicity in some cases. The most significant challenge to cancer cell reprogramming is the poor efficiency of reprogramming, partly resulting from the multifaceted heterogeneity in tumor cell populations [157,158] and formidable epigenetic barriers [159]. The use of additional supporting factors, such as NANOG and LIN28, and small molecules have been shown to improve the OSKM-induced reprogramming of neoplastic cells [141,160]. However, not all cancer cell lines, even if they are from the same tissue type, are reprogrammable [16,147]. In the context of therapeutic application, epigenetic reorganization is not always sufficient in masking the functional cancer genome, and cancer traits may reappear upon differentiation [132]. Cells may acquire de novo mutations in the process of reprogramming, and genomic instability may arise in resultant cells and their derivatives depending on underlying mutations and differentiation state [161,162].
The intrinsic oncogenic potential of iPSCs presents a confounding factor in cancer-iPSC- and CSC-related studies [163,164,165,166] and needs to be addressed carefully with proper experimental controls. Interestingly, incomplete reprogramming of kidney cells through the transient expression of OSKM factors in vivo has been shown to develop Wilm’s tumor-like neoplasia, but iPSCs generated from the tumor were non-cancerous. This observation highlights the importance of thorough epigenetic remodeling for successful reprogramming [167]. Conversely, CSC features have been reported to arise in incompletely differentiated iPSCs originally generated from non-cancerous cells. This phenomenon presents a practical concern over the safe transplantation of iPSC-derived cells, even from a benign origin [168]. Alternative reprogramming approaches, such as fibromodulin protein-based reprogramming [169], for obtaining non-tumorigenic iPSC are in development. Methods for removal of undifferentiated iPSCs before transplantation to eliminate terato- or tumorigenic cells are also sought [170,171,172,173,174,175,176,177,178].
Recently, conditional reprogramming of patient-derived cancer cells has been proposed for establishing a next-generation biobank [179]. The approach aims to achieve a “reprogrammed stem-like” state through culturing patient-derived cancer cells with murine feeder cells, Swiss 3T3-J2, and ROCK inhibitor Y-27632 [179,180]. These conditionally reprogrammed cells (CRCs) maintain their original karyotype, exhibit invasive characteristics in organoid culture, show high tumorigenic potential, and demonstrate chemosensitivity to test drugs [179,181]. CRCs can be obtained from various solid tumors [181] with few exceptions of metastatic tumors [182], and they hold great promise for transforming disease modeling and drug discovery.
In conclusion, induced pluripotent stem cell technology provides an invaluable platform for the generation of versatile cancer stem-like cells for basic and translational cancer research. With the growing knowledge of CSC-inducing tumor microenvironment factors and functional genetic and epigenetic networks within cancer-propagating cells, CSC modeling strategies will be further refined for an improved recapitulation of cancer stem cell behavior. Altogether, they will inform ongoing therapeutic advancement for realizing effective therapies against refractory malignancies.

Author Contributions

Conceptualization, D.B.S. and Q.-X.A.S.; data curation, D.B.S., Y.X., F.M. and J.A.J.; formal analysis, D.B.S., Y.X., F.M. and Q.-X.A.S.; funding acquisition, Q.-X.A.S.; investigation, D.B.S., Y.X. and F.M.; methodology, D.B.S., Y.X. and F.M.; project administration, Q.-X.A.S.; resources, Q.-X.A.S.; supervision, Q.-X.A.S.; validation, D.B.S., Y.X., F.M., J.A.J. and Q.-X.A.S.; visualization, D.B.S. and Y.X.; writing—original draft preparation, D.B.S., Y.X., F.M. and J.A.J.; writing—review and editing, D.B.S., Y.X., F.M. and Q.-X.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by Florida Department of Health (FDOH)’s Live Like Bella awards (9LA01 and 21L10), a Florida State University Council on Research and Creativity (CRC) planning grant, a Pfeiffer Professorship for Cancer Research in Chemistry and Biochemistry from the College of Arts and Sciences, and an Endowed Chair Professorship in Cancer Research from anonymous donors (to Q.-X.A.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This is a review paper and no new datasets were generated. The datasets used and analyzed are published in this paper and cited from the references.

Acknowledgments

The authors would like to thank Venkat Maddipoti for the initial scientific literature search. The figures were created with BioRender.com (accessed on 6 January 2024).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef] [PubMed]
  2. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef]
  3. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed]
  4. Yu, J.; Vodyanik, M.A.; Smuga-Otto, K.; Antosiewicz-Bourget, J.; Frane, J.L.; Tian, S.; Nie, J.; Jonsdottir, G.A.; Ruotti, V.; Stewart, R. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318, 1917–1920. [Google Scholar] [CrossRef] [PubMed]
  5. Cooper, O.; Seo, H.; Andrabi, S.; Guardia-Laguarta, C.; Graziotto, J.; Sundberg, M.; McLean, J.R.; Carrillo-Reid, L.; Xie, Z.; Osborn, T. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci. Transl. Med. 2012, 4, 141ra190. [Google Scholar] [CrossRef] [PubMed]
  6. Ito, T.; Kawai, Y.; Yasui, Y.; Iriguchi, S.; Minagawa, A.; Ishii, T.; Miyoshi, H.; Taketo, M.M.; Kawada, K.; Obama, K. The therapeutic potential of multiclonal tumoricidal T cells derived from tumor infiltrating lymphocyte-derived iPS cells. Commun. Biol. 2021, 4, 694. [Google Scholar] [CrossRef] [PubMed]
  7. Nakamura, M.; Okano, H. Cell transplantation therapies for spinal cord injury focusing on induced pluripotent stem cells. Cell Res. 2013, 23, 70–80. [Google Scholar] [CrossRef]
  8. Sugita, S.; Iwasaki, Y.; Makabe, K.; Kimura, T.; Futagami, T.; Suegami, S.; Takahashi, M. Lack of T cell response to iPSC-derived retinal pigment epithelial cells from HLA homozygous donors. Stem Cell Rep. 2016, 7, 619–634. [Google Scholar] [CrossRef]
  9. Doss, M.X.; Sachinidis, A. Current challenges of iPSC-based disease modeling and therapeutic implications. Cells 2019, 8, 403. [Google Scholar] [CrossRef]
  10. Rowe, R.G.; Daley, G.Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 2019, 20, 377–388. [Google Scholar] [CrossRef]
  11. Lazebnik, Y. What are the hallmarks of cancer? Nat. Rev. Cancer 2010, 10, 232–233. [Google Scholar] [CrossRef] [PubMed]
  12. Visvader, J.E. Cells of origin in cancer. Nature 2011, 469, 314–322. [Google Scholar] [CrossRef]
  13. Chen, L.; Kasai, T.; Li, Y.; Sugii, Y.; Jin, G.; Okada, M.; Vaidyanath, A.; Mizutani, A.; Satoh, A.; Kudoh, T. A model of cancer stem cells derived from mouse induced pluripotent stem cells. PLoS ONE 2012, 7, e33544. [Google Scholar] [CrossRef] [PubMed]
  14. Koga, T.; Chaim, I.A.; Benitez, J.A.; Markmiller, S.; Parisian, A.D.; Hevner, R.F.; Turner, K.M.; Hessenauer, F.M.; D’Antonio, M.; Nguyen, N.p.D.; et al. Longitudinal assessment of tumor development using cancer avatars derived from genetically engineered pluripotent stem cells. Nat. Commun. 2020, 11, 550. [Google Scholar] [CrossRef]
  15. Kotini, A.G.; Papapetrou, E.P. Engineering of targeted megabase-scale deletions in human induced pluripotent stem cells. Exp. Hematol. 2020, 87, 25–32. [Google Scholar] [CrossRef] [PubMed]
  16. Iskender, B.; Izgi, K.; Canatan, H. Reprogramming bladder cancer cells for studying cancer initiation and progression. Tumor Biol. 2016, 37, 13237–13245. [Google Scholar] [CrossRef] [PubMed]
  17. Portier, L.; Desterke, C.; Chaker, D.; Oudrhiri, N.; Asgarova, A.; Dkhissi, F.; Turhan, A.G.; Bennaceur-Griscelli, A.; Griscelli, F. iPSC-derived hereditary breast cancer model reveals the BRCA1-deleted tumor niche as a new culprit in disease progression. Int. J. Mol. Sci. 2021, 22, 1227. [Google Scholar] [CrossRef]
  18. Sancho-Martinez, I.; Nivet, E.; Xia, Y.; Hishida, T.; Aguirre, A.; Ocampo, A.; Ma, L.; Morey, R.; Krause, M.N.; Zembrzycki, A.; et al. Establishment of human iPSC-based models for the study and targeting of glioma initiating cells. Nat. Commun. 2016, 7, 10743. [Google Scholar] [CrossRef]
  19. Papapetrou, E.P. Patient-derived induced pluripotent stem cells in cancer research and precision oncology. Nat. Med. 2016, 22, 1392–1401. [Google Scholar] [CrossRef]
  20. Gandre-Babbe, S.; Paluru, P.; Aribeana, C.; Chou, S.T.; Bresolin, S.; Lu, L.; Sullivan, S.K.; Tasian, S.K.; Weng, J.; Favre, H. Patient-derived induced pluripotent stem cells recapitulate hematopoietic abnormalities of juvenile myelomonocytic leukemia. Blood J. Am. Soc. Hematol. 2013, 121, 4925–4929. [Google Scholar] [CrossRef]
  21. Oshima, N.; Yamada, Y.; Nagayama, S.; Kawada, K.; Hasegawa, S.; Okabe, H.; Sakai, Y.; Aoi, T. Induction of cancer stem cell properties in colon cancer cells by defined factors. PLoS ONE 2014, 9, e101735. [Google Scholar] [CrossRef] [PubMed]
  22. Yu, Z.; Pestell, T.G.; Lisanti, M.P.; Pestell, R.G. Cancer stem cells. Int. J. Biochem. Cell Biol. 2012, 44, 2144–2151. [Google Scholar] [CrossRef] [PubMed]
  23. Miyoshi, N.; Ishii, H.; Nagai, K.-i.; Hoshino, H.; Mimori, K.; Tanaka, F.; Nagano, H.; Sekimoto, M.; Doki, Y.; Mori, M. Defined factors induce reprogramming of gastrointestinal cancer cells. Proc. Natl. Acad. Sci. USA 2010, 107, 40–45. [Google Scholar] [CrossRef] [PubMed]
  24. Utikal, J.; Maherali, N.; Kulalert, W.; Hochedlinger, K. Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J. Cell Sci. 2009, 122, 3502–3510. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, Q.; Berglund, A.E.; MacAulay, R.J.; Etame, A.B. A novel role of BIRC3 in stemness reprogramming of glioblastoma. Int. J. Mol. Sci. 2021, 23, 297. [Google Scholar] [CrossRef] [PubMed]
  26. Riggi, N.; Suvà, M.-L.; De Vito, C.; Provero, P.; Stehle, J.-C.; Baumer, K.; Cironi, L.; Janiszewska, M.; Petricevic, T.; Suvà, D. EWS-FLI-1 modulates miRNA145 and SOX2 expression to initiate mesenchymal stem cell reprogramming toward Ewing sarcoma cancer stem cells. Genes Dev. 2010, 24, 916–932. [Google Scholar] [CrossRef]
  27. Wuputra, K.; Ku, C.-C.; Kato, K.; Wu, D.-C.; Saito, S.; Yokoyama, K.K. Translational models of 3-D organoids and cancer stem cells in gastric cancer research. Stem Cell Res. Ther. 2021, 12, 492. [Google Scholar] [CrossRef]
  28. Suzuka, J.; Tsuda, M.; Wang, L.; Kohsaka, S.; Kishida, K.; Semba, S.; Sugino, H.; Aburatani, S.; Frauenlob, M.; Kurokawa, T. Rapid reprogramming of tumour cells into cancer stem cells on double-network hydrogels. Nat. Biomed. Eng. 2021, 5, 914–925. [Google Scholar] [CrossRef]
  29. Hochedlinger, K.; Blelloch, R.; Brennan, C.; Yamada, Y.; Kim, M.; Chin, L.; Jaenisch, R. Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev. 2004, 18, 1875–1885. [Google Scholar] [CrossRef]
  30. Pesce, M.; Schöler, H.R. Oct-4: Gatekeeper in the beginnings of mammalian development. Stem Cells 2001, 19, 271–278. [Google Scholar] [CrossRef]
  31. Chen, Z.; Xu, W.R.; Qian, H.; Zhu, W.; Bu, X.F.; Wang, S.; Yan, Y.M.; Mao, F.; Gu, H.B.; Cao, H.L. Oct4, a novel marker for human gastric cancer. J. Surg. Oncol. 2009, 99, 414–419. [Google Scholar] [CrossRef]
  32. Karoubi, G.; Gugger, M.; Schmid, R.; Dutly, A. OCT4 expression in human non-small cell lung cancer: Implications for therapeutic intervention. Interact. Cardiovasc. Thorac. Surg. 2009, 8, 393–397. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, R.-J.; Nam, J.-S. OCT4 expression enhances features of cancer stem cells in a mouse model of breast cancer. Lab. Anim. Res. 2011, 27, 147–152. [Google Scholar] [CrossRef]
  34. Luo, W.; Li, S.; Peng, B.; Ye, Y.; Deng, X.; Yao, K. Embryonic stem cells markers SOX2, OCT4 and Nanog expression and their correlations with epithelial-mesenchymal transition in nasopharyngeal carcinoma. PLoS ONE 2013, 8, e56324. [Google Scholar]
  35. Hatefi, N.; Nouraee, N.; Parvin, M.; Ziaee, S.-A.M.; Mowla, S.J. Evaluating the expression of oct4 as a prognostic tumor marker in bladder cancer. Iran. J. Basic Med. Sci. 2012, 15, 1154. [Google Scholar] [PubMed]
  36. Lambis-Anaya, L.; Fernández-Ruiz, M.; Liscano, Y.; Suarez-Causado, A. High OCT4 expression might be associated with an aggressive phenotype in rectal cancer. Cancers 2023, 15, 3740. [Google Scholar] [CrossRef]
  37. Hua, T.; Zeng, Z.; Chen, J.; Xue, Y.; Li, Y.; Sang, Q. Human Malignant Rhabdoid Tumor Antigens as Biomarkers and Potential Therapeutic Targets. Cancers 2022, 14, 3685. [Google Scholar] [CrossRef]
  38. Robinson, M.; Gilbert, S.F.; Waters, J.A.; Lujano-Olazaba, O.; Lara, J.; Alexander, L.J.; Green, S.E.; Burkeen, G.A.; Patrus, O.; Sarwar, Z. Characterization of SOX2, OCT4 and NANOG in ovarian cancer tumor-initiating cells. Cancers 2021, 13, 262. [Google Scholar] [CrossRef]
  39. Lu, C.-S.; Shieh, G.-S.; Wang, C.-T.; Su, B.-H.; Su, Y.-C.; Chen, Y.-C.; Su, W.-C.; Wu, P.; Yang, W.-H.; Shiau, A.-L. Chemotherapeutics-induced Oct4 expression contributes to drug resistance and tumor recurrence in bladder cancer. Oncotarget 2017, 8, 30844. [Google Scholar] [CrossRef]
  40. Zhang, Q.; Han, Z.; Zhu, Y.; Chen, J.; Li, W. The role and specific mechanism of OCT4 in cancer stem cells: A review. Int. J. Stem Cells 2020, 13, 312–325. [Google Scholar] [CrossRef]
  41. Tapia, N.; MacCarthy, C.; Esch, D.; Gabriele Marthaler, A.; Tiemann, U.; Araúzo-Bravo, M.J.; Jauch, R.; Cojocaru, V.; Schöler, H.R. Dissecting the role of distinct OCT4-SOX2 heterodimer configurations in pluripotency. Sci. Rep. 2015, 5, 13533. [Google Scholar] [CrossRef] [PubMed]
  42. Bass, A.J.; Watanabe, H.; Mermel, C.H.; Yu, S.; Perner, S.; Verhaak, R.G.; Kim, S.Y.; Wardwell, L.; Tamayo, P.; Gat-Viks, I. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat. Genet. 2009, 41, 1238–1242. [Google Scholar] [CrossRef]
  43. Sanada, Y.; Yoshida, K.; Ohara, M.; Oeda, M.; Konishi, K.; Tsutani, Y. Histopathologic evaluation of stepwise progression of pancreatic carcinoma with immunohistochemical analysis of gastric epithelial transcription factor SOX2: Comparison of expression patterns between invasive components and cancerous or nonneoplastic intraductal components. Pancreas 2006, 32, 164–170. [Google Scholar] [PubMed]
  44. Mu, P.; Zhang, Z.; Benelli, M.; Karthaus, W.R.; Hoover, E.; Chen, C.-C.; Wongvipat, J.; Ku, S.-Y.; Gao, D.; Cao, Z. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53-and RB1-deficient prostate cancer. Science 2017, 355, 84–88. [Google Scholar] [CrossRef]
  45. Herreros-Villanueva, M.; Zhang, J.; Koenig, A.; Abel, E.; Smyrk, T.; Bamlet, W.; De Narvajas, A.A.; Gomez, T.; Simeone, D.; Bujanda, L. SOX2 promotes dedifferentiation and imparts stem cell-like features to pancreatic cancer cells. Oncogenesis 2013, 2, e61. [Google Scholar] [CrossRef] [PubMed]
  46. Wei, Z.; Yang, Y.; Zhang, P.; Andrianakos, R.; Hasegawa, K.; Lyu, J.; Chen, X.; Bai, G.; Liu, C.; Pera, M. Klf4 interacts directly with Oct4 and Sox2 to promote reprogramming. Stem Cells 2009, 27, 2969–2978. [Google Scholar] [CrossRef]
  47. Mitsui, K.; Tokuzawa, Y.; Itoh, H.; Segawa, K.; Murakami, M.; Takahashi, K.; Maruyama, M.; Maeda, M.; Yamanaka, S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 2003, 113, 631–642. [Google Scholar] [CrossRef]
  48. Wei, D.; Gong, W.; Kanai, M.; Schlunk, C.; Wang, L.; Yao, J.C.; Wu, T.-T.; Huang, S.; Xie, K. Drastic down-regulation of Kruppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res. 2005, 65, 2746–2754. [Google Scholar] [CrossRef]
  49. Zhao, W.; Hisamuddin, I.M.; Nandan, M.O.; Babbin, B.A.; Lamb, N.E.; Yang, V.W. Identification of Krüppel-like factor 4 as a potential tumor suppressor gene in colorectal cancer. Oncogene 2004, 23, 395–402. [Google Scholar] [CrossRef]
  50. Foster, K.W.; Frost, A.R.; McKie-Bell, P.; Lin, C.-Y.; Engler, J.A.; Grizzle, W.E.; Ruppert, J.M. Increase of GKLF messenger RNA and protein expression during progression of breast cancer. Cancer Res. 2000, 60, 6488–6495. [Google Scholar]
  51. Tetreault, M.P.; Wang, M.L.; Yang, Y.; Travis, J.; Yu, Q.C.; Klein–Szanto, A.J.; Katz, J.P. Klf4 overexpression activates epithelial cytokines and inflammation-mediated esophageal squamous cell cancer in mice. Gastroenterology 2010, 139, 2124–2134.e9. [Google Scholar] [CrossRef]
  52. He, Z.; He, J.; Xie, K. KLF4 transcription factor in tumorigenesis. Cell Death Discov. 2023, 9, 118. [Google Scholar] [CrossRef]
  53. Araki, R.; Hoki, Y.; Uda, M.; Nakamura, M.; Jincho, Y.; Tamura, C.; Sunayama, M.; Ando, S.; Sugiura, M.; Yoshida, M.A. Crucial role of c-Myc in the generation of induced pluripotent stem cells. Stem Cells 2011, 29, 1362–1370. [Google Scholar] [CrossRef]
  54. Koh, C.M.; Gurel, B.; Sutcliffe, S.; Aryee, M.J.; Schultz, D.; Iwata, T.; Uemura, M.; Zeller, K.I.; Anele, U.; Zheng, Q. Alterations in nucleolar structure and gene expression programs in prostatic neoplasia are driven by the MYC oncogene. Am. J. Pathol. 2011, 178, 1824–1834. [Google Scholar] [CrossRef]
  55. Xu, J.; Chen, Y.; Olopade, O.I. MYC and breast cancer. Genes Cancer 2010, 1, 629–640. [Google Scholar] [CrossRef]
  56. Cooper, G.W.; Hong, A.L. SMARCB1-deficient cancers: Novel molecular insights and therapeutic vulnerabilities. Cancers 2022, 14, 3645. [Google Scholar] [CrossRef] [PubMed]
  57. Shachaf, C.M.; Kopelman, A.M.; Arvanitis, C.; Karlsson, Å.; Beer, S.; Mandl, S.; Bachmann, M.H.; Borowsky, A.D.; Ruebner, B.; Cardiff, R.D. MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature 2004, 431, 1112–1117. [Google Scholar] [CrossRef]
  58. Lu, X.; Mazur, S.J.; Lin, T.; Appella, E.; Xu, Y. The pluripotency factor nanog promotes breast cancer tumorigenesis and metastasis. Oncogene 2014, 33, 2655–2664. [Google Scholar] [CrossRef] [PubMed]
  59. Jeter, C.R.; Liu, B.; Liu, X.; Chen, X.; Liu, C.; Calhoun-Davis, T.; Repass, J.; Zaehres, H.; Shen, J.; Tang, D.G. NANOG promotes cancer stem cell characteristics and prostate cancer resistance to androgen deprivation. Oncogene 2011, 30, 3833–3845. [Google Scholar] [CrossRef]
  60. Bao, B.; Ahmad, A.; Kong, D.; Ali, S.; Azmi, A.S.; Li, Y.; Banerjee, S.; Padhye, S.; Sarkar, F.H. Hypoxia induced aggressiveness of prostate cancer cells is linked with deregulated expression of VEGF, IL-6 and miRNAs that are attenuated by CDF. PLoS ONE 2012, 7, e43726. [Google Scholar] [CrossRef] [PubMed]
  61. Bao, B.; Ali, S.; Ahmad, A.; Azmi, A.S.; Li, Y.; Banerjee, S.; Kong, D.; Sethi, S.; Aboukameel, A.; Padhye, S.B. Hypoxia-induced aggressiveness of pancreatic cancer cells is due to increased expression of VEGF, IL-6 and miR-21, which can be attenuated by CDF treatment. PLoS ONE 2012, 7, e50165. [Google Scholar] [CrossRef]
  62. Schmidt, R.; Plath, K. The roles of the reprogramming factors Oct4, Sox2 and Klf4 in resetting the somatic cell epigenome during induced pluripotent stem cell generation. Genome Biol. 2012, 13, 251. [Google Scholar] [CrossRef]
  63. Scoville, D.W.; Kang, H.S.; Jetten, A.M. GLIS1-3: Emerging roles in reprogramming, stem and progenitor cell differentiation and maintenance. Stem Cell Investig. 2017, 4, 80. [Google Scholar] [CrossRef] [PubMed]
  64. Vadnais, C.; Shooshtarizadeh, P.; Rajadurai, C.V.; Lesurf, R.; Hulea, L.; Davoudi, S.; Cadieux, C.; Hallett, M.; Park, M.; Nepveu, A. Autocrine activation of the Wnt/β-catenin pathway by CUX1 and GLIS1 in breast cancers. Biol. Open 2014, 3, 937–946. [Google Scholar] [CrossRef]
  65. Kim, M.J.; Jung, D.; Park, J.Y.; Lee, S.M.; An, H.J. GLIS1 in cancer-associated fibroblasts regulates the migration and invasion of ovarian cancer cells. Int. J. Mol. Sci. 2022, 23, 2218. [Google Scholar] [CrossRef] [PubMed]
  66. Tsialikas, J.; Romer-Seibert, J. LIN28: Roles and regulation in development and beyond. Development 2015, 142, 2397–2404. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, L.; Su, Y.; Huang, C.; Yin, Y.; Chu, A.; Knupp, A.; Tang, Y. NANOG and LIN28 dramatically improve human cell reprogramming by modulating LIN41 and canonical WNT activities. Biol. Open 2019, 8, bio047225. [Google Scholar] [CrossRef]
  68. Hamano, R.; Miyata, H.; Yamasaki, M.; Sugimura, K.; Tanaka, K.; Kurokawa, Y.; Nakajima, K.; Takiguchi, S.; Fujiwara, Y.; Mori, M. High expression of Lin28 is associated with tumour aggressiveness and poor prognosis of patients in oesophagus cancer. Br. J. Cancer 2012, 106, 1415–1423. [Google Scholar] [CrossRef] [PubMed]
  69. Nguyen, L.H.; Robinton, D.A.; Seligson, M.T.; Wu, L.; Li, L.; Rakheja, D.; Comerford, S.A.; Ramezani, S.; Sun, X.; Parikh, M.S. Lin28b is sufficient to drive liver cancer and necessary for its maintenance in murine models. Cancer Cell 2014, 26, 248–261. [Google Scholar] [CrossRef]
  70. Wang, L.; Yuan, C.; Lv, K.; Xie, S.; Fu, P.; Liu, X.; Chen, Y.; Qin, C.; Deng, W.; Hu, W. Lin28 mediates radiation resistance of breast cancer cells via regulation of caspase, H2A. X and Let-7 signaling. PLoS ONE 2013, 8, e67373. [Google Scholar]
  71. Shi, J.; Jin, X.; Wang, Y.; Zhu, T.; Zhang, D.; Li, Q.; Zhong, X.; Deng, Y.; Shen, J.; Fan, X. LIN28B inhibition sensitizes cells to p53-restoring PPI therapy through unleashed translational suppression. Oncogenesis 2022, 11, 37. [Google Scholar] [CrossRef]
  72. Phi, L.T.H.; Sari, I.N.; Yang, Y.-G.; Lee, S.-H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef]
  73. Conheim, J. Congenitales, quergestreiftes muskelsarkon der nireren. Virchows Arch 1875, 65, 64. [Google Scholar] [CrossRef]
  74. Ratajczak, M.Z.; Bujko, K.; Mack, A.; Kucia, M.; Ratajczak, J. Cancer from the perspective of stem cells and misappropriated tissue regeneration mechanisms. Leukemia 2018, 32, 2519–2526. [Google Scholar] [CrossRef] [PubMed]
  75. Sell, S. On the stem cell origin of cancer. Am. J. Pathol. 2010, 176, 2584–2594. [Google Scholar] [CrossRef] [PubMed]
  76. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [PubMed]
  78. Feinberg, A.P.; Ohlsson, R.; Henikoff, S. The epigenetic progenitor origin of human cancer. Nat. Rev. Genet. 2006, 7, 21–33. [Google Scholar] [CrossRef]
  79. Franco, S.S.; Szczesna, K.; Iliou, M.S.; Al-Qahtani, M.; Mobasheri, A.; Kobolák, J.; Dinnyés, A. In vitro models of cancer stem cells and clinical applications. BMC Cancer 2016, 16, 23–49. [Google Scholar] [CrossRef] [PubMed]
  80. Kai, K.; Nagano, O.; Sugihara, E.; Arima, Y.; Sampetrean, O.; Ishimoto, T.; Nakanishi, M.; Ueno, N.T.; Iwase, H.; Saya, H. Maintenance of HCT116 colon cancer cell line conforms to a stochastic model but not a cancer stem cell model. Cancer Sci. 2009, 100, 2275–2282. [Google Scholar] [CrossRef] [PubMed]
  81. Klevebring, D.; Rosin, G.; Ma, R.; Lindberg, J.; Czene, K.; Kere, J.; Fredriksson, I.; Bergh, J.; Hartman, J. Sequencing of breast cancer stem cell populations indicates a dynamic conversion between differentiation states in vivo. Breast Cancer Res. 2014, 16, R72. [Google Scholar] [CrossRef] [PubMed]
  82. Ischenko, I.; Zhi, J.; Moll, U.M.; Nemajerova, A.; Petrenko, O. Direct reprogramming by oncogenic Ras and Myc. Proc. Natl. Acad. Sci. USA 2013, 110, 3937–3942. [Google Scholar] [CrossRef]
  83. Schwitalla, S.; Fingerle, A.A.; Cammareri, P.; Nebelsiek, T.; Göktuna, S.I.; Ziegler, P.K.; Canli, O.; Heijmans, J.; Huels, D.J.; Moreaux, G. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell 2013, 152, 25–38. [Google Scholar] [CrossRef]
  84. Vermeulen, L.; De Sousa E Melo, F.; Van Der Heijden, M.; Cameron, K.; De Jong, J.H.; Borovski, T.; Tuynman, J.B.; Todaro, M.; Merz, C.; Rodermond, H. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol. 2010, 12, 468–476. [Google Scholar] [CrossRef] [PubMed]
  85. Landsberg, J.; Kohlmeyer, J.; Renn, M.; Bald, T.; Rogava, M.; Cron, M.; Fatho, M.; Lennerz, V.; Wölfel, T.; Hölzel, M. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 2012, 490, 412–416. [Google Scholar] [CrossRef] [PubMed]
  86. Yin, X.; Zhang, B.-H.; Zheng, S.-S.; Gao, D.-M.; Qiu, S.-J.; Wu, W.-Z.; Ren, Z.-G. Coexpression of gene Oct4 and Nanog initiates stem cell characteristics in hepatocellular carcinoma and promotes epithelial-mesenchymal transition through activation of Stat3/Snail signaling. J. Hematol. Oncol. 2015, 8, 23. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, X.; Ye, Y.; Zhu, L.; Xiao, X.; Zhou, B.; Gu, Y.; Si, H.; Liang, H.; Liu, M.; Li, J. Niche stiffness sustains cancer stemness via TAZ and NANOG phase separation. Nat. Commun. 2023, 14, 238. [Google Scholar] [CrossRef]
  88. Qi, X.-T.; Li, Y.-L.; Zhang, Y.-Q.; Xu, T.; Lu, B.; Fang, L.; Gao, J.-Q.; Yu, L.-S.; Zhu, D.-F.; Yang, B. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharmacol. Sin. 2019, 40, 546–555. [Google Scholar] [CrossRef]
  89. Kim, M.; Singh, M.; Lee, B.-K.; Hibbs, M.; Richardson, K.; Ellies, L.; Wintle, L.; Stuart, L.M.; Wang, J.Y.; Voon, D.C. A MYC-ZNF148-ID1/3 regulatory axis modulating cancer stem cell traits in aggressive breast cancer. Oncogenesis 2022, 11, 60. [Google Scholar] [CrossRef]
  90. Zhang, H.L.; Wang, P.; Lu, M.Z.; Zhang, S.D.; Zheng, L. c-Myc maintains the self-renewal and chemoresistance properties of colon cancer stem cells. Oncol. Lett. 2019, 17, 4487–4493. [Google Scholar] [CrossRef]
  91. Haag, D.; Mack, N.; da Silva, P.B.G.; Statz, B.; Clark, J.; Tanabe, K.; Sharma, T.; Jäger, N.; Jones, D.T.; Kawauchi, D. H3. 3-K27M drives neural stem cell-specific gliomagenesis in a human iPSC-derived model. Cancer Cell 2021, 39, 407–422.e413. [Google Scholar] [CrossRef] [PubMed]
  92. Parisian, A.D.; Koga, T.; Miki, S.; Johann, P.D.; Kool, M.; Crawford, J.R.; Furnari, F.B. SMARCB1 loss interacts with neuronal differentiation state to block maturation and impact cell stability. Genes Dev. 2020, 34, 1316–1329. [Google Scholar] [CrossRef] [PubMed]
  93. Friedmann-Morvinski, D.; Bushong, E.A.; Ke, E.; Soda, Y.; Marumoto, T.; Singer, O.; Ellisman, M.H.; Verma, I.M. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science 2012, 338, 1080–1084. [Google Scholar] [CrossRef] [PubMed]
  94. Booth, B.; Boulanger, C.; Anderson, L.; Smith, G. The normal mammary microenvironment suppresses the tumorigenic phenotype of mouse mammary tumor virus-neu-transformed mammary tumor cells. Oncogene 2011, 30, 679–689. [Google Scholar] [CrossRef] [PubMed]
  95. Marotta, L.L.C.; Polyak, K. Cancer stem cells: A model in the making. Curr. Opin. Genet. Dev. 2009, 19, 44–50. [Google Scholar] [CrossRef]
  96. Ajani, J.A.; Song, S.; Hochster, H.S.; Steinberg, I.B. Cancer stem cells: The promise and the potential. Semin. Oncol. 2015, 42 (Suppl. S1), S3–S17. [Google Scholar] [CrossRef]
  97. Afify, S.M.; Sanchez Calle, A.; Hassan, G.; Kumon, K.; Nawara, H.M.; Zahra, M.H.; Mansour, H.M.; Khayrani, A.C.; Alam, M.J.; Du, J. A novel model of liver cancer stem cells developed from induced pluripotent stem cells. Br. J. Cancer 2020, 122, 1378–1390. [Google Scholar] [CrossRef]
  98. Calle, A.S.; Nair, N.; Oo, A.K.; Prieto-Vila, M.; Koga, M.; Khayrani, A.C.; Hussein, M.; Hurley, L.; Vaidyanath, A.; Seno, A. A new PDAC mouse model originated from iPSCs-converted pancreatic cancer stem cells (CSCcm). Am. J. Cancer Res. 2016, 6, 2799. [Google Scholar]
  99. Prieto-Vila, M.; Yan, T.; Calle, A.S.; Nair, N.; Hurley, L.; Kasai, T.; Kakuta, H.; Masuda, J.; Murakami, H.; Mizutani, A. iPSC-derived cancer stem cells provide a model of tumor vasculature. Am. J. Cancer Res. 2016, 6, 1906. [Google Scholar]
  100. Nair, N.; Calle, A.S.; Zahra, M.H.; Prieto-Vila, M.; Oo, A.K.K.; Hurley, L.; Vaidyanath, A.; Seno, A.; Masuda, J.; Iwasaki, Y. A cancer stem cell model as the point of origin of cancer-associated fibroblasts in tumor microenvironment. Sci. Rep. 2017, 7, 6838. [Google Scholar] [CrossRef]
  101. Chen, W.-J.; Ho, C.-C.; Chang, Y.-L.; Chen, H.-Y.; Lin, C.-A.; Ling, T.-Y.; Yu, S.-L.; Yuan, S.-S.; Louisa Chen, Y.-J.; Lin, C.-Y. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat. Commun. 2014, 5, 3472. [Google Scholar] [CrossRef] [PubMed]
  102. Afify, S.M.; Hassan, G.; Zahra, M.H.; Nawara, H.M.; Quora, H.A.A.; Osman, A.; Mansour, H.; Kumon, K.; Seno, A.; Chen, L. Cancer stem cells as the source of tumor associated myoepithelial cells in the tumor microenvironment developing ductal carcinoma in situ. Biomaterials 2023, 301, 122249. [Google Scholar] [CrossRef] [PubMed]
  103. Oo, A.K.K.; Calle, A.S.; Nair, N.; Mahmud, H.; Vaidyanath, A.; Yamauchi, J.; Khayrani, A.C.; Du, J.; Alam, M.J.; Seno, A. Up-regulation of PI 3-kinases and the activation of PI3K-Akt signaling pathway in cancer stem-like cells through DNA hypomethylation mediated by the cancer microenvironment. Transl. Oncol. 2018, 11, 653–663. [Google Scholar] [CrossRef] [PubMed]
  104. Hossini, A.M.; Quast, A.S.; Plötz, M.; Grauel, K.; Exner, T.; Küchler, J.; Stachelscheid, H.; Eberle, J.; Rabien, A.; Makrantonaki, E. PI3K/AKT signaling pathway is essential for survival of induced pluripotent stem cells. PLoS ONE 2016, 11, e0154770. [Google Scholar] [CrossRef]
  105. Minematsu, H.; Afify, S.M.; Sugihara, Y.; Hassan, G.; Zahra, M.H.; Seno, A.; Adachi, M.; Seno, M. Cancer stem cells induced by chronic stimulation with prostaglandin E2 exhibited constitutively activated PI3K axis. Sci. Rep. 2022, 12, 15628. [Google Scholar] [CrossRef]
  106. Yan, T.; Mizutani, A.; Chen, L.; Takaki, M.; Hiramoto, Y.; Matsuda, S.; Shigehiro, T.; Kasai, T.; Kudoh, T.; Murakami, H. Characterization of cancer stem-like cells derived from mouse induced pluripotent stem cells transformed by tumor-derived extracellular vesicles. J. Cancer 2014, 5, 572. [Google Scholar] [CrossRef]
  107. Qian, Y.; Wu, X.; Yokoyama, Y.; Okuzaki, D.; Taguchi, M.; Hirose, H.; Wang, J.; Hata, T.; Inoue, A.; Hiraki, M. E-cadherin-Fc chimera protein matrix enhances cancer stem-like properties and induces mesenchymal features in colon cancer cells. Cancer Sci. 2019, 110, 3520–3532. [Google Scholar] [CrossRef]
  108. Rich, J.N. Cancer stem cells: Understanding tumor hierarchy and heterogeneity. Medicine 2016, 95 (Suppl. S1), S2–S7. [Google Scholar] [CrossRef]
  109. Yasmin, I.A.; Dharmarajan, A.; Warrier, S. iPSC-Derived Glioblastoma Cells Have Enhanced Stemness Wnt/β-Catenin Activity Which Is Negatively Regulated by Wnt Antagonist sFRP4. Cancers 2023, 15, 3622. [Google Scholar] [CrossRef]
  110. Hassan, G.; Ohara, T.; Afify, S.M.; Kumon, K.; Zahra, M.H.; Fu, X.; Al Kadi, M.; Seno, A.; Salomon, D.S.; Seno, M. Different pancreatic cancer microenvironments convert iPSCs into cancer stem cells exhibiting distinct plasticity with altered gene expression of metabolic pathways. J. Exp. Clin. Cancer Res. 2022, 41, 29. [Google Scholar] [CrossRef]
  111. Sato, M.; Kawana, K.; Adachi, K.; Fujimoto, A.; Yoshida, M.; Nakamura, H.; Nishida, H.; Inoue, T.; Taguchi, A.; Ogishima, J. Regeneration of cervical reserve cell-like cells from human induced pluripotent stem cells (iPSCs): A new approach to finding targets for cervical cancer stem cell treatment. Oncotarget 2017, 8, 40935. [Google Scholar] [CrossRef]
  112. Kometani, T.; Kamo, K.; Kido, T.; Hiraoka, N.; Chibazakura, T.; Unno, K.; Sekine, K. Development of a novel co-culture system using human pancreatic cancer cells and human iPSC-derived stellate cells to mimic the characteristics of pancreatic ductal adenocarcinoma in vitro. Biochem. Biophys. Res. Commun. 2023, 658, 1–9. [Google Scholar] [CrossRef] [PubMed]
  113. Bloor, A.J.; Patel, A.; Griffin, J.E.; Gilleece, M.H.; Radia, R.; Yeung, D.T.; Drier, D.; Larson, L.S.; Uenishi, G.I.; Hei, D. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: A phase I, multicenter, open-label, dose-escalation study. Nat. Med. 2020, 26, 1720–1725. [Google Scholar] [CrossRef] [PubMed]
  114. Jeon, O.H.; Panicker, L.M.; Lu, Q.; Chae, J.J.; Feldman, R.A.; Elisseeff, J.H. Human iPSC-derived osteoblasts and osteoclasts together promote bone regeneration in 3D biomaterials. Sci. Rep. 2016, 6, 26761. [Google Scholar] [CrossRef] [PubMed]
  115. Zhao, Q.; Hai, B.; Kelly, J.; Wu, S.; Liu, F. Extracellular vesicle mimics made from iPS cell-derived mesenchymal stem cells improve the treatment of metastatic prostate cancer. Stem Cell Res. Ther. 2021, 12, 29. [Google Scholar] [CrossRef]
  116. Chao, H.-M.; Chern, E. Patient-derived induced pluripotent stem cells for models of cancer and cancer stem cell research. J. Formos. Med. Assoc. 2018, 117, 1046–1057. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, Z.; Chen, H.; Wang, P.; Zhou, M.; Li, G.; Hu, Z.; Hu, Q.; Zhao, J.; Liu, X.; Wu, L. Site-specific integration of TRAIL in iPSC-derived mesenchymal stem cells for targeted cancer therapy. Stem Cells Transl. Med. 2022, 11, 297–309. [Google Scholar] [CrossRef]
  118. Zhao, Q.; Gregory, C.A.; Lee, R.H.; Reger, R.L.; Qin, L.; Hai, B.; Park, M.S.; Yoon, N.; Clough, B.; McNeill, E. MSCs derived from iPSCs with a modified protocol are tumor-tropic but have much less potential to promote tumors than bone marrow MSCs. Proc. Natl. Acad. Sci. USA 2015, 112, 530–535. [Google Scholar] [CrossRef]
  119. Li, H.-J.; Reinhardt, F.; Herschman, H.R.; Weinberg, R.A. Cancer-stimulated mesenchymal stem cells create a carcinoma stem cell niche via prostaglandin E2 signaling. Cancer Discov. 2012, 2, 840–855. [Google Scholar] [CrossRef]
  120. El-Badawy, A.; Ghoneim, M.A.; Gabr, M.M.; Salah, R.A.; Mohamed, I.K.; Amer, M.; El-Badri, N. Cancer cell-soluble factors reprogram mesenchymal stromal cells to slow cycling, chemoresistant cells with a more stem-like state. Stem Cell Res. Ther. 2017, 8, 254. [Google Scholar] [CrossRef]
  121. Jing, Y.; Liang, W.; Zhang, L.; Tang, J.; Huang, Z. The role of mesenchymal stem cells in the induction of cancer-stem cell phenotype. Front. Oncol. 2022, 12, 817971. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, Z.; Liu, W.; Wang, Z.; Zeng, B.; Peng, G.; Niu, H.; Chen, L.; Liu, C.; Hu, Q.; Zhang, Y. Mesenchymal stem cells derived from iPSCs expressing interleukin-24 inhibit the growth of melanoma in the tumor-bearing mouse model. Cancer Cell Int. 2020, 20, 33. [Google Scholar] [CrossRef] [PubMed]
  123. Griscelli, F.; Oudrhiri, N.; Feraud, O.; Divers, D.; Portier, L.; Turhan, A.G.; Griscelli, A.B. Generation of induced pluripotent stem cell (iPSC) line from a patient with triple negative breast cancer with hereditary exon 17 deletion of BRCA1 gene. Stem Cell Res. 2017, 24, 135–138. [Google Scholar] [CrossRef] [PubMed]
  124. Lee, D.-F.; Su, J.; Kim, H.S.; Chang, B.; Papatsenko, D.; Zhao, R.; Yuan, Y.; Gingold, J.; Xia, W.; Darr, H. Modeling familial cancer with induced pluripotent stem cells. Cell 2015, 161, 240–254. [Google Scholar] [CrossRef] [PubMed]
  125. Hwang, J.W.; Desterke, C.; Féraud, O.; Richard, S.; Ferlicot, S.; Verkarre, V.; Patard, J.J.; Loisel-Duwattez, J.; Foudi, A.; Griscelli, F. iPSC-derived embryoid bodies as models of c-met-mutated hereditary papillary renal cell carcinoma. Int. J. Mol. Sci. 2019, 20, 4867. [Google Scholar] [CrossRef] [PubMed]
  126. Hiew, M.S.Y.; Cheng, H.P.; Huang, C.-J.; Chong, K.Y.; Cheong, S.K.; Choo, K.B.; Kamarul, T. Incomplete cellular reprogramming of colorectal cancer cells elicits an epithelial/mesenchymal hybrid phenotype. J. Biomed. Sci. 2018, 25, 57. [Google Scholar] [CrossRef]
  127. Mulero-Navarro, S.; Sevilla, A.; Roman, A.C.; Lee, D.-F.; D’Souza, S.L.; Pardo, S.; Riess, I.; Su, J.; Cohen, N.; Schaniel, C. Myeloid dysregulation in a human induced pluripotent stem cell model of PTPN11-associated juvenile myelomonocytic leukemia. Cell Rep. 2015, 13, 504–515. [Google Scholar] [CrossRef]
  128. Tasian, S.K.; Casas, J.A.; Posocco, D.; Gandre-Babbe, S.; Gagne, A.L.; Liang, G.; Loh, M.L.; Weiss, M.J.; French, D.L.; Chou, S.T. Mutation-specific signaling profiles and kinase inhibitor sensitivities of juvenile myelomonocytic leukemia revealed by induced pluripotent stem cells. Leukemia 2019, 33, 181–190. [Google Scholar] [CrossRef]
  129. Kumano, K.; Arai, S.; Hosoi, M.; Taoka, K.; Takayama, N.; Otsu, M.; Nagae, G.; Ueda, K.; Nakazaki, K.; Kamikubo, Y. Generation of induced pluripotent stem cells from primary chronic myelogenous leukemia patient samples. Blood J. Am. Soc. Hematol. 2012, 119, 6234–6242. [Google Scholar] [CrossRef]
  130. Carette, J.E.; Pruszak, J.; Varadarajan, M.; Blomen, V.A.; Gokhale, S.; Camargo, F.D.; Wernig, M.; Jaenisch, R.; Brummelkamp, T.R. Generation of iPSCs from cultured human malignant cells. Blood J. Am. Soc. Hematol. 2010, 115, 4039–4042. [Google Scholar] [CrossRef]
  131. Kotini, A.G.; Carcamo, S.; Cruz-Rodriguez, N.; Olszewska, M.; Wang, T.; Demircioglu, D.; Chang, C.-J.; Bernard, E.; Chao, M.P.; Majeti, R. Patient-Derived iPSCs Faithfully Represent the Genetic Diversity and Cellular Architecture of Human Acute Myeloid Leukemia. Blood Cancer Discov. 2023, 4, 318–335. [Google Scholar] [CrossRef] [PubMed]
  132. Chao, M.P.; Gentles, A.J.; Chatterjee, S.; Lan, F.; Reinisch, A.; Corces, M.R.; Xavy, S.; Shen, J.; Haag, D.; Chanda, S. Human AML-iPSCs reacquire leukemic properties after differentiation and model clonal variation of disease. Cell Stem Cell 2017, 20, 329–344.e327. [Google Scholar] [CrossRef] [PubMed]
  133. Tu, J.; Huo, Z.; Yu, Y.; Zhu, D.; Xu, A.; Huang, M.-F.; Hu, R.; Wang, R.; Gingold, J.A.; Chen, Y.-H. Hereditary retinoblastoma iPSC model reveals aberrant spliceosome function driving bone malignancies. Proc. Natl. Acad. Sci. USA 2022, 119, e2117857119. [Google Scholar] [CrossRef] [PubMed]
  134. Suknuntha, K.; Ishii, Y.; Tao, L.; Hu, K.; McIntosh, B.E.; Yang, D.; Swanson, S.; Stewart, R.; Wang, J.Y.; Thomson, J. Discovery of survival factor for primitive chronic myeloid leukemia cells using induced pluripotent stem cells. Stem Cell Res. 2015, 15, 678–693. [Google Scholar] [CrossRef]
  135. Golubeva, D.; Porras, D.P.; Doyle, M.; Reid, J.C.; Tanasijevic, B.; Boyd, A.L.; Vojnits, K.; Elrafie, A.; Qiao, A.; Bhatia, M. Reprogramming of Acute Myeloid Leukemia Patients Cells: Harboring Cancer Mutations Requires Targeting of AML Hierarchy. Stem Cells Transl. Med. 2023, 12, 334–354. [Google Scholar] [CrossRef] [PubMed]
  136. Kotini, A.G.; Chang, C.-J.; Boussaad, I.; Delrow, J.J.; Dolezal, E.K.; Nagulapally, A.B.; Perna, F.; Fishbein, G.A.; Klimek, V.M.; Hawkins, R.D. Functional analysis of a chromosomal deletion associated with myelodysplastic syndromes using isogenic human induced pluripotent stem cells. Nat. Biotechnol. 2015, 33, 646–655. [Google Scholar] [CrossRef] [PubMed]
  137. Ye, Z.; Liu, C.F.; Lanikova, L.; Dowey, S.N.; He, C.; Huang, X.; Brodsky, R.A.; Spivak, J.L.; Prchal, J.T.; Cheng, L. Differential sensitivity to JAK inhibitory drugs by isogenic human erythroblasts and hematopoietic progenitors generated from patient-specific induced pluripotent stem cells. Stem Cells 2014, 32, 269–278. [Google Scholar] [CrossRef]
  138. Čančer, M.; Hutter, S.; Holmberg, K.O.; Rosén, G.; Sundström, A.; Tailor, J.; Bergström, T.; Garancher, A.; Essand, M.; Wechsler-Reya, R.J.; et al. Humanized Stem Cell Models of Pediatric Medulloblastoma Reveal an Oct4/mTOR Axis that Promotes Malignancy. Cell Stem Cell 2019, 25, 855–870. [Google Scholar] [CrossRef]
  139. Casciati, A.; Tanori, M.; Manczak, R.; Saada, S.; Tanno, B.; Giardullo, P.; Porcù, E.; Rampazzo, E.; Persano, L.; Viola, G. Human medulloblastoma cell lines: Investigating on cancer stem cell-like phenotype. Cancers 2020, 12, 226. [Google Scholar] [CrossRef]
  140. Azzarelli, R.; Simons, B.D.; Philpott, A. The developmental origin of brain tumours: A cellular and molecular framework. Development 2018, 145, dev162693. [Google Scholar] [CrossRef]
  141. Vatanmakanian, M.; Yousefi, H.; Mashouri, L.; Aref, A.R.; Khamisipour, G.; Bitaraf, A.; Alizadeh, S. Generation of Induced Pluripotent Cancer Cells from Glioblastoma Multiform Cell Lines. Cell. Reprogr. 2019, 21, 238–248. [Google Scholar] [CrossRef]
  142. Carrió, M.; Mazuelas, H.; Richaud-Patin, Y.; Gel, B.; Terribas, E.; Rosas, I.; Jimenez-Delgado, S.; Biayna, J.; Vendredy, L.; Blanco, I. Reprogramming captures the genetic and tumorigenic properties of neurofibromatosis type 1 plexiform neurofibromas. Stem Cell Rep. 2019, 12, 411–426. [Google Scholar] [CrossRef]
  143. Mazuelas, H.; Carrio, M.; Serra, E. Modeling tumors of the peripheral nervous system associated with Neurofibromatosis type 1: Reprogramming plexiform neurofibroma cells. Stem Cell Res. 2020, 49, 102068. [Google Scholar] [CrossRef] [PubMed]
  144. Bernhardt, M.; Novak, D.; Assenov, Y.; Orouji, E.; Knappe, N.; Weina, K.; Reith, M.; Larribere, L.; Gebhardt, C.; Plass, C. Melanoma-derived iPCCs show differential tumorigenicity and therapy response. Stem Cell Rep. 2017, 8, 1379–1391. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, H.J.; Jeong, J.; Park, S.; Jin, Y.-W.; Lee, S.-S.; Lee, S.B.; Choi, D. Establishment of hepatocellular cancer induced pluripotent stem cells using a reprogramming technique. Gut Liver 2017, 11, 261. [Google Scholar] [CrossRef]
  146. Saito, A.; Ochiai, H.; Okada, S.; Miyata, N.; Azuma, T. Suppression of Lefty expression in induced pluripotent cancer cells. FASEB J. 2013, 27, 2165–2174. [Google Scholar] [CrossRef]
  147. Noguchi, K.; Eguchi, H.; Konno, M.; Kawamoto, K.; Nishida, N.; Koseki, J.; Wada, H.; Marubashi, S.; Nagano, H.; Doki, Y. Susceptibility of pancreatic cancer stem cells to reprogramming. Cancer Sci. 2015, 106, 1182–1187. [Google Scholar] [CrossRef] [PubMed]
  148. Hoshino, H.; Nagano, H.; Haraguchi, N.; Nishikawa, S.; Tomokuni, A.; Kano, Y.; Fukusumi, T.; Saito, T.; Ozaki, M.; Sakai, D. Hypoxia and TP53 deficiency for induced pluripotent stem cell-like properties in gastrointestinal cancer. Int. J. Oncol. 2012, 40, 1423–1430. [Google Scholar]
  149. Kim, J.; Hoffman, J.P.; Alpaugh, R.K.; Rhim, A.D.; Reichert, M.; Stanger, B.Z.; Furth, E.E.; Sepulveda, A.R.; Yuan, C.-X.; Won, K.-J. An iPSC line from human pancreatic ductal adenocarcinoma undergoes early to invasive stages of pancreatic cancer progression. Cell Rep. 2013, 3, 2088–2099. [Google Scholar] [CrossRef]
  150. Chiou, S.-H.; Wang, M.-L.; Chou, Y.-T.; Chen, C.-J.; Hong, C.-F.; Hsieh, W.-J.; Chang, H.-T.; Chen, Y.-S.; Lin, T.-W.; Hsu, H.-S. Coexpression of Oct4 and Nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell–like properties and epithelial–mesenchymal transdifferentiation. Cancer Res. 2010, 70, 10433–10444. [Google Scholar] [CrossRef]
  151. Marin Navarro, A.; Susanto, E.; Falk, A.; Wilhelm, M. Modeling cancer using patient-derived induced pluripotent stem cells to understand development of childhood malignancies. Cell Death Discov. 2018, 4, 7. [Google Scholar] [CrossRef] [PubMed]
  152. Hosain, S.B.; Khiste, S.K.; Uddin, M.B.; Vorubindi, V.; Ingram, C.; Zhang, S.; Hill, R.A.; Gu, X.; Liu, Y.-Y. Inhibition of glucosylceramide synthase eliminates the oncogenic function of p53 R273H mutant in the epithelial-mesenchymal transition and induced pluripotency of colon cancer cells. Oncotarget 2016, 7, 60575. [Google Scholar] [CrossRef]
  153. O’Brien, C.A.; Kreso, A.; Jamieson, C.H. Cancer stem cells and self-renewal. Clin. Cancer Res. 2010, 16, 3113–3120. [Google Scholar] [CrossRef]
  154. Huang, S.-D.; Yuan, Y.; Tang, H.; Liu, X.-H.; Fu, C.-G.; Cheng, H.-Z.; Bi, J.-W.; Yu, Y.-W.; Gong, D.-J.; Zhang, W. Tumor cells positive and negative for the common cancer stem cell markers are capable of initiating tumor growth and generating both progenies. PLoS ONE 2013, 8, e54579. [Google Scholar] [CrossRef] [PubMed]
  155. Kusienicka, A.; Bukowska-Strakova, K.; Cieśla, M.; Nowak, W.N.; Bronisz-Budzyńska, I.; Seretny, A.; Żukowska, M.; Jeż, M.; Krutyhołowa, R.; Taha, H. Heme Oxygenase-1 Has a Greater Effect on Melanoma Stem Cell Properties Than the Expression of Melanoma-Initiating Cell Markers. Int. J. Mol. Sci. 2022, 23, 3596. [Google Scholar] [CrossRef]
  156. Stewart, J.M.; Shaw, P.A.; Gedye, C.; Bernardini, M.Q.; Neel, B.G.; Ailles, L.E. Phenotypic heterogeneity and instability of human ovarian tumor-initiating cells. Proc. Natl. Acad. Sci. USA 2011, 108, 6468–6473. [Google Scholar] [CrossRef] [PubMed]
  157. Corominas-Faja, B.; Cufi, S.; Oliveras-Ferraros, C.; Cuyas, E.; López-Bonet, E.; Lupu, R.; Alarcon, T.; Vellon, L.; Iglesias, J.M.; Leis, O. Nuclear reprogramming of luminal-like breast cancer cells generates Sox2-overexpressing cancer stem-like cellular states harboring transcriptional activation of the mTOR pathway. Cell Cycle 2013, 12, 3109. [Google Scholar] [CrossRef]
  158. Gilson, P.; Merlin, J.-L.; Harlé, A. Deciphering tumour heterogeneity: From tissue to liquid biopsy. Cancers 2022, 14, 1384. [Google Scholar] [CrossRef]
  159. Bang, J.S.; Choi, N.Y.; Lee, M.; Ko, K.; Park, Y.S.; Ko, K. Reprogramming of cancer cells into induced pluripotent stem cells questioned. Int. J. Stem Cells 2019, 12, 430–439. [Google Scholar] [CrossRef]
  160. Hu, K.; Yu, J.; Suknuntha, K.; Tian, S.; Montgomery, K.; Choi, K.-D.; Stewart, R.; Thomson, J.A.; Slukvin, I.I. Efficient generation of transgene-free induced pluripotent stem cells from normal and neoplastic bone marrow and cord blood mononuclear cells. Blood J. Am. Soc. Hematol. 2011, 117, e109–e119. [Google Scholar] [CrossRef]
  161. Konishi, H.; Mohseni, M.; Tamaki, A.; Garay, J.P.; Croessmann, S.; Karnan, S.; Ota, A.; Wong, H.Y.; Konishi, Y.; Karakas, B. Mutation of a single allele of the cancer susceptibility gene BRCA1 leads to genomic instability in human breast epithelial cells. Proc. Natl. Acad. Sci. USA 2011, 108, 17773–17778. [Google Scholar] [CrossRef] [PubMed]
  162. Soyombo, A.A.; Wu, Y.; Kolski, L.; Rios, J.J.; Rakheja, D.; Chen, A.; Kehler, J.; Hampel, H.; Coughran, A.; Ross, T.S. Analysis of induced pluripotent stem cells from a BRCA1 mutant family. Stem Cell Rep. 2013, 1, 336–349. [Google Scholar] [CrossRef] [PubMed]
  163. Shankar, A.S.; Du, Z.; Tejeda Mora, H.; Boers, R.; Cao, W.; van den Bosch, T.P.; Korevaar, S.S.; Boers, J.; van IJcken, W.F.; Bindels, E.M. Kidney organoids are capable of forming tumors, but not teratomas. Stem Cells 2022, 40, 577–591. [Google Scholar] [CrossRef] [PubMed]
  164. Tan, Y.; Ooi, S.; Wang, L. Immunogenicity and tumorigenicity of pluripotent stem cells and their derivatives: Genetic and epigenetic perspectives. Curr. Stem Cell Res. Ther. 2014, 9, 63–72. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, Y.; Wang, D.; Chen, M.; Yang, B.; Zhang, F.; Cao, K. Intramyocardial transplantation of undifferentiated rat induced pluripotent stem cells causes tumorigenesis in the heart. PLoS ONE 2011, 6, e19012. [Google Scholar] [CrossRef]
  166. Griscelli, F.; Féraud, O.; Oudrhiri, N.; Gobbo, E.; Casal, I.; Chomel, J.-C.; Biéche, I.; Duvillard, P.; Opolon, P.; Turhan, A.G. Malignant Germ Cell–Like Tumors, Expressing Ki-1 Antigen (CD30), Are Revealed during in Vivo Differentiation of Partially Reprogrammed Human-Induced Pluripotent Stem Cells. Am. J. Pathol. 2012, 180, 2084–2096. [Google Scholar] [CrossRef] [PubMed]
  167. Ohnishi, K.; Semi, K.; Yamamoto, T.; Shimizu, M.; Tanaka, A.; Mitsunaga, K.; Okita, K.; Osafune, K.; Arioka, Y.; Maeda, T. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 2014, 156, 663–677. [Google Scholar] [CrossRef]
  168. Nishi, M.; Sakai, Y.; Akutsu, H.; Nagashima, Y.; Quinn, G.; Masui, S.; Kimura, H.; Perrem, K.; Umezawa, A.; Yamamoto, N. Induction of cells with cancer stem cell properties from nontumorigenic human mammary epithelial cells by defined reprogramming factors. Oncogene 2014, 33, 643–652. [Google Scholar] [CrossRef]
  169. Zheng, Z.; Jian, J.; Zhang, X.; Zara, J.N.; Yin, W.; Chiang, M.; Liu, Y.; Wang, J.; Pang, S.; Ting, K. Reprogramming of human fibroblasts into multipotent cells with a single ECM proteoglycan, fibromodulin. Biomaterials 2012, 33, 5821–5831. [Google Scholar] [CrossRef]
  170. Ben-David, U.; Nudel, N.; Benvenisty, N. Immunologic and chemical targeting of the tight-junction protein Claudin-6 eliminates tumorigenic human pluripotent stem cells. Nat. Commun. 2013, 4, 1992. [Google Scholar] [CrossRef]
  171. Kim, A.; Lee, S.-Y.; Chung, S.-K. Caffeic acid selectively eliminates teratogenic human-induced pluripotent stem cells via apoptotic cell death. Phytomedicine 2022, 102, 154144. [Google Scholar] [CrossRef]
  172. Tang, C.; Lee, A.S.; Volkmer, J.-P.; Sahoo, D.; Nag, D.; Mosley, A.R.; Inlay, M.A.; Ardehali, R.; Chavez, S.L.; Pera, R.R. An antibody against SSEA-5 glycan on human pluripotent stem cells enables removal of teratoma-forming cells. Nat. Biotechnol. 2011, 29, 829–834. [Google Scholar] [CrossRef]
  173. Kim, A.; Lee, S.-Y.; Kim, B.-Y.; Chung, S.-K. Elimination of teratogenic human induced pluripotent stem cells by bee venom via calcium-calpain pathway. Int. J. Mol. Sci. 2020, 21, 3265. [Google Scholar] [CrossRef]
  174. Itakura, G.; Kawabata, S.; Ando, M.; Nishiyama, Y.; Sugai, K.; Ozaki, M.; Iida, T.; Ookubo, T.; Kojima, K.; Kashiwagi, R. Fail-safe system against potential tumorigenicity after transplantation of iPSC derivatives. Stem Cell Rep. 2017, 8, 673–684. [Google Scholar] [CrossRef] [PubMed]
  175. Kim, A.; Lee, S.-Y.; Seo, C.-S.; Chung, S.-K. Ethanol extract of Magnoliae cortex (EEMC) limits teratoma formation of pluripotent stem cells by selective elimination of undifferentiated cells through the p53-dependent mitochondrial apoptotic pathway. Phytomedicine 2020, 69, 153198. [Google Scholar] [CrossRef] [PubMed]
  176. Itakura, G.; Kobayashi, Y.; Nishimura, S.; Iwai, H.; Takano, M.; Iwanami, A.; Toyama, Y.; Okano, H.; Nakamura, M. Controlling immune rejection is a fail-safe system against potential tumorigenicity after human iPSC-derived neural stem cell transplantation. PLoS ONE 2015, 10, e0116413. [Google Scholar] [CrossRef]
  177. Isoda, M.; Sanosaka, T.; Tomooka, R.; Mabuchi, Y.; Shinozaki, M.; Andoh-Noda, T.; Banno, S.; Mizota, N.; Yamaguchi, R.; Okano, H. Mesenchymal properties of iPSC-derived neural progenitors that generate undesired grafts after transplantation. Commun. Biol. 2023, 6, 611. [Google Scholar] [CrossRef] [PubMed]
  178. Okubo, T.; Iwanami, A.; Kohyama, J.; Itakura, G.; Kawabata, S.; Nishiyama, Y.; Sugai, K.; Ozaki, M.; Iida, T.; Matsubayashi, K. Pretreatment with a γ-secretase inhibitor prevents tumor-like overgrowth in human iPSC-derived transplants for spinal cord injury. Stem Cell Rep. 2016, 7, 649–663. [Google Scholar] [CrossRef]
  179. Palechor-Ceron, N.; Krawczyk, E.; Dakic, A.; Simic, V.; Yuan, H.; Blancato, J.; Wang, W.; Hubbard, F.; Zheng, Y.-L.; Dan, H. Conditional reprogramming for patient-derived cancer models and next-generation living biobanks. Cells 2019, 8, 1327. [Google Scholar] [CrossRef]
  180. Liu, X.; Krawczyk, E.; Suprynowicz, F.A.; Palechor-Ceron, N.; Yuan, H.; Dakic, A.; Simic, V.; Zheng, Y.-L.; Sripadhan, P.; Chen, C. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat. Protoc. 2017, 12, 439–451. [Google Scholar] [CrossRef]
  181. Zhong, M.; Fu, L. Culture and application of conditionally reprogrammed primary tumor cells. Gastroenterol. Rep. 2020, 8, 224–233. [Google Scholar] [CrossRef] [PubMed]
  182. Sette, G.; Salvati, V.; Giordani, I.; Pilozzi, E.; Quacquarini, D.; Duranti, E.; De Nicola, F.; Pallocca, M.; Fanciulli, M.; Falchi, M. Conditionally reprogrammed cells (CRC) methodology does not allow the in vitro expansion of patient-derived primary and metastatic lung cancer cells. Int. J. Cancer 2018, 143, 88–99. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exploiting shared molecular characteristics between iPSCs and cancer cells for the generation of cancer stem cells. (A) The shared factors not only support the self-renewal, pluripotency, and maintenance of iPSCs but also partake in oncogenesis in various cancers (details in Section 2.1). (B) The direct reprogramming of bulk cancer cells into cancer stem cells can occur through two main methods—one involves techniques like that of transfection that induces the expression of stemness-promoting proteins [25,26] (details in Section 3) or triggers factor-dependent reprogramming [27] (details in Section 5). The alternative exposure-based method relies on hydrogel-activated reprogramming [28] or nuclear transfer techniques [29] to drive CSC formation (details in Section 5).
Figure 1. Exploiting shared molecular characteristics between iPSCs and cancer cells for the generation of cancer stem cells. (A) The shared factors not only support the self-renewal, pluripotency, and maintenance of iPSCs but also partake in oncogenesis in various cancers (details in Section 2.1). (B) The direct reprogramming of bulk cancer cells into cancer stem cells can occur through two main methods—one involves techniques like that of transfection that induces the expression of stemness-promoting proteins [25,26] (details in Section 3) or triggers factor-dependent reprogramming [27] (details in Section 5). The alternative exposure-based method relies on hydrogel-activated reprogramming [28] or nuclear transfer techniques [29] to drive CSC formation (details in Section 5).
Cells 13 00125 g001
Figure 2. Utilizing iPSC technology in cancer stem cell modeling. (A) Exposure-based approaches to CSC model derivation from iPSCs are presented. The first approach involves co-culturing normal iPSCs with cancer cells, while the second strategy entails culturing iPSCs in conditioned media from cancer cells. The iPSCs exposed to tumor microenvironment factors acquire distinct CSC features and demonstrate the ability to form tumors in vivo. (B) The reprogramming of cancer cells is depicted, acknowledging the hierarchical structure of tumors. Cell sorting enables the separation of CSC marker-expressing cells from bulk tumor cells, though the presence of CSC markers may not always correlate with CSC features (non-CSCs). Following the reprogramming of non-stem cancer cells, some cancer-derived iPSCs (cancer-iPSCs) may exhibit enhanced CSC features. The reprogrammed cell population is called cancer-iPSCs, induced pluripotent cancer cells (iPCCs), or induced pluripotent cancer stem cells (iPCSCs).
Figure 2. Utilizing iPSC technology in cancer stem cell modeling. (A) Exposure-based approaches to CSC model derivation from iPSCs are presented. The first approach involves co-culturing normal iPSCs with cancer cells, while the second strategy entails culturing iPSCs in conditioned media from cancer cells. The iPSCs exposed to tumor microenvironment factors acquire distinct CSC features and demonstrate the ability to form tumors in vivo. (B) The reprogramming of cancer cells is depicted, acknowledging the hierarchical structure of tumors. Cell sorting enables the separation of CSC marker-expressing cells from bulk tumor cells, though the presence of CSC markers may not always correlate with CSC features (non-CSCs). Following the reprogramming of non-stem cancer cells, some cancer-derived iPSCs (cancer-iPSCs) may exhibit enhanced CSC features. The reprogrammed cell population is called cancer-iPSCs, induced pluripotent cancer cells (iPCCs), or induced pluripotent cancer stem cells (iPCSCs).
Cells 13 00125 g002
Table 1. Molecular commonalities between iPSCs and cancer cells.
Table 1. Molecular commonalities between iPSCs and cancer cells.
ProteinPutative Function in iPSCs Function in Cancer Cells Refs.
Oct4Reprogramming, gene expression regulation, maintenance of pluripotency, activation of Nanog Tumor promotion and maintenance, chemoresistance [30,31,32,33,34,35,36,37,38,39,40]
Sox2Cooperation with OCT4, reprogramming, gene regulation, regulation of pluripotency, activation of Nanog Oncogenic initiation and maintenance, chemoresistance [41,42,43,44,45]
Klf4Stem cell renewal and maintenance, cooperation with OCT4 & SOX2, activation of Nanog Suppression of oncogenic activity, promotion of cancer-related inflammation [46,47,48,49,50,51,52]
c-MycEnhancing efficiency of iPSC generationTumor initiation, disruption of transcription [53,54,55,56,57]
NanogMaintenance of pluripotencyChemoresistance, cancer stem cell regeneration, hypoxia-induced angiogenesis [58,59,60,61]
Glis1Pro-reprogramming functionCell growth, enhancement of invasiveness and migration [62,63,64,65]
LIN28Enhancement of reprogramming frequency Oncogenic initiation and maintenance, metastasis, chemoresistance, suppression of tumor suppressor genes [66,67,68,69,70,71]
Table 2. In vitro conversion of iPSCs into CSC-like cells through exposure to the tumor microenvironment.
Table 2. In vitro conversion of iPSCs into CSC-like cells through exposure to the tumor microenvironment.
Identity of iPSC Cancer Cell Lines (Tissue Origin) CSC Features Examined (Method) Findings Refs.
Mouse iPSCs (miPSCs)Huh7 (liver)CSC marker expression (RT-qPCR) High expressions of CD24, CD133, and CD44 [97]
In vivo tumorigenicity (inhalation and liver orthotopic injection) miPS Huh7-CM cells gave rise to nine malignant tumors out of nine mice
Self-renewal potential (sphere-formation assay) Self-renewal potential was confirmed
In vitro invasion and migration capacity (transwell and wound-healing assays) Invasive ability of miPS Huh7-CM cells was enhanced upon engraftment
miPSCsPK-8 and KLM-1 (pancreas)CSC marker expression (RT-qPCR) Upregulation of CD133, CD24a, and EpCAM [98]
In vivo tumorigenicity (subcutaneous transplantation) CM-based CSCs generated tumors in nine out of nine mice
miPSCsLLC (lung), P19 (embryonal), B16 (melanoma), MC.E12 (mammary gland) In vivo tumorigenicity (subcutaneous transplantation) Mouse allografts formed undifferentiated carcinomas [13]
Self-renewal potential (sphere-formation assay) Self-renewal potential was confirmed
miPSCsLLC (lung) In vivo tumorigenicity (subcutaneous transplantation) Tumors derived from miPS LLC-CM grew without necrotic features[99]
Self-renewal potential (sphere-formation assay) Self-renewal potential was confirmed
miPSCsT47D (breast), BT549 (breast)CSC marker expression (RT-qPCR) High expression of CD133 [100]
In vivo tumorigenicity (subcutaneous injection) Tumors with a high nuclear to cytoplasmic ratio and poorly-differentiated glandular structures
Self-renewal potential (sphere-formation assay) Self-renewal potential was confirmed
In vitro invasion and migration capacity (transwell assay) CSCcmT47D CAFLCs and CSCcm BT549 CAFLCs were highly invasive
miPSCsLLC (lung)In vivo tumorigenicity (subcutaneous injection) CSCs were capable of developing a liposarcoma that exhibited phenotypic heterogeneity [106]
In vitro invasion capacity (Matrigel invasion assay) Compared with the parental miPS LLCev cells, the invasive capacities of miPS LLCevPT (primary tumor) and miPS LLCevDT (disseminated liposarcoma) cells were significantly higher
Self-renewal potential (sphere-formation assay) Self-renewal potential was confirmed
Human iPSCs (hiPSCs) U87MG (brain) CSC marker expression (immunocytochemistry and RT-qPCR) Overexpression of CD133, CD44, ABCG2, and ABCC2 [109]
Table 3. Cancer cell reprogramming strategies and the evaluation of pluripotency induction.
Table 3. Cancer cell reprogramming strategies and the evaluation of pluripotency induction.
Species Cancer Cells Reprogramming Method Reprogramming Factors Pluripotency Features TumorigenicityRefs.
Human HCT-15 and SK-CO-1 cell lines (colorectal adenocarcinoma) Retroviral transduction OCT4, SOX2, KLF4, and c-MYC Trilineage differentiation, downregulation of pluripotency genes (incomplete reprogramming) - [126]
Human Patient-derived juvenile myelomonocytic leukemia (JMML) cells Doxycycline-inducible lentivirus OCT4, SOX2, KLF4, and c-MYCEndogenous pluripotency markers (NANOG, OCT4, DNMT3B, REX1), formation of three germ-cell layers in teratomas - [20]
Human Patient-derived imatinib-sensitive chronic myelogenous leukemia (CML) cells Retroviral transduction Oct4, Sox2, Klf4, and c-Myc Pluripotency markers (SSEA-4 and Tra-1-60), teratoma formation capacity was confirmed - [129]
Human KBM7 cell line (blast crisis stage of CML) Retroviral transduction OCT4, SOX2, KLF4, and c-MYC Pluripotency markers (Tra-1-81 and OCT4 and CD9), formation of all three germ-cell layers - [130]
Human Patient-derived acute myeloid leukemia (AML) cells Non-integrating Sendai virus Oct4, Sox2, Klf4, and c-Myc Pluripotency markers (SSEA-4 & TRA-1-81), formation of teratoma - [132]
MouseR545 cell line (melanoma)Doxycycline-inducible lentivirusOct4, Klf4, and c-MycESC-like colonies, demethylation of Oct4 and Nanog promoters, and teratoma formationChimeric mice developed from pluripotent cancer cells remained tumor-free [24]
HumanT731 and T653 cell lines (glioblastoma)Retroviral vectorOct4, Sox2, and Klf4Pluripotency marker expression-[141]
Human HT-144 and A375 (melanoma cell lines), WM266.4 (BRAFV600D mutant cell line), SK-MEL147 (NRAS mutant cell line), Mewo (BRAF and NRAS wild-type cell line) Lentiviral polycistronic vector OCT4, SOX2, and KLF4 Pluripotency markers (NANOG, SOX2, and SALL4), formation of teratoma iPCCs showed variable tumorigenicity and differentiated into non-tumorigenic lineages[144]
Mouse RAS+/ink4a/Arf-/- melanoma cell Melanoma neuclei transferred to enucleated oocyte --High tumor incidence in chimeric mice [29]
Human DLD-1, HT-29, TE-10, MKN45, MIAPaCa-2, PANC-1, PLC, and HuCCT-1 cell lines (various gastrointestinal cancers)Lentiviral and retroviral vectors OCT4, SOX2, KLF4, and c-MYC NANOG expression, Ssea-4, Tra-1-60, Tra-1-81, and Tra-2-49 surface antigens Differentiated iPCCs showed decreased in vivo tumorigenicity[23]
Human SW480 and DLD-1 cell lines (colorectal cancer) Retroviral vectorOCT4, SOX2, and KLF4 Self-renewal (in terms of CSC properties) Sustained tumorigenicity[21]
HumanHepG2, Hep3B, Huh7, and PLC cell lines (liver cancer) Retroviral vectorOCT4, SOX2, KLF4, and c-MYC Hep3B cells acquired similar characteristics to pluripotent stem cells-[145]
HumanPLC/PRF/5 cell line (hepatoma)Lentiviral and retroviral vectors OCT4, SOX2, KLF4, and c-MYC Rex1 and Nanog expression, trilineage differentiation potentialColony-forming tumorigenic iPCCs[146]
Human PANC1 cell line (pancreatic ductal adenocarcinoma)Retroviral or lentiviral vectors OCT4, SOX2, KLF4, and c-MYC Endogenous Nanog and Tra-1-60 positive for ALP activity, differentiation into germ layer derivatives -[147]
HumanHTB-9 and T24 cell lines (bladder cancer)Sendai virusOCT4, SOX2, KLF4, and c-MYC Reprogrammed T24 cells showed epithelial-like morphology, colony-forming ability, expression of pluripotency-associated markers, and differentiation capacity-[16]
HumanPatient-derived pancreatic ductal adenocarcinoma (PDAC) cellsLentiviral vectorsOct4, Sox2, Klf4, and c-MycExpression of pluripotency markers NANOG, OCT4, and SSEA4, teratoma formation, and expression of three germ layer markersOne iPSC line progressed from early to late stages of PDAC[149]
HumanA549 cell line (lung carcinoma)Lentiviral vectorOct4 and NanogStemness-related gene expression, self-renewalIn vivo tumorigenic and metastatic abilities[150]
Human Patient-derived plexiform neurofibroma cellsRetroviral vector OCT4, SOX2, KLF4, and c-MYC Pluripotency markers (TRA-1-81, SSEA3, and SSEA4), teratoma formation NF1−/− iPSC-derived Schwann cells exhibited a continuous high proliferation rate and formed 3D spheres [142]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sarker, D.B.; Xue, Y.; Mahmud, F.; Jocelyn, J.A.; Sang, Q.-X.A. Interconversion of Cancer Cells and Induced Pluripotent Stem Cells. Cells 2024, 13, 125. https://doi.org/10.3390/cells13020125

AMA Style

Sarker DB, Xue Y, Mahmud F, Jocelyn JA, Sang Q-XA. Interconversion of Cancer Cells and Induced Pluripotent Stem Cells. Cells. 2024; 13(2):125. https://doi.org/10.3390/cells13020125

Chicago/Turabian Style

Sarker, Drishty B., Yu Xue, Faiza Mahmud, Jonathan A. Jocelyn, and Qing-Xiang Amy Sang. 2024. "Interconversion of Cancer Cells and Induced Pluripotent Stem Cells" Cells 13, no. 2: 125. https://doi.org/10.3390/cells13020125

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

Article Metrics

Back to TopTop