Tumor suppressor genes (TSGs) are thought to suppress tumor development primarily via cancer cell-autonomous mechanisms. However, the tumor microenvironment (TME) also significantly influences tumorigenesis. In this context, a role for TSGs in the various cell types of the TME in regulating tumor growth is emerging. Indeed, expression analyses of TSGs in clinical samples, along with data from mouse models in which TSGs were deleted selectively in the TME, indicate a functional role for them in tumor development. In this Perspective, using TP53 and PTEN as examples, we posit that TSGs play a significant role in cells of the TME in regulating tumor development, and postulate both a ‘pro-active’ and ‘reactive’ model for their contribution to tumor growth, dependent on the temporal sequence of initiating events. Finally, we discuss the need to consider a 2-in-1 cancer-treatment strategy to improve the efficacy of clearance of cancer cells and the cancer-promoting TME.

Oncogene activation and tumor suppressor gene (TSG; Box 1) inactivation are considered to be the primary causal events in the transformation of normal cells en route to malignancy (Solomon et al., 2010; Hanahan and Weinberg, 2011). Alterations in their expression are often sufficient to promote cellular proliferation or inhibit apoptosis, eventually resulting in cellular transformation, thereby underscoring their cell-autonomous roles in the tumorigenic process (Hanahan and Weinberg, 2011; Zhu et al., 2015). Not surprisingly, most cancer genome sequencing efforts have focused on cataloguing such alterations, and molecular therapies target these tumor-cell-specific alterations (Guo et al., 2014). However, it is increasingly clear that the tumor microenvironment (TME; Box 1) that surrounds tumor cells (Anderson and Simon, 2020) impacts on tumor initiation, progression, metastasis and therapy resistance (Valkenburg et al., 2018; de Visser and Joyce, 2023). Recent studies have also alluded to a significant role for adipocytes and myofibroblasts in prognosticating overall survival in breast cancers (Li et al., 2021; Lau et al., 2023). Interestingly, modifications in stromal gene expression, especially of TSGs, have been postulated to affect neighboring tumor cells (Moinfar et al., 2000; Scherz-Shouval et al., 2014; Wu et al., 2022). For instance, loss of tumor suppressor functions in the stroma (Box 1) may either directly affect tumor cell properties or, alternatively, lead to pre-malignant changes in the adjacent epithelial cells (Bhowmick et al., 2004; Wu et al., 2022), although these effects could be cell-type specific. Whether the expression and functions of oncogenes and TSGs in the stromal cells (Box 1) of the TME play a significant role in the tumorigenic process has not been unequivocally established. We focus here on the role of two commonly mutated TSGs in the TME, i.e. TP53 and PTEN (Table 1; Box 2), and highlight their significant contribution to the tumor development process. Since fibroblasts are the most abundant cell type in the TME, most studies have investigated the role of TP53 and PTEN in these cells, as a representative of the TME. Although TP53 and PTEN are the most extensively researched TSGs in this context, similar but limited findings have been made with other TSGs, such as RB1 (Mercier et al., 2008; Pickard et al., 2012; Kitajima et al., 2020).

Box 1. Tumor microenvironment

Cancer-associated fibroblasts (CAFs): Fibroblasts or cells with mesenchymal-like features that are the most common cell type of TME in solid tumors. Their presence is often associated with poor prognosis, resistance to therapies, and disease recurrence.

Stroma: Supporting or connective tissue consisting of mesenchymal cells such as fibroblasts, immune cells, vasculature network, extracellular matrix, pericytes and adipocytes. In this Perspective, stroma refers to the niche surrounding the epithelial cells or tumor cells. In some cases, the stromal element is specified like stromal fibroblast. Otherwise, stroma refers to the whole compartment around the epithelial or tumor cells.

Stromal cells of TME: Major class of cellular components in TME including fibroblasts, mesenchymal stem cells, adipocytes, endothelial cells, pericytes, osteoblasts and astrocytes.

Tumor-associated stroma: Stromal niche around tumor cells.

Tumor microenvironment (TME): Complex cellular system that surrounds a tumor assisting its growth and expansion. It comprises the non-malignant cellular and non-cellular components, including immune cells, stromal cells, extracellular matrix, blood vessels, lymphatic vessels, cytokines and other non-cellular components.

Tumor suppressor genes (TSGs): Genes that prevent cells from growing uncontrollably and becoming cancerous.

Box 2. Glossary

APCmin-driven colon cancer model: Multiple intestinal neoplasia (min), is a mutation in the murine adenomatous polyposis coli (APC) gene that when engineered in a mouse model predisposes the animals to colorectal and intestinal tumors.

APR-246 (also known as eprenetapopt): Small molecule that reactivates mutant TP53 and selectively induces apoptosis in TP53-mutant cancer cells.

ATM (ataxia telangiectasia mutated): Serine/threonine protein kinase that is involved in the repair of double-strand DNA breaks. Mutations in ATM are associated with increased cancer development.

ATR (ataxia telangiectasia and Rad3-related): Serine/threonine protein kinase that is active during the S phase of the cell cycle and orchestrates the response to replication stress to maintain genome integrity.

ERBB2 (erb-b2 receptor tyrosine kinase 2; also known as HER2, NEU): Receptor tyrosine-protein kinase that controls cell growth and division.

KrasG12D:Kras proto-oncogene with a mutation in codon 12. The GTPase KRAS plays role in cell proliferation and survival.

Carbon tetrachloride-induced liver cancer model: Continual administration of carbon tetrachloride results in hepatocyte injury that eventually leads to liver cancer development.

Lysyl oxidases: Family of enzymes that are involved in shaping the structure of the extracellular matrix. Their enzymatic activity is also associated with regulation of many cellular functions including proliferation, survival and differentiation.

MCF-7: Human breast cancer cell line that comprises estrogen, progesterone and glucocorticoid receptors.

miR-21: MicroRNA (miRNA)-21 is one of the most abundant microRNAs in many mammalian cells. It is an oncogenic miRNA that targets many tumor suppressors. MicroRNAs are short non-coding RNA molecules that regulate eukaryotic gene expression at the post-transcriptional level.

PARP (poly [ADP-ribose] polymerase 1): DNA-binding enzyme involved in the DNA-repair pathway for single strand breaks.

PLK1 (polo-like kinase 1): PLKs are a family of serine/threonine protein kinases. PLK1 plays an important role in the initiation, maintenance and completion of mitosis. Overexpression of PLK1 has been found in a variety of human cancers.

TP53 (tumor protein p53): Well-characterized tumor suppressor protein that is encoded by TP53, the most frequently mutated gene in human cancers.

PTEN (phosphatase and tensin homolog): Protein phosphatase with tumor suppressor activity. PTEN plays role in cell growth and survival and is commonly mutated or deleted in different types of cancers.

PTEN−miR-320−ETS2 axis: A regulatory axis discovered in the stroma of breast cancer tissue, where PTEN regulates the expression of mir-320 microRNA (miRNA-320). miR-320 consequently regulates gene expression of the ETS2 transcription factor. This axis is shown to regulate fibroblast secretome.

WEE1 (WEE1 G2 checkpoint kinase): Member of a serine/threonine protein kinases, causing G2-M cell-cycle arrest in response to DNA damage.

Synthetic lethality: Phenomenon whereby the concurrent deficiency of two genes results in cellular death, whereas deficiency of one individual gene alone does not cause cell death. Synthetic lethality is one of the most effective treatment strategies in cancer therapy.

Y220C mutant: Hot spot mutation in codon 220 of TP53 that results in decreased DNA binding and reduced TP53 transcriptional activity.

Table 1.

Consequences of TP53 and PTEN inactivation in different cells of the stroma on tumor growth

Inactivation of TP53 within the stroma
Cell type with TP53 deletion/mutationEffect on tumorType of cancerReferences
MEFs supportive Prostate, breast Addadi et al., 2010; Kiaris et al., 2005; Lafkas et al., 2008; Liu et al., 2020  
Fibroblasts supportive Breast Wu et al., 2022  
CAFs suppressive Lung Arandkar et al., 2018  
Hepatic stellate cells supportive Liver Lujambio et al., 2013  
Microphages supportive Colorectal He et al., 2015  
Inactivation of TP53 within the stroma
Cell type with TP53 deletion/mutationEffect on tumorType of cancerReferences
MEFs supportive Prostate, breast Addadi et al., 2010; Kiaris et al., 2005; Lafkas et al., 2008; Liu et al., 2020  
Fibroblasts supportive Breast Wu et al., 2022  
CAFs suppressive Lung Arandkar et al., 2018  
Hepatic stellate cells supportive Liver Lujambio et al., 2013  
Microphages supportive Colorectal He et al., 2015  
Inactivation of PTEN within the stroma
Cell type with PTEN deletionEffect on tumorType of cancerReferences
Fibroblasts supportive Prostate, breast Trimboli et al., 2009  
Macrophages supportive Pancreatic Wang et al., 2018  
Endothelial supportive Lung, melanoma Hamada et al., 2005  
Tregs suppressive Lung, melanoma, lymphoma Sharma et al., 2015  
Inactivation of PTEN within the stroma
Cell type with PTEN deletionEffect on tumorType of cancerReferences
Fibroblasts supportive Prostate, breast Trimboli et al., 2009  
Macrophages supportive Pancreatic Wang et al., 2018  
Endothelial supportive Lung, melanoma Hamada et al., 2005  
Tregs suppressive Lung, melanoma, lymphoma Sharma et al., 2015  

CAFs, cancer-associated fibroblasts; MEFs, mouse embryonic fibroblasts; Treg, regulatory T cells.

Several studies have demonstrated differential gene expression in tumor-associated stroma (Box 1) relative to normal stroma (Finak et al., 2008; Bauer et al., 2010; Navab et al., 2011). These differential expressions are associated with alterations, such as loss of heterozygosity, microsatellite instability and mutations (Eng et al., 2009), in a variety of solid tumors, including breast (Moinfar et al., 2000; Kurose et al., 2002), ovarian (Tuhkanen et al., 2004), head and neck (Bian et al., 2007), and bladder cancers (Paterson et al., 2003). For instance, frequent somatic mutations are found in TP53 and PTEN in the stroma of invasive breast carcinomas (Kurose et al., 2002). Besides genetic alterations, epigenetic changes − such as histone modification and miRNA regulation − also control gene expression in the stroma (Marks et al., 2016; Yang et al., 2023), such as in cancer-associated fibroblasts (CAFs; Box 1) (Vizoso et al., 2015; Xiao et al., 2016), immune cells (Janson et al., 2008) and endothelial cells (Chung et al., 2007). For example, stromal PTEN has been shown to be regulated by both hypermethylation and miRNA (Bian et al., 2012; Yang et al., 2014). In this Perspective, we will discuss in more detail the influence alterations in TP53 and PTEN within the TME have on tumor development.

TP53

TP53 is mutated or lost in >50% of solid tumors (Sabapathy and Lane, 2018) and is also mutated in the stromal compartment (Kurose et al., 2002; Hill et al., 2005; Patocs et al., 2007). For example, TP53 protein expression and function are often suppressed in CAFs (Hawsawi et al., 2008; Bar et al., 2009) and suppression of TP53 gene expression in stromal fibroblasts induces their transition to CAFs (Procopio et al., 2015). Silencing TP53 gene expression in cultured fibroblasts increased production of stromal cell-derived factor 1 (SDF-1, officially known as CXCL12) in these cells and promoted the migration and invasion of leukemic and osteosarcoma cells in vitro (Moskovits et al., 2006). Furthermore, co-injection of the human prostate epithelial cancer cell line PC3 with fibroblasts of different TP53 status into xenograft models also showed that fibroblast-specific loss of TP53 expression promotes tumor growth and metastasis in an SDF-1 dependent manner (Addadi et al., 2010). Likewise, the presence of mutant TP53 in fibroblasts promoted prostate cancer cell growth and metastasis (Liu et al., 2020). Similar findings were noted when using the human breast cancer cell line MCF-7 (Box 2) (Kiaris et al., 2005). However, TP53-null fibroblasts sensitize tumor cells to chemotherapeutic agents, such as doxorubicin and cisplatin (Lafkas et al., 2008), and radiotherapy (Burdelya et al., 2006), indicating that stromal TP53 also influences the response to therapy.

Consistent with the ex vivo findings, selective deletion of TP53 in fibroblasts, together with the expression of mutated oncogene KrasG12D (Box 2) in the epithelium, promoted the formation of mammary tumors in genetically modified mice by creating an immunosuppressive TME (Wu et al., 2022). This effect was not observed when mammary epithelial cells expressed the oncogene ErbB2 (Box 2) (Wu et al., 2022), indicating that the effects of ablation in stromal TP53 occur in an oncogene-specific manner. In particular, loss of TP53 in fibroblasts led to the reprogramming of gene expression in not only the fibroblasts but, more interestingly, in the adjacent epithelial cells (Wu et al., 2022), supporting the idea that loss of tumor suppressor functions in the TME is sufficient to promote genetic alteration in neighboring tumor cells. Similar results have been noted with TP53 deletion in other cell types of the TME, such as macrophages (He et al., 2015) or hepatic stellate cells (Lujambio et al., 2013), in an APCmin-driven colon cancer model (Box 2) or a carbon-tetrachloride-induced liver cancer model (Box 2), respectively.

Contradictory to the established tumor-suppressive role of TP53, an intriguing observation indicated that TP53 function is required to maintain the tumor-promoting phenotype induced by CAFs, in which the secretome is significantly altered by TP53. In this case, CAF TP53 is re-wired to become tumor-supportive, as its depletion in CAFs significantly reduced lung-tumor growth in a xenograft model (Arandkar et al., 2018). This finding highlights a different function of TP53 in naive versus activated fibroblasts, where not only tumor-suppressive functionality of TP53 is quenched but TP53 undergoes alterations that confer it a tumor-supportive function.

PTEN

PTEN is frequently disrupted in sporadic tumors and has profound impacts on tumor progression (Thies et al., 2019). Besides tumor cells, loss of PTEN gene expression has been reported in the stroma of breast (Kurose et al., 2002; Trimboli et al., 2009), prostate (Ashida et al., 2012) and pancreatic cancers (Pitarresi et al., 2018), and is associated with worse outcome in the patients. Immunohistochemical staining has shown that PTEN is lost in stroma of ∼25% of breast cancer patients (Trimboli et al., 2009). Indeed, loss of heterozygosity and point mutations of PTEN have been reported in stromal fibroblasts of breast cancers (Kurose et al., 2001, 2002). Similarly, loss of PTEN expression has been reported in 20% of stromal fibroblasts of pancreatic cancer samples associated with hemizygous deletion of PTEN and chromosome 10 monosomy (Wartenberg et al., 2016). Besides genetic alterations, upregulation of miR-21 (Box 2) was reported as another mechanism leading to loss of PTEN protein expression in the collective stromal compartment of pancreatic cancer (Wartenberg et al., 2016).

As well as the above observations in patients, several studies have modeled PTEN loss in the stroma and investigated the impact on tumor progression. PTEN knockdown in patient-derived CAFs accelerated human pancreatic ductal adenocarcinoma tumor cell growth in an orthotopic co-injection mouse model (Pitarresi et al., 2018). Conditional knockout of PTEN in fibroblasts of a mouse breast cancer model (MMTV-ErbB2) altered the expression profile of these cells and accelerated initiation and progression of mammary tumor (Trimboli et al., 2009). Mechanistically, deletion of PTEN in stromal fibroblasts led to increased extracellular matrix deposition and innate immune cell infiltration via the PTEN−miR-320−ETS2 axis (Box 2) (Trimboli et al., 2009; Bronisz et al., 2011).

PTEN deletion in fibroblasts has also been shown to affect adjacent epithelial cells and lead to the expansion of mammary epithelial stem cell enriched populations (Sizemore et al., 2017). This occurs together with the downregulation of the DNA-repair response in the adjacent mammary epithelial cells, thereby promoting genome instability in the epithelial compartment (Sizemore et al., 2018). These effects on mammary epithelial cells, although insufficient to drive tumorigenesis, increase the risk of breast cancer initiation after a second carcinogenic hit (Sizemore et al., 2018). Similarly, tissue-specific deletion of PTEN in transgenic mice showed that collective stromal PTEN expression restrains endometrial carcinogenesis in mice in which PTEN has been deleted within uterine epithelial (Liang et al., 2018).

PTEN deletion in other components of the TME also impacts tumorigenesis. In macrophages, loss of PTEN induced a pro-tumorigenic M2 polarization, which led to increased pancreatic cell metastasis to lung (Wang et al., 2018). Furthermore, heterozygous PTEN deficiency in endothelial cells of transgenic mice enhanced tumor angiogenesis in melanoma and lung tumors (Hamada et al., 2005).

In contrast to the studies highlighted above, in which stromal PTEN has an inhibitory impact on tumor progression, PTEN expression in regulatory T cells has been shown to stabilize them within the tumor and maintain an immunosuppressive TME (Sharma et al., 2015). In this case, PTEN deletion in regulatory T cells reduces the growth of lung cancer, melanoma and lymphoma. Therefore, stromal PTEN has cell-type specific functions in tumor suppression and, surprisingly, tumor progression.

The currently available literature indicates that, in most cell types of the stroma, TSG inactivation promotes tumor development. However, whether this is a pro-active initiating event or a reactive consequence of the tumor milieu, is yet to be unequivocally established (Fig. 1). Whereas the reactive model is easier to envision because of DNA-damage or carcinogen-induced cellular transformation that then affects the stroma, the pro-active model is probably due to changes in gradients induced by metabolites that may then affect TSG activity (Kong et al., 2024). As experimental validation of both contexts to establish a temporal series of events is currently limited, future work will shed light on support for these models.

Available literature provides arguments in favor of the possibility of both contexts of TSG inactivation in stromal cells: either in response to the oncogenic stress imposed by adjacent tumor cells resulting in a strong selective pressure for a TSG-deficient stroma during tumor progression or, alternatively, in the pro-active context by loss of TSGs in stromal cells that may precede their loss in tumor cells

Available literature provides arguments in favor of the possibility of both contexts of TSG inactivation in stromal cells: either in response to the oncogenic stress imposed by adjacent tumor cells resulting in a strong selective pressure for a TSG-deficient stroma during tumor progression or, alternatively, in the pro-active context by loss of TSGs in stromal cells that may precede their loss in tumor cells (Palumbo et al., 2015). In the reactive model, it can be envisaged that the initial presence of a pre-cancerous lesion leads to the transformation of the surrounding stroma through shuttling of oncogenic miRNAs − or even mutant proteins, such as mutant TP53 − via extra-cellular vesicles (Cooks et al., 2018; Bhatta et al., 2021) that, in turn, facilitate the malignant growth. Significant evidence for such influence of the tumor on the stromal transcriptome has been demonstrated (Aran et al., 2017; Lau et al., 2023). In the pro-active model, a permissive TME with reduced TSG activity can also be a fertile ground for easier transformation of embedded epithelial cells, as often seen in the case of familial cancers (Moinfar et al., 2000; Brosseau and Le, 2019). For example, somatic TP53 mutations in stromal fibroblasts have been shown to increase genome instability in adjacent epithelia and are associated with breast cancer and increased regional nodal metastasis (Moinfar et al., 2000; Patocs et al., 2007).

Fig. 1.

A ‘pro-active’ and ‘reactive’ role for TSGs in TME. In the ‘reactive model’, malignant tumor cells initiate a series of events that lead to the transformation and conversion of the stroma (via extracellular vesicles, soluble factors, etc.) to inactivate the TSGs in the cells of the stroma. This is a reaction from the stroma to the presence of the tumor cells, which then generally supports the growth of the tumor. In the ‘pro-active’ model, the initiating event is the inactivation of the TSG within the stromal cells (through germline mutations, epigenetic changes, carcinogenic signals, transient effects by metabolites, etc.), which then provides the signals to initiate changes that lead to the transformation of neighboring epithelial cells. Once initiated, the epithelial cells undergo transformation and continue their growth through the reactive model. Both models differ in the initiating event and, thus, are dictated by the temporal sequence of events. Treg, regulatory T cell.

Fig. 1.

A ‘pro-active’ and ‘reactive’ role for TSGs in TME. In the ‘reactive model’, malignant tumor cells initiate a series of events that lead to the transformation and conversion of the stroma (via extracellular vesicles, soluble factors, etc.) to inactivate the TSGs in the cells of the stroma. This is a reaction from the stroma to the presence of the tumor cells, which then generally supports the growth of the tumor. In the ‘pro-active’ model, the initiating event is the inactivation of the TSG within the stromal cells (through germline mutations, epigenetic changes, carcinogenic signals, transient effects by metabolites, etc.), which then provides the signals to initiate changes that lead to the transformation of neighboring epithelial cells. Once initiated, the epithelial cells undergo transformation and continue their growth through the reactive model. Both models differ in the initiating event and, thus, are dictated by the temporal sequence of events. Treg, regulatory T cell.

Long-term organoid co-culture models could be exploited to examine both possibilities experimentally, with the genetic manipulation of either the epithelial cells or stromal fibroblasts, together with single-cell genomic and transcriptomic analyses. Overall, we propose that both pro-active and reactive models are likely to be operational, depending on the contexts and initiating events.

Given the key alterations in the TSGs of TME cells, it is imperative that they are considered during cancer management […]. Targeting the malignant cells alone is insufficient to completely eradicate the disease.

Given the key alterations in the TSGs of TME cells, it is imperative that they are considered during cancer management, which is not the current clinical practice especially with molecular targeted therapies. Targeting the malignant cells alone is insufficient to completely eradicate the disease. Thus, a 2-in-1 approach of simultaneously targeting both the tumor and the stroma should be considered. To achieve that, more work needs to be done to establish the alterations in the TME, as has been done for the malignant cells. This will set the stage for molecular targeting of the TME. Given the continuing advances in genomic approaches, single-cell RNA and DNA analysis can provide the necessary information on the altered TSG and related pathways that can then be therapeutically targeted in the TME.

Currently, loss of TSG function, regardless of the cell type, is difficult to restore therapeutically. However, several approaches have been proposed to target inactivated TSGs, including synthetic lethality (Box 2) to induce death in cells with inactivated TSGs, as well as compounds that could restore functionality of the mutant protein. In the former case, synthetic lethal interactions between loss of TP53 function and inhibition of ATR (Box 2) (Reaper et al., 2011), or inhibition of PLK1 (Box 2) (Meng et al., 2013) or inhibition WEE1 (Box 2) (Bridges et al., 2011), amongst others have been noted. Similarly, loss of PTEN activity was synthetically lethal when coinciding with inhibition of PARP (Box 2) (Mendes-Pereira et al., 2009), ATM (Box 2) (McCabe et al., 2015) or lysyl oxidases (Box 2) (Chen et al., 2019), amongst others. As such, some of these inhibitors could be used in combination with both regular cancer therapy and inhibitors that target specific alterations found in cancer cells. In the latter, compounds that might restore functionality in mutant TSGs have been identified, primarily targeting mutant TP53. Some of them − such as APR-246 (Box 2) that targets many TP53 mutants, and rezatapopt that specifically targets the Y220C mutant TP53 (Box 2) − were shown to have efficacy in pre-clinical studies (Synnott et al., 2018; Dumbrava et al., 2022; Carter et al., 2023). These drugs could be combined with cancer treatment protocols if the molecular signatures of the TME alterations are known.

More effort is required to identify and classify factors and drugs that significantly impact TSG alterations. The combination of the ability to detect and the availability of drugs to target the TSG alterations is necessary to develop 2-in-1 approach during which both the tumor and TME cells are targeted concomitantly. This, we think is likely to result in eradication of cancer in its entirety.

TME is a key contributor to tumor development and significantly influences therapeutic response. […] TSGs have a significant contributory role in TME alteration and, consequently, tumor properties.

It is becoming increasingly evident that the TME is a key contributor to tumor development and significantly influences therapeutic response. By using TP53 and PTEN as prominent examples, this Perspective has highlighted that TSGs have a significant contributory role in TME alteration and, consequently, tumor properties. We also envisage that similar alterations in other TSGs, such as RB1, and oncogenes will contribute significantly to the altered TME. Thus, establishing whether alterations in TSGs (and oncogenes) in the TME are a prevalent and universal phenomenon in cancers is a critical first step in furthering efforts to target cancer for therapy. Determining the contexts in which the stroma can be a pro-active component that promotes tumorigenesis will allow the development of potential prophylactic strategies that slow down or even prevent the progression of malignancies.

Author contributions

K.S. developed the concept for the Perspective, and wrote it together with B.T., with inputs from H.L.

Funding

This work was supported by grants from Nanyang Technological University and the National Medical Research Council of Singapore to KS. Deposited in PMC for immediate release.

Addadi
,
Y.
,
Moskovits
,
N.
,
Granot
,
D.
,
Lozano
,
G.
,
Carmi
,
Y.
,
Apte
,
R. N.
,
Neeman
,
M.
and
Oren
,
M.
(
2010
).
p53 status in stromal fibroblasts modulates tumor growth in an SDF1-dependent manner
.
Cancer Res.
70
,
9650
-
9658
.
Anderson
,
N. M.
and
Simon
,
M. C.
(
2020
).
The tumor microenvironment
.
Curr. Biol.
30
,
R921
-
R925
.
Aran
,
D.
,
Camarda
,
R.
,
Odegaard
,
J.
,
Paik
,
H.
,
Oskotsky
,
B.
,
Krings
,
G.
,
Goga
,
A.
,
Sirota
,
M.
and
Butte
,
A. J.
(
2017
).
Comprehensive analysis of normal adjacent to tumor transcriptomes
.
Nat. Commun.
8
,
1077
.
Arandkar
,
S.
,
Furth
,
N.
,
Elisha
,
Y.
,
Nataraj
,
N. B.
,
Van Der Kuip
,
H.
,
Yarden
,
Y.
,
Aulitzky
,
W.
,
Ulitsky
,
I.
,
Geiger
,
B.
and
Oren
,
M.
(
2018
).
Altered p53 functionality in cancer-associated fibroblasts contributes to their cancer-supporting features
.
Proc. Natl. Acad. Sci. USA
115
,
6410
-
6415
.
Ashida
,
S.
,
Orloff
,
M. S.
,
Bebek
,
G.
,
Zhang
,
L.
,
Zheng
,
P.
,
Peehl
,
D. M.
and
Eng
,
C.
(
2012
).
Integrated analysis reveals critical genomic regions in prostate tumor microenvironment associated with clinicopathologic phenotypes
.
Clin. Cancer Res.
18
,
1578
-
1587
.
Bar
,
J.
,
Feniger-Barish
,
R.
,
Lukashchuk
,
N.
,
Shaham
,
H.
,
Moskovits
,
N.
,
Goldfinger
,
N.
,
Simansky
,
D.
,
Perlman
,
M.
,
Papa
,
M.
,
Yosepovich
,
A.
et al.
(
2009
).
Cancer cells suppress p53 in adjacent fibroblasts
.
Oncogene
28
,
933
-
936
.
Bauer
,
M.
,
Su
,
G.
,
Casper
,
C.
,
He
,
R.
,
Rehrauer
,
W.
and
Friedl
,
A.
(
2010
).
Heterogeneity of gene expression in stromal fibroblasts of human breast carcinomas and normal breast
.
Oncogene
29
,
1732
-
1740
.
Bhatta
,
B.
,
Luz
,
I.
,
Krueger
,
C.
,
Teo
,
F. X.
,
Lane
,
D. P.
,
Sabapathy
,
K.
and
Cooks
,
T.
(
2021
).
Cancer cells shuttle extracellular vesicles containing oncogenic mutant p53 proteins to the tumor microenvironment
.
Cancers
13
,
2985
.
Bhowmick
,
N. A.
,
Chytil
,
A.
,
Plieth
,
D.
,
Gorska
,
A. E.
,
Dumont
,
N.
,
Shappell
,
S.
,
Washington
,
M. K.
,
Neilson
,
E. G.
and
Moses
,
H. L.
(
2004
).
TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia
.
Science
303
,
848
-
851
.
Bian
,
Y.
,
Knobloch
,
T. J.
,
Sadim
,
M.
,
Kaklamani
,
V.
,
Raji
,
A.
,
Yang
,
G. Y.
,
Weghorst
,
C. M.
and
Pasche
,
B.
(
2007
).
Somatic acquisition of TGFBR1*6A by epithelial and stromal cells during head and neck and colon cancer development
.
Hum. Mol. Genet.
16
,
3128
-
3135
.
Bian
,
E. B.
,
Huang
,
C.
,
Ma
,
T. T.
,
Tao
,
H.
,
Zhang
,
H.
,
Cheng
,
C.
,
Lv
,
X. W.
and
Li
,
J.
(
2012
).
DNMT1-mediated PTEN hypermethylation confers hepatic stellate cell activation and liver fibrogenesis in rats
.
Toxicol. Appl. Pharmacol.
264
,
13
-
22
.
Bridges
,
K. A.
,
Hirai
,
H.
,
Buser
,
C. A.
,
Brooks
,
C.
,
Liu
,
H.
,
Buchholz
,
T. A.
,
Molkentine
,
J. M.
,
Mason
,
K. A.
and
Meyn
,
R. E.
(
2011
).
MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells
.
Clin. Cancer Res.
17
,
5638
-
5648
.
Bronisz
,
A.
,
Godlewski
,
J.
,
Wallace
,
J. A.
,
Merchant
,
A. S.
,
Nowicki
,
M. O.
,
Mathsyaraja
,
H.
,
Srinivasan
,
R.
,
Trimboli
,
A. J.
,
Martin
,
C. K.
,
Li
,
F.
et al.
(
2011
).
Reprogramming of the tumour microenvironment by stromal PTEN-regulated miR-320
.
Nat. Cell Biol.
14
,
159
-
167
.
Brosseau
,
J.-P.
and
Le
,
L. Q.
(
2019
).
Heterozygous tumor suppressor microenvironment in cancer development
.
Trends Cancer
5
,
541
-
546
.
Burdelya
,
L. G.
,
Komarova
,
E. A.
,
Hill
,
J. E.
,
Browder
,
T.
,
Tararova
,
N. D.
,
Mavrakis
,
L.
,
Dicorleto
,
P. E.
,
Folkman
,
J.
and
Gudkov
,
A. V.
(
2006
).
Inhibition of p53 response in tumor stroma improves efficacy of anticancer treatment by increasing antiangiogenic effects of chemotherapy and radiotherapy in mice
.
Cancer Res.
66
,
9356
-
9361
.
Carter
,
B. Z.
,
Mak
,
P. Y.
,
Ke
,
B.
,
Nishida
,
Y.
,
Boettcher
,
S.
,
Bedoy
,
A.
,
Ostermann
,
L. B.
,
Dinardo
,
C. D.
,
Puzio-Kuter
,
A.
and
Levine
,
A.
et al.
(
2023
).
Selective targeting of TP53-Y220C mutant AML by PC14586 results in TP53 wild-type conformation and synergistical apoptosis induction by concomitant inhibition of Xpo-1, MDM2, or Bcl-2
.
Blood
142
,
2261
.
Chen
,
P.
,
Zhao
,
D.
,
Li
,
J.
,
Liang
,
X.
,
Li
,
J.
,
Chang
,
A.
,
Henry
,
V. K.
,
Lan
,
Z.
,
Spring
,
D. J.
and
Rao
,
G.
et al.
(
2019
).
Symbiotic macrophage-glioma cell interactions reveal synthetic lethality in PTEN-null glioma
.
Cancer Cell
35
,
868
-
884.e6
.
Chung
,
I.
,
Karpf
,
A. R.
,
Muindi
,
J. R.
,
Conroy
,
J. M.
,
Nowak
,
N. J.
,
Johnson
,
C. S.
and
Trump
,
D. L.
(
2007
).
Epigenetic silencing of CYP24 in tumor-derived endothelial cells contributes to selective growth inhibition by calcitriol
.
J. Biol. Chem.
282
,
8704
-
8714
.
Cooks
,
T.
,
Pateras
,
I. S.
,
Jenkins
,
L. M.
,
Patel
,
K. M.
,
Robles
,
A. I.
,
Morris
,
J.
,
Forshew
,
T.
,
Appella
,
E.
,
Gorgoulis
,
V. G.
and
Harris
,
C. C.
(
2018
).
Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246
.
Nat. Commun.
9
,
771
.
De Visser
,
K. E.
and
Joyce
,
J. A.
(
2023
).
The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth
.
Cancer Cell
41
,
374
-
403
.
Dumbrava
,
E. E.
,
Johnson
,
M. L.
,
Tolcher
,
A. W.
,
Shapiro
,
G.
,
Thompson
,
J. A.
,
El-Khoueiry
,
A. B.
,
Vandross
,
A. L.
,
Kummar
,
S.
,
Parikh
,
A. R.
and
Munster
,
P. N.
(
2022
).
First-in-human study of PC14586, a small molecule structural corrector of Y220C mutant p53, in patients with advanced solid tumors harboring a TP53 Y220C mutation
.
J. Clin. Oncol.
40
,
3003
.
Eng
,
C.
,
Leone
,
G.
,
Orloff
,
M. S.
and
Ostrowski
,
M. C.
(
2009
).
Genomic alterations in tumor stroma
.
Cancer Res.
69
,
6759
-
6764
.
Finak
,
G.
,
Bertos
,
N.
,
Pepin
,
F.
,
Sadekova
,
S.
,
Souleimanova
,
M.
,
Zhao
,
H.
,
Chen
,
H.
,
Omeroglu
,
G.
,
Meterissian
,
S.
,
Omeroglu
,
A.
et al.
(
2008
).
Stromal gene expression predicts clinical outcome in breast cancer
.
Nat. Med.
14
,
518
-
527
.
Guo
,
X. E.
,
Ngo
,
B.
,
Modrek
,
A. S.
and
Lee
,
W. H.
(
2014
).
Targeting tumor suppressor networks for cancer therapeutics
.
Curr. Drug Targets
15
,
2
-
16
.
Hamada
,
K.
,
Sasaki
,
T.
,
Koni
,
P. A.
,
Natsui
,
M.
,
Kishimoto
,
H.
,
Sasaki
,
J.
,
Yajima
,
N.
,
Horie
,
Y.
,
Hasegawa
,
G.
,
Naito
,
M.
et al.
(
2005
).
The PTEN/PI3K pathway governs normal vascular development and tumor angiogenesis
.
Genes Dev.
19
,
2054
-
2065
.
Hanahan
,
D.
and
Weinberg
,
R. A.
(
2011
).
Hallmarks of cancer: the next generation
.
Cell
144
,
646
-
674
.
Hawsawi
,
N. M.
,
Ghebeh
,
H.
,
Hendrayani
,
S. F.
,
Tulbah
,
A.
,
Al-Eid
,
M.
,
Al-Tweigeri
,
T.
,
Ajarim
,
D.
,
Alaiya
,
A.
,
Dermime
,
S.
and
Aboussekhra
,
A.
(
2008
).
Breast carcinoma-associated fibroblasts and their counterparts display neoplastic-specific changes
.
Cancer Res.
68
,
2717
-
2725
.
He
,
X.-Y.
,
Xiang
,
C.
,
Zhang
,
C.-X.
,
Xie
,
Y.-Y.
,
Chen
,
L.
,
Zhang
,
G.-X.
,
Lu
,
Y.
and
Liu
,
G.
(
2015
).
p53 in the myeloid lineage modulates an inflammatory microenvironment limiting initiation and invasion of intestinal tumors
.
Cell Rep.
13
,
888
-
897
.
Hill
,
R.
,
Song
,
Y.
,
Cardiff
,
R. D.
and
Van Dyke
,
T.
(
2005
).
Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis
.
Cell
123
,
1001
-
1011
.
Janson
,
P. C.
,
Marits
,
P.
,
Thörn
,
M.
,
Ohlsson
,
R.
and
Winqvist
,
O.
(
2008
).
CpG methylation of the IFNG gene as a mechanism to induce immunosuppression [correction of immunosupression] in tumor-infiltrating lymphocytes
.
J. Immunol.
181
,
2878
-
2886
.
Kiaris
,
H.
,
Chatzistamou
,
I.
,
Trimis
,
G.
,
Frangou-Plemmenou
,
M.
,
Pafiti-Kondi
,
A.
and
Kalofoutis
,
A.
(
2005
).
Evidence for nonautonomous effect of p53 tumor suppressor in carcinogenesis
.
Cancer Res.
65
,
1627
-
1630
.
Kitajima
,
S.
,
Li
,
F.
and
Takahashi
,
C.
(
2020
).
Tumor milieu controlled by RB tumor suppressor
.
Int. J. Mol. Sci.
21
,
2450
.
Kong
,
L. R.
,
Gupta
,
K.
,
Wu
,
A. J.
,
Perera
,
D.
,
Ivanyi-Nagy
,
R.
,
Ahmed
,
S. M.
,
Tan
,
T. Z.
,
Tan
,
S. L.-W.
,
Fuddin
,
A.
and
Sundaramoorthy
,
E.
et al.
(
2024
).
A glycolytic metabolite bypasses “two-hit” tumor suppression by BRCA2
.
Cell
187
,
2269
-
2287.e16
.
Kurose
,
K.
,
Hoshaw-Woodard
,
S.
,
Adeyinka
,
A.
,
Lemeshow
,
S.
,
Watson
,
P. H.
and
Eng
,
C.
(
2001
).
Genetic model of multi-step breast carcinogenesis involving the epithelium and stroma: clues to tumour-microenvironment interactions
.
Hum. Mol. Genet.
10
,
1907
-
1913
.
Kurose
,
K.
,
Gilley
,
K.
,
Matsumoto
,
S.
,
Watson
,
P. H.
,
Zhou
,
X. P.
and
Eng
,
C.
(
2002
).
Frequent somatic mutations in PTEN and TP53 are mutually exclusive in the stroma of breast carcinomas
.
Nat. Genet.
32
,
355
-
357
.
Lafkas
,
D.
,
Trimis
,
G.
,
Papavassiliou
,
A. G.
and
Kiaris
,
H.
(
2008
).
P53 mutations in stromal fibroblasts sensitize tumors against chemotherapy
.
Int. J. Cancer
123
,
967
-
971
.
Lau
,
H. S. H.
,
Tan
,
V. K. M.
,
Tan
,
B. K. T.
,
Sim
,
Y.
,
Quist
,
J.
,
Thike
,
A. A.
,
Tan
,
P. H.
,
Pervaiz
,
S.
,
Grigoriadis
,
A.
and
Sabapathy
,
K.
(
2023
).
Adipose-enriched peri-tumoral stroma, in contrast to myofibroblast-enriched stroma, prognosticates poorer survival in breast cancers
.
NPJ Breast Cancer
9
,
84
.
Li
,
B.
,
Pei
,
G.
,
Yao
,
J.
,
Ding
,
Q.
,
Jia
,
P.
and
Zhao
,
Z.
(
2021
).
Cell-type deconvolution analysis identifies cancer-associated myofibroblast component as a poor prognostic factor in multiple cancer types
.
Oncogene
40
,
4686
-
4694
.
Liang
,
X.
,
Daikoku
,
T.
,
Terakawa
,
J.
,
Ogawa
,
Y.
,
Joshi
,
A. R.
,
Ellenson
,
L. H.
,
Sun
,
X.
and
Dey
,
S. K.
(
2018
).
The uterine epithelial loss of Pten is inefficient to induce endometrial cancer with intact stromal Pten
.
PLoS Genet.
14
,
e1007630
.
Liu
,
Q.
,
Yu
,
B.
,
Tian
,
Y.
,
Dan
,
J.
,
Luo
,
Y.
and
Wu
,
X.
(
2020
).
P53 mutant p53(N236S) regulates cancer-associated fibroblasts properties through Stat3 pathway
.
Onco Targets Ther.
13
,
1355
-
1363
.
Lujambio
,
A.
,
Akkari
,
L.
,
Simon
,
J.
,
Grace
,
D.
,
Tschaharganeh
,
D. F.
,
Bolden
,
J. E.
,
Zhao
,
Z.
,
Thapar
,
V.
,
Joyce
,
J. A.
and
Krizhanovsky
,
V.
et al.
(
2013
).
Non-cell-autonomous tumor suppression by p53
.
Cell
153
,
449
-
460
.
Marks
,
D. L.
,
Olson
,
R. L.
and
Fernandez-Zapico
,
M. E.
(
2016
).
Epigenetic control of the tumor microenvironment
.
Epigenomics
8
,
1671
-
1687
.
McCabe
,
N.
,
Hanna
,
C.
,
Walker
,
S. M.
,
Gonda
,
D.
,
Li
,
J.
,
Wikstrom
,
K.
,
Savage
,
K. I.
,
Butterworth
,
K. T.
,
Chen
,
C.
and
Harkin
,
D. P.
et al.
(
2015
).
Mechanistic rationale to target PTEN-deficient tumor cells with inhibitors of the DNA damage response kinase ATM
.
Cancer Res.
75
,
2159
-
2165
.
Mendes-Pereira
,
A.
,
Martin
,
S.
,
Brough
,
R.
,
McCarthy
,
A.
,
Taylor
,
J.
,
Kim
,
J.
,
Waldman
,
T.
,
Lord
,
C.
and
Ashworth
,
A.
(
2009
).
Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors
.
EMBO Mol. Med.
1
,
315
-
322
.
Meng
,
X.
,
Laidler
,
L. L.
,
Kosmacek
,
E. A.
,
Yang
,
S.
,
Xiong
,
Z.
,
Zhu
,
D.
,
Wang
,
X.
,
Dai
,
D.
,
Zhang
,
Y.
and
Wang
,
X.
et al.
(
2013
).
Induction of mitotic cell death by overriding G2/M checkpoint in endometrial cancer cells with non-functional p53
.
Gynecol. Oncol.
128
,
461
-
469
.
Mercier
,
I.
,
Casimiro
,
M. C.
,
Wang
,
C.
,
Rosenberg
,
A. L.
,
Quong
,
J.
,
Minkeu
,
A.
,
Allen
,
K. G.
,
Danilo
,
C.
,
Sotgia
,
F.
,
Bonuccelli
,
G.
et al.
(
2008
).
Human breast cancer-associated fibroblasts (CAFs) show caveolin-1 downregulation and RB tumor suppressor functional inactivation: implications for the response to hormonal therapy
.
Cancer Biol. Ther.
7
,
1212
-
1225
.
Moinfar
,
F.
,
Man
,
Y. G.
,
Arnould
,
L.
,
Bratthauer
,
G. L.
,
Ratschek
,
M.
and
Tavassoli
,
F. A.
(
2000
).
Concurrent and independent genetic alterations in the stromal and epithelial cells of mammary carcinoma: implications for tumorigenesis
.
Cancer Res.
60
,
2562
-
2566
.
Moskovits
,
N.
,
Kalinkovich
,
A.
,
Bar
,
J.
,
Lapidot
,
T.
and
Oren
,
M.
(
2006
).
p53 Attenuates cancer cell migration and invasion through repression of SDF-1/CXCL12 expression in stromal fibroblasts
.
Cancer Res.
66
,
10671
-
10676
.
Navab
,
R.
,
Strumpf
,
D.
,
Bandarchi
,
B.
,
Zhu
,
C. Q.
,
Pintilie
,
M.
,
Ramnarine
,
V. R.
,
Ibrahimov
,
E.
,
Radulovich
,
N.
,
Leung
,
L.
,
Barczyk
,
M.
et al.
(
2011
).
Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer
.
Proc. Natl. Acad. Sci. USA
108
,
7160
-
7165
.
Palumbo
,
A.
Jr
,
de Oliveira Meireles Da Costa
,
N.
,
Bonamino
,
M. H.
,
Pinto
,
L. F.
and
Nasciutti
,
L. E.
(
2015
).
Genetic instability in the tumor microenvironment: a new look at an old neighbor
.
Mol. Cancer
14
,
145
.
Paterson
,
R. F.
,
Ulbright
,
T. M.
,
MacLennan
,
G. T.
,
Zhang
,
S.
,
Pan
,
C. X.
,
Sweeney
,
C. J.
,
Moore
,
C. R.
,
Foster
,
R. S.
,
Koch
,
M. O.
,
Eble
,
J. N.
et al.
(
2003
).
Molecular genetic alterations in the laser-capture-microdissected stroma adjacent to bladder carcinoma
.
Cancer
98
,
1830
-
1836
.
Patocs
,
A.
,
Zhang
,
L.
,
Xu
,
Y.
,
Weber
,
F.
,
Caldes
,
T.
,
Mutter
,
G. L.
,
Platzer
,
P.
and
Eng
,
C.
(
2007
).
Breast-cancer stromal cells with TP53 mutations and nodal metastases
.
N. Engl. J. Med.
357
,
2543
-
2551
.
Pickard
,
A.
,
Cichon
,
A. C.
,
Barry
,
A.
,
Kieran
,
D.
,
Patel
,
D.
,
Hamilton
,
P.
,
Salto-Tellez
,
M.
,
James
,
J.
and
McCance
,
D. J.
(
2012
).
Inactivation of Rb in stromal fibroblasts promotes epithelial cell invasion
.
EMBO J.
31
,
3092
-
3103
.
Pitarresi
,
J. R.
,
Liu
,
X.
,
Avendano
,
A.
,
Thies
,
K. A.
,
Sizemore
,
G. M.
,
Hammer
,
A. M.
,
Hildreth
,
B. E.
, 3rd
,
Wang
,
D. J.
,
Steck
,
S. A.
,
Donohue
,
S.
et al.
(
2018
).
Disruption of stromal hedgehog signaling initiates RNF5-mediated proteasomal degradation of PTEN and accelerates pancreatic tumor growth
.
Life Sci. Alliance
1
,
e201800190
.
Procopio
,
M. G.
,
Laszlo
,
C.
,
Al Labban
,
D.
,
Kim
,
D. E.
,
Bordignon
,
P.
,
Jo
,
S. H.
,
Goruppi
,
S.
,
Menietti
,
E.
,
Ostano
,
P.
,
Ala
,
U.
et al.
(
2015
).
Combined CSL and p53 downregulation promotes cancer-associated fibroblast activation
.
Nat. Cell Biol.
17
,
1193
-
1204
.
Reaper
,
P. M.
,
Griffiths
,
M. R.
,
Long
,
J. M.
,
Charrier
,
J.-D.
,
MacCormick
,
S.
,
Charlton
,
P. A.
,
Golec
,
J. M.
and
Pollard
,
J. R.
(
2011
).
Selective killing of ATM-or p53-deficient cancer cells through inhibition of ATR
.
Nat. Chem. Biol.
7
,
428
-
430
.
Sabapathy
,
K.
and
Lane
,
D. P.
(
2018
).
Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others
.
Nat. Rev. Clin. Oncol.
15
,
13
-
30
.
Scherz-Shouval
,
R.
,
Santagata
,
S.
,
Mendillo
,
M. L.
,
Sholl
,
L. M.
,
Ben-Aharon
,
I.
,
Beck
,
A. H.
,
Dias-Santagata
,
D.
,
Koeva
,
M.
,
Stemmer
,
S. M.
,
Whitesell
,
L.
et al.
(
2014
).
The reprogramming of tumor stroma by HSF1 is a potent enabler of malignancy
.
Cell
158
,
564
-
578
.
Sharma
,
M. D.
,
Shinde
,
R.
,
McGaha
,
T. L.
,
Huang
,
L.
,
Holmgaard
,
R. B.
,
Wolchok
,
J. D.
,
Mautino
,
M. R.
,
Celis
,
E.
,
Sharpe
,
A. H.
,
Francisco
,
L. M.
et al.
(
2015
).
The PTEN pathway in Tregs is a critical driver of the suppressive tumor microenvironment
.
Sci. Adv.
1
,
e1500845
.
Sizemore
,
G. M.
,
Balakrishnan
,
S.
,
Hammer
,
A. M.
,
Thies
,
K. A.
,
Trimboli
,
A. J.
,
Wallace
,
J. A.
,
Sizemore
,
S. T.
,
Kladney
,
R. D.
,
Woelke
,
S. A.
,
Yu
,
L.
et al.
(
2017
).
Stromal PTEN inhibits the expansion of mammary epithelial stem cells through Jagged-1
.
Oncogene
36
,
2297
-
2308
.
Sizemore
,
G. M.
,
Balakrishnan
,
S.
,
Thies
,
K. A.
,
Hammer
,
A. M.
,
Sizemore
,
S. T.
,
Trimboli
,
A. J.
,
Cuitiño
,
M. C.
,
Steck
,
S. A.
,
Tozbikian
,
G.
,
Kladney
,
R. D.
et al.
(
2018
).
Stromal PTEN determines mammary epithelial response to radiotherapy
.
Nat. Commun.
9
,
2783
.
Solomon
,
H.
,
Brosh
,
R.
,
Buganim
,
Y.
and
Rotter
,
V.
(
2010
).
Inactivation of the p53 tumor suppressor. gene and activation of the Ras oncogene: cooperative events in tumorigenesis
.
Discov. Med.
9
,
448
-
454
.
Synnott
,
N. C.
,
Madden
,
S. F.
,
Bykov
,
V. J.
,
Crown
,
J.
,
Wiman
,
K. G.
and
Duffy
,
M. J.
(
2018
).
The mutant p53-targeting compound APR-246 induces ROS-modulating genes in breast cancer cells
.
Transl. Oncol.
11
,
1343
-
1349
.
Thies
,
K. A.
,
Lefler
,
J. E.
,
Leone
,
G.
and
Ostrowski
,
M. C.
(
2019
).
PTEN in the Stroma
.
Cold Spring Harb. Perspect. Med.
9
,
a036111
.
Trimboli
,
A. J.
,
Cantemir-Stone
,
C. Z.
,
Li
,
F.
,
Wallace
,
J. A.
,
Merchant
,
A.
,
Creasap
,
N.
,
Thompson
,
J. C.
,
Caserta
,
E.
,
Wang
,
H.
,
Chong
,
J. L.
et al.
(
2009
).
Pten in stromal fibroblasts suppresses mammary epithelial tumours
.
Nature
461
,
1084
-
1091
.
Tuhkanen
,
H.
,
Anttila
,
M.
,
Kosma
,
V. M.
,
Ylä-Herttuala
,
S.
,
Heinonen
,
S.
,
Kuronen
,
A.
,
Juhola
,
M.
,
Tammi
,
R.
,
Tammi
,
M.
and
Mannermaa
,
A.
(
2004
).
Genetic alterations in the peritumoral stromal cells of malignant and borderline epithelial ovarian tumors as indicated by allelic imbalance on chromosome 3p
.
Int. J. Cancer
109
,
247
-
252
.
Valkenburg
,
K. C.
,
De Groot
,
A. E.
and
Pienta
,
K. J.
(
2018
).
Targeting the tumour stroma to improve cancer therapy
.
Nat. Rev. Clin. Oncol.
15
,
366
-
381
.
Vizoso
,
M.
,
Puig
,
M.
,
Carmona
,
F. J.
,
Maqueda
,
M.
,
Velásquez
,
A.
,
Gómez
,
A.
,
Labernadie
,
A.
,
Lugo
,
R.
,
Gabasa
,
M.
,
Rigat-Brugarolas
,
L. G.
et al.
(
2015
).
Aberrant DNA methylation in non-small cell lung cancer-associated fibroblasts
.
Carcinogenesis
36
,
1453
-
1463
.
Wang
,
X.
,
Luo
,
G.
,
Zhang
,
K.
,
Cao
,
J.
,
Huang
,
C.
,
Jiang
,
T.
,
Liu
,
B.
,
Su
,
L.
and
Qiu
,
Z.
(
2018
).
Hypoxic tumor-derived exosomal miR-301a mediates M2 macrophage polarization via PTEN/PI3Kγ to promote pancreatic cancer metastasis
.
Cancer Res.
78
,
4586
-
4598
.
Wartenberg
,
M.
,
Centeno
,
I.
,
Haemmig
,
S.
,
Vassella
,
E.
,
Zlobec
,
I.
,
Galván
,
J. A.
,
Neuenschwander
,
M.
,
Schlup
,
C.
,
Gloor
,
B.
,
Lugli
,
A.
et al.
(
2016
).
PTEN alterations of the stromal cells characterise an aggressive subpopulation of pancreatic cancer with enhanced metastatic potential
.
Eur. J. Cancer
65
,
80
-
90
.
Wu
,
J.
,
Liu
,
X.
,
Reeser
,
J. A. W.
,
Trimboli
,
A. J.
,
Pécot
,
T.
,
Sizemore
,
G. M.
,
Naidu
,
S. K.
,
Fernandez
,
S. A.
,
Yu
,
L.
,
Hallett
,
M.
et al.
(
2022
).
Stromal p53 regulates breast cancer development, the immune landscape, and survival in an oncogene-specific manner
.
Mol. Cancer Res.
20
,
1233
-
1246
.
Xiao
,
Q.
,
Zhou
,
D.
,
Rucki
,
A. A.
,
Williams
,
J.
,
Zhou
,
J.
,
Mo
,
G.
,
Murphy
,
A.
,
Fujiwara
,
K.
,
Kleponis
,
J.
,
Salman
,
B.
et al.
(
2016
).
Cancer-associated fibroblasts in pancreatic cancer are reprogrammed by tumor-induced alterations in genomic DNA methylation
.
Cancer Res.
76
,
5395
-
5404
.
Yang
,
T. S.
,
Yang
,
X. H.
,
Chen
,
X.
,
Wang
,
X. D.
,
Hua
,
J.
,
Zhou
,
D. L.
,
Zhou
,
B.
and
Song
,
Z. S.
(
2014
).
MicroRNA-106b in cancer-associated fibroblasts from gastric cancer promotes cell migration and invasion by targeting PTEN
.
FEBS Lett.
588
,
2162
-
2169
.
Yang
,
J.
,
Xu
,
J.
,
Wang
,
W.
,
Zhang
,
B.
,
Yu
,
X.
and
Shi
,
S.
(
2023
).
Epigenetic regulation in the tumor microenvironment: molecular mechanisms and therapeutic targets
.
Signal Transduct. Target Ther.
8
,
210
.
Zhu
,
K.
,
Liu
,
Q.
,
Zhou
,
Y.
,
Tao
,
C.
,
Zhao
,
Z.
,
Sun
,
J.
and
Xu
,
H.
(
2015
).
Oncogenes and tumor suppressor genes: comparative genomics and network perspectives
.
BMC Genomics
16 Suppl 7
,
S8
.

Competing interests

The authors declare no competing or financial interests.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.