Abstract
Our understanding of the function of the transcriptional regulators YAP/TAZ in cancer is advancing. In this Review, we provide an update on recent progress in YAP/TAZ biology, their regulation by Hippo signaling and mechanotransduction, and highlight open questions. YAP/TAZ signaling is an addiction shared by multiple tumor types and their microenvironments, providing many malignant attributes. As such, it represents an important vulnerability that may offer a broad window of therapeutic efficacy, and here we give an overview of the current treatment strategies and pioneering clinical trials.
Multiple solid malignancies display accumulation and activation of YAP/TAZ, paralogous genes encoding for transcriptional co-activators related to Drosophila Yorkie. Their activation positively correlates with malignancy, relapse, metastasis, lower overall survival and chemoresistance. Genetic evidence in mouse models and human cancer cells reveals that YAP/TAZ activity is essential for tumor initiation and progression (Table 1). These studies suggest that YAP/TAZ may represent master transcriptional regulators of cancer and their engagement by cancer cells may serve as an epigenetic "switch" to enable phenotypic plasticity, a fluid cell state essential to adapt to nutrient deprivation, escape immune attack, and resist stresses associated with the metastatic cascade. Accumulating evidence implies that YAP/TAZ also act as a nexus by which tumor cells can reprogram their surrounding ecosystem, including fibroblasts, immune and endothelial cells, into a resilient, growth-promoting and immunologically cold tumor microenvironment (TME). As such, YAP/TAZ are appealing candidates as orchestrators of “tumor morphogenesis” by acting on the tumor's epithelial and stromal components.
Table 1. Consequences of YAP/TAZ activation across cancer types.
| Patient data | Murine cancer models | Human cancer models |
|---|---|---|
| Breast Cancer (BC) | ||
| Gene signatures for YAP/TAZ activity correlate with poor differentiation, enrichment of cancer stem cell signatures, metastasis proclivity, and poor patient outcome (reviewed in 42). TAZ protein expression and nuclear localization is associated to poor differentiation and it is prognostic of poor clinical outcome (reviewed in 42). |
Genetic loss of Yap greatly increases the latency while reducing the growth and metastatic proclivity of mammary tumors arising in MMTV-PyMT mice6. YAP/TAZ deletion abolishes tumor initiation triggered by APC knockout in the mammary gland2. YAP overexpression confers metastatic abilities to otherwise benign mouse mammary cancer cells122. |
TAZ knockdown suppresses tumor formation by MCF7 cells (reviewed in 42). TAZ overactivation confers CSC and malignant properties to otherwise benign MCF10AT cells, whereas TAZ knockdown reduces tumor growth of the malignant MCF10ACA1 cell line57. TAZ is required for metastasis formation by primary human BC cells injected in the orthotopic site107; YAP is required for metastasis formation by intracardiacal-injected HCC1954 or MDA-MB-231 cells131. |
| Non-Small Cell Lung Cancer (NSCLC) | ||
| Gene signatures for YAP/TAZ activity correlate with poor patient outcome (reviewed in 42). Cells with elevated YAP and TAZ protein expression are enriched in lung adenocarcinomas (LUADs, 76% of cases) and lung squamous cell carcinomas (LUSCCs, 65%of cases), respectively, and are associated with shorter patient survival (reviewed in 42). |
In mice, YAP overexpression in KrasG12D-induced lung adenomas promotes their progression to LUAD; conversely, YAP knockout inhibits tumor progression from adenoma to LUAD and LUSCC in the KrasG12D/Lkb1KO mouse model59. | TAZ depletion represses lung tumor formation by tail vein-injected A549 LUAD cells (reviewed in 42). YAP depletion suppresses brain metastasis formation by intracardiacal-injected H2030 LUAD cells131. |
| Colo-Rectal Cancer (CRC) | ||
| Gene signatures for YAP/TAZ activity correlate with poor patient outcome (reviewed in 191 and in 42). YAP/TAZ mRNA and protein expression are upregulated in CRCs compared to the normal tissue, and elevated YAP/TAZ expression is prognostic of shorter patient survival (reviewed in 42). | YAP or TAZ knockout severely compromises tumor initiation in the intestine of APCmin mice63,69. | YAP is required for the growth of tumors formed by HCT116 cells in the orthotopic site and by SW620 cells in xenografts146,147. |
| Gastric cancer (GC) | ||
| YAP/TAZ protein expression is upregulated in GCs compared to the normal tissue, and elevated YAP/TAZ expression is prognostic of shorter patient survival (reviewed in 42). | A peptide inhibitor of YAP/TAZ-TEAD interaction strongly decreases gastric adenoma formation induced by the mutagen MNNG combined with Helicobacter pylori infection (reviewed in 42). | YAP depletion inhibits tumor growth and metastatic dissemination of SGC7901 cells injected in the orthotopic site192. |
| Hepatocellular Carcinoma (HCC) | ||
| High levels of nuclear YAP are found in about 66% of cases, and this is associated with poor differentiation and short patient survival (reviewed in 42). | In vivo depletion of YAP via siRNAs causes regression of advanced HCC lesions induced by sporadic MST1/2 knockout in hepatocytes (reviewed in 42). | YAP depletion reduces tumor growth in a PDX model of HCC 193. |
| Pancreatic Adenocarcinoma (PDAC) | ||
| Nuclear YAP is detected in 90% of cases and its overexpression correlates with shorter patient survival194. | YAP/TAZ are required for pancreatic tumor initiation and progression to PDAC in the KRasG12D/p53KO mouse model27,104 | YAP depletion suppresses tumor initiation by xenografts of BxPC-3 cells194. |
| Prostate Cancer (PC) | ||
| YAP and TAZ are nuclear in the majority of PCs (>60%), and the frequency of cells with nuclear YAP/TAZ increases with grade; elevated YAP expression is predictive of poor patient outcome195,196 | YAP overexpression in the prostate epithelium is sufficient to induce age-related PC induction (reviewed in 42). | YAP depletion impairs the growth of xenografted C4-2 cells (reviewed in 42). TAZ depletion impairs the growth of xenografted PC-3 cells195. |
| Cutaneous Squamous Cell Carcinoma (CSCC) | ||
| YAP/TAZ are nuclear in the majority of cases197,198 | YAP/TAZ knockout impedes CSCC formation induced either by chemical carcinogenesis or by forced expression of oncogenic KRAS in the epidermis91,197. | YAP depletion suppresses the growth of tumors formed by xenografted A431 cells198. |
| Basal Cell Carcinoma (BCC) | ||
| YAP and TAZ are found in the nucleus of BCC cells in >90% of patients197,198 | YAP/TAZ are required for the development of BCCs triggered by overactivation of the shh pathway in epidermal basal cells197,199. | |
| Melanoma (MEL) | ||
| Cutaneous (CMEL): YAP is found in the nuclei of benign nevi and CMELs (reviewed in 42). In lymph node (LN)-positive CMELs, elevated nuclear YAP in the cells of the invasive front correlates with distant metastasis proclivity125. The pro-metastatic invasive cell state of CMEL is driven by YAP/TAZ/TEAD (reviewed in 42). Uveal (UMEL): nuclear YAP is found in the majority of cells of Gαq/11-mutant tumors, enlisting about 66% of UMELs26,46 |
YAP depletion impairs lymph node metastasis of the mouse CMEL cell line B16F10125 | YAP or TAZ depletion inhibits metastatic colonization of the lung by tail-vein injected 1205Lu CMEL cells(reviewed in 42), and the growth of some PDX models of CMEL200 YAP depletion impairs the growth of tumors formed by xenografted Gαq/11-mutant UMEL cells26,46 |
| Glioblastoma Multiforme (GBM) | ||
| TAZ expression is prognostic of shorter patient survival (reviewed in 42). Nuclear TAZ is invariably found in the cells surrounding the necrotic core of GBMs; expression of a YAP/TAZ-driven GBM stem cell signature is prognostic of shorter patient survival101 |
YAP/TAZ depletion abolishes GBM initiation in the orthotopic site by oncogene-transformed glioma cells, and by GL261 and CT2A mouse GBM cell lines101 | YAP/TAZ depletion abolishes tumor initiation in the orthotopic site by primary GBM cell lines 101 (see also 42). |
| Osteosarcoma (OS) | ||
| Nuclear YAP is found in 46% of OSs and is prognostic of worse patient outcome201. | YAP depletion inhibits subcutaneous growth of primary mouse OS cells (reviewed in 42) | YAP depletion inhibits subcutaneous growth of MG-63 and of HOS cells (reviewed in 42). |
| Soft-Tissue Sarcoma (STS) | ||
| Nuclear YAP or TAZ staining is found in 55% and 33% of STSs, respectively202. | YAP depletion reduces subcutaneous growth of tumors formed by primary mouse STS cells (reviewed in 42). | |
| Rhabdomyosarcoma (RMS) | ||
| Nuclear YAP is detected in 36% of Alveolar RMS (ARMS) and in 73% of Embryonal RMS (ERMS) (reviewed in 42) | YAP overexpression in satellite cells induces the formation of ERMS (reviewed in 42). | YAP depletion reduces tumor growth by xenografted RD ERMS cells (reviewed in42). |
YAP/TAZ as transcriptional determinants
YAP and TAZ do not bind DNA directly but require DNA-binding partners in order to associate to cognate cis-regulatory elements on chromatin. In the large majority of normal and cancer cells, these partners are the TEAD family members (reviewed in Ref. 1). The YAP/TAZ-TEAD complexes are mainly recruited to enhancer elements, and, in a minority of cases, to promoters1 (Figure 1a). These regulatory elements are typically co-bound by AP-1 transcriptional complexes (heterodimers of FOS and JUN family members), which are required for the transcriptional activation of a relevant portion of YAP/TAZ target genes1. YAP/TAZ promote the recruitment of general transcriptional coactivators, such as MED1, BRD4 and CDK9, to enhancers (Figure 1a). This induces the formation of liquid-liquid phase separated bodies, essential for transcriptional responses1. Once bound to enhancers, YAP/TAZ induces the recruitment of RNA polymerase II (POLII) at the promoters of YAP/TAZ target genes, at least in part by favouring the recruitment of BRD4 at the same loci. In fact, YAP/TAZ dictate the genome-wide association of BRD4 to chromatin, endowing YAP/TAZ-bound enhancers the same functional properties of superenhancers2. In addition, recent evidence indicates that YAP/TAZ can feed their own transcriptional responses by Tet1-mediated rewiring of TEAD-responsive enhancers3.
Figure 1. YAP/TAZ regulation inside and outside the nucleus.
a) Regulation of YAP/TAZ transcriptional activities inside the nucleus. YAP/TAZ are transcriptional co-factors that are recruited to enhancer elements on chromatin by TEAD proteins, and act in concert with other transcription factors (e.g. AP-1) to assemble chromatin modifiers and readers, ultimately promoting Pol II elongation on the promoter elements of cognate genes. In absence of mechanical stimulation, the SWI/SNF complex interacts with YAP/TAZ, impeding their binding to TEADs. Mechanical tension on the cells induces the formation of nuclear actin filaments that sequester SWI/SNF complexes away from YAP/TAZ, thus contributing to YAP/TAZ activation.
b) YAP/TAZ regulation by mechanotransduction and Hippo signaling. Cells respond to ECM rigidity by adjusting their tensional state through integrin-mediated cell–ECM Focal adhesions (FAs). FAs connect the ECM with the intracellular F-actin cytoskeleton in a manner involving the FAK and Src kinases and cells increases actomyosin tension and restructures its entire cytoskeleton in a process requiring Rho-GTPases (such as Rho or Rac1), myosin motors and ROCK. The actomyosin tension transduces physical inputs into modulation of YAP/TAZ nuclear–cytoplasmic shuttling. The F-actin cytoskeleton impacts also on the mechanics and shape of the nucleus through LINC complexes, favoring YAP/TAZ nuclear entry by inducing nuclear deformation. RhoGEFs, Rho guanidine nucleotide exchange factors; ROCK, Rho-associated protein kinase; FAK, Focal adhesion Kinase; LINC, Linker of nucleoskeleton and cytoskeleton.
The Hippo pathway inhibits YAP/TAZ either through direct phosphorylation of these proteins by LATS1/2, or indirectly, by influencing the actin cytoskeleton. In turn, acto-myosin contractility can affect LATS1/2 activity trough the GTPase RAP2.
Regulation of YAP/TAZ by the Hippo pathway
The Hippo pathway is a cascade composed of two kinases, that is MST1/2 and LATS1/2, and of their respective cofactors, Sav1 and Mob1A/B4 (Figure 1b). Other components of the Hippo pathway include the adaptor protein NF2, and the MAP4K4 family and the PTPN14 phosphatase. MST1/2 or MAP4K4 phosphorylate and activate LATS1/2, which in turn phosphorylate YAP/TAZ; this is considered an inhibitory step promoting YAP/TAZ cytoplasmic sequestration and protein degradation4. Yet, to what extent the direct phosphorylation of YAP/TAZ by LATS1/2 is relevant for their inhibition at endogenous protein levels and in vivo still awaits genetic validation5.
As shown in Table 2, some human tumor types display genetic alterations in components of the Hippo pathway (reviewed in Ref. 4). This includes NF2 mutations or deletions in mesotheliomas, medulloblastomas and schwannomas; NF2 and LATS1/2 mutations in renal cell carcinomas; and Sav deletions in cholangiocarcinomas. These genetic alterations occur at various frequencies and despite consistently elevated YAP/TAZ activation, even in these tumors types genetic alterations are absent in the majority of cases. For all other tumors, in spite of pervasive and widespread YAP/TAZ hyperactivation, genetic loss of the Hippo pathway is either extremely rare or non-existing; and, irrespectively of genetics, there is actually scant evidence of changes in the level of active, phosphorylated LATS1/2 in tumors compared to normal tissues. Thus, YAP/TAZ activity is induced in human tumors without requiring the inactivation of Hippo pathway components. Of note, even in mouse models, loss of Hippo pathway components is either inconsequential or insufficient for tumor formation in several adult tissues, such as intestine, mammary gland, pancreas, lung, and skin6–11. And yet, tumors induced by transgenic oncogenes in these same tissues display elevated YAP/TAZ activity and require YAP/TAZ for their emergence.
Table 2. Human tumors displaying genetic alterations in Hippo pathway components or in YAP/TAZ.
| Affected gene | Consequence on YAP/TAZ enrichment | Functional Evidence |
|---|---|---|
| Human tumors displaying genetic alterations in Hippo pathway components | ||
| Malignant Mesothelioma (MM) | ||
|
NF2 (19% inactivating mutations; 19% CNL; 7% inactivating fusions) STK4 (MST1; 16% CNL) SAV1 (12% CNL) LATS1 (20% CNL) LATS2 (1% inactivating mutations; 5% CNL) (reviewed in 4). |
YAP staining is negative in normal pleural samples, but positive in 93% of MM samples203. | Mice develop MMs after combined ablation of Nf2 and Cdkn2a or Nf2 and Tp53 in mesothelial cells204. The development of asbestos-induced MMs is greatly accelerated in mice lacking one allele of Nf2205. YAP depletion inhibits the invasive and clonogenic abilities of H290 and H2052 NF2-mutant human MM cells203. |
| Sporadic Meningioma | ||
| NF2 (~45% inactivating mutations combined with LOH) (reviewed in 4) | YAP is highly expressed and localizes in the nucleus of tumor cells in 100% of human meningiomas of all grades106. | Patients with germline heterozygous mutation of NF2 develop neurofibromatosis type 2, which includes the development of meningiomas associated to LOH of NF2
206. In mice, biallelic loss of Nf2 in cells of the arachnoid leads to development of meningiomas207. YAP knockdown suppresses in vitro proliferation and migration of human meningioma cells106. |
| Sporadic Schwannoma | ||
| NF2 (77% inactivating mutations and/or CNL) ((reviewed in 4). | YAP is found in the nucleus of schwannoma cells in 100% of cases (reviewed in 42). | Patients with germline heterozygous mutation of NF2 develop neurofibromatosis type 2, which includes the development of schwannomas associated to LOH of NF2. 206 Mice with biallelic loss of Nf2 in Schwann cells develop manifestations of neurofibromatosis type 2, including development of schwannomas208. |
| Sporadic Spine-Ependymoma (SP-EPN) | ||
| NF2 (39% inactivating mutations) (reviewed in 4). | Patients with germline heterozygous mutation of NF2 develop neurofibromatosis type 2, which includes the development of ependymomas associated to LOH of NF2.206 In mice, loss of Lats1/2 in Neural Stem Cells (NSCs) induces formation of EPNs in a YAP/TAZ-dependent manner209. |
|
| Renal-Cell Carcinoma (RCC) | ||
| Papillary (pRCC): NF2 (6% inactivating mutations) SAV1 (3% inactivating mutations) (reviewed in 4) Sarcomatoid clear cell (sccRCC): NF2 (12% inactivating mutations) LATS2 (6% inactivating mutations)210 Mucinous tubular and spindle cell (mtscRCC): NF2 (23% biallelic inactivating mutations) SAV1 (4.5% biallelic inactivating mutations) PTPN14 (32% biallelic inactivating mutations)211. |
YAP is found nuclear in 90% of mtscRCC cases211. | In mice, biallelic loss of Nf2 or deletion of both Lats1 and Lats2 in adult kidney epithelia induces formation of malignant RCCs212,213. NF2 reconstitution or YAP knockdown in NF2-deficient sccRCC cells restrains proliferation and invasion in vitro and tumor growth in vivo210. |
| Intrahepatic Cholangiocarcinoma (ICC) | ||
| SAV1 (~10% CNL) NF2 (9% inactivating mutations) (reviewed in 4) | YAP is found nuclear in 71% of ICC cases, and this is predictive of poor patient survival214. | In mice, homozygous knockout of Sav1 or Nf2 in liver cells causes biliary cell reaction, later developing in liver tumors, including ICCs. These events are rescued by concomitant deletion of Yap1. (reviewed in 42). |
| Human tumors displaying genetic alterations in YAP and/or TAZ (WWTR1) | ||
| Cervical Carcinoma | ||
| YAP1 (12-16% focal amplifications) WWTR1 (8% focal amplifications)75 | YAP is weakly expressed by normal cervical tissue, but it is overexpressed by 91% of cervical carcinomas215. | Expression of transgenic YAP-S127A in the basal cells of the cervix induces development of invasive cervical carcinomas in mice215. |
| Head and Neck Squamous Cell Carcinoma (HNSCC) | ||
| YAP1 (6% focal amplifications) WWTR1 (9% focal amplifications) 75 | TAZ is overexpressed and nuclear in 50% of oral HNSCC samples, and is predictive of poor patient survival (reviewed in 42). | YAP and TAZ are required for tumor formation by HNSCC cells transplanted in the orthotopic site (tongue) (reviewed in 42). YAP and TAZ are required for the fitness of distinct types of oral HNSCC cell lines, with TAZ being required in cells displaying amplification of WWTR1216 |
| Esophageal Squamous Cell Carcinoma (ESCC) | ||
| YAP1 (6% focal amplifications) WWTR1 (27% focal amplifications) 75 | 60-90% of ESCC samples display nuclear YAP and TAZ staining. Nuclear expression of YAP is predictive of poor survival in ESCC patients (reviewed in 42). | YAP is required for proliferation of ESCC in vitro (reviewed in 42). |
| Ovarian Serous Cystadenocarcinoma (OV) | ||
| YAP1 (8% focal amplifications) 75 | 49% of OV display elevated expression of YAP, mostly in the nucleus217 | Overexpression of YAP converts cells of the fallopian tube (the cells-of-origin of ovarian cancer) into high-grade serous carcer cells able to form tumors in vivo (reviewed in 42). |
| Hemangio-Endothelioma (HE) | ||
| Epithelioid (EHE): WWTR1 (~90% gene fusion with CAMTA1). YAP1 (8% gene fusion with TFE3) (reviewed in 79). Retinoid (RHE): YAP1 (38% fusion with MAML2) (reviewed in 79). Composite (CHE): YAP1 (27% fusion with MAML2) (reviewed in 79). |
TAZ-CAMTA1 is found in the nucleus in ~90% of EHEs. 7% of EHEs display nuclear YAP-TFE3 (reviewed in 79). | Transgenic WWTR1-CAMTA1 expression in endothelial cells induces development of HE in mice77,78. |
| Supratentorial Ependymoma (ST-EPN) | ||
| YAP1 (5% gene fusions, 86% of which with MAMLD1, and 14% with FAM118B) (reviewed in 79). | Transgenic expression of the YAP-MAMLD1 or YAP-FAM118B in NSCs induces formation of ST-EPN in mice (reviewed in 79). | |
| Poroma/Porocarcinoma | ||
| Poroma: YAP1 (20.2% fusion with NUTM1, 68.3% fusion with MAML2) WWTR1 (1% fusion with NUTM1) Porocarcinoma: YAP1 (54.6% fusion with NUTM1, 9.1% fusion with MAML2) (reviewed in 79). | Nuclear YAP/TAZ is detected in 96% of poromas and 64% of porocarcinomas (reviewed in 79). | YAP1-MAML2, YAP1-NUTM1, and WWTR1-NUTM1 can promote fibroblast transformation in soft agar assay (reviewed in 79). |
| Hybrid Low-grade Fibromyxoid Sarcoma/Sclerosing Epithelioid Fibrosarcoma | ||
| YAP1 (75% fusion with KMT2A) (reviewed in 79). | ||
Why are Hippo pathway mutations so rare in epithelial tumors? A plausible answer to this riddle is that mutations in the Hippo pathway may be in fact detrimental for cancer cells. This could entail Hippo-dependent, but YAP/TAZ-independent regulation, including the requirement of LATS1/2 as checkpoints of anti-tumor immunity12, as regulators of mitotic stress through phosphorylation of Aurora B kinase, or in the context of lipid metabolism and epigenetics13,14.
Regulation of YAP/TAZ by mechanotransduction
YAP/TAZ are overarchingly regulated by mechanotransduction, a tissue-level informational system based on the exchange of physical forces between cells and their extracellular matrix (ECM)15,16. Via mechanical signals cells are informed of perturbations of their surroundings, and of their own shape, polarity and cytoskeletal organization. Mechanical forces in living tissues are the product of local patterns of geometrical and mechanical strains, which are related to the positioning of the cell within the 3D tissue architecture and to the composition of the stromal milieu16,17. Mechanical signals are generated by the interplay between cell-generated traction forces and resisting forces within the ECM. Integrins are key mediators of such interplay, connecting, on their cytoplasmic side, with the F-actin cytoskeleton through Focal Adhesions, in a manner involving the ILK, FAK and Src proteins18 (Figure 1b). As response to extracellular forces, the cell increases F-actin contractility and restructures its entire cytoskeleton in a process requiring Rho-GTPases (such as Rho or Rac1), myosin activity and ROCK19. This Integrin/FA/Rho-Rac/ROCK/F-actin core mechanosignaling engine is also an appealing target of therapeutic interventions, as discussed at the end of this review.
A missing link in the field is how to connect the F-actin architecture of mechanically activated cells with YAP/TAZ nuclear localization and activation (Figure 1b). A key permissive step in YAP/TAZ nuclear entry is the opening of the nuclear pores by nucleo-cytoskeletal coupling20. In addition, mechanosignaling also enhances YAP/TAZ transcriptional responses by releasing their association with components of the SWI/SNF complex21 (Figure 1a). Besides these regulations, how mechanically modulated F-actin in the cytoplasm controls YAP/TAZ nuclear entry remains a matter of speculation; for example, cytoplasmic F-actin may trap or promote degradation of sequestering factor(s) that prevent YAP/TAZ nuclear accrual.
In addition, LATS1/2 have been proposed as elements by which mechanically modulated F-actin regulates YAP/TAZ22–24. However, if LATS1/2 were downstream of F-actin, then LATS1/2 activity, as measured by LATS phosphorylation, should be patterned by mechanotransduction. In contrast, there is limited evidence of such modulation. Moreover, if cell mechanics were upstream of LATS1/2, cells depleted of LATS1/2 should be insensitive to mechano-modulations. In contrast, LATS1/2 inactivation can hardly rescue YAP/TAZ function when mechanosignaling is overtly inhibited15,20,25–34; intriguingly, LATS1/2 in fact requires at least some residual F-actin cytoskeleton in order to blunt YAP/TAZ activity32. Our interpretation of these data is that LATS1/2 represent a tonic checkpoint that is formally dispensable for the F-actin/YAP/TAZ axis, but that remains relevant for inhibition of such axis. Of note, LATS kinases have been shown to regulate components of the F-actin regulatory machinery, such as ENA and AMOT35–39 and these, in principle, may represent effective means by which LATS could interfere with YAP/TAZ activity, although indirectly, through the cytoskeleton (Figure 1b). Changes in YAP phosphorylation at LATS1/2 sites are consistently detected upon modulation of mechanosignaling4. However, it should be noted that such phosphorylation does not automatically imply a direct involvement of LATS1/2 in YAP/TAZ mechanotransduction, as this may represent a reinforcing step, secondary to any YAP/TAZ inhibitory mechanism that leads to YAP/TAZ cytoplasmic accumulation (where LATS1/2 mainly operate). A conservative interpretation of these results is that YAP/TAZ protumorigenic effects requires sufficient mechanosignaling and concomitant attenuation of Hippo kinase activity. To conclude, a truly comprehensive molecular understanding of the link between mechanosignaling and YAP/TAZ regulation is still lacking.
Nevertheless, if inactivation of the Hippo pathway is an unlikely culprit for the widespread YAP/TAZ hyperactivation in solid tumors, the question of whether mechanosignaling serves such a purpose emerges. Addressing this question remains a major quest in cancer biology. Recent evidence suggests that normal tissue architecture is a potent tumor suppressor that keeps YAP/TAZ at bay by opposing their nuclear accumulation16,24,27,34,40. Indeed, during injury and regeneration (in skin, pancreas, lung, heart, intestine and liver), cells react to mechanical changes in their surrounding by activating YAP/TAZ and initiating a highly choreographed cellular and supracellular program that drives tissue healing back to homeostatic mechanical equilibrium until the wound is resolved and YAP/TAZ inactivated (reviewed in 41–43). In contrast, tumors are “chronic wounds”, tissues with distorted architecture that never resolves, characterized by increased ECM deposition and stiffness, inflammation, edema, and altered vascular blood flow44. These environmental changes may profoundly affect the mechanics, contributing to persistent YAP/TAZ activation16,24,27,34,40 (Figure 2).
Figure 2. Synergistic effects of oncogenes and mechano-signaling on YAP/TAZ activation in cancer.
Oncogenic mutations promote the assembly of robust acto-myosin contractile structures in cooperation with mechanical stimulations from the microenvironment, eventually leading to YAP/TAZ activation and cell transformation. In the absence of either oncogenes or substantial mechanical stimulation, cells do not accumulate contractile microfilaments and nuclear YAP/TAZ; thus transformation does not occur.
In addition to extrinsic mechanical cues, recent mechanistic inspections revealed that mechanosignaling contributes to YAP/TAZ activation in cancer also downstream of key oncogenes, soluble factors, and GPCR signaling, as described in the following section.
Regulation of YAP/TAZ by oncogenic alterations
In the following sections we will discuss in detail how YAP/TAZ are regulated by oncogenic alterations that aberrantly activate intracellular signaling.
GPCR signaling
A number of growth factors operate through G-protein-coupled receptors (GPCRs), in turn either inducing or suppressing YAP/TAZ activity23. This suggests that oncogenic mutations in G-protein α-subunits in different types of cancer may in fact work by promoting YAP/TAZ activity. A case in point is uveal melanoma, where gain-of-function mutations of Gαq (occurring in 90% of cases) drive tumor growth in vivo. Prerequisite for this regulation is a sufficient induction of Integrin/FAK mechanosignaling26,45; GPCR signaling in uveal melanoma entails both LATS-independent rewiring of the actin cytoskeleton through Rho (Figure 2), but also MOB phosphorylation and LATS inhibition26,45,46. Intriguingly, recent work found that tissue-specific oncogenic GPCR ligands can be secreted by stromal cells to promote YAP activity in epithelial cells. A prominent example is prostaglandin E2 secreted by intestinal mesenchymal cells, that signals through the GPCR Ptger4 expressed in epithelial cells, causing YAP nuclear accumulation and hyperactivation47.
RTK/RAS/RAC1 signaling
A main oncogenic driver contributing to YAP/TAZ activity is receptor tyrosine kinase (RTK)-Ras signaling27,34. Oncogenic mutations in RTK signaling, such as those affecting K-Ras or Erbb2, indeed change cellular mechanosensing by increasing F-actin stress fibers and contractility through their downstream effector Rac127 (Figure 2). This sensitizes epithelial cells to increases in ECM stiffness akin those measured in tumors. In doing so, oncogenes become integral to mechanotransduction and exploit YAP/TAZ to drive cell fate reprogramming and transformation27. In metastatic colorectal cancer (CRC), blockade of hyperactive EGFR reduces YAP activity and tumor growth48. Notably, YAP/TAZ regulation through cytoskeletal remodeling might also incorporate the effects of other oncogenic lesions. For instance, Src has been reported to be upstream of YAP/TAZ30; then mutant-p53 has been recently linked to the activation of Rho/Rac1/ROCK, as such impacting cell mechanics and YAP/TAZ30,34,49. Downstream of oncogenic RTK signaling, YAP/TAZ activity accounts for a large fraction of oncogene-induced transcriptional responses27.
In spite of oncogenic RTK/Ras/Rac1 being able to boost mechanosignaling, the normal compliance of healthy epithelial layers is far below the threshold required to turn on YAP/TAZ mechanotransduction (Figure 2); in these conditions, oncogene-bearing cells do not transform and remain functionally and transcriptionally indistinguishable from their normal counterparts27,50. It is tempting to speculate that this phenomenon may account for the observation in genome sequencing data that ostensibly healthy human tissues can display the same type and number of oncogenic mutations found in malignancies of the same tissue51–53.
SWI/SNF
Inactivation of components of the nuclear SWI/SNF complex, and in particular of ARID1A, occurs at very high frequencies in a variety of human malignancies54. ARID1a binds and inhibits YAP/TAZ in the nucleus, anchoring them to the BAF-SWI/SNF complex. This nuclear buffering of YAP/TAZ activity is also dependent on the tensional state of the cells, as the BAF-SWI/SNF complex preferentially binds nuclear F-Actin in conditions of elevated mechanical strain, leading to release of nuclear YAP/TAZ from ARID1a inhibition21 (Figure 1a). Thus, increased mechanical stimulation does not only promote YAP/TAZ nuclear entry, but also contributes to relieving nuclear inhibitors. Intriguingly, it has recently been reported that treatment of triple negative breast cancer (TNBC) cells with FGFR inhibitors can suppress the function of the SWI/SNF complex, enabling YAP/TAZ recruitment to cognate enhancers and expression of YAP/TAZ-driven therapy resistance genes55.
Epithelial-to-mesenchymal transition (EMT) and cell polarity
EMT has been associated with increased cancer malignancy and stemness56 and its activation by Twist or Snail precedes TAZ activation and stemness induction by disrupting the localization of the epithelial polarity factor Scribble57. In turn, Scribble is also controlled by the oncosuppressor LKB1, and loss of LKB1 favors emergence of non-small cell lung cancer in a YAP/TAZ-driven manner58,59. Inactivation of FAT1, a protocadherin frequently mutated in human cancers60, promotes malignant skin squamous carcinomas (SCCs) in mouse models in part by installing a hybrid EMT61. This process involves CAMK2/CD44/Src-mediated activation of YAP/TAZ mechanotransduction and FAT1-knockout tumor cells behave on soft substrates as if they were exposed to a stiff ECM61. Of note, FAT1 truncating mutations have been identified in head and neck SCCs as the top genetic alterations associated with increased YAP/TAZ activity60.
Wnt signaling
YAP and TAZ can bind to components of the β-catenin destruction complex Axin, APC and β-catenin itself62–64. This configures a crosstalk between YAP/TAZ, Hippo and Wnt signaling, such that cytoplasmic YAP/TAZ (in a Hippo ON scenario) can inhibit Wnt stimulation62,64–66. Conversely, Wnt can lead, at least in certain cellular contexts, to the activation of YAP/TAZ, either through canonical inhibition of the destruction complex or through non-canonical Wnt signaling. For example, strong accumulation of nuclear YAP/TAZ has been reported in Wnt1-induced mammary tumors67,68, and in APC-mutant intestinal epithelium, where YAP/TAZ are required for APC-driven transformation62,63,69,70.
Viral oncogenes
YAP/TAZ activation also occurs downstream of oncogenic viral infection. This is the case for cervical carcinoma driven by Human papilloma virus, whose E6 and E7 oncogenes can promote YAP protein stabilization through a compiled mechanism involving Scribble inhibition and regulation of cytoskeletal dynamics71–73. Another example is Kaposi sarcoma-associated herpesvirus, triggering YAP/TAZ activation through Gαq/11 and Gα12/1374.
Regulation of YAP/TAZ by alteration of their coding-genes
The most common genetic alterations targeting YAP/TAZ-coding genes (YAP1 and WWTR1, respectively) in cancer are amplifications, mostly occurring in a fraction of squamous cell carcinomas (cervical, head and neck and esophageal SCCs) and in ovarian cancers75 (Table 2).
Other genetic alterations targeting YAP/TAZ in cancer are gene fusions. These events occur at high frequency only in rare tumor types (Table 2)76. The chimeric proteins generated by these fusions invariably contain the TEAD-binding domain of YAP or TAZ, combined with a new C-terminal portion derived from a different transcription factor (Table2 for specifics) containing a nuclear localization signal and a transcriptional activation domain. As result, these fusion proteins accumulate in the nucleus, and promote the transcription of YAP/TAZ target genes77–79. A case in point is YAP/TAZ oncogenic fusions that cause epithelioid hemangioendothelioma (EHE). This monogenic tumor type may benefit from anti-YAP/TAZ therapy, in analogy to other monogenic tumor types (e.g., BCR-Abl-driven cancers). Of note, simvastatin, inhibiting Rho-GTPases and YAP/TAZ mechanosignaling, had dramatic inhibitory effects on endothelial cells transformed by TAZ-CAMTA1 fusion oncogene78. It is also worth discussing that YAP, but not TAZ, display several splicing variants, with some short isoforms of YAP1 being able to form heterodimers with TAZ endowing specific transcriptional responses80,81.
YAP/TAZ as tumor suppressors
In contrast to the widespread activation of YAP/TAZ in the vast majority of solid tumors (YAP/TAZON-type), the opposite behavior, with inhibited YAP/TAZ (YAP/TAZOFF-type), has been detected in hematological malignancies82. Myelomas, Lymphomas, and Leukemias display very low transcriptional expression of YAP and TAZ mRNAs82–84, which, at least in part, is related to focal deletion of YAP1 in a fraction of these tumors83,84. Survival analyses indicate that residual YAP expression is prognostic of better survival for patients with Multiple Myeloma or Acute Myeloid Leukemia (AML)83,84, suggesting a tumor suppressive role for YAP in these contexts. Supporting this notion, YAP reconstitution dampens cell proliferation and induces cell death in human Multiple Myeloma and AML cells83.
Solid tumors displaying very low YAP/TAZ protein expression and transcriptional activity have also been reported; intriguingly, these are cases carrying genetic inactivation of the Rb locus and displaying molecular traits related either to neural cells (e.g., Retinoblastoma), or to neuroendocrine cells (e.g., Small Cell Lung Cancer (SCLC))82. Functionally, forced YAP/TAZ activation induces growth arrest and apoptosis in cell lines originated from these tumors and genetic inactivation of YAP/TAZ promotes tumor growth and progression, and reduces survival in mouse models82, indicating that YAP/TAZ act as tumor suppressors. Mechanistically, YAP/TAZOFF-type of tumors appear to be characterized by a peculiar set of TEAD-bound enhancers; contrasting YAP/TAZON tumors, which are void of AP1 cooperating elements82, at least suggesting the potential involvement of other TEAD partners with tumor suppressive functions.
Hallmarks of YAP/TAZ activity in cancer
Once the above-described regulations raise nuclear YAP/TAZ accumulation above a critical threshold, the gene expression programs induced by these factors empower a number of fundamental attributes in cancer cells85. These include phenotypic plasticity, drug resistance, cell proliferation, and gain of metastatic abilities; other attributes represent non-cell autonomous mechanisms, including the control of stromal cells, inflammation, senescence, immunity, angiogenesis and cell competition, some of which have been described as “enablers” of the cancer phenotype85.
Cancer cell plasticity
Exogenous expression of YAP or TAZ in healthy, differentiated cells, is sufficient to convert them into cells operationally and molecularly resembling tissue-specific stem cells (SCs)86. Similarly, overexpression of TAZ can convert non-stem tumor cells into cancer stem cells (CSCs)57 (Figure 3a). These findings, while highlighting the potent epigenetic effects of YAP/TAZ, also raise questions as to when YAP/TAZ-mediated cell fate reprogramming becomes important in living tissues. YAP/TAZ are in fact dispensable in healthy, adult epithelia, including normal SCs, but essential for tissue regeneration, tumorigenesis and ex vivo growth as organoids6,27,31,41–43,62,87–93. Thus, SC-like cells induced by YAP/TAZ under regenerative/tumorigenic conditions are different from normal SCs that do not rely on YAP/TAZ. Collective work in the intestine started to shed light on these questions: during intestinal regeneration, YAP/TAZ are key to generate, even from residual differentiated cells, progenitor cells that are in fact very special. These regenerating SCs indeed display traits typical of the fetal intestine, bearing no resemblance with any of the cell types of the adult (including adult SCs)69,92–95. It is thus temping to speculate, given the parallel requirement of YAP/TAZ for tumor emergence, that these special progenitor cells may also represent the cancer cell of origin, generated through YAP/TAZ-mediated reprogramming of normal, differentiated cells (Figure 3a). Although this hypothesis remains to be functionally tested, it should be noted that YAP/TAZ-driven cell fate reprogramming has been observed in different cellular contexts, in mammary luminal cells, astrocytes, hepatocytes, pancreatic acinar cells and Schwann cells86,96–101, that is in cells that also correspond to the tumor cell of origin in their corresponding tissues. This at least suggests the potential for a general involvement of YAP/TAZ-mediated reprogramming in tumor initiation.
Figure 3. The hallmarks of YAP/TAZ activity in cancer.
a) YAP/TAZ activity (red nuclei) can promote differentiate cells to acquire a stem-like or a cancer stem cell identity, as depicted by cell-fate transition in the Waddington epigenetic landscape. Conversely, loss of YAP/TAZ activity in cancer stem cells would cause differentiation toward a less aggressive phenotype.
b) High mechanical stimulation promotes resistance against a number of targeted therapies and chemotherapeutic regimens through activation of YAP/TAZ. Conversely, YAP/TAZ inhibition sensitizes cancer cells to these same treatments.
c) Schematic representation of the metastatic cascade, showing the involvement of YAP/TAZ in the various steps of the metastatic journey, including cell migration from the primary tumor, growth of local metastases in the lymph nodes, intravasation, survival in the circulation trough mechanical crosstalk with platelets, extravasation, spreading in the metastatic site, and awakening from dormancy.
d) YAP/TAZ activation (red nuclei) in tumor stromal components leads to neo-angiogenesis and conversion of fibroblasts and pericytes into Cancer-Associated Fibroblasts (CAFs). In turn, the restructuring of the tumor microenvironment by CAFs induces mechano-activation of YAP/TAZ in tumor cells, which secrete factors further promoting these feed-forward interactions.
e) YAP/TAZ-mediated crosstalk between tumor cells and the immune cells in the tumor ecosystem. YAP/TAZ activity in cancer cells promotes immunosuppression through the recruitment of Myeloid-Derived Suppressor Cells (MDSCs) and Tumor-Associated Macrophages (TAMs). Immunosuppression is further promoted by YAP/TAZ activation in Tregs. YAP/TAZ activity in TAMs also promotes tumor growth by fostering ECM deposition.
Consistent with the ability of YAP/TAZ to favor progenitor cell states, recent single cell analyses of glioblastoma multiforme (GBM) also showed that YAP/TAZ sit at the apex of a gene-regulatory networks that maintains plasticity and aggressiveness of GBM cells101. Blocked differentiation by YAP/TAZ would also be consistent with the association of YAP/TAZ activation with less differentiated and more aggressive tumor types, including breast cancer, colon cancer, lung squamous cell carcinoma, pancreatic cancer, CRC, head and neck squamous cell carcinoma and osteosarcoma42 (Table 1).
Cell proliferation
Enhancing cellular proliferation is a well characterized effect of YAP/TAZ activation, at least in vitro. As reviewed elsewhere42,102, in cancer cell lines, YAP/TAZ sustain proliferation by direct transcriptional induction of a number of mitotic and DNA replication factors, promotion of the cell cycle, and by sustaining expression of oncogenic transcription factors, such as AP1 (in turn feeding on YAP/TAZ-transcription) or, depending on the cell type, c-Myc91,103–105. It is worth noting that enhancing cell proliferation is an element of YAP/TAZ-induced stemness that may only be induced in presence of other cooperating transcription factors and contextual signals91. For example, YAP/TAZ inactivation in glioma affects stemness properties, yet cell proliferation is only affected indirectly101.
Drug resistance
Induction of phenotypic plasticity by YAP/TAZ also ensures adaptability to stressful conditions and/or adoption of quiescent fates that enable cancer cells to escape therapeutic treatments (Figure 3b). YAP/TAZ can promote resistance to both cytotoxic (e.g. paclitaxel, doxorubicin, cisplatin, UV, radiation) and targeted therapies (RAF, MEK, ER, CDK4/6 and Erbb2 inhibitors) in diverse tumor types55,57,106–116 and also controls entry and exit of tumor cells from therapy-induced dormancy, as observed in non-small cell lung cancer after treatment with EGFR inhibitors117,118, or in CRC after chemotherapy119. Intriguingly, in the latter case, disease relapse is promoted by chemotherapy-induced FAK-dependent YAP activation, involving a restructuring of cell-ECM adhesion and, likely, of the cytoskeleton119. Additionally, YAP/TAZ have been recently proposed to drive resistance to the multi-kinase inhibitor Sorafenib in liver cancer by preventing Sorafenib-induced ferroptosis120.
Activation of YAP/TAZ after pharmacological inhibition of driving oncogenes has been connected to increased mechanotransduction in resistant cells. This is the case for vemurafenib-resistant BRAF-mutant melanoma cells112 and lapatinib-resistant HER-2 positive breast cancer cells113. These results are compatible with the view that ECM remodeling and changes in tissue stiffness may activate mechanosensing pathways promoting YAP/TAZ activation after these treatments were initially successful in tumor control. This may further imply that a combinatorial therapeutic strategy that combines oncogene-specific targeted therapy with inhibition of mechanotransduction might be a powerful means to overcome resistance mechanisms.
Metastasis
During metastasis, malignant cells must withstand a stressful journey, escape immune surveillance and co-opt local and systemic factors for recurrence after therapy. Our molecular understanding of these processes, and of their interconnections, remains limited. Intriguingly, experimental gain-of-YAP/TAZ is sufficient to provide full metastatic potential to non-metastatic cells, from dissemination to organ colonization, while YAP/TAZ inactivation impedes metastasis (reviewed in Ref. 121). The available evidence indicates that YAP/TAZ contribute to distinct aspects of the metastatic cascade (Figure 3c). To start, YAP/TAZ activity can foster metastatic dissemination through cell migration and invasion61,122–126, survival in the circulation127 and endothelial transmigration128–130. Disseminated cells can spread in a pericyte-like behavior through L1CAM-medited adhesion to vessel walls in the host tissue; such spreading mechanically activates YAP/TAZ that awaken metastatic cells form dormancy131. These YAP/TAZ-regulated processes may represent special attributes, acquired de novo and distinct from those establishing malignancy at the primary tumor site; or alternatively, may be additional adaptations of the same YAP/TAZ-orchestrated cell plasticity program at play in primary tumors. More work is required to deepen our understanding of cell states in aggressive tumors and their metastatic lesions in order to dissect these alternatives.
Metastasis is mainly a non-genetic process, highly dependent on instructing the host microenvironment into a nurturing niche. Stromal cells provide metastatic cells with soluble growth factors, such as Wnt or TGFβ, but also mechanical cues critical for outgrowth into lethal metastatic nodules. An example is the remodeling of ECM promoted by proteases secreted by neutrophils within DNA-scaffolded extracellular “traps” (i.e., NETosis), resulting in an increase in integrin signaling, which in turn promotes YAP/TAZ-dependent reawakening from dormancy and initiation of colonization132 (Figure 3c). ECM stiffening of pancreatic cancer can also promote metastatic growth through YAP/TAZ-dependent metabolic reprogramming133.
It has been noticed that metastasis in fact also represents a remarkable mechanical journey through very diverse mechanical strains, such as compression, stretching, intravasation, circulation, adhesion, extravasation and regrowth within an alien organ architecture134. All these mechanical stresses necessarily impact on the cells’ own mechanical state and feedback on the ability to adapt to such strains134. We predict that YAP/TAZ may be an integral element in such interplay. For example, loss of mechanosignaling in malignant cells entering into the bloodstream may contribute to their metastatic inefficiency. Conversely, mechanical activation of YAP/TAZ may represent a means to avoid nuclear damage by extreme mechanical cell deformations135,136,137. In line with this hypothesis, evidence exists that YAP/TAZ are not only downstream of cytoskeletal inputs but also actively contribute to F-actin dynamics through expression of cytoskeletal proteins in metastatic cells126,138,139.
YAP/TAZ and the tumor microenvironment
Activation of YAP/TAZ in tumor cells initiates iterative and bidirectional interactions with other elements of the tumor microenvironment (Figures 3d-e), that also entails YAP/TAZ activation in fibroblasts, endothelial cells and neighboring healthy epithelial cells. An emerging and particularly exciting area of investigation relates to the role of YAP/TAZ in controlling immune cell composition and activity.
Cell competition
In healthy tissues, cells that are “perceived as different” are actively eliminated through a process called “cell competition”. This phenomenon was initially described in Drosophila in fact in association with loss-of-function mutations in the Hippo pathway and was then recognized in mammalian systems140,141. Cell competition in tumors is bidirectional, with healthy tissue inhibiting cancer cell growth, and, vice-versa, with cancer cells fostering the demise of non-malignant cells to find new soil for their expansion142. Intriguingly, in a mouse model of cholangiocarcinoma, experimental hyperactivation of YAP/TAZ in peritumoral normal hepatocytes causes them to acquire a competitive advantage vs. tumor cells143. Similarly, in glioblastoma, high YAP expressing clones of normal cells acquire dominance over surrounding tumor cells144. These non-cell autonomous interactions might also help to reconcile recent provocative findings on APC-mutant intestinal tumor models, in which conditional depletion of YAP/TAZ mediated by Adenoviral-Cre injection in the colon mucosae has been reported to favor tumor growth145. Although at odds with other reports on YAP/TAZ being required for intestinal tumorigenesis69,88,146–148, this result might be potentially explained by an overarching effect of YAP/TAZ loss in non-malignant cells loosing competitive capacity against APC-mutant clones.
Angiogenesis
Increased YAP/TAZ function also occurs in stromal cells, including cancer associated fibroblasts (CAFs), Endothelial cells (ECs) and pericytes (reviewed in Ref. 43; Figure 3d). This suggests that YAP/TAZ activation may represent a trait shared by different cells of the tumor ecosystem. In development, YAP/TAZ are fundamental drivers of vascularization149, and this property is hijacked in cancer to support tumor survival and expansion (Figure 3d). In lung cancer allografts, high expression of YAP in the tumor endothelium induced tumor growth150, and ECs exposed to YAP-positive tumor cells activated their own YAP/TAZ for neo-angiogenesis151,152. Conversely, loss of YAP/TAZ in renal cell carcinomas inhibited EC proliferation, through modulation of VEGF signaling153. Thus, YAP/TAZ are the centerpiece of the communication between tumor cells and tumor angiogenesis. To do so, YAP/TAZ integrate a number of inputs, such as hypoxia, through HIF1α-dependent and -independent mechanisms, and angiogenic and inflammatory cytokines. For example, the VEGF pro-angiogenic effect has been reported to signal through the Hippo kinases to increase YAP/TAZ154. In turn, tumor ECs express high level of Endothelin1, that induces YAP/SMAD association and activation of inflammatory responses155. ECM stiffening and increased matrix metalloproteases (MMP)-mediated ECM remodeling in tumors also fuel sprouting angiogenesis149. Notably, increased fibrosis around tumor cells is mediated by YAP activation in CAFs, which are known to promote malignancy and angiogenesis40 (Figure 3d). In line, treatment of CRC liver metastases with a combination of VEGF and angiotensin inhibitors reduced myofibroblast activity, lowered ECM stiffness and prevented angiogenesis, thereby limiting metastatic aggressiveness156.
Tumor vessels derive from an aberrant and unphysiological sprouting, forming immature and disorganized networks157, characterized by disturbed blood flow that can further contribute to YAP/TAZ activation in ECs and to endothelial inflammatory responses158,159 (Figure 3d). In addition, tumor vessels are leaky, leading to accumulation of interstitial fluid pressure, contributing, through cell compression and stretching, to YAP/TAZ mechanical modulation149. Thus, targeting YAP/TAZ function in ECs and/or CAFs may be sufficient to curtail ECM-mediated, YAP/TAZ self-feeding loops, reducing fibrosis, normalizing tumor vasculature and improving effectiveness of therapies. In this aspect, it is worth considering that YAP/TAZ transcriptionally control matricellular proteins such as CTGF and Cyr61, whose targeting may be beneficial to improve chemosensitivity160.
Crosstalk with the immune system
Immune-checkpoint inhibitors (ICI) are pillars of cancer treatment. However, a large fraction of patients does not achieve long-lasting benefits from ICI161,162. Intriguingly, YAP/TAZ activation in tumor cells has been shown to rewire immune responses in the TME, enabling tumor cells to escape immune surveillance (Figure 3e). As such, targeting YAP/TAZ in the TME may represent an efficient route to boost the effect of immune-directed therapies. To start, YAP overexpression in the mouse liver fosters infiltration of protumorigenic macrophages163,164. Similarly, in nonalcoholic steatohepatitis, a condition that precedes hepatocellular carcinoma, TAZ activity also associates to macrophage recruitment, although functional validation of this observation is still pending165. In pancreatic and prostate tumors, YAP/TAZ also cause recruitment of Myeloid-derived-suppressor-cells (MDSCs), immature myeloid cells that favor anergy of effector T cells and tumor tolerance166,167. Vice-versa, YAP-defective tumor cells reduce MDSC recruitment. These findings are also reflected in patient data, as CRC patients with overactive YAP display increased density of MDSCs and adverse clinical features.
Similarly to CAFs and ECs, immune cell populations are also subject to the same mechanical and biochemical cues that induce YAP/TAZ activation in tumor cells, raising their own YAP/TAZ levels to promote malignancy (Figure 3e). For example, DAB2-expressing tumor-associated macrophages (TAMs) are a population of exquisitely mechanosensitive cells in which YAP/TAZ are key to “escort” tumor cells for metastatic dissemination168. DAB2 expression is predictive of overall patient survival in luminal B breast cancer but not in the more malignant subtypes of mammary tumors168, potentially because less aggressive tumors are more dependent on cellular aid to invade nearby tissues. In addition, YAP/TAZ are undetectable in naïve CD4+ T cells but upregulated in intra-tumoral Tregs where they are required to generate an immunologically cold and tolerant microenvironment169,170. Coherently with the above discussion, YAP genetic ablation in effector CD4+ and CD8+ T cells results in enhanced T-cell activation and tumor infiltration171,172. Intriguingly, YAP activation on effector T-cells is under the control of mechanotransduction, as stiffening of draining lymph nodes promotes YAP/TAZ activity and opposes the amplification of effector T-cells173.
Targeting YAP/TAZ in cancer
In the past ten years many efforts were directed to the identification of pharmacological approaches to inhibit YAP/TAZ activity, as summarized below (Figure 4a).
Figure 4. Targeting YAP/TAZ in cancer.
a) Schematics of the most promising molecular drugs targeting YAP/TAZ activity. Red boxes highlight inhibitors of specific elements of YAP/TAZ mechanotransduction (in light blue boxes).
b) Ribbon diagram showing the conserved carboxy-terminal YAP-binding domain of TEAD protein. YAP is shown in yellow and TEAD in green. Image from the RCSB PDB (rcsb.org) of PDB ID 3KYS188, created using Mol* Viewer189.
c) Ribbon diagram showing the hydrophobic pocket of TEAD protein hosting a palmitoyl group covalently bound to a cysteine. Palmitoylated cysteine is shown in red and TEAD in green. Image from the RCSB PDB (rcsb.org) of PDB ID 5emv190, created using Mol* Viewer189.
Targeting YAP/TAZ expression
YAP/TAZ have been depicted as undruggable molecules; however, their expression can be experimentally decreased using RNA-interference methods. Recent chemical developments on the backbone of antisense oligonucleotides (ASOs) allowed to create reagents that are sufficiently stable to induce depletion of the desired target genes in vivo. ION537, an anti-YAP DNA ASO created by Ionis Pharmaceuticals, has been shown to effectively inhibit YAP expression and to blunt the growth of tumor xenografts174, and it is currently undergoing a phase I trial in patients with advanced solid tumors (NCT04659096).
Targeting YAP/TAZ interaction with TEAD
The first example for this therapeutic entry point is verteporfin, a drug able to restrain YAP/TAZ binding to TEADs, dampening YAP/TAZ-induced transcription and activity175, although its cytotoxic effects may be YAP-independent176,177. Several other molecules (compounds and peptides) have been identified as inhibitors of the YAP/TAZ-TEAD interaction (Figure 4b) (reviewed in Ref. 174). IAG933, an inhibitor of YAP/TAZ-mediated transcription developed by Novartis that possibly belongs to this category, is currently being evaluated in a phase I clinical trial against tumors bearing mutations on the Hippo pathway components or YAP/TAZ gene fusions (NCT04857372).
Targeting TEADs
Structural studies on TEAD proteins revealed that they contain a hydrophobic pocket, harboring a palmitoyl group covalently bound to a cysteine (Figure 4c)178,179. Palmitoylation is relevant for TEAD folding and stability, and thus represents a molecular entry point to blunt YAP/TAZ activity. Several YAP/TAZ-inhibitors have been indeed identified as targeting the hydrophobic pocket of TEADs (reviewed in Ref. 174). Of these, VT3989, a drug developed by Vivace Therapeutics, has entered a phase I clinical trial (NCT04665206) in solid tumors and mesotheliomas with NF2 mutations. Intriguingly, TEAD inhibitors have been shown to be effective at causing regression of pre-established NF2 or LATS deficient mesothelioma models180,181.
Targeting BRD4 and other BET proteins
BET-inhibitors (BETi, i.e., drugs targeting BRD4 and its family members) are promising anti-YAP/TAZ therapeutic agents, given that YAP/TAZ recruit BRD4 on chromatin. In line, BETi suppress YAP/TAZ-driven tumorigenesis in vivo2. A number of clinical trials have been initiated with BETi, but only few have been concluded so far (reviewed in Ref. 182), and data remain too scarce and discontinuous to draw firm conclusions.
Targeting Rho-GTPases and ROCK
Several drugs affecting the mevalonate biosynthetic pathway interfere with the geranyl-geranylation of Rho-GTPases, a modification required for their membrane anchoring. As such, these compounds may represent mechano-inhibitory drugs30, although their effectiveness as YAP/TAZ inhibitors at pharmacological concentrations remains undetermined. Nevertheless, statins have already been noted to have beneficial effects in cancer patients, and their usage correlates with decreased cancer incidence in the general population (reviewed in Ref. 183). Of note, some clinical trials with statins also found that these drugs can enhance the effects of chemotherapy183, warranting an in-depth clinical exploration of these “off-the-shelf” and safe compounds. Another appealing therapeutic target is ROCK1/2, that is effective at blunting YAP/TAZ in vitro and in animal models. The development of ROCK-inhibitors is a field of interest, intriguingly connected to treatment of immune disorders and fibrosis184.
Targeting SRC family members
Compounds affecting the activity SRC and its family members (e.g., Dasatinib) are candidate mechanodrugs, and have been shown to inhibit YAP/TAZ activity in vitro and in vivo. Several of these drugs have already been tested in clinical trials on cancer patients with mixed results (reviewed in Ref. 185). Of note, pancreatic and colon cancers display the greatest propensity to respond to these treatments, although it remains unclear whether these responses could be ascribed to YAP/TAZ inhibition.
Conclusions and Perspectives
Despite remarkable insights gained in recent decades, many outstanding questions remain. First, a remarkable property of YAP/TAZ activation is to reprogram cell fates, a feature that has been studied in some detail in the regenerating intestine, in intestinal and other tissues’ organoids17,27,31,62,69,86,88,92–95,186. Is this exploited in other contexts, including tumors and if yes, by which means?
Second, and as evident in this Review, we still remain with quite superficial descriptions of phenotypes controlled by these transcriptional regulators, and molecular insights are limited. For example, YAP/TAZ conditional inactivation in individual cells of the tumor microenvironment, followed by longitudinal single-cell characterization of the whole tumor ecosystem may inform on YAP/TAZ-dependent changes in cell fates, dynamics and cell-cell communication. Moreover, if incorporated into spatial transcriptomic technologies, signatures of YAP/TAZ activity in individual cells may shed light on tumor 3D self-organization and maintenance, on the spatial contexture of tumor-specific cellular networks, and on the relative positioning of tumor and stromal cells into “tissue motifs”. We believe that this may generate hypotheses on the supracellular principles that are central to tumor growth, and on the role of mechanical strains in orchestrating tumor morphogenesis.
Third, genetic analysis have focused on the role of YAP/TAZ in tumor initiation. A main unknown is whether metastases are still addicted to YAP/TAZ or whether other mechanisms may be at play to sustain an advanced tumor ecosystem. Sophisticated mouse models, combining conditional activation of oncogenes in specific cell types and subsequent conditional YAP/TAZ deletions in the same cells, are required to investigate the consequences of YAP/TAZ inactivation within highly malignant lesions.
Moreover, a particularly attractive aspect that has recently emerged is the interplay of YAP/TAZ mechanosignaling in tumor cells with myeloid and lymphoid components of the stroma. The available evidence at least suggests that targeting mechanotransduction may hit, at once, a vulnerability shared by all these cells, and turn immunologically cold tumors into hot. If true, this scenario would incentivize testing combinations of YAP/TAZ inhibitors and ICI in the clinical setting.
The emergence of novel, first in class therapeutics targeting YAP/TAZ is, given all the above, an exciting news for the field. However, there are two notes of caution. First, our knowledge on YAP/TAZ biology is largely centered on epithelial cells and epithelial-specific conditional mutants in mouse models; these studies revealed the inconsequentiality of YAP/TAZ for normal cells, and their paramount relevance for tumor cells. It is from these results that the idea of targeting YAP/TAZ was proposed as a candidate “silver bullet” for cancer therapy. However, this is an oversimplification and potentially an overstatement: we recently reported that endogenous YAP/TAZ mechanotransduction does play physiological roles in fibroblasts and smooth muscle cells, preventing aging-related degeneration in a variety of organs137. This may result in toxicities associated with long-term treatment with YAP/TAZ inhibitors. The second potential concern relates to the emerging role of YAP/TAZ as supporters of Tregs and inhibitors of effector T-cells. Systemic inactivation of YAP/TAZ may outbalance immune tolerance in multiple organs, favoring the rise of auto-immunity and inflammatory diseases. Tempering these concerns may require a regimen of YAP/TAZ inhibition either very short or delivered to targeted cell types.
Finally, although we have so far dealt with “YAP/TAZ” as if they were a unique entity, as justified by the vast redundancy between two factors, we also believe that time is finally ripe to discriminate the specificities of YAP vs. TAZ in the multifaceted tumor biology, and pioneering efforts in this direction are starting to emerge80,81,187. Targeting just one of these two factors, for example by using gene-specific RNA-therapeutics, could allow to limit unwanted effects on healthy tissues while retaining effectiveness against cancer.
Acknowledgements
We are grateful to SP lab members for critical reading of the MS and inputs for its inception. Work in SP lab is funded by Fondazione AIRC under 5 per mille 2019 - ID. 22759 program - P.I. Piccolo Stefano; Fondazione AIRC, IG 2019 - ID. 23307 project – P.I. Piccolo Stefano; PRIN-MIUR to S.P., and T.P (2017HWTP2K and 2017L8FWY8) and Bando Ricerca Scientifica di Eccellenza 2018 Fondazione Cariparo" nr. 52008.
Footnotes
Competing Interests
The authors declare no competing interests.
References
- 1.Battilana G, Zanconato F, Piccolo S. Mechanisms of YAP/TAZ transcriptional control. Cell Stress. 2021;5:167–172. doi: 10.15698/cst2021.11.258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zanconato F, et al. Transcriptional addiction in cancer cells is mediated by YAP/TAZ through BRD4. Nat Med. 2018;24:1599–1610. doi: 10.1038/s41591-018-0158-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wu BK, Mei SC, Chen EH, Zheng Y, Pan D. YAP induces an oncogenic transcriptional program through TET1-mediated epigenetic remodeling in liver growth and tumorigenesis. Nat Genet. 2022 doi: 10.1038/s41588-022-01119-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell. 2019;50:264–282. doi: 10.1016/j.devcel.2019.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chen Q, et al. Homeostatic control of Hippo signaling activity revealed by an endogenous activating mutation in YAP. Genes Dev. 2015;29:1285–1297. doi: 10.1101/gad.264234.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Chen Q, et al. A temporal requirement for Hippo signaling in mammary gland differentiation, growth, and tumorigenesis. Genes Dev. 2014;28:432–437. doi: 10.1101/gad.233676.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.George NM, Day CE, Boerner BP, Johnson RL, Sarvetnick NE. Hippo signaling regulates pancreas development through inactivation of Yap. Mol Cell Biol. 2012;32:5116–5128. doi: 10.1128/MCB.01034-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Schlegelmilch K, et al. Yap1 acts downstream of alpha-catenin to control epidermal proliferation. Cell. 2011;144:782–795. doi: 10.1016/j.cell.2011.02.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lange AW, et al. Hippo/Yap signaling controls epithelial progenitor cell proliferation and differentiation in the embryonic and adult lung. J Mol Cell Biol. 2015;7:35–47. doi: 10.1093/jmcb/mju046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li Q, et al. Lats1/2 Sustain Intestinal Stem Cells and Wnt Activation through TEAD-Dependent and Independent Transcription. Cell Stem Cell. 2020;26:675–692.:e678. doi: 10.1016/j.stem.2020.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chung C, et al. Hippo-Foxa2 signaling pathway plays a role in peripheral lung maturation and surfactant homeostasis. Proc Natl Acad Sci U S A. 2013;110:7732–7737. doi: 10.1073/pnas.1220603110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Moroishi T, et al. The Hippo Pathway Kinases LATS1/2 Suppress Cancer Immunity. Cell. 2016;167:1525–1539.:e1517. doi: 10.1016/j.cell.2016.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Aylon Y, et al. The LATS2 tumor suppressor inhibits SREBP and suppresses hepatic cholesterol accumulation. Genes Dev. 2016;30:786–797. doi: 10.1101/gad.274167.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Furth N, Aylon Y. The LATS1 and LATS2 tumor suppressors: beyond the Hippo pathway. Cell Death Differ. 2017;24:1488–1501. doi: 10.1038/cdd.2017.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dupont S, et al. Role of YAP/TAZ in mechanotransduction. Nature. 2011;474:179–183. doi: 10.1038/nature10137. [DOI] [PubMed] [Google Scholar]
- 16.Panciera T, Azzolin L, Cordenonsi M, Piccolo S. Mechanobiology of YAP and TAZ in physiology and disease. Nat Rev Mol Cell Biol. 2017;18:758–770. doi: 10.1038/nrm.2017.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Brusatin G, Panciera T, Gandin A, Citron A, Piccolo S. Biomaterials and engineered microenvironments to control YAP/TAZ-dependent cell behaviour. Nat Mater. 2018;17:1063–1075. doi: 10.1038/s41563-018-0180-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wolfenson H, Yang B, Sheetz MP. Steps in Mechanotransduction Pathways that Control Cell Morphology. Annu Rev Physiol. 2019;81:585–605. doi: 10.1146/annurev-physiol-021317-121245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Butcher DT, Alliston T, Weaver VM. A tense situation: forcing tumour progression. Nat Rev Cancer. 2009;9:108–122. doi: 10.1038/nrc2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Elosegui-Artola A, et al. Force Triggers YAP Nuclear Entry by Regulating Transport across Nuclear Pores. Cell. 2017;171:1397–1410.:e1314. doi: 10.1016/j.cell.2017.10.008. [DOI] [PubMed] [Google Scholar]
- 21.Chang L, et al. The SWI/SNF complex is a mechanoregulated inhibitor of YAP and TAZ. Nature. 2018;563:265–269. doi: 10.1038/s41586-018-0658-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Codelia VA, Sun G, Irvine KD. Regulation of YAP by mechanical strain through Jnk and Hippo signaling. Curr Biol. 2014;24:2012–2017. doi: 10.1016/j.cub.2014.07.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yu FX, et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150:780–791. doi: 10.1016/j.cell.2012.06.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Meng Z, et al. RAP2 mediates mechanoresponses of the Hippo pathway. Nature. 2018;560:655–660. doi: 10.1038/s41586-018-0444-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Das A, Fischer RS, Pan D, Waterman CM. YAP Nuclear Localization in the Absence of Cell-Cell Contact Is Mediated by a Filamentous Actin-dependent, Myosin II- and Phospho-YAP-independent Pathway during Extracellular Matrix Mechanosensing. J Biol Chem. 2016;291:6096–6110. doi: 10.1074/jbc.M115.708313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Feng X, et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell. 2014;25:831–845. doi: 10.1016/j.ccr.2014.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Panciera T, et al. Reprogramming normal cells into tumour precursors requires ECM stiffness and oncogene-mediated changes of cell mechanical properties. Nat Mater. 2020;19:797–806. doi: 10.1038/s41563-020-0615-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Reginensi A, et al. Yap- and Cdc42-dependent nephrogenesis and morphogenesis during mouse kidney development. PLoS Genet. 2013;9:e1003380. doi: 10.1371/journal.pgen.1003380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Silvis MR, et al. alpha-catenin is a tumor suppressor that controls cell accumulation by regulating the localization and activity of the transcriptional coactivator Yap1. Sci Signal. 2011;4:ra33. doi: 10.1126/scisignal.2001823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sorrentino G, et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat Cell Biol. 2014;16:357–366. doi: 10.1038/ncb2936. [DOI] [PubMed] [Google Scholar]
- 31.Taniguchi K, et al. A gp130-Src-YAP module links inflammation to epithelial regeneration. Nature. 2015;519:57–62. doi: 10.1038/nature14228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Aragona M, et al. A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell. 2013;154:1047–1059. doi: 10.1016/j.cell.2013.07.042. [DOI] [PubMed] [Google Scholar]
- 33.Wada K, Itoga K, Okano T, Yonemura S, Sasaki H. Hippo pathway regulation by cell morphology and stress fibers. Development. 2011;138:3907–3914. doi: 10.1242/dev.070987. [DOI] [PubMed] [Google Scholar]
- 34.Esposito D, et al. ROCK1 mechano-signaling dependency of human malignancies driven by TEAD/YAP activation. Nat Commun. 2022;13:703. doi: 10.1038/s41467-022-28319-3. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 35.Lucas EP, et al. The Hippo pathway polarizes the actin cytoskeleton during collective migration of Drosophila border cells. J Cell Biol. 2013;201:875–885. doi: 10.1083/jcb.201210073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Adler JJ, et al. Serum deprivation inhibits the transcriptional co-activator YAP and cell growth via phosphorylation of the 130-kDa isoform of Angiomotin by the LATS1/2 protein kinases. Proc Natl Acad Sci U S A. 2013;110:17368–17373. doi: 10.1073/pnas.1308236110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chan SW, et al. Actin-binding and cell proliferation activities of angiomotin family members are regulated by Hippo pathway-mediated phosphorylation. J Biol Chem. 2013;288:37296–37307. doi: 10.1074/jbc.M113.527598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dai X, et al. Phosphorylation of angiomotin by Lats1/2 kinases inhibits F-actin binding, cell migration, and angiogenesis. J Biol Chem. 2013;288:34041–34051. doi: 10.1074/jbc.M113.518019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mana-Capelli S, Paramasivam M, Dutta S, McCollum D. Angiomotins link F-actin architecture to Hippo pathway signaling. Mol Biol Cell. 2014;25:1676–1685. doi: 10.1091/mbc.E13-11-0701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Calvo F, et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat Cell Biol. 2013;15:637–646. doi: 10.1038/ncb2756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Moya IM, Halder G. Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat Rev Mol Cell Biol. 2019;20:211–226. doi: 10.1038/s41580-018-0086-y. [DOI] [PubMed] [Google Scholar]
- 42.Zanconato F, Cordenonsi M, Piccolo S. YAP/TAZ at the Roots of Cancer. Cancer Cell. 2016;29:783–803. doi: 10.1016/j.ccell.2016.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zanconato F, Cordenonsi M, Piccolo S. YAP and TAZ: a signalling hub of the tumour microenvironment. Nat Rev Cancer. 2019;19:454–464. doi: 10.1038/s41568-019-0168-y. [DOI] [PubMed] [Google Scholar]
- 44.Northey JJ, Przybyla L, Weaver VM. Tissue Force Programs Cell Fate and Tumor Aggression. Cancer Discov. 2017;7:1224–1237. doi: 10.1158/2159-8290.CD-16-0733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Feng X, et al. A Platform of Synthetic Lethal Gene Interaction Networks Reveals that the GNAQ Uveal Melanoma Oncogene Controls the Hippo Pathway through FAK. Cancer Cell. 2019;35:457–472.:e455. doi: 10.1016/j.ccell.2019.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yu FX, et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell. 2014;25:822–830. doi: 10.1016/j.ccr.2014.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Roulis M, et al. Paracrine orchestration of intestinal tumorigenesis by a mesenchymal niche. Nature. 2020;580:524–529. doi: 10.1038/s41586-020-2166-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lupo B, et al. Colorectal cancer residual disease at maximal response to EGFR blockade displays a druggable Paneth cell-like phenotype. Sci Transl Med. 2020;12 doi: 10.1126/scitranslmed.aax8313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Moon SH, et al. p53 Represses the Mevalonate Pathway to Mediate Tumor Suppression. Cell. 2019;176:564–580.:e519. doi: 10.1016/j.cell.2018.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Almagro J, Messal HA, Elosegui-Artola A, van Rheenen J, Behrens A. Tissue architecture in tumor initiation and progression. Trends Cancer. 2022 doi: 10.1016/j.trecan.2022.02.007. [DOI] [PubMed] [Google Scholar]
- 51.Lee-Six H, et al. The landscape of somatic mutation in normal colorectal epithelial cells. Nature. 2019;574:532–537. doi: 10.1038/s41586-019-1672-7. [DOI] [PubMed] [Google Scholar]
- 52.Martincorena I, et al. Tumor evolution. High burden and pervasive positive selection of somatic mutations in normal human skin. Science. 2015;348:880–886. doi: 10.1126/science.aaa6806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yokoyama A, et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature. 2019;565:312–317. doi: 10.1038/s41586-018-0811-x. [DOI] [PubMed] [Google Scholar]
- 54.Kadoch C, Crabtree GR. Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics. Sci Adv. 2015;1:e1500447. doi: 10.1126/sciadv.1500447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Li Y, et al. FGFR-inhibitor-mediated dismissal of SWI/SNF complexes from YAP-dependent enhancers induces adaptive therapeutic resistance. Nat Cell Biol. 2021;23:1187–1198. doi: 10.1038/s41556-021-00781-z. [DOI] [PubMed] [Google Scholar]
- 56.Lambert AW, Weinberg RA. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer. 2021;21:325–338. doi: 10.1038/s41568-021-00332-6. [DOI] [PubMed] [Google Scholar]
- 57.Cordenonsi M, et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell. 2011;147:759–772. doi: 10.1016/j.cell.2011.09.048. [DOI] [PubMed] [Google Scholar]
- 58.Mohseni M, et al. A genetic screen identifies an LKB1-MARK signalling axis controlling the Hippo-YAP pathway. Nat Cell Biol. 2014;16:108–117. doi: 10.1038/ncb2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhang W, et al. YAP promotes malignant progression of Lkb1-deficient lung adenocarcinoma through downstream regulation of survivin. Cancer Res. 2015;75:4450–4457. doi: 10.1158/0008-5472.CAN-14-3396. [DOI] [PubMed] [Google Scholar]
- 60.Martin D, et al. Assembly and activation of the Hippo signalome by FAT1 tumor suppressor. Nat Commun. 2018;9:2372. doi: 10.1038/s41467-018-04590-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pastushenko I, et al. Fat1 deletion promotes hybrid EMT state, tumour stemness and metastasis. Nature. 2021;589:448–455. doi: 10.1038/s41586-020-03046-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Azzolin L, et al. YAP/TAZ incorporation in the beta-catenin destruction complex orchestrates the Wnt response. Cell. 2014;158:157–170. doi: 10.1016/j.cell.2014.06.013. [DOI] [PubMed] [Google Scholar]
- 63.Cai J, Maitra A, Anders RA, Taketo MM, Pan D. beta-Catenin destruction complex-independent regulation of Hippo-YAP signaling by APC in intestinal tumorigenesis. Genes Dev. 2015;29:1493–1506. doi: 10.1101/gad.264515.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Imajo M, Miyatake K, Iimura A, Miyamoto A, Nishida E. A molecular mechanism that links Hippo signalling to the inhibition of Wnt/beta-catenin signalling. EMBO J. 2012;31:1109–1122. doi: 10.1038/emboj.2011.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Heallen T, et al. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science. 2011;332:458–461. doi: 10.1126/science.1199010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Nowell CS, et al. Chronic inflammation imposes aberrant cell fate in regenerating epithelia through mechanotransduction. Nat Cell Biol. 2016;18:168–180. doi: 10.1038/ncb3290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Park HW, et al. Alternative Wnt Signaling Activates YAP/TAZ. Cell. 2015;162:780–794. doi: 10.1016/j.cell.2015.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Serrano I, McDonald PC, Lock F, Muller WJ, Dedhar S. Inactivation of the Hippo tumour suppressor pathway by integrin-linked kinase. Nat Commun. 2013;4:2976. doi: 10.1038/ncomms3976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Gregorieff A, Liu Y, Inanlou MR, Khomchuk Y, Wrana JL. Yap-dependent reprogramming of Lgr5(+) stem cells drives intestinal regeneration and cancer. Nature. 2015;526:715–718. doi: 10.1038/nature15382. [DOI] [PubMed] [Google Scholar]
- 70.Camargo FD, et al. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr Biol. 2007;17:2054–2060. doi: 10.1016/j.cub.2007.10.039. [DOI] [PubMed] [Google Scholar]
- 71.He C, et al. The Hippo/YAP pathway interacts with EGFR signaling and HPV oncoproteins to regulate cervical cancer progression. EMBO Mol Med. 2015;7:1426–1449. doi: 10.15252/emmm.201404976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Messa L, et al. The Dimeric Form of HPV16 E6 Is Crucial to Drive YAP/TAZ Upregulation through the Targeting of hScrib. Cancers (Basel) 2021;13 doi: 10.3390/cancers13164083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Matarrese P, Vona R, Ascione B, Paggi MG, Mileo AM. Physical Interaction between HPV16E7 and the Actin-Binding Protein Gelsolin Regulates Epithelial-Mesenchymal Transition via HIPPO-YAP Axis. Cancers (Basel) 2021;13 doi: 10.3390/cancers13020353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Liu G, et al. Kaposi sarcoma-associated herpesvirus promotes tumorigenesis by modulating the Hippo pathway. Oncogene. 2015;34:3536–3546. doi: 10.1038/onc.2014.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sanchez-Vega F, et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell. 2018;173:321–337.:e310. doi: 10.1016/j.cell.2018.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Szulzewsky F, Holland EC, Vasioukhin V. YAP1 and its fusion proteins in cancer initiation, progression and therapeutic resistance. Dev Biol. 2021;475:205–221. doi: 10.1016/j.ydbio.2020.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Seavey CN, et al. WWTR1(TAZ)-CAMTA1 gene fusion is sufficient to dysregulate YAP/TAZ signaling and drive epithelioid hemangioendothelioma tumorigenesis. Genes Dev. 2021;35:512–527. doi: 10.1101/gad.348220.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Driskill JH, et al. WWTR1(TAZ)-CAMTA1 reprograms endothelial cells to drive epithelioid hemangioendothelioma. Genes Dev. 2021;35:495–511. doi: 10.1101/gad.348221.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Szulzewsky F, et al. Comparison of tumor-associated YAP1 fusions identifies a recurrent set of functions critical for oncogenesis. Genes Dev. 2020;34:1051–1064. doi: 10.1101/gad.338681.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ben C, et al. Alternative splicing reverses the cell-intrinsic and cell-extrinsic pro-oncogenic potentials of YAP1. J Biol Chem. 2020;295:13965–13980. doi: 10.1074/jbc.RA120.013820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Vrbsky J, et al. Evidence for discrete modes of YAP1 signaling via mRNA splice isoforms in development and diseases. Genomics. 2021;113:1349–1365. doi: 10.1016/j.ygeno.2021.03.009. [DOI] [PubMed] [Google Scholar]
- 82.Pearson JD, et al. Binary pan-cancer classes with distinct vulnerabilities defined by pro-or anti-cancer YAP/TEAD activity. Cancer Cell. 2021;39:1115–1134.:e1112. doi: 10.1016/j.ccell.2021.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cottini F, et al. Rescue of Hippo coactivator YAP1 triggers DNA damage-induced apoptosis in hematological cancers. Nat Med. 2014;20:599–606. doi: 10.1038/nm.3562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zheng B, et al. Integrated Transcriptomic Analysis Reveals a Distinctive Role of YAP1 in Extramedullary Invasion and Therapeutic Sensitivity of Multiple Myeloma. Front Oncol. 2021;11:787814. doi: 10.3389/fonc.2021.787814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022;12:31–46. doi: 10.1158/2159-8290.CD-21-1059. [DOI] [PubMed] [Google Scholar]
- 86.Panciera T, et al. Induction of Expandable Tissue-Specific Stem/Progenitor Cells through Transient Expression of YAP/TAZ. Cell Stem Cell. 2016;19:725–737. doi: 10.1016/j.stem.2016.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bai H, et al. Yes-associated protein regulates the hepatic response after bile duct ligation. Hepatology. 2012;56:1097–1107. doi: 10.1002/hep.25769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Cai J, et al. The Hippo signaling pathway restricts the oncogenic potential of an intestinal regeneration program. Genes Dev. 2010;24:2383–2388. doi: 10.1101/gad.1978810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lee MJ, Byun MR, Furutani-Seiki M, Hong JH, Jung HS. YAP and TAZ regulate skin wound healing. J Invest Dermatol. 2014;134:518–525. doi: 10.1038/jid.2013.339. [DOI] [PubMed] [Google Scholar]
- 90.Su T, et al. Two-signal requirement for growth-promoting function of Yap in hepatocytes. Elife. 2015;4 doi: 10.7554/eLife.02948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zanconato F, et al. Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth. Nat Cell Biol. 2015;17:1218–1227. doi: 10.1038/ncb3216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Yui S, et al. YAP/TAZ-Dependent Reprogramming of Colonic Epithelium Links ECM Remodeling to Tissue Regeneration. Cell Stem Cell. 2018;22:35–49.:e37. doi: 10.1016/j.stem.2017.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Serra D, et al. Self-organization and symmetry breaking in intestinal organoid development. Nature. 2019;569:66–72. doi: 10.1038/s41586-019-1146-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ohara TE, Colonna M, Stappenbeck TS. Adaptive differentiation promotes intestinal villus recovery. Dev Cell. 2022;57:166–179.:e166. doi: 10.1016/j.devcel.2021.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Heuberger J, et al. High Yap and Mll1 promote a persistent regenerative cell state induced by Notch signaling and loss of p53. Proc Natl Acad Sci U S A. 2021;118 doi: 10.1073/pnas.2019699118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Planas-Paz L, et al. YAP, but Not RSPO-LGR4/5, Signaling in Biliary Epithelial Cells Promotes a Ductular Reaction in Response to Liver Injury. Cell Stem Cell. 2019;25:39–53.:e10. doi: 10.1016/j.stem.2019.04.005. [DOI] [PubMed] [Google Scholar]
- 97.Yimlamai D, et al. Hippo pathway activity influences liver cell fate. Cell. 2014;157:1324–1338. doi: 10.1016/j.cell.2014.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Robledinos-Anton N, Escoll M, Guan KL, Cuadrado A. TAZ Represses the Neuronal Commitment of Neural Stem Cells. Cells. 2020;9 doi: 10.3390/cells9102230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Talwar S, Kant A, Xu T, Shenoy VB, Assoian RK. Mechanosensitive smooth muscle cell phenotypic plasticity emerging from a null state and the balance between Rac and Rho. Cell Rep. 2021;35:109019. doi: 10.1016/j.celrep.2021.109019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Xu Z, Orkwis JA, Harris GM. Cell Shape and Matrix Stiffness Impact Schwann Cell Plasticity via YAP/TAZ and Rho GTPases. Int J Mol Sci. 2021;22 doi: 10.3390/ijms22094821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Castellan M, et al. Single-cell analyses reveal YAP/TAZ as regulators of stemness and cell plasticity in Glioblastoma. Nat Cancer. 2021;2:174–188. doi: 10.1038/s43018-020-00150-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Totaro A, Panciera T, Piccolo S. YAP/TAZ upstream signals and downstream responses. Nat Cell Biol. 2018;20:888–899. doi: 10.1038/s41556-018-0142-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jang W, Kim T, Koo JS, Kim SK, Lim DS. Mechanical cue-induced YAP instructs Skp2-dependent cell cycle exit and oncogenic signaling. EMBO J. 2017;36:2510–2528. doi: 10.15252/embj.201696089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kapoor A, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell. 2014;158:185–197. doi: 10.1016/j.cell.2014.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mizuno T, et al. YAP induces malignant mesothelioma cell proliferation by upregulating transcription of cell cycle-promoting genes. Oncogene. 2012;31:5117–5122. doi: 10.1038/onc.2012.5. [DOI] [PubMed] [Google Scholar]
- 106.Baia GS, et al. Yes-associated protein 1 is activated and functions as an oncogene in meningiomas. Mol Cancer Res. 2012;10:904–913. doi: 10.1158/1541-7786.MCR-12-0116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bartucci M, et al. TAZ is required for metastatic activity and chemoresistance of breast cancer stem cells. Oncogene. 2015;34:681–690. doi: 10.1038/onc.2014.5. [DOI] [PubMed] [Google Scholar]
- 108.Ciamporcero E, et al. YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene. 2016;35:1541–1553. doi: 10.1038/onc.2015.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fernandez LA, et al. Oncogenic YAP promotes radioresistance and genomic instability in medulloblastoma through IGF2-mediated Akt activation. Oncogene. 2012;31:1923–1937. doi: 10.1038/onc.2011.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hall CA, et al. Hippo pathway effector Yap is an ovarian cancer oncogene. Cancer Res. 2010;70:8517–8525. doi: 10.1158/0008-5472.CAN-10-1242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kim H, et al. YAP, CTGF and Cyr61 are overexpressed in tamoxifen-resistant breast cancer and induce transcriptional repression of ERalpha. J Cell Sci. 2021;134 doi: 10.1242/jcs.256503. [DOI] [PubMed] [Google Scholar]
- 112.Kim MH, et al. Actin remodeling confers BRAF inhibitor resistance to melanoma cells through YAP/TAZ activation. EMBO J. 2016;35:462–478. doi: 10.15252/embj.201592081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lin CH, et al. Microenvironment rigidity modulates responses to the HER2 receptor tyrosine kinase inhibitor lapatinib via YAP and TAZ transcription factors. Mol Biol Cell. 2015;26:3946–3953. doi: 10.1091/mbc.E15-07-0456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Lin L, et al. The Hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet. 2015;47:250–256. doi: 10.1038/ng.3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Mao B, et al. SIRT1 regulates YAP2-mediated cell proliferation and chemoresistance in hepatocellular carcinoma. Oncogene. 2014;33:1468–1474. doi: 10.1038/onc.2013.88. [DOI] [PubMed] [Google Scholar]
- 116.Cheng H, et al. Functional genomics screen identifies YAP1 as a key determinant to enhance treatment sensitivity in lung cancer cells. Oncotarget. 2016;7:28976–28988. doi: 10.18632/oncotarget.6721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ercan D, et al. Reactivation of ERK signaling causes resistance to EGFR kinase inhibitors. Cancer Discov. 2012;2:934–947. doi: 10.1158/2159-8290.CD-12-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kurppa KJ, et al. Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway. Cancer Cell. 2020;37:104–122.:e112. doi: 10.1016/j.ccell.2019.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ohta Y, et al. Cell-matrix interface regulates dormancy in human colon cancer stem cells. Nature. 2022 doi: 10.1038/s41586-022-05043-y. [DOI] [PubMed] [Google Scholar]
- 120.Gao R, et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol Med. 2021;13:e14351. doi: 10.15252/emmm.202114351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Warren JSA, Xiao Y, Lamar JM. YAP/TAZ Activation as a Target for Treating Metastatic Cancer. Cancers (Basel) 2018;10 doi: 10.3390/cancers10040115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lamar JM, et al. The Hippo pathway target, YAP, promotes metastasis through its TEAD-interaction domain. Proc Natl Acad Sci U S A. 2012;109:E2441–2450. doi: 10.1073/pnas.1212021109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Nallet-Staub F, et al. Pro-invasive activity of the Hippo pathway effectors YAP and TAZ in cutaneous melanoma. J Invest Dermatol. 2014;134:123–132. doi: 10.1038/jid.2013.319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Liu J, et al. Synaptopodin-2 suppresses metastasis of triple-negative breast cancer via inhibition of YAP/TAZ activity. J Pathol. 2018;244:71–83. doi: 10.1002/path.4995. [DOI] [PubMed] [Google Scholar]
- 125.Lee CK, et al. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science. 2019;363:644–649. doi: 10.1126/science.aav0173. [DOI] [PubMed] [Google Scholar]
- 126.Mason DE, et al. YAP and TAZ limit cytoskeletal and focal adhesion maturation to enable persistent cell motility. J Cell Biol. 2019;218:1369–1389. doi: 10.1083/jcb.201806065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Haemmerle M, et al. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat Commun. 2017;8:310. doi: 10.1038/s41467-017-00411-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sharif GM, et al. Cell growth density modulates cancer cell vascular invasion via Hippo pathway activity and CXCR2 signaling. Oncogene. 2015;34:5879–5889. doi: 10.1038/onc.2015.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Huang JL, Urtatiz O, Van Raamsdonk CD. Oncogenic G Protein GNAQ Induces Uveal Melanoma and Intravasation in Mice. Cancer Res. 2015;75:3384–3397. doi: 10.1158/0008-5472.CAN-14-3229. [DOI] [PubMed] [Google Scholar]
- 130.Gu JJ, et al. Inactivation of ABL kinases suppresses non-small cell lung cancer metastasis. JCI Insight. 2016;1:e89647. doi: 10.1172/jci.insight.89647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Er EE, et al. Pericyte-like spreading by disseminated cancer cells activates YAP and MRTF for metastatic colonization. Nat Cell Biol. 2018;20:966–978. doi: 10.1038/s41556-018-0138-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Albrengues J, et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018;361 doi: 10.1126/science.aao4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Papalazarou V, et al. The creatine-phosphagen system is mechanoresponsive in pancreatic adenocarcinoma and fuels invasion and metastasis. Nat Metab. 2020;2:62–80. doi: 10.1038/s42255-019-0159-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gensbittel V, et al. Mechanical Adaptability of Tumor Cells in Metastasis. Dev Cell. 2021;56:164–179. doi: 10.1016/j.devcel.2020.10.011. [DOI] [PubMed] [Google Scholar]
- 135.Denais CM, et al. Nuclear envelope rupture and repair during cancer cell migration. Science. 2016;352:353–358. doi: 10.1126/science.aad7297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Raab M, et al. ESCRT III repairs nuclear envelope ruptures during cell migration to limit DNA damage and cell death. Science. 2016;352:359–362. doi: 10.1126/science.aad7611. [DOI] [PubMed] [Google Scholar]
- 137.Sladitschek-Martens HL, et al. YAP/TAZ activity in stromal cells prevents ageing by controlling cGAS-STING. Nature. 2022;607:790–798. doi: 10.1038/s41586-022-04924-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Qiao Y, et al. YAP Regulates Actin Dynamics through ARHGAP29 and Promotes Metastasis. Cell Rep. 2017;19:1495–1502. doi: 10.1016/j.celrep.2017.04.075. [DOI] [PubMed] [Google Scholar]
- 139.Kidiyoor GR, et al. ATR is essential for preservation of cell mechanics and nuclear integrity during interstitial migration. Nat Commun. 2020;11:4828. doi: 10.1038/s41467-020-18580-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Chen CL, Schroeder MC, Kango-Singh M, Tao C, Halder G. Tumor suppression by cell competition through regulation of the Hippo pathway. Proc Natl Acad Sci U S A. 2012;109:484–489. doi: 10.1073/pnas.1113882109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Norman M, et al. Loss of Scribble causes cell competition in mammalian cells. J Cell Sci. 2012;125:59–66. doi: 10.1242/jcs.085803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Baker NE. Emerging mechanisms of cell competition. Nat Rev Genet. 2020;21:683–697. doi: 10.1038/s41576-020-0262-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Moya IM, et al. Peritumoral activation of the Hippo pathway effectors YAP and TAZ suppresses liver cancer in mice. Science. 2019;366:1029–1034. doi: 10.1126/science.aaw9886. [DOI] [PubMed] [Google Scholar]
- 144.Liu Z, et al. Differential YAP expression in glioma cells induces cell competition and promotes tumorigenesis. J Cell Sci. 2019;132 doi: 10.1242/jcs.225714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cheung P, et al. Regenerative Reprogramming of the Intestinal Stem Cell State via Hippo Signaling Suppresses Metastatic Colorectal Cancer. Cell Stem Cell. 2020;27:590–604.:e599. doi: 10.1016/j.stem.2020.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Konsavage WM, Jr, Kyler SL, Rennoll SA, Jin G, Yochum GS. Wnt/beta-catenin signaling regulates Yes-associated protein (YAP) gene expression in colorectal carcinoma cells. J Biol Chem. 2012;287:11730–11739. doi: 10.1074/jbc.M111.327767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Rosenbluh J, et al. beta-Catenin-driven cancers require a YAP1 transcriptional complex for survival and tumorigenesis. Cell. 2012;151:1457–1473. doi: 10.1016/j.cell.2012.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zhou D, et al. Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc Natl Acad Sci U S A. 2011;108:E1312–1320. doi: 10.1073/pnas.1110428108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Boopathy GTK, Hong W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front Cell Dev Biol. 2019;7:49. doi: 10.3389/fcell.2019.00049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.He J, et al. Yes-Associated Protein Promotes Angiogenesis via Signal Transducer and Activator of Transcription 3 in Endothelial Cells. Circ Res. 2018;122:591–605. doi: 10.1161/CIRCRESAHA.117.311950. [DOI] [PubMed] [Google Scholar]
- 151.Yan Y, Song Q, Yao L, Zhao L, Cai H. YAP Overexpression in Breast Cancer Cells Promotes Angiogenesis Through Activating Yap Signaling in Vascular Endothelial Cells. ResearchSquare. 2020 doi: 10.1155/2022/5942379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Marti P, et al. YAP promotes proliferation, chemoresistance, and angiogenesis in human cholangiocarcinoma through TEAD transcription factors. Hepatology. 2015;62:1497–1510. doi: 10.1002/hep.27992. [DOI] [PubMed] [Google Scholar]
- 153.Xu S, Zhang H, Chong Y, Guan B, Guo P. YAP Promotes VEGFA Expression and Tumor Angiogenesis Though Gli2 in Human Renal Cell Carcinoma. Arch Med Res. 2019;50:225–233. doi: 10.1016/j.arcmed.2019.08.010. [DOI] [PubMed] [Google Scholar]
- 154.Wang X, et al. YAP/TAZ Orchestrate VEGF Signaling during Developmental Angiogenesis. Dev Cell. 2017;42:462–478.:e467. doi: 10.1016/j.devcel.2017.08.002. [DOI] [PubMed] [Google Scholar]
- 155.Tocci P, Blandino G, Bagnato A. YAP and endothelin-1 signaling: an emerging alliance in cancer. J Exp Clin Cancer Res. 2021;40:27. doi: 10.1186/s13046-021-01827-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Shen Y, et al. Reduction of Liver Metastasis Stiffness Improves Response to Bevacizumab in Metastatic Colorectal Cancer. Cancer Cell. 2020;37:800–817.:e807. doi: 10.1016/j.ccell.2020.05.005. [DOI] [PubMed] [Google Scholar]
- 157.Weis SM, Cheresh DA. Pathophysiological consequences of VEGF-induced vascular permeability. Nature. 2005;437:497–504. doi: 10.1038/nature03987. [DOI] [PubMed] [Google Scholar]
- 158.Wang KC, et al. Flow-dependent YAP/TAZ activities regulate endothelial phenotypes and atherosclerosis. Proc Natl Acad Sci U S A. 2016;113:11525–11530. doi: 10.1073/pnas.1613121113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wang L, et al. Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature. 2016;540:579–582. doi: 10.1038/nature20602. [DOI] [PubMed] [Google Scholar]
- 160.Shen YW, Zhou YD, Luan X, Zhang WD. Blocking CTGF-Mediated Tumor-Stroma Interplay in Pancreatic Cancer. Trends Mol Med. 2020;26:1064–1067. doi: 10.1016/j.molmed.2020.08.005. [DOI] [PubMed] [Google Scholar]
- 161.Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355. doi: 10.1126/science.aar4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018;8:1069–1086. doi: 10.1158/2159-8290.CD-18-0367. [DOI] [PubMed] [Google Scholar]
- 163.Guo X, et al. Single tumor-initiating cells evade immune clearance by recruiting type II macrophages. Genes Dev. 2017;31:247–259. doi: 10.1101/gad.294348.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Kim W, et al. Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma. Gut. 2018;67:1692–1703. doi: 10.1136/gutjnl-2017-314061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wang X, et al. Hepatocyte TAZ/WWTR1 Promotes Inflammation and Fibrosis in Nonalcoholic Steatohepatitis. Cell Metab. 2016;24:848–862. doi: 10.1016/j.cmet.2016.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Murakami S, et al. Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma. Oncogene. 2017;36:1232–1244. doi: 10.1038/onc.2016.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Wang G, et al. Targeting YAP-Dependent MDSC Infiltration Impairs Tumor Progression. Cancer Discov. 2016;6:80–95. doi: 10.1158/2159-8290.CD-15-0224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Marigo I, et al. Disabled Homolog 2 Controls Prometastatic Activity of Tumor-Associated Macrophages. Cancer Discov. 2020;10:1758–1773. doi: 10.1158/2159-8290.CD-20-0036. [DOI] [PubMed] [Google Scholar]
- 169.Ni X, et al. YAP Is Essential for Treg-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2018;8:1026–1043. doi: 10.1158/2159-8290.CD-17-1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Geng J, et al. The transcriptional coactivator TAZ regulates reciprocal differentiation of TH17 cells and Treg cells. Nat Immunol. 2017;18:800–812. doi: 10.1038/ni.3748. [DOI] [PubMed] [Google Scholar]
- 171.Lebid A, Chung L, Pardoll DM, Pan F. YAP Attenuates CD8 T Cell-Mediated Antitumor Response. Front Immunol. 2020;11:580. doi: 10.3389/fimmu.2020.00580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Stampouloglou E, et al. Yap suppresses T-cell function and infiltration in the tumor microenvironment. PLoS Biol. 2020;18:e3000591. doi: 10.1371/journal.pbio.3000591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Meng KP, Majedi FS, Thauland TJ, Butte MJ. Mechanosensing through YAP controls T cell activation and metabolism. J Exp Med. 2020;217 doi: 10.1084/jem.20200053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Barry ER, Simov V, Valtingojer I, Venier O. Recent Therapeutic Approaches to Modulate the Hippo Pathway in Oncology and Regenerative Medicine. Cells. 2021;10 doi: 10.3390/cells10102715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Liu-Chittenden Y, et al. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012;26:1300–1305. doi: 10.1101/gad.192856.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Dasari VR, et al. Verteporfin exhibits YAP-independent anti-proliferative and cytotoxic effects in endometrial cancer cells. Oncotarget. 2017;8:28628–28640. doi: 10.18632/oncotarget.15614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zhang H, et al. Tumor-selective proteotoxicity of verteporfin inhibits colon cancer progression independently of YAP1. Sci Signal. 2015;8:ra98. doi: 10.1126/scisignal.aac5418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Chan P, et al. Autopalmitoylation of TEAD proteins regulates transcriptional output of the Hippo pathway. Nat Chem Biol. 2016;12:282–289. doi: 10.1038/nchembio.2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Pobbati AV, et al. Targeting the Central Pocket in Human Transcription Factor TEAD as a Potential Cancer Therapeutic Strategy. Structure. 2015;23:2076–2086. doi: 10.1016/j.str.2015.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kaneda A, et al. The novel potent TEAD inhibitor, K-975, inhibits YAP1/TAZ-TEAD protein-protein interactions and exerts an anti-tumor effect on malignant pleural mesothelioma. Am J Cancer Res. 2020;10:4399–4415. [PMC free article] [PubMed] [Google Scholar]
- 181.Tang TT, et al. Small Molecule Inhibitors of TEAD Auto-palmitoylation Selectively Inhibit Proliferation and Tumor Growth of NF2-deficient Mesothelioma. Mol Cancer Ther. 2021;20:986–998. doi: 10.1158/1535-7163.MCT-20-0717. [DOI] [PubMed] [Google Scholar]
- 182.Shorstova T, Foulkes WD, Witcher M. Achieving clinical success with BET inhibitors as anti-cancer agents. Br J Cancer. 2021;124:1478–1490. doi: 10.1038/s41416-021-01321-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Jiang W, Hu JW, He XR, Jin WL, He XY. Statins: a repurposed drug to fight cancer. J Exp Clin Cancer Res. 2021;40:241. doi: 10.1186/s13046-021-02041-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Kim S, Kim SA, Han J, Kim IS. Rho-Kinase as a Target for Cancer Therapy and Its Immunotherapeutic Potential. Int J Mol Sci. 2021;22 doi: 10.3390/ijms222312916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Martellucci S, et al. Src Family Kinases as Therapeutic Targets in Advanced Solid Tumors: What We Have Learned so Far. Cancers (Basel) 2020;12 doi: 10.3390/cancers12061448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Gjorevski N, et al. Designer matrices for intestinal stem cell and organoid culture. Nature. 2016;539:560–564. doi: 10.1038/nature20168. [DOI] [PubMed] [Google Scholar]
- 187.Lee Y, et al. Common and Unique Transcription Signatures of YAP and TAZ in Gastric Cancer Cells. Cancers (Basel) 2020;12 doi: 10.3390/cancers12123667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Li Z, et al. Structural insights into the YAP and TEAD complex. Genes Dev. 2010;24:235–240. doi: 10.1101/gad.1865810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Sehnal D, et al. Mol* Viewer: modern web app for 3D visualization and analysis of large biomolecular structures. Nucleic Acids Res. 2021;49:W431–W437. doi: 10.1093/nar/gkab314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Noland CL, et al. Palmitoylation of TEAD Transcription Factors Is Required for Their Stability and Function in Hippo Pathway Signaling. Structure. 2016;24:179–186. doi: 10.1016/j.str.2015.11.005. [DOI] [PubMed] [Google Scholar]
- 191.Mouillet-Richard S, Laurent-Puig P. YAP/TAZ Signalling in Colorectal Cancer: Lessons from Consensus Molecular Subtypes. Cancers (Basel) 2020;12 doi: 10.3390/cancers12113160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Zhou Z, et al. siRNA targeting YAP gene inhibits gastric carcinoma growth and tumor metastasis in SCID mice. Oncol Lett. 2016;11:2806–2814. doi: 10.3892/ol.2016.4319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Wang X, Wu B, Zhong Z. Downregulation of YAP inhibits proliferation, invasion and increases cisplatin sensitivity in human hepatocellular carcinoma cells. Oncol Lett. 2018;16:585–593. doi: 10.3892/ol.2018.8633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Jiang Z, et al. Inhibiting YAP expression suppresses pancreatic cancer progression by disrupting tumor-stromal interactions. J Exp Clin Cancer Res. 2018;37:69. doi: 10.1186/s13046-018-0740-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Lin M, et al. TAZ is overexpressed in prostate cancers and regulates the proliferation, migration and apoptosis of prostate cancer PC3 cells. Oncol Rep. 2020;44:747–756. doi: 10.3892/or.2020.7616. [DOI] [PubMed] [Google Scholar]
- 196.Marx A, et al. Up regulation of the Hippo signalling effector YAP1 is linked to early biochemical recurrence in prostate cancers. Sci Rep. 2020;10:8916. doi: 10.1038/s41598-020-65772-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Debaugnies M, et al. YAP and TAZ are essential for basal and squamous cell carcinoma initiation. EMBO Rep. 2018;19 doi: 10.15252/embr.201845809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Jia J, et al. Yes-Associated Protein Contributes to the Development of Human Cutaneous Squamous Cell Carcinoma via Activation of RAS. J Invest Dermatol. 2016;136:1267–1277. doi: 10.1016/j.jid.2016.02.005. [DOI] [PubMed] [Google Scholar]
- 199.Maglic D, et al. YAP-TEAD signaling promotes basal cell carcinoma development via a c-JUN/AP1 axis. EMBO J. 2018;37 doi: 10.15252/embj.201798642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Zhang X, et al. Somatic Hypermutation of the YAP Oncogene in a Human Cutaneous Melanoma. Mol Cancer Res. 2019;17:1435–1449. doi: 10.1158/1541-7786.MCR-18-0407. [DOI] [PubMed] [Google Scholar]
- 201.Zucchini C, et al. ROCK2 deprivation leads to the inhibition of tumor growth and metastatic potential in osteosarcoma cells through the modulation of YAP activity. J Exp Clin Cancer Res. 2019;38:503. doi: 10.1186/s13046-019-1506-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Isfort I, et al. Prevalence of the Hippo Effectors YAP1/TAZ in Tumors of Soft Tissue and Bone. Sci Rep. 2019;9:19704. doi: 10.1038/s41598-019-56247-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Zhang WQ, et al. Targeting YAP in malignant pleural mesothelioma. J Cell Mol Med. 2017;21:2663–2676. doi: 10.1111/jcmm.13182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Jongsma J, et al. A conditional mouse model for malignant mesothelioma. Cancer Cell. 2008;13:261–271. doi: 10.1016/j.ccr.2008.01.030. [DOI] [PubMed] [Google Scholar]
- 205.Altomare DA, et al. A mouse model recapitulating molecular features of human mesothelioma. Cancer Res. 2005;65:8090–8095. doi: 10.1158/0008-5472.CAN-05-2312. [DOI] [PubMed] [Google Scholar]
- 206.Coy S, Rashid R, Stemmer-Rachamimov A, Santagata S. An update on the CNS manifestations of neurofibromatosis type 2. Acta Neuropathol. 2020;139:643–665. doi: 10.1007/s00401-019-02029-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Kalamarides M, et al. Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse. Genes Dev. 2002;16:1060–1065. doi: 10.1101/gad.226302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Giovannini M, et al. Conditional biallelic Nf2 mutation in the mouse promotes manifestations of human neurofibromatosis type 2. Genes Dev. 2000;14:1617–1630. [PMC free article] [PubMed] [Google Scholar]
- 209.Eder N, et al. YAP1/TAZ drives ependymoma-like tumour formation in mice. Nat Commun. 2020;11:2380. doi: 10.1038/s41467-020-16167-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Malouf GG, et al. Molecular characterization of sarcomatoid clear cell renal cell carcinoma unveils new candidate oncogenic drivers. Sci Rep. 2020;10:701. doi: 10.1038/s41598-020-57534-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Mehra R, et al. Biallelic Alteration and Dysregulation of the Hippo Pathway in Mucinous Tubular and Spindle Cell Carcinoma of the Kidney. Cancer Discov. 2016;6:1258–1266. doi: 10.1158/2159-8290.CD-16-0267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Morris ZS, McClatchey AI. Aberrant epithelial morphology and persistent epidermal growth factor receptor signaling in a mouse model of renal carcinoma. Proc Natl Acad Sci U S A. 2009;106:9767–9772. doi: 10.1073/pnas.0902031106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Carter P, et al. Deletion of Lats1/2 in adult kidney epithelia leads to renal cell carcinoma. J Clin Invest. 2021;131 doi: 10.1172/JCI144108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Sugiura K, et al. The Expression of Yes-Associated Protein (YAP) Maintains Putative Cancer Stemness and Is Associated with Poor Prognosis in Intrahepatic Cholangiocarcinoma. Am J Pathol. 2019;189:1863–1877. doi: 10.1016/j.ajpath.2019.05.014. [DOI] [PubMed] [Google Scholar]
- 215.He C, et al. A Human Papillomavirus-Independent Cervical Cancer Animal Model Reveals Unconventional Mechanisms of Cervical Carcinogenesis. Cell Rep. 2019;26:2636–2650.:e2635. doi: 10.1016/j.celrep.2019.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Chai AWY, et al. Genome-wide CRISPR screens of oral squamous cell carcinoma reveal fitness genes in the Hippo pathway. Elife. 2020;9 doi: 10.7554/eLife.57761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Cho SY, et al. Expression of Yes-associated protein 1 and its clinical significance in ovarian serous cystadenocarcinoma. Oncol Rep. 2017;37:2620–2632. doi: 10.3892/or.2017.5517. [DOI] [PMC free article] [PubMed] [Google Scholar]




