Next Article in Journal
Hidden Blood Loss and Its Possible Risk Factors in Full Endoscopic Lumbar Interbody Fusion
Previous Article in Journal
Influence of the Psychological Effect of COVID-19 on Saudi Student Nurses’ Attitudes toward Nursing and Their Future Career Plans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Familial CCM Genes Might Not Be Main Drivers for Pathogenesis of Sporadic CCMs-Genetic Similarity between Cancers and Vascular Malformations

Departments of Molecular & Translational Medicine (MTM), Texas Tech University Health Science Center El Paso (TTUHSCEP), El Paso, TX 79905, USA
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(4), 673; https://doi.org/10.3390/jpm13040673
Submission received: 20 March 2023 / Revised: 5 April 2023 / Accepted: 15 April 2023 / Published: 17 April 2023

Abstract

:
Cerebral cavernous malformations (CCMs) are abnormally dilated intracranial capillaries that form cerebrovascular lesions with a high risk of hemorrhagic stroke. Recently, several somatic “activating” gain-of-function (GOF) point mutations in PIK3CA (phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit p110α) were discovered as a dominant mutation in the lesions of sporadic forms of cerebral cavernous malformation (sCCM), raising the possibility that CCMs, like other types of vascular malformations, fall in the PIK3CA-related overgrowth spectrum (PROS). However, this possibility has been challenged with different interpretations. In this review, we will continue our efforts to expound the phenomenon of the coexistence of gain-of-function (GOF) point mutations in the PIK3CA gene and loss-of-function (LOF) mutations in CCM genes in the CCM lesions of sCCM and try to delineate the relationship between mutagenic events with CCM lesions in a temporospatial manner. Since GOF PIK3CA point mutations have been well studied in reproductive cancers, especially breast cancer as a driver oncogene, we will perform a comparative meta-analysis for GOF PIK3CA point mutations in an attempt to demonstrate the genetic similarities shared by both cancers and vascular anomalies.

1. Introduction

Cerebral cavernous malformations (CCMs) and developmental venous anomalies (DVAs) are two types of cerebral vascular malformations characterized by abnormal blood vessels in the brain. CCMs are clusters of abnormal capillaries, while DVAs are irregular arrangements of cerebral veins. While most familial cases of CCM (fCCM) are caused by gene mutations, the cause of most sporadic cases (sCCM) remains unknown. However, activating mutations in the gene PIK3CA have been recently linked to the pathogenesis of CCMs [1,2,3,4]. There is ongoing debate about the relationship between CCMs and DVAs and their potential interaction [5]. This review aims to examine the relationship between altered PI3K signaling pathways and the formation and progression of sporadic CCMs, drawing on data from tumorigenic genetics in reproductive cancer, specifically breast cancer.
The transforming potential of PI3K catalytic isoforms was realized decades ago in cancer research [6,7,8], and these isoforms have been defined as oncogenes [8,9]. Somatic GOF PIK3CA point mutations have been frequently described in various cancers as an oncogene [10,11,12,13,14,15], as well as in many forms of vascular anomalies, especially the well-known vascular malformations (VMs) [16,17,18,19,20]. This shared genetic similarity between tumors and vascular anomalies suggests that as benign tumors, VeMs might have similar genetic underpins as cancers caused by the exact same somatic PIK3CA GOF mutations [16,19,20,21].
CCMs (or cavernous angiomas) are clusters of tightly packed, abnormal cerebral capillaries with thin walls, while DVAs (or venous angiomas) are irregular arrangements of cerebral veins, and DVAs are the most common form of congenital cerebral venous malformations (VeMs) without any clinically symptomatic event [22]. It has been well defined that most fCCM cases are caused by mutations in one of the three CCM genes, while the majority of sCCM cases are still unknown, despite that CCM gene mutations have been found in some sCCM [23,24]. A common clinical phenomenon of sCCM is its frequent association with DVA [25,26,27]. Recently, activating PIK3CA ‘hot spot’ point mutations (DVAs/VeMs causative mutations) has been proposed as the possible fourth locus for the pathogenesis of CCMs [4]. However, the activation of PI3K pathways has not been found to be an universal regulator of vascular morphogenesis, especially for CCMs. Furthermore, following reports on the coexistence of CCMs and DVAs, more attention has been given to the potential interaction and relationship between CCMs and DVAs. One vessel structural/fluid dynamic hypothesis suggested that either CCMs or DVAs can directly or indirectly affect their adjacent area venous outflow through their malformed cerebral vessels, which could further exacerbate pre-existing structural DVAs followed by further worsened outflow. Under prolonged exposure to altered venous outflow, DVAs can eventually induce hemorrhagic events in adjacent CCM lesions [28]. Another genetic hypothesis suggested that ECs of DVAs with GOF mutations in the PIK3CA gene have high somatic mutation potential as the known tumor driver mutation. These clonal ECs of DVAs with GOF PIK3CA mutations are hyper-proliferative while migrating along vasculatures, leading to the occurrence of secondary mutations, including CCM genes, in these ECs, which then result in the formation and progression of sporadic CCM lesions at DVAs adjacent vessels [5].
This review aims to examine the relationship between altered PI3K signaling pathways and the formation and progression of sporadic CCMs, drawing on data from tumorigenic genetics in reproductive cancer, specifically breast cancer.

2. Common Somatic Activating GOF Mutation of PIK3CA Gene Is the Key for Tumorigenesis

Spontaneous somatic mutation is a common event. Genetic mutations in somatic cells are generally random and can contribute to aging, neurodegeneration, and cancer initiation [29,30]. These mutations occur in every individual tissue and accumulate over time, leading to clonal expansion of cells with driver mutations in either cancer [31,32,33] or non-cancer tissues [34,35]. Although most spontaneous mutations have no noticeable effect, some mutations can alter key cellular functions, and, with time, the accumulation of these key mutations can lead to the growth advantage of cells, which is the first step in tumorigenesis [36,37,38,39]. Hereditary oncogenic genes, which are rare germline mutations in DNA repair pathways, increase the risk of cancer [40,41], but most cancers are caused by somatic mutations in oncogenic genes [30,42,43,44,45,46,47]. Studies have shown that certain genetic mutations confer a selective growth advantage [48,49] and can be considered cancer drivers [31,33], suggesting that most cancers arise from “bad luck” random events [48,50,51,52,53,54]. Cancer driver mutations are responsible for oncogenesis, and many tumors have recurrent somatic mutations in the PI3K-AKT1-mTOR signaling pathways [9,55,56,57], making these PI3K signaling genes good candidates for cancer therapy [57,58,59,60,61,62,63,64,65,66,67,68,69,70].
PI3K/AKT/mTOR signaling pathway and cancer. The spontaneous somatic mutations of specific oncogenic genes, including the PI3K/AKT/mTOR signaling pathway, are crucial in regulating cellular processes such as survival, growth, differentiation, metabolism, and cytoskeletal reorganization [59,60,61]. These mutations are a major cause of cancer [59,71]. As an evolutionary conserved family of lipid kinases [71,72], the PI3K pathway is one of the most frequently activated in human cancer [30,42,43,44,45,46,47], where it phosphorylates phosphatidylinositides to regulate downstream signaling [73,74]. One of the most significant transforming potentials from deregulation of class I PI3K is reflected in the PIK3CA gene [12,14,15,56,75,76,77,78,79,80,81,82,83].
Somatic activating GOF PIK3CA point mutation is a tumor driver. Genomic amplification of both PIK3CA and AKT genes has been linked to early stages of tumor formation [12,80,84,85,86,87,88,89]. Activating mutations in these genes have been frequently identified in many types of cancer and have been shown to result in hyper-activation of the PI3K/AKT/mTOR signaling pathway [14,15,56,76,77,78,79,80,81,82,83]. This leads to a range of altered cellular processes, including survival, proliferation, growth, metabolism, angiogenesis, and metastasis [83,90,91,92]. GOF point mutations in the PIK3CA gene have been identified as causative oncogenic mutations in various cancer types, particularly in reproductive cancers [14,57,91,93,94,95,96], making PIK3CA one of the major cancer driver genes [14,15,56,76,77,78,79,80,81,82,83]. Of the three class-I PI3K catalytic isoforms, only PIK3CA has been shown to be essential for vascular development, and its expression is selective in endothelial PI3K signaling during angiogenesis [90,97,98,99]. This has been supported by numerous clinical reports that show the same GOF point mutations in PIK3CA gene are present in both tumors and vascular anomalies [3,4,98,99,100,101,102,103,104,105,106,107].

3. Somatic GOF PIK3CA Mutations Are Oncogenic and Driver in Breast Cancer

Oncogenic mutations in the PI3K signaling pathway and their role in breast cancer. DNA sequencing of cancer samples revealed that among the 16 members of the PI3K family, the PIK3CA gene is the only one with activating (GOF) mutations [12,75,108,109,110], making it oncogenic [12,75]. This was suggested by the high frequency of GOF PIK3CA mutations in breast cancer cases [111,112]. Additionally, activating mutations in other genes in the PI3K signaling pathway, such as AKT1, are also commonly found in breast cancer [112]. Oncogenic mutations in the PI3K kinase signaling pathway generally refer to activating mutations in key members such as PIK3CA and AKT1, as well as other well-known oncogenic genes like TP53 [91,113,114,115]. The hyper-proliferative nature of these mutations leads to genomic instability and an increased number of secondary mutations in the cancer cells during the evolution of the cancer. Some of these passenger mutations might contribute to tumor progression [116,117,118], while others are not found to have a significant impact or their genetic role is not yet fully understood [54,116,119,120,121].
PIK3CA and TP53 oncogenes as key drivers of breast cancer tumorigenesis. Both TP53 and PIK3CA somatic mutations have been recorded as the top-two recurrent oncogenes in breast cancers [111,122,123,124,125,126], with TP53 dominant in mutations (69%), followed by PIK3CA somatic GOF mutations (29%), indicating their oncogenic driver roles in breast cancer tumorigenesis [127,128].
The oncogenic effect of enhanced PI3K signaling in the reproductive system, especially in breast cancers, has been extensively studied [91,129,130,131]. Cooperation between somatic GOF PIK3CA and LOF p53 mutations have been well documented in breast cancer [19,125,132,133,134,135,136,137] and reproductive cancers in general [19,82,125,133,134,135,136,137,138,139,140,141,142].
GOF PIK3CA mutations induce the passenger mutations of members of CmPn signaling in reproductive cancers. The treatment of PIK3CA-related breast cancers has been a major focus in breast cancer research, and the combination of PI3K/AKT inhibitors with rapamycin has been explored as a potential treatment option [63,114,143,144,145,146]. Additionally, this approach has also been applied to the treatment of patients with vascular anomalies and overgrowth syndromes caused by GOF PIK3CA mutations, indicating a genetic similarity between these two conditions [16,147,148,149].
It has been defined that the three CCM proteins form the CCM signaling complex (CSC) that couples classic and non-classic progesterone (PRG) receptors (nPRs/mPRs) to form the CSC-mPR-PRG-nPR (CmPn) signaling network, which has a major impact on angiogenesis and tumorigenesis [150,151,152,153,154,155,156,157]. Key members of the CmPn network have been defined as important biomarkers for certain oncogenic conditions [150,151,152,155,156,158,159,160,161,162]. Since most common GOF PIK3CA mutations (hotspots) in breast cancers are somatic point mutations, by utilizing the GDC data portal from the National Cancer Institute (NCI), simple somatic mutations (SSM) of breast cancer lesions for TP53 and PIK3CA genes, along with all key CmPn signaling components, were assessed with Oncogrid. The distribution of SSM for breast cancer lesions clearly demonstrates that both TP53 and PIK3CA genes are indeed breast cancer drivers, while all SSM for CCM genes and other members of the CmPn network are passenger mutations (Figure 1). This finding was further validated by the similar distribution of SSM of reproductive cancer lesions for TP53 and PIK3CA genes, and key CmPn signaling components with Oncogrid (Figure 2), suggesting the oncogenic role of GOF PIK3CA mutations (hotspots) to generate randomly appearing passenger gene mutations of key CmPn signaling components. Therefore, as previously indicated, the secondary passenger mutations of CCM genes may have consequential events in sporadic CCMs [5].
Confirmation of oncogenic driver role of GOF PIK3CA mutations in mouse cancer models and their association with breast cancer subtypes and vascular phenotypes. Combining with other oncogenes, activating PIK3CA mutations in transgenic mice were utilized to create various types of cancer mouse models (such as colon, prostate, lung, and brain cancers, etc.) [163,164,165,166,167,168,169,170], in either a non-inducible [96,125,171] or conditional and inducible system [79,95,132,163,167]. Activating GOF PIK3CA mutations have been shown to predominantly predispose mouse reproductive organs/tissues to tumors [77,79,95,96,124,125,132,171,172,173,174]. Actually, the majority of mouse models with activating GOF PIK3CA mutations have been used to investigate various subtypes of breast cancer and their therapeutic strategies [77,78,79,96,124,125,132,164,171,172,173,175,176,177,178]. Interestingly, although like other tumor mouse models, breast cancer mouse models with enhanced PI3K activity targeted in epithelial cells, still exhibit vascular phenotypes such that both lymphatic (LMs) and venous malformations (VeMs) and have been reported in epithelial cancers associated with GOF PIK3CA mutations [72,101], suggesting a shared genetic mechanism between breast cancers and vascular anomalies.

4. Somatic GOF PIK3CA Mutations Are Genetic Drivers for Vascular Anomalies

Vascular anomalies are common phenotypes in the PIK3CA-Related Overgrowth Spectrum (PROS). PIK3CA mutations are widely recognized as a common human oncogene and have been detected in many types of cancer [9,12,60]. GOF point mutations in the PIK3CA gene, such as E542K, E545K, and H1047R, result in the activation of the PI3K pathway and are considered to be oncogenic [76]. However, PIK3CA mutations also cause noncancerous conditions known as PIK3CA-Related Overgrowth Spectrum (PROS) disorders, which are characterized by hyper-proliferation in various tissues and cell types [19,179,180]. It is noteworthy that vascular anomalies are a prevalent phenotype in PROS [98,101,181,182], emphasizing the critical involvement of PIK3CA mutations in their development [98,183,184,185]. The presence of these mutations has been confirmed in various forms of vascular anomalies [98,183,184,185], indicating their contribution to the maintenance of microvasculature.
Somatic GOF PIK3CA mutations is a driver of various types of vascular malformations (VMs). Vascular malformations (VMs) are a group of defects in the formation of blood vessels and are classified into lymphatic, capillary, venous, and arteriovenous malformations [186,187,188,189]. These can further be divided into simple, complex, and syndromic VMs based on their vessel composition [188,190,191]. PIK3CA, a gene affecting cellular growth, is crucial for proper vascular development [97]. Mutations in PIK3CA result in hyper-proliferative blood vessels with reduced pericyte coverage and decreased expression of arteriovenous markers, leading to the development of PIK3CA-Related Overgrowth Spectrum (PROS) disorders [180,182,192,193,194], with VMs being a common phenotype [195,196,197]. Hyper-proliferative clonal populations with enhanced PI3K/AKT signaling have been isolated from patients with simple and complex VMs and are caused by mutations in PIK3CA [16,20,101,102,198,199,200,201]. Complex and syndromic VMs often result from mosaicism in early developmental stages and can vary in manifestation depending on the timing of the mutation and the cell lineage affected [190,202]. Despite these differences, all GOF PIK3CA-associated VMs share the same pathogenesis as slow-flow VMs (SFVMs), which are frequently caused by GOF PIK3CA mutations [18,190,203]. This has been supported by recent sequencing data in patients with slow-flow VMs [18].
VeM pathology can be realized in mouse models carrying GOF PIK3CA mutations. Creating mouse models with activated PIK3CA mutations for vascular phenotypes has been challenging. Early attempts using the constitutively active form of membrane-bound PIK3CA resulted in embryonic lethality, indicating that too much activation of PI3K signaling can cause lethal vascular malformation (VM) phenotypes [96]. This embryonic lethality occurred due to defective vasculogenesis and angiogenesis when activated early in embryogenesis, suggesting that over-activation of PI3K signaling too early leads to lethal VM phenotypes [124,195,202,203,204,205,206]. To overcome this, researchers have used special approaches to generate PIK3CA-related vascular phenotypes in mouse models. Using patient-derived xenograft (PDX) mouse models has allowed for the investigation of the pathogenesis of venous malformations (VeMs), as activating PIK3CA mutations in endothelial cells have been identified as the main drivers of VeMs [202,203,205,206]. Several VeMs PDX mouse models have been created by injecting human VeM-derived endothelial cells into athymic nude mice. Within 7–9 days, ectatic vascular channels can be seen in the VeM-PDX mouse models. The PDX mouse models have demonstrated that human VeM-ECs with different activating PIK3CA mutations behave like tumor cells and invade mouse tissue to form lesions with human VeM-ECs [20,98,205,207], similar to VeM phenotypes in humans [20,98,205,207].

5. Vascular Phenotypes Shared by VeMs in Cancer Mouse Models

Timing for Expression of GOF PIK3CA point mutations is a challenge. Mouse models with GOF PIK3CA point mutations have frequently been used in tumorigenesis studies, using various transgenic techniques [174]. These models have primarily focused on targeting epithelial cells, with both overexpression of the activating mutation H1047R [79] and knock-in of mutations E545K or H1047R [163] resulting in vascular phenotypes. However, properly timing the expression of these mutations in VeM mouse models remains challenging. Mouse models of heterozygous GOF PIK3CA point mutation H1047R in endothelial cells resulted in embryonic death with extraembryonic defects [195]. This phenotype is common in VeM/LM mouse models with activating PIK3CA mutations, and likely explains the absence of inherited germline PIK3CA mutations in humans. In order to overcome this issue, inducible mouse models with active PIK3CA mutations were created, which resulted in the mice dying from multiple uncontrolled growth abnormalities and visible subcutaneous vascular abnormalities [99,205,207,208], resembling typical VeM symptoms in humans [209].
VeM-associated metastatic tumor cells or VeM-Endothelial cells (VeM-ECs) carrying activating PIK3CA mutations are invasive. Activating mutations in key factors of the PI3K/AKT/mTOR pathway, such as PIK3CA, AKT, or MAP3K3, lead to the activation of PI3K signaling and uncontrolled cell proliferation in many human cancers [76]. In PROS, the timing and nature of postzygotic activation of these factors determine the distribution of highly proliferative cells in the individual, resulting in overgrowth, specific tumors, and subcutaneous vascular abnormalities [194,210]. Cancer is a common abnormality in PROS, and it is possible that metastatic forms of tumor cells with activating mutations in PIK3CA or MAP3K3 can invade tissue/vasculature [211], circulate in the blood, and reside in microvascular lesions, including CCMs [5]. Activating mutations in key factors of the PI3K/AKT/mTOR pathway, such as TIE2, PIK3CA, AKT, or MAP3K3, lead to the activation of PI3K signaling and uncontrolled cell proliferation in many human cancers [76]. In PROS, the timing and nature of postzygotic activation of these factors determine the distribution of highly proliferative cells in the individual, resulting in overgrowth, specific tumors, and subcutaneous vascular abnormalities [194,210]. As noted earlier, cancer is a frequent occurrence in PROS, and it is plausible that tumor cells with activating mutations in PIK3CA or MAP3K3 may infiltrate tissues and blood vessels in metastatic forms, leading to the formation of microvascular lesions such as CCMs [211].

6. Sporadic CCMs Caused by Somatic Passenger Mutations of CCM Genes

DVAs are a causative factor for sporadic cases of CCMs. Developmental venous anomalies (DVAs) are a type of venous malformation (VeMs) located in the brain. It is believed that both DVAs and VeMs have a similar genetic cause [98,99,101,212,213]. The coexistence of CCMs and DVAs has long been recognized and is considered a standard criteria in identifying sCCM cases [214,215,216,217,218]. Studies have found a significant link between typical DVAs and sCCMs but not with fCCMs [217,219,220]. This suggests that DVAs may play a role in the development of sCCMs. Recently, a case report highlighted the presence of long-term distant recurrence of hemorrhagic sCCM associated with a DVA, suggesting that focal inflammatory and stressful conditions generated by DVAs could contribute to CCM lesion development or clinical manifestation [221]. It has been noted that although sporadic CCMs are not hereditary, they may present multiple lesions, similar to familial CCMs, leading to misdiagnosis [27]. However, the distribution of these lesions in sporadic cases is different from that in familial cases. While familial CCMs are evenly distributed throughout the brain, sporadic CCMs are often clustered in the area drained by the collecting vein associated with DVAs. This suggests that sporadic CCMs can be considered as VeM-derived CCMs [5].
Sequential events of VeMs-derived CCM lesions in sporadic CCM cases. Like tumorigenic events observed in breast cancer, somatic point mutations in angiogenic factors such as TIE2, AKT1, MAP3K3, and PIK3CA, involved in PI3K signaling, have been identified as a cause of enhanced RTK/PI3K/AKT/mTOR signaling in ECs and play a key role in the pathogenesis of VeMs. Although genetic screening for the three CCM (1-3) genes has not found any genomic mutations in sporadic CCM cases associated with VeMs [27,222], somatic mutations in the CCM (1-3) genes have been found in CCM lesions along with GOF mutations of the PIK3CA gene [1,2,3]. This suggests that VeM-ECs with enhanced PI3K/AKT/mTOR signaling, similar to their breast cancer counterparts, show high proliferative potential, decreased genomic stability, and increased invasiveness. The VeM-derived CCM model remains the most likely explanation for sCCM and DVAs [5], which are linked by a common genetic mechanism and are characterized by bi-allelic mutations in DNA mismatch repair genes in patients with constitutional mismatch repair deficiency syndrome, even though both conditions have their own independent regulatory signaling cascades [223]. The presence of DVA-led CCM lesions around DVAs supports this hypothesis and suggests that GOF mutations of the PIK3CA gene drive the subsequent appearance of passenger mutations, including CCM genes, in ECs; which can be mirrored in breast and reproductive cancers (Figure 1 and Figure 2). A common genetic paradigm linking reproductive cancers and PROS associated vascular anomalies with the same GOF PIK3CA gene mutation should be established [224], while mutated CCM genes result in clinically observed sCCM cases.

7. Materials and Methods

In our meta-analysis, we used Oncogrid to visualize the genomic landscape of breast and reproductive cancer patients from the Cancer Genome Atlas (TCGA) PanCancer Atlas. The Oncogrid analysis integrated genomic variants and tumor samples, available through the TCGA portal web application [225,226]. For breast cancer, Oncogrid was generated for the top two somatically mutated genes (TP53 and PIK3CA), as well as another frequently mutated gene in the PI3K signaling pathway (AKT1), using 200 tumors from a multi-centric cohort. The same process was repeated for reproductive cancer, with Oncogrid generated from 200 tumors and the same three oncogenes. In this paper, our focus was on gain of function (GOF) point mutations in the PIK3CA gene, so we only retained simple somatic mutations (SSM) for Oncogrid by excluding copy number variations and alteration-based survival data. The genes were ordered based on their alteration frequency in each tumor, and SSMs were represented as green-colored dots.

8. Conclusions

In this meta-analysis, we aimed to examine the coexistence of gain-of-function (GOF) mutations in the PIK3CA gene and loss-of-function (LOF) mutations in CCM genes in sCCM lesions. We utilized genomic data from the Cancer Genome Atlas (TCGA) for breast and reproductive cancers to inform our analysis. Our goal was to understand the relationship between mutational events and CCM lesions over time. We will perform a comparative meta-analysis for GOF PIK3CA point mutations in an attempt to demonstrate the genetic similarities shared by both cancers and vascular anomalies. Given the well-known role of GOF PIK3CA mutations as a driver oncogene in reproductive cancers, particularly breast cancer, we conducted a comparative meta-analysis to highlight any genetic similarities shared between these cancers and vascular anomalies.

Author Contributions

J.Z.: Conceptualization, methodology, writing—original draft preparation, writing—reviewing and editing; J.C.: Writing—reviewing and editing; A.L.: Data analysis, illustrating. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Patient consent was waived due to the absence of patient records being reviewed.

Data Availability Statement

Data were obtained from the Cancer Genome Atlas (TCGA) and are available from the corresponding author if necessary.

Acknowledgments

We wish to thank Johnathan Abou-Fadel at Texas Tech University Health Science Center El Paso (TTUHSCEP) for providing his technical help during the experiments.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Cerebral cavernous malformations (CCMs); familial case of CCM (fCCM); sporadic case of CCM (sCCM); CCM signaling complex (CSC); endothelial cells (ECs); single nucleotide polymorphism (SNP); gain-of-function (GOF); loss of function (LOF); phosphoinositide 3-kinase (PI3K); phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit p110α (PIK3CA); AKT (Protein Kinase-B, PKB); mTOR (mechanistic target of rapamycin); Receptor tyrosine kinases (RTKs); PIK3CA-related overgrowth spectrum (PROS); vascular malformations (VM); lymphatic malformations (LMs); venous malformations (VeMs); developmental venous anomalies (DVAs); progesterone (PRG); classic nuclear progesterone receptors (nPRs); non-classic membrane progesterone receptors (mPRs); CSC-mPR-PRG-nPR signaling network (CmPn).

References

  1. Weng, J.; Yang, Y.; Song, D.; Huo, R.; Li, H.; Chen, Y.; Nam, Y.; Zhou, Q.; Jiao, Y.; Fu, W.; et al. Somatic MAP3K3 mutation defines a subclass of cerebral cavernous malformation. Am. J. Hum. Genet. 2021, 108, 942–950. [Google Scholar] [CrossRef]
  2. Ren, A.A.; Snellings, D.A.; Su, Y.R.S.; Hong, C.C.; Castro, M.; Tang, A.T.; Detter, M.R.; Hobson, N.; Girard, R.; Romanos, S.; et al. PIK3CA and CCM mutations fuel cavernomas through a cancer-like mechanism. Nature 2021, 594, 271–276. [Google Scholar] [CrossRef] [PubMed]
  3. Hong, T.; Xiao, X.; Ren, J.; Cui, B.; Zong, Y.; Zou, J.; Kou, Z.; Jiang, N.; Meng, G.; Zeng, G.; et al. Somatic MAP3K3 and PIK3CA mutations in sporadic cerebral and spinal cord cavernous malformations. Brain 2021, 144, 2648–2658. [Google Scholar] [CrossRef] [PubMed]
  4. Peyre, M.; Miyagishima, D.; Bielle, F.; Chapon, F.; Sierant, M.; Venot, Q.; Lerond, J.; Marijon, P.; Abi-Jaoude, S.; Le Van, T.; et al. Somatic PIK3CA Mutations in Sporadic Cerebral Cavernous Malformations. N. Engl. J. Med. 2021, 385, 996–1004. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, J.; Abou-Fadel, J.; Renteria, M.; Belkin, O.; Chen, B.; Zhu, Y.; Dammann, P.; Rigamonti, D. Cerebral cavernous malformations do not fall in the spectrum of PIK3CA-related overgrowth. J. Neurol. Neurosurg. Psychiatry 2022, 93, 808–815. [Google Scholar] [CrossRef]
  6. Klippel, A.; Reinhard, C.; Kavanaugh, W.M.; Apell, G.; Escobedo, M.A.; Williams, L.T. Membrane localization of phosphatidylinositol 3-kinase is sufficient to activate multiple signal-transducing kinase pathways. Mol. Cell Biol. 1996, 16, 4117–4127. [Google Scholar] [CrossRef]
  7. Zhao, J.J.; Gjoerup, O.V.; Subramanian, R.R.; Cheng, Y.; Chen, W.; Roberts, T.M.; Hahn, W.C. Human mammary epithelial cell transformation through the activation of phosphatidylinositol 3-kinase. Cancer Cell 2003, 3, 483–495. [Google Scholar] [CrossRef]
  8. Kang, S.; Bader, A.G.; Vogt, P.K. Phosphatidylinositol 3-kinase mutations identified in human cancer are oncogenic. Proc. Natl. Acad. Sci. USA 2005, 102, 802–807. [Google Scholar] [CrossRef]
  9. Samuels, Y.; Ericson, K. Oncogenic PI3K and its role in cancer. Curr. Opin. Oncol. 2006, 18, 77–82. [Google Scholar] [CrossRef]
  10. Zhao, J.J.; Liu, Z.; Wang, L.; Shin, E.; Loda, M.F.; Roberts, T.M. The oncogenic properties of mutant p110alpha and p110beta phosphatidylinositol 3-kinases in human mammary epithelial cells. Proc. Natl. Acad. Sci. USA 2005, 102, 18443–18448. [Google Scholar] [CrossRef]
  11. Zhang, H.; Liu, G.; Dziubinski, M.; Yang, Z.; Ethier, S.P.; Wu, G. Comprehensive analysis of oncogenic effects of PIK3CA mutations in human mammary epithelial cells. Breast Cancer Res. Treat. 2008, 112, 217–227. [Google Scholar] [CrossRef] [PubMed]
  12. Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S.M.; Riggins, G.J.; et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004, 304, 554. [Google Scholar] [CrossRef] [PubMed]
  13. Oda, K.; Okada, J.; Timmerman, L.; Rodriguez-Viciana, P.; Stokoe, D.; Shoji, K.; Taketani, Y.; Kuramoto, H.; Knight, Z.A.; Shokat, K.M.; et al. PIK3CA cooperates with other phosphatidylinositol 3’-kinase pathway mutations to effect oncogenic transformation. Cancer Res. 2008, 68, 8127–8136. [Google Scholar] [CrossRef]
  14. Isakoff, S.J.; Engelman, J.A.; Irie, H.Y.; Luo, J.; Brachmann, S.M.; Pearline, R.V.; Cantley, L.C.; Brugge, J.S. Breast cancer-associated PIK3CA mutations are oncogenic in mammary epithelial cells. Cancer Res. 2005, 65, 10992–11000. [Google Scholar] [CrossRef]
  15. Ikenoue, T.; Kanai, F.; Hikiba, Y.; Obata, T.; Tanaka, Y.; Imamura, J.; Ohta, M.; Jazag, A.; Guleng, B.; Tateishi, K.; et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res. 2005, 65, 4562–4567. [Google Scholar] [CrossRef] [PubMed]
  16. Le Cras, T.D.; Boscolo, E. Cellular and molecular mechanisms of PIK3CA-related vascular anomalies. Vasc. Biol. 2019, 1, H33–H40. [Google Scholar] [CrossRef]
  17. Limaye, N.; Kangas, J.; Mendola, A.; Godfraind, C.; Schlogel, M.J.; Helaers, R.; Eklund, L.; Boon, L.M.; Vikkula, M. Somatic Activating PIK3CA Mutations Cause Venous Malformation. Am. J. Hum. Genet. 2015, 97, 914–921. [Google Scholar] [CrossRef]
  18. Nozawa, A.; Fujino, A.; Yuzuriha, S.; Suenobu, S.; Kato, A.; Shimizu, F.; Aramaki-Hattori, N.; Kuniyeda, K.; Sakaguchi, K.; Ohnishi, H.; et al. Comprehensive targeted next-generation sequencing in patients with slow-flow vascular malformations. J. Hum. Genet. 2022, 67, 721–728. [Google Scholar] [CrossRef]
  19. Marechal, E.; Poliard, A.; Henry, K.; Moreno, M.; Legrix, M.; Macagno, N.; Mondielli, G.; Fauquier, T.; Barlier, A.; Etchevers, H.C. Multiple congenital malformations arise from somatic mosaicism for constitutively active Pik3ca signaling. Front. Cell Dev. Biol. 2022, 10, 1013001. [Google Scholar] [CrossRef]
  20. Le Cras, T.D.; Goines, J.; Lakes, N.; Pastura, P.; Hammill, A.M.; Adams, D.M.; Boscolo, E. Constitutively active PIK3CA mutations are expressed by lymphatic and vascular endothelial cells in capillary lymphatic venous malformation. Angiogenesis 2020, 23, 425–442. [Google Scholar] [CrossRef]
  21. Aoki, R.; Srivatanakul, K. Developmental Venous Anomaly: Benign or Not Benign. Neurol. Med. Chir. 2016, 56, 534–543. [Google Scholar] [CrossRef]
  22. Brzegowy, K.; Kowalska, N.; Solewski, B.; Musial, A.; Kasprzycki, T.; Herman-Sucharska, I.; Walocha, J.A. Prevalence and anatomical characteristics of developmental venous anomalies: An MRI study. Neuroradiology 2021, 63, 1001–1008. [Google Scholar] [CrossRef]
  23. Padarti, A.; Zhang, J. Recent advances in cerebral cavernous malformation research. Vessel. Plus. 2018, 2, 21. [Google Scholar] [CrossRef]
  24. Zafar, A.; Quadri, S.A.; Farooqui, M.; Ikram, A.; Robinson, M.; Hart, B.L.; Mabray, M.C.; Vigil, C.; Tang, A.T.; Kahn, M.L.; et al. Familial Cerebral Cavernous Malformations. Stroke 2019, 50, 1294–1301. [Google Scholar] [CrossRef]
  25. Petersen, T.A.; Morrison, L.A.; Schrader, R.M.; Hart, B.L. Familial versus sporadic cavernous malformations: Differences in developmental venous anomaly association and lesion phenotype. AJNR Am. J. Neuroradiol. 2010, 31, 377–382. [Google Scholar] [CrossRef] [PubMed]
  26. Kumar, S.; Lanzino, G.; Brinjikji, W.; Hocquard, K.W.; Flemming, K.D. Infratentorial Developmental Venous Abnormalities and Inflammation Increase Odds of Sporadic Cavernous Malformation. J. Stroke Cereb. Dis. 2019, 28, 1662–1667. [Google Scholar] [CrossRef]
  27. Dammann, P.; Wrede, K.; Zhu, Y.; Matsushige, T.; Maderwald, S.; Umutlu, L.; Quick, H.H.; Hehr, U.; Rath, M.; Ladd, M.E.; et al. Correlation of the venous angioarchitecture of multiple cerebral cavernous malformations with familial or sporadic disease: A susceptibility-weighted imaging study with 7-Tesla MRI. J. Neurosurg. 2017, 126, 570–577. [Google Scholar] [CrossRef] [PubMed]
  28. Maish, W.N. Developmental venous anomalies and brainstem cavernous malformations: A proposed physiological mechanism for haemorrhage. Neurosurg. Rev. 2019, 42, 663–670. [Google Scholar] [CrossRef]
  29. Vijg, J. Somatic mutations, genome mosaicism, cancer and aging. Curr. Opin. Genet. Dev. 2014, 26, 141–149. [Google Scholar] [CrossRef]
  30. Garcia-Nieto, P.E.; Morrison, A.J.; Fraser, H.B. The somatic mutation landscape of the human body. Genome Biol. 2019, 20, 298. [Google Scholar] [CrossRef]
  31. Martincorena, I. Somatic mutation and clonal expansions in human tissues. Genome Med. 2019, 11, 35. [Google Scholar] [CrossRef]
  32. Zhang, J.; Zhang, S. Discovery of cancer common and specific driver gene sets. Nucleic Acids Res. 2017, 45, e86. [Google Scholar] [CrossRef] [PubMed]
  33. Jolly, C.; Van Loo, P. Timing somatic events in the evolution of cancer. Genome Biol. 2018, 19, 95. [Google Scholar] [CrossRef] [PubMed]
  34. Kandarakov, O.; Belyavsky, A. Clonal Hematopoiesis, Cardiovascular Diseases and Hematopoietic Stem Cells. Int. J. Mol. Sci. 2020, 21, 7902. [Google Scholar] [CrossRef]
  35. Kakiuchi, N.; Ogawa, S. Clonal expansion in non-cancer tissues. Nat. Rev. Cancer 2021, 21, 239–256. [Google Scholar] [CrossRef] [PubMed]
  36. Forbes, S.A.; Beare, D.; Bindal, N.; Bamford, S.; Ward, S.; Cole, C.G.; Jia, M.; Kok, C.; Boutselakis, H.; De, T.; et al. COSMIC: High-Resolution Cancer Genetics Using the Catalogue of Somatic Mutations in Cancer. Curr. Protoc. Hum. Genet. 2016, 91, 10.11.11–10.11.37. [Google Scholar] [CrossRef]
  37. Forbes, S.A.; Beare, D.; Boutselakis, H.; Bamford, S.; Bindal, N.; Tate, J.; Cole, C.G.; Ward, S.; Dawson, E.; Ponting, L.; et al. COSMIC: Somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017, 45, D777–D783. [Google Scholar] [CrossRef]
  38. Tate, J.G.; Bamford, S.; Jubb, H.C.; Sondka, Z.; Beare, D.M.; Bindal, N.; Boutselakis, H.; Cole, C.G.; Creatore, C.; Dawson, E.; et al. COSMIC: The Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019, 47, D941–D947. [Google Scholar] [CrossRef]
  39. Greenman, C.; Stephens, P.; Smith, R.; Dalgliesh, G.L.; Hunter, C.; Bignell, G.; Davies, H.; Teague, J.; Butler, A.; Stevens, C.; et al. Patterns of somatic mutation in human cancer genomes. Nature 2007, 446, 153–158. [Google Scholar] [CrossRef]
  40. Shiovitz, S.; Korde, L.A. Genetics of breast cancer: A topic in evolution. Ann. Oncol. 2015, 26, 1291–1299. [Google Scholar] [CrossRef]
  41. Rousset-Jablonski, C.; Gompel, A. Screening for familial cancer risk: Focus on breast cancer. Maturitas 2017, 105, 69–77. [Google Scholar] [CrossRef] [PubMed]
  42. Vural, S.; Wang, X.; Guda, C. Classification of breast cancer patients using somatic mutation profiles and machine learning approaches. BMC Syst. Biol. 2016, 10 (Suppl. 3), 62. [Google Scholar] [CrossRef]
  43. Pereira, B.; Chin, S.F.; Rueda, O.M.; Vollan, H.K.; Provenzano, E.; Bardwell, H.A.; Pugh, M.; Jones, L.; Russell, R.; Sammut, S.J.; et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat. Commun. 2016, 7, 11479. [Google Scholar] [CrossRef] [PubMed]
  44. Perduca, V.; Alexandrov, L.B.; Kelly-Irving, M.; Delpierre, C.; Omichessan, H.; Little, M.P.; Vineis, P.; Severi, G. Stem cell replication, somatic mutations and role of randomness in the development of cancer. Eur. J. Epidemiol. 2019, 34, 439–445. [Google Scholar] [CrossRef]
  45. Gatenby, R.A.; Brown, J. Mutations, evolution and the central role of a self-defined fitness function in the initiation and progression of cancer. Biochim. Biophys Acta Rev. Cancer 2017, 1867, 162–166. [Google Scholar] [CrossRef] [PubMed]
  46. Przytycki, P.F.; Singh, M. Differential analysis between somatic mutation and germline variation profiles reveals cancer-related genes. Genome Med. 2017, 9, 79. [Google Scholar] [CrossRef]
  47. Bokhari, Y.; Alhareeri, A.; Arodz, T. QuaDMutNetEx: A method for detecting cancer driver genes with low mutation frequency. BMC Bioinform. 2020, 21, 122. [Google Scholar] [CrossRef]
  48. Martinez-Jimenez, F.; Muinos, F.; Sentis, I.; Deu-Pons, J.; Reyes-Salazar, I.; Arnedo-Pac, C.; Mularoni, L.; Pich, O.; Bonet, J.; Kranas, H.; et al. A compendium of mutational cancer driver genes. Nat. Rev. Cancer 2020, 20, 555–572. [Google Scholar] [CrossRef]
  49. Bostrom, M.; Larsson, E. Somatic mutation distribution across tumour cohorts provides a signal for positive selection in cancer. Nat. Commun. 2022, 13, 7023. [Google Scholar] [CrossRef]
  50. Yang, H.; Wei, Q.; Zhong, X.; Yang, H.; Li, B. Cancer driver gene discovery through an integrative genomics approach in a non-parametric Bayesian framework. Bioinformatics 2017, 33, 483–490. [Google Scholar] [CrossRef]
  51. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef] [PubMed]
  52. Tamborero, D.; Gonzalez-Perez, A.; Perez-Llamas, C.; Deu-Pons, J.; Kandoth, C.; Reimand, J.; Lawrence, M.S.; Getz, G.; Bader, G.D.; Ding, L.; et al. Comprehensive identification of mutational cancer driver genes across 12 tumor types. Sci. Rep. 2013, 3, 2650. [Google Scholar] [CrossRef] [PubMed]
  53. Gough, N.R. Focus issue: From genomic mutations to oncogenic pathways. Sci. Signal 2013, 6, eg3. [Google Scholar] [CrossRef]
  54. De, S.; Ganesan, S. Looking beyond drivers and passengers in cancer genome sequencing data. Ann. Oncol. 2017, 28, 938–945. [Google Scholar] [CrossRef] [PubMed]
  55. Lien, E.C.; Dibble, C.C.; Toker, A. PI3K signaling in cancer: Beyond AKT. Curr. Opin. Cell Biol. 2017, 45, 62–71. [Google Scholar] [CrossRef]
  56. Martini, M.; Ciraolo, E.; Gulluni, F.; Hirsch, E. Targeting PI3K in Cancer: Any Good News? Front Oncol. 2013, 3, 108. [Google Scholar] [CrossRef]
  57. Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef]
  58. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef]
  59. Wu, P.; Liu, T.; Hu, Y. PI3K inhibitors for cancer therapy: What has been achieved so far? Curr. Med. Chem. 2009, 16, 916–930. [Google Scholar] [CrossRef]
  60. Lu, Y.; Wang, H.; Mills, G.B. Targeting PI3K-AKT pathway for cancer therapy. Rev. Clin. Exp. Hematol. 2003, 7, 205–228. [Google Scholar]
  61. Courtney, K.D.; Corcoran, R.B.; Engelman, J.A. The PI3K pathway as drug target in human cancer. J. Clin. Oncol. 2010, 28, 1075–1083. [Google Scholar] [CrossRef] [PubMed]
  62. Tanaka, H.; Yoshida, M.; Tanimura, H.; Fujii, T.; Sakata, K.; Tachibana, Y.; Ohwada, J.; Ebiike, H.; Kuramoto, S.; Morita, K.; et al. The selective class I PI3K inhibitor CH5132799 targets human cancers harboring oncogenic PIK3CA mutations. Clin. Cancer Res. 2011, 17, 3272–3281. [Google Scholar] [CrossRef] [PubMed]
  63. Costa, R.L.B.; Han, H.S.; Gradishar, W.J. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: A review. Breast Cancer Res. Treat. 2018, 169, 397–406. [Google Scholar] [CrossRef]
  64. Wolin, E.M. PI3K/Akt/mTOR pathway inhibitors in the therapy of pancreatic neuroendocrine tumors. Cancer Lett. 2013, 335, 1–8. [Google Scholar] [CrossRef] [PubMed]
  65. Rafii, S.; Roda, D.; Geuna, E.; Jimenez, B.; Rihawi, K.; Capelan, M.; Yap, T.A.; Molife, L.R.; Kaye, S.B.; de Bono, J.S.; et al. Higher Risk of Infections with PI3K-AKT-mTOR Pathway Inhibitors in Patients with Advanced Solid Tumors on Phase I Clinical Trials. Clin. Cancer Res. 2015, 21, 1869–1876. [Google Scholar] [CrossRef] [PubMed]
  66. Yuan, Y.; Long, H.; Zhou, Z.; Fu, Y.; Jiang, B. PI3K-AKT-Targeting Breast Cancer Treatments: Natural Products and Synthetic Compounds. Biomolecules 2023, 13, 93. [Google Scholar] [CrossRef]
  67. Rodon, J.; Dienstmann, R.; Serra, V.; Tabernero, J. Development of PI3K inhibitors: Lessons learned from early clinical trials. Nat. Rev. Clin. Oncol. 2013, 10, 143–153. [Google Scholar] [CrossRef]
  68. LoRusso, P.M. Inhibition of the PI3K/AKT/mTOR Pathway in Solid Tumors. J. Clin. Oncol. 2016, 34, 3803–3815. [Google Scholar] [CrossRef]
  69. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug. Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef]
  70. Cho, D.C. Targeting the PI3K/Akt/mTOR pathway in malignancy: Rationale and clinical outlook. BioDrugs 2014, 28, 373–381. [Google Scholar] [CrossRef]
  71. Bauer, T.M.; Patel, M.R.; Infante, J.R. Targeting PI3 kinase in cancer. Pharmacol. Ther. 2015, 146, 53–60. [Google Scholar] [CrossRef] [PubMed]
  72. Fruman, D.A.; Chiu, H.; Hopkins, B.D.; Bagrodia, S.; Cantley, L.C.; Abraham, R.T. The PI3K Pathway in Human Disease. Cell 2017, 170, 605–635. [Google Scholar] [CrossRef] [PubMed]
  73. Ersahin, T.; Tuncbag, N.; Cetin-Atalay, R. The PI3K/AKT/mTOR interactive pathway. Mol. Biosyst. 2015, 11, 1946–1954. [Google Scholar] [CrossRef] [PubMed]
  74. Korkolopoulou, P.; Levidou, G.; Trigka, E.A.; Prekete, N.; Karlou, M.; Thymara, I.; Sakellariou, S.; Fragkou, P.; Isaiadis, D.; Pavlopoulos, P.; et al. A comprehensive immunohistochemical and molecular approach to the PI3K/AKT/mTOR (phosphoinositide 3-kinase/v-akt murine thymoma viral oncogene/mammalian target of rapamycin) pathway in bladder urothelial carcinoma. BJU Int. 2012, 110, E1237–E1248. [Google Scholar] [CrossRef] [PubMed]
  75. Samuels, Y.; Waldman, T. Oncogenic mutations of PIK3CA in human cancers. Curr. Top Microbiol. Immunol. 2010, 347, 21–41. [Google Scholar] [CrossRef]
  76. Lai, K.; Killingsworth, M.C.; Lee, C.S. Gene of the month: PIK3CA. J. Clin. Pathol. 2015, 68, 253–257. [Google Scholar] [CrossRef]
  77. Koren, S.; Reavie, L.; Couto, J.P.; De Silva, D.; Stadler, M.B.; Roloff, T.; Britschgi, A.; Eichlisberger, T.; Kohler, H.; Aina, O.; et al. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 2015, 525, 114–118. [Google Scholar] [CrossRef]
  78. Meyer, D.S.; Brinkhaus, H.; Muller, U.; Muller, M.; Cardiff, R.D.; Bentires-Alj, M. Luminal expression of PIK3CA mutant H1047R in the mammary gland induces heterogeneous tumors. Cancer Res. 2011, 71, 4344–4351. [Google Scholar] [CrossRef]
  79. Liu, P.; Cheng, H.; Santiago, S.; Raeder, M.; Zhang, F.; Isabella, A.; Yang, J.; Semaan, D.J.; Chen, C.; Fox, E.A.; et al. Oncogenic PIK3CA-driven mammary tumors frequently recur via PI3K pathway-dependent and PI3K pathway-independent mechanisms. Nat. Med. 2011, 17, 1116–1120. [Google Scholar] [CrossRef]
  80. Shayesteh, L.; Lu, Y.; Kuo, W.L.; Baldocchi, R.; Godfrey, T.; Collins, C.; Pinkel, D.; Powell, B.; Mills, G.B.; Gray, J.W. PIK3CA is implicated as an oncogene in ovarian cancer. Nat. Genet. 1999, 21, 99–102. [Google Scholar] [CrossRef]
  81. Karakas, B.; Bachman, K.E.; Park, B.H. Mutation of the PIK3CA oncogene in human cancers. Br. J. Cancer 2006, 94, 455–459. [Google Scholar] [CrossRef] [PubMed]
  82. Bienfait, L.; D’Haene, N.; Catteau, X.; Noel, J.C. PIK3CA and p53 Mutations by Next Generation Sequencing in Lymphoepithelioma-Like Carcinoma of the Endometrium. Case Rep. Pathol. 2018, 2018, 5894589. [Google Scholar] [CrossRef] [PubMed]
  83. Herberts, C.; Murtha, A.J.; Fu, S.; Wang, G.; Schonlau, E.; Xue, H.; Lin, D.; Gleave, A.; Yip, S.; Angeles, A.; et al. Activating AKT1 and PIK3CA Mutations in Metastatic Castration-Resistant Prostate Cancer. Eur. Urol. 2020, 78, 834–844. [Google Scholar] [CrossRef] [PubMed]
  84. Vivanco, I.; Sawyers, C.L. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat. Rev. Cancer 2002, 2, 489–501. [Google Scholar] [CrossRef]
  85. Bellacosa, A.; de Feo, D.; Godwin, A.K.; Bell, D.W.; Cheng, J.Q.; Altomare, D.A.; Wan, M.; Dubeau, L.; Scambia, G.; Masciullo, V.; et al. Molecular alterations of the AKT2 oncogene in ovarian and breast carcinomas. Int. J. Cancer 1995, 64, 280–285. [Google Scholar] [CrossRef]
  86. Knobbe, C.B.; Reifenberger, G. Genetic alterations and aberrant expression of genes related to the phosphatidyl-inositol-3’-kinase/protein kinase B (Akt) signal transduction pathway in glioblastomas. Brain Pathol. 2003, 13, 507–518. [Google Scholar] [CrossRef]
  87. Bastian, B.C.; LeBoit, P.E.; Hamm, H.; Brocker, E.B.; Pinkel, D. Chromosomal gains and losses in primary cutaneous melanomas detected by comparative genomic hybridization. Cancer Res. 1998, 58, 2170–2175. [Google Scholar]
  88. Actor, B.; Cobbers, J.M.; Buschges, R.; Wolter, M.; Knobbe, C.B.; Lichter, P.; Reifenberger, G.; Weber, R.G. Comprehensive analysis of genomic alterations in gliosarcoma and its two tissue components. Genes Chromosom. Cancer 2002, 34, 416–427. [Google Scholar] [CrossRef]
  89. Yuan, T.L.; Cantley, L.C. PI3K pathway alterations in cancer: Variations on a theme. Oncogene 2008, 27, 5497–5510. [Google Scholar] [CrossRef]
  90. Osborn, A.J.; Dickie, P.; Neilson, D.E.; Glaser, K.; Lynch, K.A.; Gupta, A.; Dickie, B.H. Activating PIK3CA alleles and lymphangiogenic phenotype of lymphatic endothelial cells isolated from lymphatic malformations. Hum. Mol. Genet. 2015, 24, 926–938. [Google Scholar] [CrossRef]
  91. Polo, M.L.; Riggio, M.; May, M.; Rodriguez, M.J.; Perrone, M.C.; Stallings-Mann, M.; Kaen, D.; Frost, M.; Goetz, M.; Boughey, J.; et al. Activation of PI3K/Akt/mTOR signaling in the tumor stroma drives endocrine therapy-dependent breast tumor regression. Oncotarget 2015, 6, 22081–22097. [Google Scholar] [CrossRef] [PubMed]
  92. El Ahanidi, H.; El Azzouzi, M.; Arrouchi, H.; Alaoui, C.H.; Tetou, M.; Bensaid, M.; Oukabli, M.; Ameur, A.; Al Bouzidi, A.; El Mzibri, M.; et al. AKT1 and PIK3CA activating mutations in Moroccan bladder cancer patients biopsies and matched urine. Pan. Afr. Med. J. 2022, 41, 59. [Google Scholar] [CrossRef] [PubMed]
  93. Ilic, N.; Roberts, T.M. Comparing the roles of the p110alpha and p110beta isoforms of PI3K in signaling and cancer. Curr. Top Microbiol. Immunol. 2010, 347, 55–77. [Google Scholar] [CrossRef] [PubMed]
  94. Vanhaesebroeck, B.; Guillermet-Guibert, J.; Graupera, M.; Bilanges, B. The emerging mechanisms of isoform-specific PI3K signalling. Nat. Rev. Mol. Cell Biol. 2010, 11, 329–341. [Google Scholar] [CrossRef] [PubMed]
  95. Kinross, K.M.; Montgomery, K.G.; Kleinschmidt, M.; Waring, P.; Ivetac, I.; Tikoo, A.; Saad, M.; Hare, L.; Roh, V.; Mantamadiotis, T.; et al. An activating Pik3ca mutation coupled with Pten loss is sufficient to initiate ovarian tumorigenesis in mice. J. Clin. Invest. 2012, 122, 553–557. [Google Scholar] [CrossRef]
  96. Renner, O.; Blanco-Aparicio, C.; Grassow, M.; Canamero, M.; Leal, J.F.; Carnero, A. Activation of phosphatidylinositol 3-kinase by membrane localization of p110alpha predisposes mammary glands to neoplastic transformation. Cancer Res. 2008, 68, 9643–9653. [Google Scholar] [CrossRef] [PubMed]
  97. Graupera, M.; Guillermet-Guibert, J.; Foukas, L.C.; Phng, L.K.; Cain, R.J.; Salpekar, A.; Pearce, W.; Meek, S.; Millan, J.; Cutillas, P.R.; et al. Angiogenesis selectively requires the p110alpha isoform of PI3K to control endothelial cell migration. Nature 2008, 453, 662–666. [Google Scholar] [CrossRef]
  98. Canaud, G.; Hammill, A.M.; Adams, D.; Vikkula, M.; Keppler-Noreuil, K.M. A review of mechanisms of disease across PIK3CA-related disorders with vascular manifestations. Orphanet J. Rare Dis. 2021, 16, 306. [Google Scholar] [CrossRef]
  99. Castillo, S.D.; Tzouanacou, E.; Zaw-Thin, M.; Berenjeno, I.M.; Parker, V.E.; Chivite, I.; Mila-Guasch, M.; Pearce, W.; Solomon, I.; Angulo-Urarte, A.; et al. Somatic activating mutations in Pik3ca cause sporadic venous malformations in mice and humans. Sci. Transl. Med. 2016, 8, 332ra343. [Google Scholar] [CrossRef]
  100. di Blasio, L.; Puliafito, A.; Gagliardi, P.A.; Comunanza, V.; Somale, D.; Chiaverina, G.; Bussolino, F.; Primo, L. PI3K/mTOR inhibition promotes the regression of experimental vascular malformations driven by PIK3CA-activating mutations. Cell Death Dis. 2018, 9, 45. [Google Scholar] [CrossRef]
  101. Castillo, S.D.; Baselga, E.; Graupera, M. PIK3CA mutations in vascular malformations. Curr. Opin. Hematol. 2019, 26, 170–178. [Google Scholar] [CrossRef] [PubMed]
  102. Fereydooni, A.; Nassiri, N. Evaluation and management of the lateral marginal vein in Klippel-Trenaunay and other PIK3CA-related overgrowth syndromes. J. Vasc. Surg. Venous Lymphat Disord. 2020, 8, 482–493. [Google Scholar] [CrossRef]
  103. Hoeger, P.H.; Martinez, A.; Maerker, J.; Harper, J.I. Vascular anomalies in Proteus syndrome. Clin. Exp. Derm. 2004, 29, 222–230. [Google Scholar] [CrossRef] [PubMed]
  104. Schwefel, K.; Spiegler, S.; Ameling, S.; Much, C.D.; Pilz, R.A.; Otto, O.; Volker, U.; Felbor, U.; Rath, M. Biallelic CCM3 mutations cause a clonogenic survival advantage and endothelial cell stiffening. J. Cell Mol. Med. 2019, 23, 1771–1783. [Google Scholar] [CrossRef] [PubMed]
  105. Meng, G.L.; Bai, C.F.; Yu, T.F.; Wu, Z.; Liu, X.; Zhang, J.T.; Zhao, J.Z. The association between cerebral developmental venous anomaly and concomitant cavernous malformation: An observational study using magnetic resonance imaging. BMC Neurol. 2014, 14, 1–6. [Google Scholar] [CrossRef] [PubMed]
  106. McDonald, D.A.; Shenkar, R.; Shi, C.B.; Stockton, R.A.; Akers, A.L.; Kucherlapati, M.H.; Kucherlapati, R.; Brainer, J.; Ginsberg, M.H.; Awad, I.A.; et al. A novel mouse model of cerebral cavernous malformations based on the two-hit mutation hypothesis recapitulates the human disease. Hum. Mol. Genet. 2011, 20, 211–222. [Google Scholar] [CrossRef]
  107. Docker, D.; Schubach, M.; Menzel, M.; Spaich, C.; Gabriel, H.D.; Zenker, M.; Bartholdi, D.; Biskup, S. Germline PTPN11 and somatic PIK3CA variant in a boy with megalencephaly-capillary malformation syndrome (MCAP)--pure coincidence? Eur. J. Hum. Genet. 2015, 23, 409–412. [Google Scholar] [CrossRef]
  108. Wood, L.D.; Parsons, D.W.; Jones, S.; Lin, J.; Sjoblom, T.; Leary, R.J.; Shen, D.; Boca, S.M.; Barber, T.; Ptak, J.; et al. The genomic landscapes of human breast and colorectal cancers. Science 2007, 318, 1108–1113. [Google Scholar] [CrossRef]
  109. Stephens, M.; Sloan, J.S.; Robertson, P.D.; Scheet, P.; Nickerson, D.A. Automating sequence-based detection and genotyping of SNPs from diploid samples. Nat. Genet. 2006, 38, 375–381. [Google Scholar] [CrossRef]
  110. Board, R.E.; Thelwell, N.J.; Ravetto, P.F.; Little, S.; Ranson, M.; Dive, C.; Hughes, A.; Whitcombe, D. Multiplexed assays for detection of mutations in PIK3CA. Clin. Chem. 2008, 54, 757–760. [Google Scholar] [CrossRef]
  111. Cancer Genome Atlas, N. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [PubMed]
  112. Ciriello, G.; Sinha, R.; Hoadley, K.A.; Jacobsen, A.S.; Reva, B.; Perou, C.M.; Sander, C.; Schultz, N. The molecular diversity of Luminal A breast tumors. Breast Cancer Res. Treat. 2013, 141, 409–420. [Google Scholar] [CrossRef] [PubMed]
  113. Stemke-Hale, K.; Gonzalez-Angulo, A.M.; Lluch, A.; Neve, R.M.; Kuo, W.L.; Davies, M.; Carey, M.; Hu, Z.; Guan, Y.; Sahin, A.; et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 2008, 68, 6084–6091. [Google Scholar] [CrossRef]
  114. Xing, Y.; Lin, N.U.; Maurer, M.A.; Chen, H.; Mahvash, A.; Sahin, A.; Akcakanat, A.; Li, Y.; Abramson, V.; Litton, J.; et al. Phase II trial of AKT inhibitor MK-2206 in patients with advanced breast cancer who have tumors with PIK3CA or AKT mutations, and/or PTEN loss/PTEN mutation. Breast Cancer Res. 2019, 21, 78. [Google Scholar] [CrossRef] [PubMed]
  115. Forde, K.; Resta, N.; Ranieri, C.; Rea, D.; Kubassova, O.; Hinton, M.; Andrews, K.A.; Semple, R.; Irvine, A.D.; Dvorakova, V. Clinical experience with the AKT1 inhibitor miransertib in two children with PIK3CA-related overgrowth syndrome. Orphanet. J. Rare Dis. 2021, 16, 109. [Google Scholar] [CrossRef] [PubMed]
  116. McFarland, C.D.; Yaglom, J.A.; Wojtkowiak, J.W.; Scott, J.G.; Morse, D.L.; Sherman, M.Y.; Mirny, L.A. The Damaging Effect of Passenger Mutations on Cancer Progression. Cancer Res. 2017, 77, 4763–4772. [Google Scholar] [CrossRef] [PubMed]
  117. Castro-Giner, F.; Ratcliffe, P.; Tomlinson, I. The mini-driver model of polygenic cancer evolution. Nat. Rev. Cancer 2015, 15, 680–685. [Google Scholar] [CrossRef] [PubMed]
  118. Burgess, D.J. Tumour evolution: Weighed down by passengers? Nat. Rev. Cancer 2013, 13, 219. [Google Scholar] [CrossRef]
  119. McFarland, C.D.; Mirny, L.A.; Korolev, K.S. Tug-of-war between driver and passenger mutations in cancer and other adaptive processes. Proc. Natl. Acad. Sci. USA 2014, 111, 15138–15143. [Google Scholar] [CrossRef]
  120. Wodarz, D.; Newell, A.C.; Komarova, N.L. Passenger mutations can accelerate tumour suppressor gene inactivation in cancer evolution. J. R. Soc. Interface 2018, 15. [Google Scholar] [CrossRef]
  121. Alexandrov, L.B.; Stratton, M.R. Mutational signatures: The patterns of somatic mutations hidden in cancer genomes. Curr. Opin. Genet. Dev. 2014, 24, 52–60. [Google Scholar] [CrossRef] [PubMed]
  122. Holmes, F.A.; Levin, M.K.; Cao, Y.; Balasubramanian, S.; Ross, J.S.; Krekow, L.; McIntyre, K.; Osborne, C.; Espina, V.; Liotta, L.; et al. Comutation of PIK3CA and TP53 in Residual Disease After Preoperative Anti-HER2 Therapy in ERBB2 (HER2)-Amplified Early Breast Cancer. Jco. Precis Oncol. 2019, 3. [Google Scholar] [CrossRef] [PubMed]
  123. You, C.; Sandalcioglu, I.E.; Dammann, P.; Felbor, U.; Sure, U.; Zhu, Y. Loss of CCM3 impairs DLL4-Notch signalling: Implication in endothelial angiogenesis and in inherited cerebral cavernous malformations. J. Cell Mol. Med. 2013, 17, 407–418. [Google Scholar] [CrossRef] [PubMed]
  124. Sheen, M.R.; Marotti, J.D.; Allegrezza, M.J.; Rutkowski, M.; Conejo-Garcia, J.R.; Fiering, S. Constitutively activated PI3K accelerates tumor initiation and modifies histopathology of breast cancer. Oncogenesis 2016, 5, e267. [Google Scholar] [CrossRef]
  125. Adams, J.R.; Xu, K.; Liu, J.C.; Agamez, N.M.; Loch, A.J.; Wong, R.G.; Wang, W.; Wright, K.L.; Lane, T.F.; Zacksenhaus, E.; et al. Cooperation between Pik3ca and p53 mutations in mouse mammary tumor formation. Cancer Res. 2011, 71, 2706–2717. [Google Scholar] [CrossRef]
  126. Boyault, S.; Drouet, Y.; Navarro, C.; Bachelot, T.; Lasset, C.; Treilleux, I.; Tabone, E.; Puisieux, A.; Wang, Q. Mutational characterization of individual breast tumors: TP53 and PI3K pathway genes are frequently and distinctively mutated in different subtypes. Breast Cancer Res. Treat. 2012, 132, 29–39. [Google Scholar] [CrossRef]
  127. Ng, C.K.Y.; Piscuoglio, S.; Geyer, F.C.; Burke, K.A.; Pareja, F.; Eberle, C.A.; Lim, R.S.; Natrajan, R.; Riaz, N.; Mariani, O.; et al. The Landscape of Somatic Genetic Alterations in Metaplastic Breast Carcinomas. Clin. Cancer Res. 2017, 23, 3859–3870. [Google Scholar] [CrossRef]
  128. Davis, N.M.; Sokolosky, M.; Stadelman, K.; Abrams, S.L.; Libra, M.; Candido, S.; Nicoletti, F.; Polesel, J.; Maestro, R.; D’Assoro, A.; et al. Deregulation of the EGFR/PI3K/PTEN/Akt/mTORC1 pathway in breast cancer: Possibilities for therapeutic intervention. Oncotarget 2014, 5, 4603–4650. [Google Scholar] [CrossRef]
  129. Jiang, Q.; Jin, M. Feature Selection for Breast Cancer Classification by Integrating Somatic Mutation and Gene Expression. Front Genet. 2021, 12, 629946. [Google Scholar] [CrossRef]
  130. Siegel, M.B.; He, X.; Hoadley, K.A.; Hoyle, A.; Pearce, J.B.; Garrett, A.L.; Kumar, S.; Moylan, V.J.; Brady, C.M.; Van Swearingen, A.E.; et al. Integrated RNA and DNA sequencing reveals early drivers of metastatic breast cancer. J. Clin. Invest. 2018, 128, 1371–1383. [Google Scholar] [CrossRef]
  131. Mosele, F.; Stefanovska, B.; Lusque, A.; Tran Dien, A.; Garberis, I.; Droin, N.; Le Tourneau, C.; Sablin, M.P.; Lacroix, L.; Enrico, D.; et al. Outcome and molecular landscape of patients with PIK3CA-mutated metastatic breast cancer. Ann. Oncol. 2020, 31, 377–386. [Google Scholar] [CrossRef] [PubMed]
  132. Yuan, W.; Stawiski, E.; Janakiraman, V.; Chan, E.; Durinck, S.; Edgar, K.A.; Kljavin, N.M.; Rivers, C.S.; Gnad, F.; Roose-Girma, M.; et al. Conditional activation of Pik3ca(H1047R) in a knock-in mouse model promotes mammary tumorigenesis and emergence of mutations. Oncogene 2013, 32, 318–326. [Google Scholar] [CrossRef] [PubMed]
  133. Yuan, W.; Goldstein, L.D.; Durinck, S.; Chen, Y.J.; Nguyen, T.T.; Kljavin, N.M.; Sokol, E.S.; Stawiski, E.W.; Haley, B.; Ziai, J.; et al. S100a4 upregulation in Pik3caH1047R;Trp53R270H;MMTV-Cre-driven mammary tumors promotes metastasis. Breast Cancer Res. 2019, 21, 152. [Google Scholar] [CrossRef] [PubMed]
  134. Yu, X.; Zhang, Y.; Xiong, S.; McDaniel, J.M.; Sun, C.; Chau, G.P.; Gencel-Augusto, J.; Chachad, D.; Morrissey, R.L.; Rao, X.; et al. Omics analyses of a somatic Trp53(R245W/+) breast cancer model identify cooperating driver events activating PI3K/AKT/mTOR signaling. Proc. Natl. Acad. Sci. USA 2022, 119, e2210618119. [Google Scholar] [CrossRef] [PubMed]
  135. Pang, B.; Sun, S.P.; Gao, L.; Zhu, R.L.; Zhang, L.X.; An, C.; Liu, Z.Y.; Liu, G.J. A single nucleotide polymorphism in PIK3CA gene is inversely associated with P53 protein expression in breast cancer. Med. Oncol. 2014, 31, 30. [Google Scholar] [CrossRef] [PubMed]
  136. Kim, J.S.; Lee, C.; Bonifant, C.L.; Ressom, H.; Waldman, T. Activation of p53-dependent growth suppression in human cells by mutations in PTEN or PIK3CA. Mol. Cell Biol. 2007, 27, 662–677. [Google Scholar] [CrossRef] [PubMed]
  137. Astanehe, A.; Arenillas, D.; Wasserman, W.W.; Leung, P.C.; Dunn, S.E.; Davies, B.R.; Mills, G.B.; Auersperg, N. Mechanisms underlying p53 regulation of PIK3CA transcription in ovarian surface epithelium and in ovarian cancer. J. Cell Sci. 2008, 121, 664–674. [Google Scholar] [CrossRef]
  138. Bozhanov, S.S.; Angelova, S.G.; Krasteva, M.E.; Markov, T.L.; Christova, S.L.; Gavrilov, I.G.; Georgieva, E.I. Alterations in p53, BRCA1, ATM, PIK3CA, and HER2 genes and their effect in modifying clinicopathological characteristics and overall survival of Bulgarian patients with breast cancer. J. Cancer Res. Clin. Oncol. 2010, 136, 1657–1669. [Google Scholar] [CrossRef]
  139. Astanehe, A.; Arenillas, D.; Wasserman, W.W.; Leung, P.C.K.; Dunn, S.E.; Davies, B.R.; Mills, G.B.; Auersperg, N. Expression of Concern: Mechanisms underlying p53 regulation of PIK3CA transcription in ovarian surface epithelium and in ovarian cancer. J. Cell Sci. 2020, 133. [Google Scholar] [CrossRef]
  140. Singh, B.; Reddy, P.G.; Goberdhan, A.; Walsh, C.; Dao, S.; Ngai, I.; Chou, T.C.; P, O.C.; Levine, A.J.; Rao, P.H.; et al. p53 regulates cell survival by inhibiting PIK3CA in squamous cell carcinomas. Genes Dev. 2002, 16, 984–993. [Google Scholar] [CrossRef]
  141. Italiano, A.; Chen, C.L.; Thomas, R.; Breen, M.; Bonnet, F.; Sevenet, N.; Longy, M.; Maki, R.G.; Coindre, J.M.; Antonescu, C.R. Alterations of the p53 and PIK3CA/AKT/mTOR pathways in angiosarcomas: A pattern distinct from other sarcomas with complex genomics. Cancer 2012, 118, 5878–5887. [Google Scholar] [CrossRef]
  142. Chen, S.M.Y.; Li, B.; Nicklawsky, A.G.; Krinsky, A.L.; Brunetti, T.; Woolaver, R.A.; Wang, X.; Chen, Z.; Young, C.D.; Gao, D.; et al. Deletion of p53 and Hyper-Activation of PIK3CA in Keratin-15(+) Stem Cells Lead to the Development of Spontaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2020, 21, 6585. [Google Scholar] [CrossRef] [PubMed]
  143. Coussy, F.; El Botty, R.; Lavigne, M.; Gu, C.; Fuhrmann, L.; Briaux, A.; de Koning, L.; Dahmani, A.; Montaudon, E.; Morisset, L.; et al. Combination of PI3K and MEK inhibitors yields durable remission in PDX models of PIK3CA-mutated metaplastic breast cancers. J. Hematol. Oncol. 2020, 13, 13. [Google Scholar] [CrossRef] [PubMed]
  144. Kaboli, P.J.; Imani, S.; Jomhori, M.; Ling, K.H. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am. J. Cancer Res. 2021, 11, 5155–5183. [Google Scholar] [PubMed]
  145. Elkabets, M.; Vora, S.; Juric, D.; Morse, N.; Mino-Kenudson, M.; Muranen, T.; Tao, J.; Campos, A.B.; Rodon, J.; Ibrahim, Y.H.; et al. mTORC1 inhibition is required for sensitivity to PI3K p110alpha inhibitors in PIK3CA-mutant breast cancer. Sci. Transl. Med. 2013, 5, 196ra199. [Google Scholar] [CrossRef]
  146. Bloom, B.S.; Nelson, J.S.; Geronemus, R.G. Topical rapamycin combined with pulsed dye laser (PDL) in the treatment of capillary vascular malformations-Anatomical differences in response to PDL are relevant to interpretation of study results. J. Am. Acad Derm. 2015, 73, e71. [Google Scholar] [CrossRef] [PubMed]
  147. Seront, E.; Van Damme, A.; Boon, L.M.; Vikkula, M. Rapamycin and treatment of venous malformations. Curr. Opin. Hematol. 2019, 26, 185–192. [Google Scholar] [CrossRef]
  148. Marques, L.; Nunez-Cordoba, J.M.; Aguado, L.; Pretel, M.; Boixeda, P.; Nagore, E.; Baselga, E.; Redondo, P. Topical rapamycin combined with pulsed dye laser in the treatment of capillary vascular malformations in Sturge-Weber syndrome: Phase II, randomized, double-blind, intraindividual placebo-controlled clinical trial. J. Am. Acad Derm. 2015, 72, 151–158 e151. [Google Scholar] [CrossRef]
  149. Akgumus, G.; Chang, F.; Li, M.M. Overgrowth Syndromes Caused by Somatic Variants in the Phosphatidylinositol 3-Kinase/AKT/Mammalian Target of Rapamycin Pathway. J. Mol. Diagn. 2017, 19, 487–497. [Google Scholar] [CrossRef]
  150. Abou-Fadel, J.; Bhalli, M.; Grajeda, B.; Zhang, J. CmP Signaling Network Leads to Identification of Prognostic Biomarkers for Triple-Negative Breast Cancer in Caucasian Women. Genet. Test Mol. Biomark. 2022, 26, 198–219. [Google Scholar] [CrossRef]
  151. Abou-Fadel, J.; Grajeda, B.; Jiang, X.; Cailing-De La, O.A.; Flores, E.; Padarti, A.; Bhalli, M.; Le, A.; Zhang, J. CmP signaling network unveils novel biomarkers for triple negative breast cancer in African American women. Cancer Biomark 2022, 34, 607–636. [Google Scholar] [CrossRef] [PubMed]
  152. Abou-Fadel, J.; Jiang, X.; Grajeda, B.; Padarti, A.; Ellis, C.C.; Flores, E.; Cailing-De La, O.A.; Zhang, J. CCM signaling complex (CSC) couples both classic and non-classic Progesterone receptor signaling. Cell Commun. Signal 2022, 20, 120. [Google Scholar] [CrossRef]
  153. Abou-Fadel, J.; Jiang, X.; Padarti, A.; Goswami, D.; Smith, M.; Grajeda, B.; Walker, W.; Zhang, J. CCM signaling complex (CSC) is a master regulator governing homeostasis of progestins and their mediated signaling cascades. bioRxiv 2020. [Google Scholar] [CrossRef]
  154. Abou-Fadel, J.; Jiang, X.; Padarti, A.; Goswami, D.G.; Smith, M.; Grajeda, B.; Bhalli, M.; Le, A.; Walker, W.E.; Zhang, J. mPR-Specific Actions Influence Maintenance of the Blood-Brain Barrier (BBB). Int. J. Mol. Sci. 2022, 23, 9684. [Google Scholar] [CrossRef] [PubMed]
  155. Renteria, M.; Belkin, O.; Aickareth, J.; Jang, D.; Hawwar, M.; Zhang, J. Zinc’s Association with the CmPn/CmP Signaling Network in Breast Cancer Tumorigenesis. Biomolecules 2022, 12, 1672. [Google Scholar] [CrossRef]
  156. Renteria, M.; Belkin, O.; Jang, D.; Aickareth, J.; Bhalli, M.; Zhang, J. CmPn signaling networks in the tumorigenesis of breast cancer. Front Endocrinol. 2022, 13, 1013892. [Google Scholar] [CrossRef]
  157. Zhang, J.; Abou-Fadel, J. Calm the raging hormone—A new therapeutic strategy involving progesterone-signaling for hemorrhagic CCMs. Vessel. Plus. 2021, 5, 23. [Google Scholar] [CrossRef]
  158. Abou-Fadel, J.; Qu, Y.; Gonzalez, E.; Smith, M.; Zhang, J. Emerging roles of CCM genes during tumorigenesis with potential application as novel biomarkers across major types of cancers. Oncol. Rep. 2020, 3, 1945–1963. [Google Scholar] [CrossRef]
  159. Abou-Fadel, J.; Reid, V.; Le, A.; Zhang, J. Differential expression of key CmPn members distinguishes histological and immune subtypes of hepatic cancers. Qeios 2022. [Google Scholar] [CrossRef]
  160. Abou-Fadel, J.; Vasquez, M.; Grajeda, B.; Ellis, C.; Zhang, J. Systems-wide analysis unravels the new roles of CCM signal complex (CSC). Heliyon 2019, 5, e02899. [Google Scholar] [CrossRef]
  161. Aickareth, J.; Hawwar, M.; Sanchez, N.; Gnanasekaran, R.; Zhang, J. Membrane Progesterone Receptors (mPRs/PAQRs) Are Going beyond Its Initial Definitions. Membranes 2023, 13, 260. [Google Scholar] [CrossRef] [PubMed]
  162. Abou-Fadel, J.; Reid, V.; Le, A.; Croft, J.; Zhang, J. Key Members of the CmPn as Biomarkers Distinguish Histological and Immune Subtypes of Hepatic Cancers. Diagnostics 2023, 13, 1012. [Google Scholar] [CrossRef]
  163. Robinson, G.; Parker, M.; Kranenburg, T.A.; Lu, C.; Chen, X.; Ding, L.; Phoenix, T.N.; Hedlund, E.; Wei, L.; Zhu, X.; et al. Novel mutations target distinct subgroups of medulloblastoma. Nature 2012, 488, 43–48. [Google Scholar] [CrossRef] [PubMed]
  164. Renner, O.; Fominaya, J.; Alonso, S.; Blanco-Aparicio, C.; Leal, J.F.; Carnero, A. Mst1, RanBP2 and eIF4G are new markers for in vivo PI3K activation in murine and human prostate. Carcinogenesis 2007, 28, 1418–1425. [Google Scholar] [CrossRef] [PubMed]
  165. Leystra, A.A.; Deming, D.A.; Zahm, C.D.; Farhoud, M.; Olson, T.J.; Hadac, J.N.; Nettekoven, L.A.; Albrecht, D.M.; Clipson, L.; Sullivan, R.; et al. Mice expressing activated PI3K rapidly develop advanced colon cancer. Cancer Res. 2012, 72, 2931–2936. [Google Scholar] [CrossRef]
  166. Hare, L.M.; Phesse, T.J.; Waring, P.M.; Montgomery, K.G.; Kinross, K.M.; Mills, K.; Roh, V.; Heath, J.K.; Ramsay, R.G.; Ernst, M.; et al. Physiological expression of the PI3K-activating mutation Pik3ca(H1047R) combines with Apc loss to promote development of invasive intestinal adenocarcinomas in mice. Biochem. J. 2014, 458, 251–258. [Google Scholar] [CrossRef]
  167. Engelman, J.A.; Chen, L.; Tan, X.; Crosby, K.; Guimaraes, A.R.; Upadhyay, R.; Maira, M.; McNamara, K.; Perera, S.A.; Song, Y.; et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat. Med. 2008, 14, 1351–1356. [Google Scholar] [CrossRef]
  168. Deming, D.A.; Leystra, A.A.; Nettekoven, L.; Sievers, C.; Miller, D.; Middlebrooks, M.; Clipson, L.; Albrecht, D.; Bacher, J.; Washington, M.K.; et al. PIK3CA and APC mutations are synergistic in the development of intestinal cancers. Oncogene 2014, 33, 2245–2254. [Google Scholar] [CrossRef]
  169. Therkildsen, C.; Bergmann, T.K.; Henrichsen-Schnack, T.; Ladelund, S.; Nilbert, M. The predictive value of KRAS, NRAS, BRAF, PIK3CA and PTEN for anti-EGFR treatment in metastatic colorectal cancer: A systematic review and meta-analysis. Acta Oncol. 2014, 53, 852–864. [Google Scholar] [CrossRef]
  170. Pearson, H.B.; Li, J.; Meniel, V.S.; Fennell, C.M.; Waring, P.; Montgomery, K.G.; Rebello, R.J.; Macpherson, A.A.; Koushyar, S.; Furic, L.; et al. Identification of Pik3ca Mutation as a Genetic Driver of Prostate Cancer That Cooperates with Pten Loss to Accelerate Progression and Castration-Resistant Growth. Cancer Discov. 2018, 8, 764–779. [Google Scholar] [CrossRef]
  171. Meyer, D.S.; Koren, S.; Leroy, C.; Brinkhaus, H.; Muller, U.; Klebba, I.; Muller, M.; Cardiff, R.D.; Bentires-Alj, M. Expression of PIK3CA mutant E545K in the mammary gland induces heterogeneous tumors but is less potent than mutant H1047R. Oncogenesis 2013, 2, e74. [Google Scholar] [CrossRef]
  172. Van Keymeulen, A.; Lee, M.Y.; Ousset, M.; Brohee, S.; Rorive, S.; Giraddi, R.R.; Wuidart, A.; Bouvencourt, G.; Dubois, C.; Salmon, I.; et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature 2015, 525, 119–123. [Google Scholar] [CrossRef] [PubMed]
  173. Tikoo, A.; Roh, V.; Montgomery, K.G.; Ivetac, I.; Waring, P.; Pelzer, R.; Hare, L.; Shackleton, M.; Humbert, P.; Phillips, W.A. Physiological levels of Pik3ca(H1047R) mutation in the mouse mammary gland results in ductal hyperplasia and formation of ERalpha-positive tumors. PLoS ONE 2012, 7, e36924. [Google Scholar] [CrossRef] [PubMed]
  174. Mitchell, C.B.; Phillips, W.A. Mouse Models for Exploring the Biological Consequences and Clinical Significance of PIK3CA Mutations. Biomolecules 2019, 9, 158. [Google Scholar] [CrossRef]
  175. Stratikopoulos, E.E.; Kiess, N.; Szabolcs, M.; Pegno, S.; Kakit, C.; Wu, X.; Poulikakos, P.I.; Cheung, P.; Schmidt, H.; Parsons, R. Mouse ER+/PIK3CA(H1047R) breast cancers caused by exogenous estrogen are heterogeneously dependent on estrogen and undergo BIM-dependent apoptosis with BH3 and PI3K agents. Oncogene 2019, 38, 47–59. [Google Scholar] [CrossRef]
  176. Stratikopoulos, E.E.; Dendy, M.; Szabolcs, M.; Khaykin, A.J.; Lefebvre, C.; Zhou, M.M.; Parsons, R. Kinase and BET Inhibitors Together Clamp Inhibition of PI3K Signaling and Overcome Resistance to Therapy. Cancer Cell 2015, 27, 837–851. [Google Scholar] [CrossRef] [PubMed]
  177. Koren, S.; Bentires-Alj, M. Mouse models of PIK3CA mutations: One mutation initiates heterogeneous mammary tumors. FEBS J. 2013, 280, 2758–2765. [Google Scholar] [CrossRef]
  178. Hanker, A.B.; Pfefferle, A.D.; Balko, J.M.; Kuba, M.G.; Young, C.D.; Sanchez, V.; Sutton, C.R.; Cheng, H.; Perou, C.M.; Zhao, J.J.; et al. Mutant PIK3CA accelerates HER2-driven transgenic mammary tumors and induces resistance to combinations of anti-HER2 therapies. Proc. Natl. Acad. Sci. USA 2013, 110, 14372–14377. [Google Scholar] [CrossRef]
  179. Mirzaa, G.; Graham, J.M., Jr.; Keppler-Noreuil, K. PIK3CA-Related Overgrowth Spectrum. In GeneReviews®; Adam, M.P., Everman, D.B., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  180. Keppler-Noreuil, K.M.; Rios, J.J.; Parker, V.E.; Semple, R.K.; Lindhurst, M.J.; Sapp, J.C.; Alomari, A.; Ezaki, M.; Dobyns, W.; Biesecker, L.G. PIK3CA-related overgrowth spectrum (PROS): Diagnostic and testing eligibility criteria, differential diagnosis, and evaluation. Am. J. Med. Genet. A 2015, 167A, 287–295. [Google Scholar] [CrossRef]
  181. Wang, S.K.; Drucker, N.A.; Gupta, A.K.; Marshalleck, F.E.; Dalsing, M.C. Diagnosis and management of the venous malformations of Klippel-Trenaunay syndrome. J. Vasc. Surg. Venous Lymphat. Disord. 2017, 5, 587–595. [Google Scholar] [CrossRef]
  182. Hughes, M.; Hao, M.; Luu, M. PIK3CA vascular overgrowth syndromes: An update. Curr. Opin. Pediatr. 2020, 32, 539–546. [Google Scholar] [CrossRef] [PubMed]
  183. Su, L.X.; Sun, Y.; Wang, Z.; Wang, D.; Yang, X.; Zheng, L.; Wen, M.; Fan, X.; Cai, R. Complex vascular anomalies and tissue overgrowth of limbs associated with increased skin temperature and peripheral venous dilatation: Parks weber syndrome or PROS? Hereditas 2022, 159, 1. [Google Scholar] [CrossRef] [PubMed]
  184. McCuaig, C.C. Update on classification and diagnosis of vascular malformations. Curr. Opin. Pediatr. 2017, 29, 448–454. [Google Scholar] [CrossRef] [PubMed]
  185. Kirkorian, A.Y.; Grossberg, A.L.; Puttgen, K.B. Genetic basis for vascular anomalies. Semin. Cutan. Med. Surg. 2016, 35, 128–136. [Google Scholar] [CrossRef] [PubMed]
  186. Rose, A.L.; Cathey, S.S. Genetic Causes of Vascular Malformations and Common Signaling Pathways Involved in Their Formation. Derm. Clin. 2022, 40, 449–459. [Google Scholar] [CrossRef]
  187. Richter, G.T.; Friedman, A.B. Hemangiomas and vascular malformations: Current theory and management. Int. J. Pediatr. 2012, 2012, 645678. [Google Scholar] [CrossRef]
  188. Kunimoto, K.; Yamamoto, Y.; Jinnin, M. ISSVA Classification of Vascular Anomalies and Molecular Biology. Int. J. Mol. Sci. 2022, 23, 2358. [Google Scholar] [CrossRef]
  189. Donnelly, L.F.; Adams, D.M.; Bisset, G.S., 3rd. Vascular malformations and hemangiomas: A practical approach in a multidisciplinary clinic. AJR Am. J. Roentgenol. 2000, 174, 597–608. [Google Scholar] [CrossRef]
  190. Diociaiuti, A.; Paolantonio, G.; Zama, M.; Alaggio, R.; Carnevale, C.; Conforti, A.; Cesario, C.; Dentici, M.L.; Buonuomo, P.S.; Rollo, M.; et al. Vascular Birthmarks as a Clue for Complex and Syndromic Vascular Anomalies. Front. Pediatr. 2021, 9, 730393. [Google Scholar] [CrossRef]
  191. Andrews, L.; Shope, C.; Lee, L.W.; Hochman, M. Vascular Anomalies: Nomenclature and Diagnosis. Derm. Clin. 2022, 40, 339–343. [Google Scholar] [CrossRef]
  192. Manor, J.; Lalani, S.R. Overgrowth Syndromes-Evaluation, Diagnosis, and Management. Front. Pediatr. 2020, 8, 574857. [Google Scholar] [CrossRef] [PubMed]
  193. Lacerda Lda, S.; Alves, U.D.; Zanier, J.F.; Machado, D.C.; Camilo, G.B.; Lopes, A.J. Differential diagnoses of overgrowth syndromes: The most important clinical and radiological disease manifestations. Radiol. Res. Pr. 2014, 2014, 947451. [Google Scholar] [CrossRef] [PubMed]
  194. Madsen, R.R.; Vanhaesebroeck, B.; Semple, R.K. Cancer-Associated PIK3CA Mutations in Overgrowth Disorders. Trends Mol. Med. 2018, 24, 856–870. [Google Scholar] [CrossRef] [PubMed]
  195. Hare, L.M.; Schwarz, Q.; Wiszniak, S.; Gurung, R.; Montgomery, K.G.; Mitchell, C.A.; Phillips, W.A. Heterozygous expression of the oncogenic Pik3ca(H1047R) mutation during murine development results in fatal embryonic and extraembryonic defects. Dev. Biol. 2015, 404, 14–26. [Google Scholar] [CrossRef]
  196. Swift, M.R.; Weinstein, B.M. Arterial-venous specification during development. Circ. Res. 2009, 104, 576–588. [Google Scholar] [CrossRef]
  197. Kume, T. Specification of arterial, venous, and lymphatic endothelial cells during embryonic development. Histol. Histopathol. 2010, 25, 637–646. [Google Scholar] [CrossRef]
  198. Blei, F. Overgrowth syndromes with vascular anomalies. Curr. Probl. Pediatr. Adolesc. Health Care 2015, 45, 118–131. [Google Scholar] [CrossRef]
  199. Marty, M.; Bonnaud, C.; Jones, N.; Longy, M.; Vaysse, F.; Bieth, E.; Bailleul-Forestier, I. Gingival Biopsy to Detect Mosaicism in Overgrowth Syndromes: Report of Two Cases of Megalencephaly-Capillary Malformation Syndrome with Periodontal Anomalies. Case Rep. Dent. 2020, 2020, 8826945. [Google Scholar] [CrossRef]
  200. Horbach, S.E.; Jolink, F.; van der Horst, C.M. Oral sildenafil as a treatment option for lymphatic malformations in PIK3CA-related tissue overgrowth syndromes. Dermatol. Ther. 2016, 29, 466–469. [Google Scholar] [CrossRef]
  201. Castel, P.; Carmona, F.J.; Grego-Bessa, J.; Berger, M.F.; Viale, A.; Anderson, K.V.; Bague, S.; Scaltriti, M.; Antonescu, C.R.; Baselga, E.; et al. Somatic PIK3CA mutations as a driver of sporadic venous malformations. Sci. Transl. Med. 2016, 8, 332ra342. [Google Scholar] [CrossRef]
  202. Mahajan, P.; Bergstrom, K.L.; Phung, T.L.; Metry, D.W. The genetics of vascular birthmarks. Clin Derm. 2022, 40, 313–321. [Google Scholar] [CrossRef] [PubMed]
  203. Luks, V.L.; Kamitaki, N.; Vivero, M.P.; Uller, W.; Rab, R.; Bovee, J.V.; Rialon, K.L.; Guevara, C.J.; Alomari, A.I.; Greene, A.K.; et al. Lymphatic and other vascular malformative/overgrowth disorders are caused by somatic mutations in PIK3CA. J. Pediatr. 2015, 166, 1048–1054.e1-5. [Google Scholar] [CrossRef] [PubMed]
  204. Sheen, M.R.; Warner, S.L.; Fields, J.L.; Conejo-Garcia, J.R.; Fiering, S. Myristoylated p110alpha Causes Embryonic Death Due to Developmental and Vascular Defects. Open Life Sci. 2015, 10, 461–478. [Google Scholar] [CrossRef] [PubMed]
  205. Schrenk, S.; Goines, J.; Boscolo, E. A Patient-Derived Xenograft Model for Venous Malformation. J. Vis. Exp. 2020. [Google Scholar] [CrossRef]
  206. Schonning, M.J.; Koh, S.; Sun, R.W.; Richter, G.T.; Edwards, A.K.; Shawber, C.J.; Wu, J.K. Venous malformation vessels are improperly specified and hyperproliferative. PLoS ONE 2021, 16, e0252342. [Google Scholar] [CrossRef]
  207. Goines, J.; Li, X.; Cai, Y.; Mobberley-Schuman, P.; Metcalf, M.; Fishman, S.J.; Adams, D.M.; Hammill, A.M.; Boscolo, E. A xenograft model for venous malformation. Angiogenesis 2018, 21, 725–735. [Google Scholar] [CrossRef]
  208. Goines, J.; Boscolo, E. A Xenograft Model for Venous Malformation. Methods Mol. Biol. 2021, 2206, 179–192. [Google Scholar] [CrossRef]
  209. Venot, Q.; Blanc, T.; Rabia, S.H.; Berteloot, L.; Ladraa, S.; Duong, J.P.; Blanc, E.; Johnson, S.C.; Hoguin, C.; Boccara, O.; et al. Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature 2018, 558, 540–546. [Google Scholar] [CrossRef]
  210. Loconte, D.C.; Grossi, V.; Bozzao, C.; Forte, G.; Bagnulo, R.; Stella, A.; Lastella, P.; Cutrone, M.; Benedicenti, F.; Susca, F.C.; et al. Molecular and Functional Characterization of Three Different Postzygotic Mutations in PIK3CA-Related Overgrowth Spectrum (PROS) Patients: Effects on PI3K/AKT/mTOR Signaling and Sensitivity to PIK3 Inhibitors. PLoS ONE 2015, 10, e0123092. [Google Scholar] [CrossRef]
  211. Saal, L.H.; Holm, K.; Maurer, M.; Memeo, L.; Su, T.; Wang, X.; Yu, J.S.; Malmstrom, P.O.; Mansukhani, M.; Enoksson, J.; et al. PIK3CA mutations correlate with hormone receptors, node metastasis, and ERBB2, and are mutually exclusive with PTEN loss in human breast carcinoma. Cancer Res. 2005, 65, 2554–2559. [Google Scholar] [CrossRef]
  212. Kobialka, P.; Sabata, H.; Vilalta, O.; Gouveia, L.; Angulo-Urarte, A.; Muixi, L.; Zanoncello, J.; Munoz-Aznar, O.; Olaciregui, N.G.; Fanlo, L.; et al. The onset of PI3K-related vascular malformations occurs during angiogenesis and is prevented by the AKT inhibitor miransertib. EMBO Mol. Med. 2022, 14, e15619. [Google Scholar] [CrossRef] [PubMed]
  213. Mooney, M.A.; Zabramski, J.M. Developmental venous anomalies. Handb. Clin. Neurol. 2017, 143, 279–282. [Google Scholar] [CrossRef]
  214. Das, K.K.; Rangari, K.; Singh, S.; Bhaisora, K.S.; Jaiswal, A.K.; Behari, S. Coexistent Cerebral Cavernous Malformation and Developmental Venous Anomaly: Does an Aggressive Natural History Always Call for Surgical Intervention? Asian J. Neurosurg. 2019, 14, 318–321. [Google Scholar] [CrossRef]
  215. Aydin, F.; Ogul, H. Coexistence of Cerebral Calcified Cavernous Malformation and Developmental Venous Anomaly. J. Craniofac. Surg. 2021, 32, e45–e46. [Google Scholar] [CrossRef] [PubMed]
  216. Awad, I.A.; Giannotta, S.L.; Robinson, J.R.; Chandrasoma, P.T.; Mohanty, S.; Samson, D.; Estes, M.L. Mixed Vascular Malformations of the Brain—Clinical and Pathogenetic Considerations. Neurosurgery 1993, 33, 179–188. [Google Scholar] [CrossRef]
  217. Idiculla, P.S.; Gurala, D.; Philipose, J.; Rajdev, K.; Patibandla, P. Cerebral Cavernous Malformations, Developmental Venous Anomaly, and Its Coexistence: A Review. Eur. Neurol. 2020, 83, 360–368. [Google Scholar] [CrossRef]
  218. Wu, B.; Liu, W.D.; Zhao, Y. Coexistence of Extra-Axial Cavernous Malformation and Cerebellar Developmental Venous Anomaly in the Cerebellopontine Angle. World Neurosurg. 2012, 78. [Google Scholar] [CrossRef]
  219. Maeder, P.; Gudinchet, F.; Meuli, R.; de Tribolet, N. Development of a cavernous malformation of the brain. AJNR Am. J. Neuroradiol. 1998, 19, 1141–1143. [Google Scholar]
  220. Rigamonti, D.; Hadley, M.N.; Drayer, B.P.; Johnson, P.C.; Hoenig-Rigamonti, K.; Knight, J.T.; Spetzler, R.F. Cerebral cavernous malformations. Incidence and familial occurrence. N. Engl. J. Med. 1988, 319, 343–347. [Google Scholar] [CrossRef] [PubMed]
  221. Bianconi, A.; Salvati, L.F.; Perrelli, A.; Ferraris, C.; Massara, A.; Minardi, M.; Aruta, G.; Rosso, M.; Massa Micon, B.; Garbossa, D.; et al. Distant Recurrence of a Cerebral Cavernous Malformation in the Vicinity of a Developmental Venous Anomaly: Case Report of Local Oxy-Inflammatory Events. Int. J. Mol. Sci. 2022, 23, 14643. [Google Scholar] [CrossRef] [PubMed]
  222. Mondejar, R.; Solano, F.; Rubio, R.; Delgado, M.; Perez-Sempere, A.; Gonzalez-Meneses, A.; Vendrell, T.; Izquierdo, G.; Martinez-Mir, A.; Lucas, M. Mutation prevalence of cerebral cavernous malformation genes in Spanish patients. PLoS ONE 2014, 9, e86286. [Google Scholar] [CrossRef]
  223. Shiran, S.I.; Ben-Sira, L.; Elhasid, R.; Roth, J.; Tabori, U.; Yalon, M.; Constantini, S.; Dvir, R. Multiple Brain Developmental Venous Anomalies as a Marker for Constitutional Mismatch Repair Deficiency Syndrome. AJNR Am. J. Neuroradiol. 2018, 39, 1943–1946. [Google Scholar] [CrossRef]
  224. Madsen, R.R.; Semple, R.K. PIK3CA-related overgrowth: Silver bullets from the cancer arsenal? Trends Mol. Med. 2022, 28, 255–257. [Google Scholar] [CrossRef]
  225. Heng, Y.J.; Lester, S.C.; Tse, G.M.; Factor, R.E.; Allison, K.H.; Collins, L.C.; Chen, Y.Y.; Jensen, K.C.; Johnson, N.B.; Jeong, J.C.; et al. The molecular basis of breast cancer pathological phenotypes. J. Pathol. 2017, 241, 375–391. [Google Scholar] [CrossRef]
  226. Cooper, L.A.; Demicco, E.G.; Saltz, J.H.; Powell, R.T.; Rao, A.; Lazar, A.J. PanCancer insights from The Cancer Genome Atlas: The pathologist’s perspective. J. Pathol. 2018, 244, 512–524. [Google Scholar] [CrossRef]
Figure 1. Oncogrid analysis of simple somatic mutations in known breast cancer oncogenes and key genes within the CmPn signaling network. Known driver oncogenes (red colored) and key components (black colored) of the CmPn signaling network from 200 breast cancer lesions in Genomic Data Commons (GDC) and The Cancer Genome Atlas (TCGA). Utilizing the GDC data portal from the National Cancer Institute (NCI), the distribution of simple somatic mutations (SSM) for breast cancer lesions for common breast cancer driver genes, along with SSM counted in all key CmPn players were assessed. SSM, represented with green dots, are somatic mutations that include single base substitutions, small deletions, and insertions (≤200 bp).
Figure 1. Oncogrid analysis of simple somatic mutations in known breast cancer oncogenes and key genes within the CmPn signaling network. Known driver oncogenes (red colored) and key components (black colored) of the CmPn signaling network from 200 breast cancer lesions in Genomic Data Commons (GDC) and The Cancer Genome Atlas (TCGA). Utilizing the GDC data portal from the National Cancer Institute (NCI), the distribution of simple somatic mutations (SSM) for breast cancer lesions for common breast cancer driver genes, along with SSM counted in all key CmPn players were assessed. SSM, represented with green dots, are somatic mutations that include single base substitutions, small deletions, and insertions (≤200 bp).
Jpm 13 00673 g001
Figure 2. Oncogrid analysis of simple somatic mutations in known reproductive cancer oncogenes and key genes within the CmPn signaling network. Known driver oncogenes (red colored) and key components (black colored) of the CmPn signaling network from 200 breast cancer lesions in Genomic Data Commons (GDC) and The Cancer Genome Atlas (TCGA). Utilizing the GDC data portal from the National Cancer Institute (NCI), the distribution of simple somatic mutations (SSM) for reproductive cancer lesions for common cancer driver genes, along with SSM counted in all key CmPn players were assessed. SSM, represented with green dots, are somatic mutations that include single base substitutions, small deletions, and insertions (≤200 bp).
Figure 2. Oncogrid analysis of simple somatic mutations in known reproductive cancer oncogenes and key genes within the CmPn signaling network. Known driver oncogenes (red colored) and key components (black colored) of the CmPn signaling network from 200 breast cancer lesions in Genomic Data Commons (GDC) and The Cancer Genome Atlas (TCGA). Utilizing the GDC data portal from the National Cancer Institute (NCI), the distribution of simple somatic mutations (SSM) for reproductive cancer lesions for common cancer driver genes, along with SSM counted in all key CmPn players were assessed. SSM, represented with green dots, are somatic mutations that include single base substitutions, small deletions, and insertions (≤200 bp).
Jpm 13 00673 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, J.; Croft, J.; Le, A. Familial CCM Genes Might Not Be Main Drivers for Pathogenesis of Sporadic CCMs-Genetic Similarity between Cancers and Vascular Malformations. J. Pers. Med. 2023, 13, 673. https://doi.org/10.3390/jpm13040673

AMA Style

Zhang J, Croft J, Le A. Familial CCM Genes Might Not Be Main Drivers for Pathogenesis of Sporadic CCMs-Genetic Similarity between Cancers and Vascular Malformations. Journal of Personalized Medicine. 2023; 13(4):673. https://doi.org/10.3390/jpm13040673

Chicago/Turabian Style

Zhang, Jun, Jacob Croft, and Alexander Le. 2023. "Familial CCM Genes Might Not Be Main Drivers for Pathogenesis of Sporadic CCMs-Genetic Similarity between Cancers and Vascular Malformations" Journal of Personalized Medicine 13, no. 4: 673. https://doi.org/10.3390/jpm13040673

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

Article Metrics

Back to TopTop