Next Article in Journal
Towards Personalized Sampling in Clear Cell Renal Cell Carcinomas
Next Article in Special Issue
The Therapeutic Role of Plastic and Reconstructive Surgery in the Interdisciplinary Treatment of Soft-Tissue Sarcomas in Germany—Cross-Sectional Results of a Prospective Nationwide Observational Study (PROSa)
Previous Article in Journal
Prevalence and Risk Factors for Hyposalivation and Xerostomia in Childhood Cancer Survivors Following Different Treatment Modalities—A Dutch Childhood Cancer Survivor Study Late Effects 2 Clinical Study (DCCSS LATER 2)
Previous Article in Special Issue
Oncological Safety and Recurrence in the Surgical Treatment of Atypical Fibroxanthoma and Pleomorphic Dermal Sarcoma of the Scalp
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of CDK Pathway Dysregulation and Its Therapeutic Potential in Soft Tissue Sarcoma

by
Johannes Tobias Thiel
*,
Adrien Daigeler
,
Jonas Kolbenschlag
,
Katarzyna Rachunek
and
Sebastian Hoffmann
Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, 72076 Tuebingen, Germany
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(14), 3380; https://doi.org/10.3390/cancers14143380
Submission received: 17 May 2022 / Revised: 7 July 2022 / Accepted: 9 July 2022 / Published: 12 July 2022

Abstract

:

Simple Summary

Soft tissue sarcomas (STSs) are rare malignant conditions with more than 70 subtypes that are difficult to treat, especially in advanced or metastatic states. Recently, next-generation sequencing technologies have provided comprehensive information and developed personalized medicine for treating cancer in general and STSs in particular. Growing knowledge of diverse gene alterations and biomolecular targets in various subtypes of STSs raises hope for novel treatment approaches and heralds a paradigm shift in the treatment of STSs. Activated cyclin-dependent kinases (CDKs) appear to play a critical role in sarcoma development and represent important targets for sarcoma therapy. This review discusses how CDK signaling influences STS development and its implications for STS prediction and targeted treatment.

Abstract

Soft tissue sarcomas (STSs) are tumors that are challenging to treat due to their pathologic and molecular heterogeneity and their tumor biology that is not yet fully understood. Recent research indicates that dysregulation of cyclin-dependent kinase (CDK) signaling pathways can be a strong driver of sarcogenesis. CDKs are enzyme forms that play a crucial role in cell-cycle control and transcription. They belong to the protein kinases group and to the serine/threonine kinases subgroup. Recently identified CDK/cyclin complexes and established CDK/cyclin complexes that regulate the cell cycle are involved in the regulation of gene expression through phosphorylation of critical components of transcription and pre-mRNA processing mechanisms. The current and continually growing body of data shows that CDKs play a decisive role in tumor development and are involved in the proliferation and growth of sarcoma cells. Since the abnormal expression or activation of large numbers of CDKs is considered to be characteristic of cancer development and progression, dysregulation of the CDK signaling pathways occurs in many subtypes of STSs. This review discusses how reversal and regulation can be achieved with new therapeutics and summarizes the current evidence from studies regarding CDK modulation for STS treatment.

1. Introduction

Soft tissue sarcomas (STSs) are rare, heterogeneous malignant tumors that accounts for about 1–2% of all cancers. In the United States, there are 12,750 new cases diagnosed yearly, and STSs kill 5270 people each year [1]. The crude incidence rate of STSs is 4.71 per 100,000 people in Europe, with an estimated 25,851 new cases in the European Union [2]. Soft tissue sarcoma is currently composed of approximately 80 subtypes defined by the World Health Organization (WHO), classified based on a combination of unique morphological, immunohistochemical, and molecular characteristics [3]. Although the ultimate cellular origin of sarcoma subtypes remains unclear, there is increasing evidence that they arise de novo from mesenchymal pluripotent stem cells [4,5].
The mainstay of therapy has been surgical resection with negative margins, but the prognostic impact of tumor-free margins on prognosis remains controversial [6,7]. The risk of recurrence and distant metastasis (DM) is mainly related to tumor biology. There are significant variations in the incidence of DM across different sarcoma histologies, and the tumor grade and size impact this risk significantly in STSs [8]. Overall, the estimated five-year survival for STSs is ~57–62% and can vary widely depending on the disease stage and the complex interplay between the anatomical site and STS subtype [9]. Patients with advanced STSs have a median overall survival of fewer than 18 months and require systemic therapies, which unfortunately have not been very promising so far [10,11,12].
Additionally, due to the fact that STSs are heterogeneous, responses to generalized therapy and active substances are variable and usually no longer translate among unique subtypes [13]. Therefore, the treatment for each sarcoma subtype should be individual and personalized. To accomplish this goal, biomarkers and critical points in the signaling pathways for growth and progression must be elucidated and characterized. Recent advances advise that changes in cyclin-dependent kinase (CDK) pathways are vital drivers of sarcomagenesis specifically and of cancer in general [9,10,14,15].

2. What Are Cyclin-Dependent Kinases (CDKs)?

Cyclin-dependent kinases (CDKs) were first discovered through genetic and biochemical studies in various organisms, including yeasts and complex organisms such as frogs. With their discovery came a growing understanding of the importance of CDKs in cell reproduction [16,17]. In the 1960s, the cell-cycle phase in eukaryotic cells was described as a sequence of four phases (see Figure 1). A few years later, in 1987, the first CDK was described, cell division cycle 2 (cdc2), again changing our understanding of cell-cycle progression. As scientists discovered cdc2 first, it was named CDK1 [18]. CDKs are serine-threonine kinases; they phosphorylate their substrates at serines and threonines. The enzymes regulate transcription and mRNA processing and may also be involved in neuronal differentiation [10]. CDKs have no function in resting cells because of a structural confirmation that obscures the catalytic and substrate-binding domains [10,19,20]. Their serine/threonine-specific catalytic core partners, called cyclins, inherit regulatory subunits, controlling kinase activity and substrate specificity [19]. Specific subsets of cyclins and CDKs regulate each phase transition in the cell cycle. Therefore, CDKs are essential enzymes that control the transition of the individual phases in the cell cycle through restriction points in a compassionate manner [21].
CDK–cyclin complex activity is tightly regulated by many CDK inhibitors (CKIs), which stop the cell-cycle progression under unfavorable conditions [22]. To date, 20 different CDKs (numbered from CDK1 to CDK20) and 29 human cyclins and cyclin-like proteins have been identified [18]. UniProtKB IDs list the functions, structures, sequences, and interactions of many known CDKs, accessible at the website https://www.uniprot.org/uniprot/ (accessed on 16 May 2022) [20].
CDKs were traditionally divided into two groups: CDKs of the first group can bind multiple cyclins and regulate the cell cycle progression (CDKs 1–4, 6 and 7). CDKs of the second group form complexes with a single cyclin and are involved in regulating transcription processes (CDKs 7–9,12, 13 and 19) [23,24,25,26]. CDKs 5, 10, 11, 14–18, and 20 do not fit into the abovementioned categories. They lack explicit functional annotations and have different functionalities, which are often tissue specific [23]. CDK5, for example, cannot directly control cell-cycle regulation [27]. It regulates neuronal development and post-mitotic neuronal activities by binding with p35 [28]. Substrates of CDK5, such as transcription factor p53 and myocyte enhancer factor 2 (MEF2), are involved in sarcoma progression [29,30].
Due to deregulation of the CDK pathway, uncontrolled cell proliferation often leads to cancer [31].
There is increasing evidence that the impaired activation and expression of CDKs are associated with tumors; conversely, targeting CDKs in tumor cells has become a promising therapeutic strategy [32]. The inhibition of CDKs can reduce the growth and progression of sarcoma cells and lead the diseased cells into apoptosis [20,32].

3. Selected CDKs and Their Role in Sarcoma Research and Treatment

CDKs are focused on providing targeted therapy to patients suffering from sarcoma. However, the same treatment differs in efficacy in different patients and tumors. These results reflect the unique microenvironment of each tumor. In addition to the specific microenvironment, compensatory pathways that undermine the mechanism of new CDK-related treatments have also been discovered [9,33]. Based on sequence homology, scientists have mapped and grouped CDKs, cyclins, and CKIs. As more and more information has been collected, it has become clear that the earlier, rather strict criteria for classifying these proteins are no longer correct. Recently, studies have shown that complexes of CDK and cyclin subunits are themselves highly active [19]. In the following section, we summarize the most important CDKs in STSs, potential biomarkers, and possible molecular targets of CKIs.

3.1. CDK1

CDK1 (CDC2) plays a vital role during the cell cycle. This enzyme strongly regulates the S phase and the G2 phase. The separate binding of cyclin A and cyclin B to CDK1 drives the transition from the G2 phase to the M phase [32,34]. Experiments with knockout CDK1 mice have shown that CDK1 is essential for initiating mitosis [34]. The phosphorylation of the complex of CDK1 and cyclin B by Wee1, a serine/threonine kinase, leads to the inhibition of CDK1 (see Figure 1) [32]. Therefore, by inhibiting the inhibitor Wee1, the activity of CDK1 can be increased. The proof of this principle has been shown using the CKI MK1775 (adavosertib), a Wee1 inhibitor in different cell lines, derived from human liposarcomas (LPSs) and from rhabdomyosarcomas (RMSs). Notably, in these cell lines, CDK1 is strongly expressed during the progression of the S phase and the transition from the G phase to the M phase. The proliferation ability of the cells is decreased by the inhibition of CDK1 expression or activation [35,36].

3.2. CDK2

CDK2, similar to CDK1, is a serine/threonine kinase and is involved in the transition from the G1 phase to the S phase and is closely associated with cyclins A and E (see Figure 1). For the treatment of STSs, p27, a tumor suppressor protein, is of interest. P27 inhibits CDK2. Therefore, upregulation of p27 in a human RMS cell line results in potent inhibition of CDK2, and decreases the proliferation ability of cells [37,38]. Transforming growth factor-beta (TGFβ1) was found to initiate the upregulation of p27 in RMS. It also enhances the binding affinity of p27 to the complex of CDK2 and cyclin E [37]. Due to these two mechanisms, TGFβ1 is a promising inhibitor of tumor cell proliferation in RMS, round liposarcomas, and myxoid cell lines. The cell line HS-18 derived from human LPSs highly expresses CDK2 and cyclin A and cyclin E [39,40]. This high expression of CDK2 and genetic aberrations in the coding sequences for CDK2 in sarcoma have also been associated with a bad clinical course. Therefore, CDK2 gene aberrations are considered crucial prognoses influencing factors [41].

3.3. CDK4 and CDK6

CDK4 and CDK6 both interact with D-type cyclins. Three D-type cyclins are currently known: cyclins D1, D2, and D3. Not only are binding partners identical, but also 71% of the amino acid identity in CDK4 and CDK6 is the same [32]. The crucial role of both proteins is to promote the progression of the G1 phase and the transition from the G1 phase to the S phase (see Figure 1).
P16 (also known as CDK inhibitor 2A) and p21 (also known as CDK inhibitor 1A) are familiar CKIs for CDK4 and CDK6; both p proteins are tumor suppressors. CDK4 and CDK6 can phosphorylate the retinoblastoma tumor suppressor protein (Rb1). In this way, both CDKs inactivate Rb1 and silence multiple genes [42].
The interplay of Rb1, CDK4, and CDK6 influences cancer cell proliferation, differentiation, and transformation [43]. In sarcomas, research on CDK4 and CDK6 as well as CKIs such as palbociclib is still ongoing and the most promising, even if the majority of studies are experimental. Palbociclib is a potent inhibitor (CKI) of both CDK4 and CDK6. Rb1-proficient ovarian cancer cell lines are sensitive to palbociclib; in contrast, glioblastoma multiforme cell lines are highly resistant to palbociclib [44,45]. These cell lines inherit deletions or mutations in the Rb1 gene. This highlights that active, hypophosphorylated Rb1 is key to the efficacy of palbociclib. By confronting Rb1-deficient cell lines with extremely high levels of CKI, the principle was proven. Even high concentrations of palbociclib have failed to induce G1 arrest [45]. Some authors have suggested that Rb1 is a predictive biomarker for response to CDK4 and CDK6 inhibitors [32].
A total of 85% of myxoid and round cell liposarcoma highly express both CDKs. Rb1 immunoreactivity has been reported in 66% of these sarcoma subtypes [46]. Scientists have observed a significant overexpression of CDK4 and CDK6 in mouse model, linked with the progression and occurrence of both dedifferentiated (DDLPSs) and well-differentiated liposarcomas (WDLPSs) [47,48].
CDK4 may even be used as a prognostic marker, as poor disease-specific survival was associated with high expression of CDK4 in 56 patients with LPSs [49]. Additionally, a significantly high expression of CDK4 has been found in patients with WDLPSs recurrence after surgery [49]. Complementary low expression of CDK4 in these tumors was associated with a better prognosis and a higher progression-free survival [50].
In addition to palbociclib, ribociclib is another interesting CKI, also known as LEE011, and a selective CDK4 inhibitor. This drug arrests liposarcoma tumor cells in the G0 phase and G1 phase after 24 h of incubation and limits tumor cell proliferation [51]. Few clinical trials have been designed with this drug, and the majority of registered clinical trials are still ongoing. Recently, a phase Ib study in patients with LPSs and ribociclib has been completed [52]. Together with other clinical trial results, these are discussed in Section 5. CDK4 and CDK6 are very similar from the ultrastructural point of view. Interestingly, the efficacy of dual CDK4/6 inhibitors against CDK4 and CDK6 is different. In vitro studies have found that the dual inhibitors abemaciclib, ribociclib, and trilaciclib were more powerfully inhibiting CDK4 than CDK6, while palbociclib, in contrast, had comparable efficacy against both CDK4 and CDK6 [53,54,55]. Finally, abemaciclib and trilaciclib not only inhibit CDK4/6 but also have a slight inhibitory effect on CDK5 and CDK9.
In RMS, CDK4/6 inhibitors appear to be able to arrest tumor growth. CDK4 knockdown mice receiving RMS cell lines showed impaired proliferation and poor transformation of tumor cells arrested in G1 phase. Deficient Rb1 phosphorylation induced this arrest [56]. In 2015, RMS expressing low levels of CDK4 were shown to be especially sensitive to ribociclib and, therefore, the inhibition of CDK4/6 [57].
In summary, there are few trials on CDK4 and CDK6 inhibitors in the treatment of STS, most of which are in the preclinical or early clinical stage [32,58,59,60,61].

3.4. CDK9

CDK9 is the catalytic subunit of two enzymes: one is Tat-activating kinase and the other is positive transcription elongation factor b (pTEFb) [61]. CDK9 is present and expressed during the whole cell cycle [62]. CDK9 supplies the transcriptional homeostasis and, therefore, fundamental regulation of gene transcription [63]. In malignancies, physiological homeostasis of transcription is generally absent; oncogenes take control and modulate transcription. Therefore, CDK9 is of great interest for targeted-therapy concepts because of its function as a “guardian of cellular transcriptional homeostasis” [20,32,64,65]. The interaction of CDK1/CDK2 and CDK9 can lead to apoptotic arrest in G2 and M phases causing inhibition of the whole cell cycle [66].
Alterations in CDK9 activity in RMS are suspected to inhibit the physiological differentiation of tumor cells [65,67,68]. Synovial sarcoma also shows a clear correlation between poor prognosis and high levels of CDK9 [69]. In chronic lymphocytic leukemia, small-cell lung cancer, and breast cancer, two CDK9 inhibitors (alvocidib (flavopiridol) and seliciclib) have already been applied [70,71].

3.5. CDK11

CDK11 differs from other CDKs; it is not only encoded by a single gene but by two genes. CDC2L1 (CDK11B) and CDC2L2 (CDK11A) share many bases homologously. CDC2L2 is specific to humans and is absent in mice [72,73]; both conventional groups of CDKs do not apply to CDK11 due to its variety of tasks [74]. In eukaryotic cells, a minimum of ten isoforms have been cloned already. The most common and highly active of them is CDK11p110 [75,76,77]. CDK11p110, similar to all CDK11 isoforms, is associated with RNA splicing, transcriptional regulation, and cell division [78]. Although very similar, the isoforms slightly vary in their way of functioning. CDK11p110 forms a complex with cyclin L and mainly interacts with RNA processing and transcription, whereas CDK11p58 pushes mitosis and acts kinase specific in the G2 and M phase [75,78,79,80,81].
LPS tissue microarrays analyzed by immunohistochemistry showed high levels of CDK11. Benign lipoma tissue, in contrast, expressed significantly less CDK11 [74]. First attempts with synthetic lentiviral shRNA and siRNA suppressing CDKs successfully induced and increased doxorubicin’s cytotoxic capability in LPS cells [74]. However, CDK 11 isotypes’ functions cannot be simplified. Scientists have proven the antagonistic effects of CDK11p58 and CDK11p110 in breast and prostate cancer. CDK11p58 appears to induce anti-metastatic and anti-proliferation effects. In contrast, CDK11p110 promotes the cell viability and survival abilities of cell clones [82,83,84].
Although research has intensified on CDK11 inhibitors, no single CKI has been designed yet.

4. CDK Pathway Dysregulation in Prevalent STSs

The following section provides further insight into the dysregulation of the CDK pathways, characterizes potential future molecular targets, and highlights the preclinical studies of the three most common (extra abdominal) STS subtypes in adults. In addition, two examples for rarer subtypes such as malignant peripheral nerve sheath tumors (MPNSTs) and myxofibrosarcomas (MFSs) are addressed and summarized.

4.1. Liposarcoma

Liposarcomas (LPSs) arise from adipocytes and account for a significant proportion (~13–20%) of adult sarcomas [10]. Three biological groups depend on the molecular profile and growth characteristics. The groups consist of well-differentiated (WDLPSs) and dedifferentiated liposarcomas (DDLPSs), myxoid/round cell LPS (MCRLPSs), and pleiomorphic LPSs (PLPSs) [85,86]. However, WDLPSs/DDLPSs account for around 60% of all LPSs, while PLPSs are the rarest (~5%) [87]. WDLPSs/DDLPSs are two sides of one subtype. Unlike WDLPSs, DDLPSs can multiply quickly, are aggressive, and metastasize early [88]. They both involve the amplification and overexpression of CDK4, HMGA2 (high-mobility group AT-hook 2), and mouse double minute 2 (MDM2) due to an amplification of chromosome 12q13-15. In contrast, over 95% of MCRLPSs carry a translocation of FUS and DDIT3 (CHOP) genes, whereas PLPSs often causing the loss of tumor suppressors p53 and Rb1 [89,90,91].
Treatment of locally advanced WDLPSs and DDLPSs or systemic disease is complex because these tumors are hardly chemosensitive [92,93,94]. Current clinical trials are focusing on MDM2 inhibitors, CDK4, and CDK6. Some of these trials are discussed later in Section 5. A total of 90–95% of all WDLPSs/DDLPSs have high levels of CDK4 and almost 100% have co-amplification of MDM2 and, in theory, represent a very good target for new drugs [52,86]. Palbociclib efficiently inhibits CDK4 and CDK6, and thus, cell growth of WDLPSs/DDLPSs in vitro and in xenograft models [91]. Feeding the CDK4/6 inhibitor ribociclib to mice bearing human LPS xenografts ultimately decreased tumor biomarkers, including Rb1 phosphorylation. Continued treatment inhibited the growth of tumors or even caused a regression [51].
CDK11 (see Section 3.5) is suggested to generally be crucial for the growth and proliferation of LPS cells. While CDK11 expression is significantly higher in LPSs, no CDK11 changes are detected in benign lesions (lipomas) [74]. CDK11 could be a promising therapeutic target for the treatment of LPSs. However, there have been no clinical trials of targeted CDK11 inhibition in LPSs due to the lack of a specific CKI.

4.2. Leiomyosarcoma

Leiomyosarcoma (LMS) is a neoplasm characterized by smooth-muscle differentiation, the loss of tumor suppressors, and a lack of recurrent driver mutations. It accounts for up to 20% of all sarcoma diagnoses [95,96]. As typical for most STSs, the overall frequency of LMS increases with age, peaking at 70. However, LMS sometimes also occurs in younger patients starting from 30 years on. Unfortunately, the lack of correspondence of established LMS cell lines to the original mesenchymal neoplasm limits understanding of this subtype.
Nevertheless, some preclinical models have already been developed [95]. The loss of tumor suppressor gene Rb1 leads to a lack of cell-cycle regulation at the checkpoint from the G1 phase to the S phase, leading to uncontrolled cell division [97]. Up to 90% of LMS patients hold altered Rb-cyclin D1 signaling pathways, resulting in a fatal prognosis, which could be an essential factor for proliferation, at least in a subgroup of LMS [98,99]. In three different LMS cell lines, seliciclib, a pan inhibitor of CDK1, 2, 5, and 9 combined with cisplatin and single use, caused a decline in CDK2 mRNA and protein concentration over 72 h [100,101].
Deletions and mutations of Tp53 occur in roughly 50% of cases [10]. Riva et al. presented a case of ten different LMSs affecting a single patient over three years [102]. All 10 LMSs had a deletion on chromosome p19 that inherits the gene for cyclin-dependent kinase inhibitor 2A (CDKN2A). Downregulation of CDKN2A has been described in many malignant processes and significantly correlates with shorter patient survival [102,103,104].

4.3. Undifferentiated Pleomorphic Sarcomas

Undifferentiated pleomorphic sarcomas (UPSs) are a common STS histotype. They account for 14% of all adult STSs, and about 60% occur in the extremities. Nevertheless, they can grow anywhere in the human body [105,106]. Up to 78% of UPS tumors have a deletion of the Rb1 gene due to a loss of regions within chr13q [107,108]. Furthermore, genetic mutation, deletion, or silencing of chr9p21, the region containing the CDK inhibitor p16, leads to free activation of cyclin D-CDK4/6 kinases. A total of 29% of UPSs upregulate genes encoding MDM2 and CDK4 [10,109]. The oncoprotein MDM2 ubiquitinates the tumor suppressor p53 and promotes its proteasomal degradation. Overexpression of MDM2 leads to downregulation of the CKI p21. P21 is a transcriptional target of p53, and its downregulation causes hyperactivation of CDKs [10]. The loss of p53 leads to general genetic instability and promotes additional tumor-promoting mutations. P53 plays a crucial role in the regulation of DNA repair, the cell cycle, programmed cell death, and cellular senescence [110].
Recently, it has been shown that UPSs often have copy-number alterations or mutations in the tumor suppressor genes Rb1 and Tp53. Deletion and mutation of the p53 gene, Tp53, can still be observed in 43% of all UPSs [111]. A recently published study revealed that Rb1- and p53-deficient UPSs required S phase kinase-associated protein 2 (Skp2) for survival. Skp2 can drive the proliferation of UPS cell lines by degrading p21 and p27. The loss of both Rb1 and p53 in patient-derived cell lines renders undifferentiated pleomorphic sarcoma dependent on Skp2, which could provide the basis for promising novel systemic therapies [112].
Further studies have verified that targeting the neurofibromin 1 (NF1) gene and a deletion of CDKN2A can lead to the formation of UPSs [113,114,115]. Despite the high number of experimental studies published so far, there is not a single clinical trial yet that has investigated potential CKIs in patients suffering from UPSs.

4.4. Myxofibrosarcomas

Myxofibrosarcomas (MFSs) are rare mesenchymal soft tissue sarcomas with a high local recurrence (LR) rate [116]. About 5% of all sarcomas are MFSs, the myxoid component in combination with a hypocellular appearance characterizes these sarcomas [117]. A study in 2022 revealed a 5-year LR of 12%, metastasis of 17%, and overall survival (OS) of 84% in 293 patients [118]. Whole exome sequencing of nearly 100 MFS tumors revealed frequent alterations in genes related to the tumor suppressors p53, p15, p16, and Rb1, in addition to MDM2, cyclin D1, and CDK6 [10,119]. These mutations led to the inactivation of the NF1 gene. UPSs showing this inactivation enhance Ras signaling and upregulation of cyclin D1 transcription [91,120].
Amplifying the CDK6 gene and overexpression of the protein leads to higher grading, highlighting the clinical importance of this molecular aberration in promoting disease progression in MFSs [121]. Li et al. characterized the relevance of the alpha-methylacyl coenzyme A racemase (AMACR) in MFSs. AMACR protein overexpression and gene amplification was associated with less metastasis-free survival and disease-specific survival [122]. Stable AMACR knockdown suppresses cell proliferation, growth, and cyclins D1 and T2 expression [122]. Furthermore, downregulation of CDK2 induces high tumor suppressor p12 levels, thereby, inducing MFS cell cycle arrest and apoptosis. Vice versa, a low p12 level is a poor prognostic factor in patients with MFSs [123]. So far, unfortunately, the aforementioned findings and knowledge about potential targets such as CDK6 or CDK2 have not resulted in a clinical trial in MFSs.

4.5. Malignant Peripheral Nerve Sheath Tumors

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that develop in the connective tissue surrounding the nerves and occur predominantly in the extremities [124]. MPNSTs account for 3–10% of all STS diagnoses and arise sporadically (in ~50% of cases) or in patients with the cancer predisposition syndrome, neurofibromatosis type I (NF1) [108,125]. MPNSTs constitute a significant cause of fatal outcomes in NF1 patients, with a 5-year survival rate of only 20–35%. In most MPNSTs, the loss of the tumor suppressor proteins p16 and p27 leads to overexpression of CDK2 and CDK4/6 [126]. These changes results in the inactivation of the Rb1 tumor suppressor, one of the most important tumor suppressors in human cancers, which is a meaningful event in the pathogenesis of MPNST [125,127,128,129].
Characteristic for MPNSTs are NF1-inactivating mutations and frequent genetic disorders of the tumor suppressor gene CDKN2A and polycomb repressor complex 2 (PRC2) [130,131]. In addition, several genes of the CDK metabolism, such as the genes for cyclins D1 (CCND1) and E1 (CCNE1), are suppressed by PRC2 [132,133]. Many genetically engineered mouse models have been developed to more fundamentally understand the genetic changes that occur during MPNST development. For example, mutations of NF1 and Trp53 or NF1 with CDKN2A are possible drivers of MPNST development [134,135,136].
Genomic analyses of MPNSTs have demonstrated the loss of the CDKN2A/B locus in up to 70–80% of MPNSTs [137,138,139]. The loss of CDKN2A leads to the upregulation of CDK4/6, which causes the initiation of the S phase and promotion of mitosis [125,140,141,142]. However, not all MPNSTs are CDKN2A inactivated. In some MPNSTs, only a heterozygous loss is present, which does not cause total inactivation [10,130,143]. Nevertheless, the loss of CDKN2A leads to a more potent activation of CDK4/6, suggesting CDK4/6 inhibitors (CKIs) to be potentially effective in the therapy for MPNSTs [138,144]. Again, clinical studies and evidence are lacking.
In 2020, Kohlmeyer et al. studied and reported a new oncoprotein and negative regulator of Rb1, p53 signaling, and its role in MPNST cell lines [126]. RAB-like 6 isoform A (RABL6A) is this novel protein [145,146,147,148]. Conversely, the silencing of RABL6A led to MPNST cell death and cell-cycle arrest in the G1 phase. Crucial elements for this arrest are the upregulation of p27, the downregulation of CDK4/6, and the activation of Rb1 [126]. In vitro palbociclib promoted MPNST cell death via the reactivation of Rb1 (in a RABL6A-dependent manner). In vivo, it suppressed MPNST. The antitumor effect was enhanced by low-dose combinations of drugs that inhibit multiple kinases (CDK4/6, CDK2). The combined therapy of multiple drugs targeting different signaling pathways could effectively treat MPNSTs [124,126].

4.6. Preclinical Findings in Other Rare Subtypes

BCL6 corepressor (BCOR) sarcomas are rare and defined by alterations of the BCOR gene. These alterations are caused by fusion or BCOR intern tandem duplication. In a retrospective database research, 40 uterine sarcomas in 1390 patients were identified to hold BCOR gene rearrangements [149]. Furthermore, 38% of these uterine BCOR sarcomas showed amplification of CDK4, whereas 45% enhanced MDM2. Finally, 28% were positive for the homozygous deletion of CDKN2A, and therefore, lacked the CDK4 inhibitor. In this regard, a rare clinical case of a BCOR-CCNB3 fusion sarcoma (BCS) treated with palbociclib is discussed in Section 5.

5. Lessons from Clinical Trials Regarding CDK-Directed Therapy in STSs

CDKs appear to be promising targets for cancer treatment and a range of non-oncological diseases such as autoimmune diseases, inflammatory diseases, viral infections including COVID-19, and central nervous system diseases [20]. In recent years, a steadily growing number of clinical trials with CKIs as single agents or in combination with other drugs underlines the importance of CKIs in sarcoma research. Although extensive efforts have been made to discover CKIs, only four CDK inhibitors have been approved since the early 1990s, all dual selective CDK4/6 inhibitors [150]. Yet, clinicians have apply palbociclib, ribociclib, abemaciclib, and trilaciclib for treating estrogen receptor-positive/HER2-negative metastatic breast cancer [151,152,153,154]. Most CKIs under preclinical and clinical investigation targets the cell cycle-associated subset traditionally grouped as CDK group 1. In contrast, the development of CKIs selectively targeting CDKs that regulate transcription (CDK group 2 such as CDK9 or non-groupable CDKs) has just begun.
Targeted therapy with high CDK selectivity has become a significant trend. In contrast to pan-CKIs, selective CKIs can avoid undesirable side effects. However, a significant disadvantage of selective inhibition is that, due to the very dense CDK network and multiple interactions, CDKs can compensate one another in their function and regulation of signaling pathways. For example, dual inhibition of CDK4 and CDK6 has little clinical impact on colorectal cancer and melanoma, as other CDKs such as CDK1–3 compensate for the loss of CDK4 and CDK6 [34]. These results indicate that some cancers might not be helpful to treat with a single specific CKI, which inhibits CDKs selectively.
Nevertheless, what do these preclinical results mean for STS therapy? Can these findings be transferred from the laboratory to the bedside? So far, no single CKI has been approved for therapy in STSs. Up to June 2022, more than a dozen clinical trials were registered with one or more interventional drugs for CDK-targeted therapy (see Table 1). The website www.clinicaltrials.gov (accessed on 30 June 2022) lists all updates in this field.
Most of the ongoing clinical trials are investigating the efficacy and side effects of CKIs. The focus lays mainly on CDK4/6 inhibitors in LPS, alone or combined with other agents. To the best of our knowledge, there are only two registered clinical trials (see Table 1) that have been completed and published yet [155,156,157].

5.1. Clinical Findings in LPSs

As previously stated, WDLPSs and DDLPSs are the subtypes of STSs that show the most promising results for treatment with CKIs, especially with CDK4/6 inhibitors [158].
In 2013, initial data of the first-ever reported phase II study on palbociclib showed a satisfactory progression-free rate in 30 patients suffering from WDLPSs/DDLPSs [156]. CDK4 amplification and Rb expression were closely monitored in this population of advanced tumor patients. All patients were treated with 200 mg palbociclib orally, once daily for 14 days in 21-day cycles. The treatment was generally well tolerated with no serious adverse events. Nevertheless, 24% of patients needed a dose reduction because of hematologic toxicity [156]. The 12-week progression-free survival (PFS) under palbociclib treatment was achieved at 66% with a median PFS of 17.9 weeks. PFS for the standard second-line treatment with ifosfamide was 65% and for trabectin, 40–56% [159].
In a second non-randomized trial, aiming to reduce toxicity, an additional 30 patients were treated with 125 mg palbociclib, once daily for 21 days in a 28-day cycle. The most common side effect was reversible neutropenia. The median PFS was 18 weeks, similar to the previously described study. A 125 mg dose of palbociclib is commercially available and was FDA approved in 2015 for breast cancer [46,160,161,162]. In light of positive study results, palbociclib counts as category 2A evidence in the STS National Comprehensive Cancer Network (NCCN) guidelines; however, to date, it has not been approved by the FDA for STS indication [158].
An ongoing clinical trial (NCT02846987) with abemaciclib shows favorable PFS and objective tumor response (see Table 1). The already available preliminary results show toxicity to be manageable [163]. Abemaciclib belongs to the CDK4 and 6 inhibitor group and seems to be more promising in DDLPSs than palbociclib. In this study, 30 DDLPS patients received abemaciclib 200 mg continuously two times daily, and finally, 29 patients were evaluable for the primary endpoint; 76% of the patients survived after week 12, and the median PFS was 30.4 weeks. The response to abemaciclib seemed to be only partial. Major adverse events were anemia (37%), thrombocytopenia (17%), and diarrhea (7%).
These findings led to a recently initiated multicenter phase III trial (NCT04967521) testing abemaciclib in more patients with DDLPSs. The study focuses on response rate and PFSs in patients treated with abemaciclib for five years (see Table 1, last row).
Ribociclib is a more selective CDK4 inhibitor than palbociclib and also seems to be very promising. A proof-of-concept phase Ib trial published in March 2022 demonstrated that ribociclib paired with siremadlin, a p53-MDM2 antagonist, showed initial signs of antitumor activity in WDLPS or DDLPS patients [52]. In three groups, 74 patients received siremadlin and ribociclib in different schemes and dosages for 13 weeks (median, range 1–174): Group A (n = 26) went through a 4-week cycle of 15 mg siremadlin and 400 mg ribociclib, each once daily. Two weeks of treatment followed two weeks of pause. Group B (n = 29) involved the administration of siremadlin once every three weeks (range 120–200 mg) and ribociclib (range 200–400 mg) daily for two weeks, followed by one week of pause. Finally, in Group C (n = 19), the patients underwent treatment with siremadlin once every four weeks in different dosages (range 120–200 mg) and ribociclib (range 300–400 mg) every two weeks, followed by two weeks of pause. An important aim had been to reduce the risk of bone marrow toxicities. The 3-month PFS rates were 43.8% under regimen A, 65.9% under regimen B, and 55.6% under regimen C. The recommended dose of expansion (RDE) was determined as follows: siremadlin 120 mg every three weeks plus ribociclib 200 mg for two weeks followed by two weeks of pause (regimen B) [52]. In total, three patients achieved a partial response, 38 patients achieved stable disease, and one patient died due to hematotoxicity. Next-generation sequencing (NGS) showed p53 alterations in three of 74 patients, a negative predictor of response to MDM2 inhibitors [164]. These three patients lacked MDM2 amplification, two patients had a concomitant deletion of the Rb1 gene. This deletion is a robust negative predictor of CDK4 inhibitor response. Accordingly, to the gene aberrations, all three patients showed tumor progress despite treatment [165]. In summary, this recently published trial proved the low-dose daily regimen A to be less effective than the high-dose pulsed regimens B and C, at least treating advanced WDLPSs or DDLPSs in patients with MDM2 amplification.
In a previous study, the administration of MDM2 antagonists in combination with doxorubicin resulted in a high rate of hemotoxicity, precluding further development [166]. For this reason, the combination of an MDM2 antagonist with a targeted, less cytotoxic drug, such as selective CKIs such as ribociclib, may be a more relevant approach, and further studies culminating in a phase III study are needed [52].

5.2. Clinical Findings in LMS

Patients with uterus LMS (uLMS) harboring a CDKN2A mutation can profit from a treatment with palbociclib; 19% among 279 uLMS samples inherited mutations affecting the CDK pathway according to genomic analysis [167].
In one case, a woman with uLMS was treated with 125 mg palbociclib for 21 days monthly after multiple surgeries and frustrated chemotherapy. Before starting the CKI treatment, the patient faced metastasis and tumor progression. The tumor was proven to inherit a mutation of the CDKN2A gene, resulting in upregulation of CDK 4 and 6. While under palbociclib due to pancytopenia, the doses had to be reduced from 125 mg to 75 mg. After eight months of CKI treatment, the radiological follow-up by CT scan showed only minor enlargement of the LMS tumors; the tumor had not spread further [167].
A further retrospective NGS study on 114 patients with different sarcoma subtypes, found only 15 patients (13.2%) with relevant therapeutic targets validated by NGS [168]. Furthermore, only four of these 15 patients (26.7%) showed partial response or stable disease for more than six months. Although in one patient diagnosed with LMS holding a CDKN2A/B deletion, molecular profiling suggested a therapy with palbociclib and fulvestrant, no clinical efficacy was evident.
Based on these inconsistent findings, more extensive clinical trials evaluating CKIs, such as palbociclib, for treating LMS are highly required.
In 2017, clinicians in the United States initiated a two-armed study on advanced DDLPSs and LMSs (NCT03114527, see also Table 1). In this study, patients who had received at least one prior systemic medication are put under the combinatory regimen of ribociclib and everolimus. The dose for ribociclib is 300 mg for three weeks with the following week pause, while the dose for everolimus is 2.5 mg in a 28-day cycle. To determine the response evaluation criteria in solid tumors (RECIST), CT or MRI diagnostics are run at several different time points (week 8, 16, 24, and every 12 weeks following). Combining the mTOR inhibitor (everolimus) with a CDK4/6 inhibitor (ribociclib) is a promising approach to connecting novel targeted therapy with an already established antitumor drug. Positive results could lead to a new therapeutic tool for STSs. The first results will be available approximately in late 2022.

5.3. Clinical Findings in Other STSs

A phase II trial included patients who suffered from complex malign tumors that had shown no response to conventional therapy. The primary endpoint was to stabilize the disease for 16 or more weeks. Prior to treatment with ribociclib, patients’ tumor analyzations monitored cyclin D1/D3 amplification, CDK4/CDK6 amplification, CDK4/CDK6 mutations, and p16 mutations. Tumors with amplification of the cyclin D1-CDK4/CDK6 pathway showed a response to the treatment with ribociclib. In summary, only 3 (23.1%) of 13 included STS patients (of a total of 105 included patients with diverse tumor entities) had a partial response. Nevertheless, the primary endpoint was generally not met. The authors of the 2019 study recommended further investigation and additional complementary therapies to ribociclib monotherapy [169].
In 2006, a phase II study concluded that flavopiridol (alvocidib) had few, manageable side effects in treating STSs. Nevertheless, the pan-CDK2, CDK4, CDK6, and CDK9 inhibitor showed no objective treatment response [170]. Therefore, the authors did not recommend further studies with flavopiridol as monotherapy. However, earlier studies have shown additive benefits of (pan-)CKIs such as flavopiridol, improving the efficiencies of other drugs [10]. For example, the combination of flavopiridol and doxorubicin has been well-tolerated in vitro, and in phase I studies on LPSs and MPNSTs [100,101,171].
Two registered clinical trials (see Table 1) with novel selective CDK9 inhibitors (TP-1287 and PRT2527) are currently ongoing.
Finally, palbociclib administration resulted in a complete response in a rare case of refractory pediatric BCS [172]. Several genes in the CDK4/6-RB pathway had been overexpressed, making palbociclib an optimal therapeutic candidate for which child-specific dosing information was available. The targeted therapy with palbociclib, based on a dedicated germline and somatic whole-genome DNA sequencing combined with RNA sequencing, started at the age of eight. For 25 months, there was no further tumor evidence on imaging morphology.

6. Conclusions

Growing evidence on signaling pathways, microenvironments, and interactions among the increasing number of CDKs, CKIs, and tumor cells underlines that cyclin-dependent kinases are vital in sarcoma biology. Strikingly, the plethora of predominantly experimental studies is opaque. Suitable results to date are limited, and the abundance of (experimental) data can be confusing. However, clinical research is still in its infancy, heading for the ambitious goal of targeted-tumor therapy based on each patient’s biomolecular CDK-related footprint. The main focus in CDK clinical research, after extensively characterizing CDKs and their pathways, is now to create and test suitable CKIs and evaluate their efficacy as well as their side effects in clinical trials. For example, CDK11 could be a potential target in LPS therapy; however, so far, a CDK11 inhibitor is not available. Due to their tumor-inhibiting effect in multiple preclinical studies, CDK4/6 inhibitors have become the central component in phase 1 and 2 trials for various sarcoma subtypes. CDK4/6 inhibitors such as palbociclib, abemaciclib and ribociclib currently appear to have the greatest potential for future individualized sarcoma therapy and approval. CKIs have shown mixed results across STS subtypes, suggesting that new, more reliable biomarkers for sensitivity and resistance should be identified. Recent data show that microRNA related to CDK4/6 inhibitors may be useful as predictive biomarkers without compromising sensitivity to the treatment itself [173]. Nevertheless, scientists and clinicians have initially used CKIs only as single agents. However, the current trend is to use CKIs in combination with other chemotherapeutic agents or inhibitors. One advantage of these different combinations, which interfere with different signaling pathways of the STS cell, seems to be the minimization of drug resistance. Ultimately, further phase 2 studies and especially phase 3 trials are needed to confirm the clinical efficacy of CDK inhibitors alone or in combination in the treatment of STSs.

Author Contributions

Conceptualization, J.T.T., S.H. and A.D.; writing—original draft preparation, J.T.T.; writing—review and editing, J.T.T., A.D., J.K., K.R. and S.H.; visualization, S.H. All authors have read and agreed to the published version of the manuscript.

Funding

We acknowledge support by the Open Access Publishing Fund of the University of Tuebingen.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wisdom, A.J.; Mowery, Y.M.; Riedel, R.F.; Kirsch, D.G. Rationale and emerging strategies for immune checkpoint blockade in soft tissue sarcoma. Cancer 2018, 124, 3819–3829. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, H.-W.; Chen, T.W.-W. Genomic-guided precision therapy for soft tissue sarcoma. ESMO Open 2020, 5, e000626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Gronchi, A.; Miah, A.B.; Dei Tos, A.; Abecassis, N.; Bajpai, J.; Bauer, S.; Biagini, R.; Bielack, S.; Blay, J.Y.; Bolle, S.; et al. Soft tissue and visceral sarcomas: ESMO–EURACAN–GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up☆. Ann. Oncol. 2021, 32, 1348–1365. [Google Scholar] [CrossRef]
  4. Bleloch, J.; Ballim, R.D.; Kimani, S.; Parkes, J.; Panieri, E.; Willmer, T.; Prince, S. Managing sarcoma: Where have we come from and where are we going? Ther. Adv. Med Oncol. 2017, 9, 637–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lye, K.L.; Nordin, N.; Vidyadaran, S.; Thilakavathy, K. Mesenchymal stem cells: From stem cells to sarcomas. Cell Biol. Int. 2016, 40, 610–618. [Google Scholar] [CrossRef] [PubMed]
  6. Harati, K.; Goertz, O.; Pieper, A.; Daigeler, A.; Joneidi-Jafari, H.; Niggemann, H.; Stricker, I.; Lehnhardt, M. Soft Tissue Sarcomas of the Extremities: Surgical Margins Can Be Close as Long as the Resected Tumor Has No Ink on It. Oncologist 2017, 22, 1400–1410. [Google Scholar] [CrossRef] [Green Version]
  7. Daigeler, A.; Zmarsly, I.; Hirsch, S.T.F.; Goertz, O.; Steinau, H.-U.; Lehnhardt, M.; Harati, K. Long-term outcome after local recurrence of soft tissue sarcoma: A retrospective analysis of factors predictive of survival in 135 patients with locally recurrent soft tissue sarcoma. Br. J. Cancer 2014, 110, 1456–1464. [Google Scholar] [CrossRef] [Green Version]
  8. Spolverato, G.; Callegaro, D.; Gronchi, A. Defining Which Patients Are at High Risk for Recurrence of Soft Tissue Sarcoma. Curr. Treat. Options Oncol. 2020, 21, 56. [Google Scholar] [CrossRef]
  9. Grünewald, T.G.; Alonso, M.; Avnet, S.; Banito, A.; Burdach, S.; Cidre-Aranaz, F.; Di Pompo, G.; Distel, M.; Dorado-Garcia, H.; Garcia-Castro, J.; et al. Sarcoma treatment in the era of molecular medicine. EMBO Mol. Med. 2020, 12, e11131. [Google Scholar] [CrossRef]
  10. Kohlmeyer, J.L.; Gordon, D.J.; Tanas, M.R.; Monga, V.; Dodd, R.D.; E Quelle, D. CDKs in Sarcoma: Mediators of Disease and Emerging Therapeutic Targets. Int. J. Mol. Sci. 2020, 21, 3018. [Google Scholar] [CrossRef]
  11. A Gusho, C.; Seder, C.W.; Lopez-Hisijos, N.; Blank, A.T.; Batus, M. Pulmonary metastasectomy in bone and soft tissue sarcoma with metastasis to the lung. Interact. Cardiovasc. Thorac. Surg. 2021, 33, 879–884. [Google Scholar] [CrossRef]
  12. Lucchesi, C.; Khalifa, E.; Laizet, Y.; Soubeyran, I.; Mathoulin-Pelissier, S.; Chomienne, C.; Italiano, A. Targetable Alterations in Adult Patients With Soft-Tissue Sarcomas. JAMA Oncol. 2018, 4, 1398–1404. [Google Scholar] [CrossRef]
  13. Dancsok, A.R.; Asleh-Aburaya, K.; Nielsen, T.O. Advances in sarcoma diagnostics and treatment. Oncotarget 2016, 8, 7068–7093. [Google Scholar] [CrossRef] [Green Version]
  14. Jain, S.; Xu, R.; Prieto, V.G.; Lee, P. Molecular classification of soft tissue sarcomas and its clinical applications. Int. J. Clin. Exp. Pathol. 2010, 3, 416–428. [Google Scholar]
  15. Schaefer, I.-M.; Hong, K.; Kalbasi, A. How Technology Is Improving the Multidisciplinary Care of Sarcoma. Am. Soc. Clin. Oncol. Educ. Book 2020, 40, 445–462. [Google Scholar] [CrossRef]
  16. Paris, J.; Le Guellec, R.; Couturier, A.; Omilli, F.; Camonis, J.; MacNeill, S.; Philippe, M. Cloning by differential screening of a Xenopus cDNA coding for a protein highly homologous to cdc2. Proc. Natl. Acad. Sci. USA 1991, 88, 1039–1043. [Google Scholar] [CrossRef] [Green Version]
  17. Malumbres, M. Cyclin-dependent kinases. Genome Biol. 2014, 15, 122. [Google Scholar] [CrossRef] [Green Version]
  18. Uzbekov, R.; Prigent, C. A Journey through Time on the Discovery of Cell Cycle Regulation. Cells 2022, 11, 704. [Google Scholar] [CrossRef]
  19. Lim, S.; Kaldis, P. Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development 2013, 140, 3079–3093. [Google Scholar] [CrossRef] [Green Version]
  20. Xie, Z.; Hou, S.; Yang, X.; Duan, Y.; Han, J.; Wang, Q.; Liao, C. Lessons Learned from Past Cyclin-Dependent Kinase Drug Discovery Efforts. J. Med. Chem. 2022, 65, 6356–6389. [Google Scholar] [CrossRef]
  21. Ettl, T.; Schulz, D.; Bauer, R.J. The Renaissance of Cyclin Dependent Kinase Inhibitors. Cancers 2022, 14, 293. [Google Scholar] [CrossRef]
  22. Hydbring, P.; Malumbres, M.; Sicinski, P.H.P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat. Rev. Mol. Cell Biol. 2016, 17, 280–292. [Google Scholar] [CrossRef]
  23. Chou, J.; Quigley, D.A.; Robinson, T.M.; Feng, F.Y.; Ashworth, A. Transcription-Associated Cyclin-Dependent Kinases as Targets and Biomarkers for Cancer Therapy. Cancer Discov. 2020, 10, 351–370. [Google Scholar] [CrossRef] [Green Version]
  24. Roskoski, R., Jr. Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs. Pharmacol. Res. 2018, 139, 471–488. [Google Scholar] [CrossRef]
  25. Sánchez-Martínez, C.; Lallena, M.J.; Sanfeliciano, S.G.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015–2019). Bioorganic Med. Chem. Lett. 2019, 29, 126637. [Google Scholar] [CrossRef]
  26. Sánchez-Martínez, C.; Gelbert, L.M.; Lallena, M.J.; de Dios, A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs. Bioorganic Med. Chem. Lett. 2015, 25, 3420–3435. [Google Scholar] [CrossRef]
  27. Dhavan, R.; Tsai, L.H. A Decade of CDK5. Nat. Rev. Mol. Cell Biol. 2001, 2, 749–759. [Google Scholar] [CrossRef]
  28. Patrick, G.N.; Zukerberg, L.R.; Nikolic, M.; De La Monte, S.; Dikkes, P.; Tsai, L.-H. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodegeneration. Nature 1999, 402, 615–622. [Google Scholar] [CrossRef]
  29. Zhang, J.; Krishnamurthy, P.K.; Johnson, G.V.W. Cdk5 phosphorylates p53 and regulates its activity. J. Neurochem. 2002, 81, 307–313. [Google Scholar] [CrossRef] [Green Version]
  30. Tang, X.; Wang, X.; Gong, X.; Tong, M.; Park, D.; Xia, Z.; Mao, Z. Cyclin-Dependent Kinase 5 Mediates Neurotoxin-Induced Degradation of the Transcription Factor Myocyte Enhancer Factor 2. J. Neurosci. 2005, 25, 4823–4834. [Google Scholar] [CrossRef] [Green Version]
  31. Kciuk, M.; Gielecińska, A.; Mujwar, S.; Mojzych, M.; Kontek, R. Cyclin-dependent kinases in DNA damage response. Biochim. Biophys. Acta 2022, 1877, 188716. [Google Scholar] [CrossRef]
  32. Liao, Y.; Feng, Y.; Shen, J.; Hornicek, F.J.; Duan, Z. The roles and therapeutic potential of cyclin-dependent kinases (CDKs) in sarcoma. Cancer Metastasis Rev. 2015, 35, 151–163. [Google Scholar] [CrossRef] [PubMed]
  33. Brown, H.K.; Tellez-Gabriel, M.; Cartron, P.-F.; Vallette, F.M.; Heymann, M.-F.; Heymann, D. Characterization of circulating tumor cells as a reflection of the tumor heterogeneity: Myth or reality? Drug Discov. Today 2018, 24, 763–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Santamaría, D.; Barrière, C.; Cerqueira, A.; Hunt, S.; Tardy, C.; Newton, K.; Cáceres, J.F.; Dubus, P.; Malumbres, M.; Barbacid, M. Cdk1 is sufficient to drive the mammalian cell cycle. Nature 2007, 448, 811–815. [Google Scholar] [CrossRef]
  35. Lohberger, B.; Kretschmer, N.; Bernhart, E.; Rinner, B.; Stuendl, N.; Kaltenegger, H.; Kahl, S.; Bauer, R.; Leithner, A. 25-O-acetyl-23,24-dihydro-cucurbitacin F induces cell cycle G2/M arrest and apoptosis in human soft tissue sarcoma cells. J. Ethnopharmacol. 2015, 164, 265–272. [Google Scholar] [CrossRef]
  36. Kreahling, J.M.; Gemmer, J.Y.; Reed, D.; Letson, D.; Bui, M.; Altiok, S. MK1775, a Selective Wee1 Inhibitor, Shows Single-Agent Antitumor Activity against Sarcoma Cells. Mol. Cancer Ther. 2012, 11, 174–182. [Google Scholar] [CrossRef] [Green Version]
  37. Ye, L.; Zhang, H.-Y.; Wang, H.; Yang, G.-H.; Bu, H.; Zhang, L.; Wang, S.-L. Effects of transforming growth factor beta 1 on the growth of rhabdomyosarcoma cell line RD. Chin. Med. J. 2005, 118, 678–686. [Google Scholar]
  38. Mitra, J.; Dai, C.Y.; Somasundaram, K.; El-Deiry, W.S.; Satyamoorthy, K.; Herlyn, M.; Enders, G.H. Induction of p21WAF1/CIP1 and Inhibition of Cdk2 Mediated by the Tumor Suppressor p16INK4a. Mol. Cell. Biol. 1999, 19, 3916–3928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Olofsson, A.; Willén, H.; Göransson, M.; Engström, K.; Meis-Kindblom, J.M.; Stenman, G.; Kindblom, L.-G.; Aman, P. Abnormal expression of cell cycle regulators in FUS-CHOP carrying liposarcomas. Int. J. Oncol. 2004, 25, 1349–1355. [Google Scholar] [CrossRef] [PubMed]
  40. Li, W.-W.; Cordon-Cardo, C.; Chen, Q.; Jhanwar, S.C.; Bertino, J.R. Establishment, characterization and drug sensitivity of four new human soft tissue sarcoma cell lines. Int. J. Cancer 1996, 68, 514–519. [Google Scholar] [CrossRef]
  41. Nakagawa, Y.; Numoto, K.; Yoshida, A.; Kunisada, T.; Ohata, H.; Takeda, K.; Wai, D.; Poremba, C.; Ozaki, T. Chromosomal and genetic imbalances in synovial sarcoma detected by conventional and microarray comparative genomic hybridization. J. Cancer Res. Clin. Oncol. 2006, 132, 444–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Malumbres, M.; Barbacid, M. To cycle or not to cycle: A critical decision in cancer. Nat. Cancer 2001, 1, 222–231. [Google Scholar] [CrossRef] [PubMed]
  43. Malumbres, M.; Sotillo, R.; Santamaría, D.; Galán, J.; Cerezo, A.; Ortega, S.; Dubus, P.; Barbacid, M. Mammalian Cells Cycle without the D-Type Cyclin-Dependent Kinases Cdk4 and Cdk6. Cell 2004, 118, 493–504. [Google Scholar] [CrossRef] [Green Version]
  44. Konecny, G.E.; Winterhoff, B.; Kolarova, T.; Qi, J.; Manivong, K.; Dering, J.; Yang, G.; Chalukya, M.; Wang, H.-J.; Anderson, L.; et al. Expression of p16 and Retinoblastoma Determines Response to CDK4/6 Inhibition in Ovarian Cancer. Clin. Cancer Res. 2011, 17, 1591–1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wiedemeyer, W.R.; Dunn, I.F.; Quayle, S.N.; Zhang, J.; Chheda, M.G.; Dunn, G.P.; Zhuang, L.; Rosenbluh, J.; Chen, S.; Xiao, Y.; et al. Pattern of retinoblastoma pathway inactivation dictates response to CDK4/6 inhibition in GBM. Proc. Natl. Acad. Sci. USA 2010, 107, 11501–11506. [Google Scholar] [CrossRef] [Green Version]
  46. Kraus, A.L.; Yu-Kite, M.; Mardekian, J.; Cotter, M.J.; Kim, S.; Decembrino, J.; Snow, T.; Carson, K.R.; Rockland, J.M.; Gossai, A.; et al. Real-World Data of Palbociclib in Combination With Endocrine Therapy for the Treatment of Metastatic Breast Cancer in Men. Clin. Pharmacol. Ther. 2021, 111, 302–309. [Google Scholar] [CrossRef] [PubMed]
  47. Hélias-Rodzewicz, Z.; Pédeutour, F.; Coindre, J.-M.; Terrier, P.; Aurias, A. Selective elimination of amplified CDK4 sequences correlates with spontaneous adipocytic differentiation in liposarcoma. Genes Chromosom. Cancer 2009, 48, 943–952. [Google Scholar] [CrossRef]
  48. Chung, J.; Shevchenko, A.; Lee, J.B. Evolution of a melanoma in situ to a sarcomatoid dedifferentiated melanoma. J. Cutan. Pathol. 2021, 48, 943–947. [Google Scholar] [CrossRef]
  49. Lee, S.; Park, H.; Ha, S.Y.; Paik, K.Y.; Lee, S.E.; Kim, J.M.; Park, J.B.; Kwon, C.D.; Joh, J.-W.; Choi, Y.-L.; et al. CDK4 Amplification Predicts Recurrence of Well-Differentiated Liposarcoma of the Abdomen. PLoS ONE 2014, 9, e99452. [Google Scholar] [CrossRef] [Green Version]
  50. Shin, Y.K. High level of CDK4 amplification is a poor prognostic factor in well-differentiated and dedifferentiated liposarcoma. Histol. Histopathol. 2013, 29, 127–138. [Google Scholar] [CrossRef]
  51. Zhang, Y.-X.; Sicinska, E.; Czaplinski, J.T.; Remillard, S.P.; Moss, S.; Wang, Y.; Brain, C.; Loo, A.; Snyder, E.L.; Demetri, G.D.; et al. Antiproliferative Effects of CDK4/6 Inhibition in CDK4-Amplified Human Liposarcoma In Vitro and In Vivo. Mol. Cancer Ther. 2014, 13, 2184–2193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Razak, A.R.A.; Bauer, S.; Suarez, C.; Lin, C.-C.; Quek, R.; Hütter-Krönke, M.L.; Cubedo, R.; Ferretti, S.; Guerreiro, N.; Jullion, A.; et al. Co-Targeting of MDM2 and CDK4/6 with Siremadlin and Ribociclib for the Treatment of Patients with Well-Differentiated or Dedifferentiated Liposarcoma: Results from a Proof-of-Concept, Phase Ib Study. Clin. Cancer Res. 2022, 28, 1087–1097. [Google Scholar] [CrossRef]
  53. Tripathy, D.; Bardia, A.; Sellers, W.R. Ribociclib (LEE011): Mechanism of Action and Clinical Impact of This Selective Cyclin-Dependent Kinase 4/6 Inhibitor in Various Solid Tumors. Clin. Cancer Res. 2017, 23, 3251–3262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gelbert, L.M.; Cai, S.; Lin, X.; Sanchez-Martinez, C.; Del Prado, M.; Lallena, M.J.; Torres, R.; Ajamie, R.T.; Wishart, G.N.; Flack, R.S.; et al. Preclinical characterization of the CDK4/6 inhibitor LY2835219: In-vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine. Investig. New Drugs 2014, 32, 825–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fry, D.W.; Harvey, P.J.; Keller, P.R.; Elliott, W.L.; Meade, M.; Trachet, E.; Albassam, M.; Zheng, X.; Leopold, W.R.; Pryer, N.K.; et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol. Cancer Ther. 2004, 3, 1427–1438. [Google Scholar] [CrossRef] [PubMed]
  56. Olanich, M.E.; Sun, W.; Hewitt, S.M.; Abdullaev, Z.; Pack, S.D.; Barr, F.G. CDK4 Amplification Reduces Sensitivity to CDK4/6 Inhibition in Fusion-Positive Rhabdomyosarcoma. Clin. Cancer Res. 2015, 21, 4947–4959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Liu, L.; Wu, J.; Ong, S.S.; Chen, T. Cyclin-Dependent Kinase 4 Phosphorylates and Positively Regulates PAX3-FOXO1 in Human Alveolar Rhabdomyosarcoma Cells. PLoS ONE 2013, 8, e58193. [Google Scholar] [CrossRef] [Green Version]
  58. Cetin, B.; A Wabl, C.; Gumusay, O. CDK4/6 inhibitors: Mechanisms of resistance and potential biomarkers of responsiveness in breast cancer. Futur. Oncol. 2022, 18, 1143–1157. [Google Scholar] [CrossRef]
  59. Bruno, L.; Ostinelli, A.; Waisberg, F.; Enrico, D.; Ponce, C.; Rivero, S.; Blanco, A.; Zarba, M.; Loza, M.; Fabiano, V.; et al. Cyclin-Dependent Kinase 4/6 Inhibitor Outcomes in Patients With Advanced Breast Cancer Carrying Germline Pathogenic Variants in DNA Repair–Related Genes. JCO Precis. Oncol. 2022, 6, e2100140. [Google Scholar] [CrossRef]
  60. Sheikh, M.S.; Satti, S.A. The emerging CDK4/6 inhibitor for breast cancer treatment. Mol. Cell. Pharmacol. 2021, 13, 9–12. [Google Scholar]
  61. Pirngruber, J.; A Johnsen, S. Induced G1 cell-cycle arrest controls replication-dependent histone mRNA 3′ end processing through p21, NPAT and CDK9. Oncogene 2010, 29, 2853–2863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Garriga, J.; Bhattacharya, S.; Calbó, J.; Marshall, R.M.; Truongcao, M.; Haines, D.S.; Graña, X. CDK9 Is Constitutively Expressed throughout the Cell Cycle, and Its Steady-State Expression Is Independent of SKP2. Mol. Cell. Biol. 2003, 23, 5165–5173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bacon, C.W.; D’Orso, I. CDK9: A signaling hub for transcriptional control. Transcription 2018, 10, 57–75. [Google Scholar] [CrossRef] [PubMed]
  64. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.E.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [Green Version]
  65. Cassandri, M.; Fioravanti, R.; Pomella, S.; Valente, S.; Rotili, D.; Del Baldo, G.; De Angelis, B.; Rota, R.; Mai, A. CDK9 as a Valuable Target in Cancer: From Natural Compounds Inhibitors to Current Treatment in Pediatric Soft Tissue Sarcomas. Front. Pharmacol. 2020, 11, 1230. [Google Scholar] [CrossRef]
  66. Cai, D.; Byth, K.F.; Shapiro, G.I. AZ703, an Imidazo[1,2-a]Pyridine Inhibitor of Cyclin-Dependent Kinases 1 and 2, Induces E2F-1-Dependent Apoptosis Enhanced by Depletion of Cyclin-Dependent Kinase 9. Cancer Res. 2006, 66, 435–444. [Google Scholar] [CrossRef] [Green Version]
  67. Simone, C.; Giordano, A. Abrogation of signal-dependent activation of the cdk9/cyclin T2a complex in human RD rhabdomyosarcoma cells. Cell Death Differ. 2006, 14, 192–195. [Google Scholar] [CrossRef] [Green Version]
  68. Zhang, H.; Pandey, S.; Travers, M.; Sun, H.; Morton, G.; Madzo, J.; Chung, W.; Khowsathit, J.; Perez-Leal, O.; Barrero, C.A.; et al. Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell 2018, 175, 1244–1258.e26. [Google Scholar] [CrossRef] [Green Version]
  69. Li, X.; Seebacher, N.A.; Xiao, T.; Hornicek, F.J.; Duan, Z. Targeting regulation of cyclin dependent kinase 9 as a novel therapeutic strategy in synovial sarcoma. J. Orthop. Res. 2018, 37, 510–521. [Google Scholar] [CrossRef] [Green Version]
  70. Sengupta, S.; Biarnes, M.C.; Jordan, V.C. Cyclin dependent kinase-9 mediated transcriptional de-regulation of cMYC as a critical determinant of endocrine-therapy resistance in breast cancers. Breast Cancer Res. Treat. 2013, 143, 113–124. [Google Scholar] [CrossRef] [Green Version]
  71. Shan, B.; Zhuo, Y.; Chin, D.; Morris, C.A.; Morris, G.F.; Lasky, J.A. Cyclin-dependent Kinase 9 Is Required for Tumor Necrosis Factor-α-stimulated Matrix Metalloproteinase-9 Expression in Human Lung Adenocarcinoma Cells. J. Biol. Chem. 2005, 280, 1103–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Shi, J.; Feng, Y.; Goulet, A.-C.; Vaillancourt, R.R.; Sachs, N.A.; Hershey, J.W.; Nelson, M.A. The p34 -related Cyclin-dependent kinase 11 Interacts with the p47 Subunit of Eukaryotic Initiation Factor 3 during Apoptosis. J. Biol. Chem. 2003, 278, 5062–5071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhou, Y.; Shen, J.K.; Hornicek, F.J.; Kan, Q.; Duan, Z. The emerging roles and therapeutic potential of cyclin-dependent kinase 11 (CDK11) in human cancer. Oncotarget 2016, 7, 40846–40859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Jia, B.; Choy, E.; Cote, G.; Harmon, D.; Ye, S.; Kan, Q.; Mankin, H.; Hornicek, F.; Duan, Z. Cyclin-dependent kinase 11 (CDK11) is crucial in the growth of liposarcoma cells. Cancer Lett. 2013, 342, 104–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Choi, H.-H.; Jung, S.Y.; Hyle, J.; Kim, B.-J.; Yoon, K.; Cho, E.-J.; Youn, H.-D.; Lahti, J.M.; Qin, J.; Kim, S.-T. CHK2 kinase promotes pre-mRNA splicing via phosphorylating CDK11p110. Oncogene 2012, 33, 108–115. [Google Scholar] [CrossRef] [Green Version]
  76. Sachs, N.A.; Vaillancourt, R.R. Cyclin-dependent kinase 11p110 and casein kinase 2 (CK2) inhibit the interaction between tyrosine hydroxylase and 14-3-3. J. Neurochem. 2003, 88, 51–62. [Google Scholar] [CrossRef]
  77. A Sachs, N.; Vaillancourt, R.R. Cyclin-dependent kinase 11p110 activity in the absence of CK2. Biochim. Biophys. Acta Gen. Subj. 2003, 1624, 98–108. [Google Scholar] [CrossRef]
  78. Endicott, J.A.; Noble, M.E. Structural characterization of the cyclin-dependent protein kinase family. Biochem. Soc. Trans. 2013, 41, 1008–1016. [Google Scholar] [CrossRef] [Green Version]
  79. Trembley, J.H.; Loyer, P.; Hu, D.; Li, T.; Grenet, J.; Lahti, J.M.; Kidd, V.J. Cyclin Dependent Kinase 11 in RNA Transcription and Splicing. Prog. Nucleic Acid Res. Mol. Biol. 2004, 77, 263–288. [Google Scholar] [CrossRef]
  80. Hu, D.; Mayeda, A.; Trembley, J.H.; Lahti, J.M.; Kidd, V.J. CDK11 Complexes Promote Pre-mRNA Splicing. J. Biol. Chem. 2003, 278, 8623–8629. [Google Scholar] [CrossRef] [Green Version]
  81. Loyer, P.; Trembley, J.H.; Grenet, J.A.; Busson, A.; Corlu, A.; Zhao, W.; Kocak, M.; Kidd, V.J.; Lahti, J.M. Characterization of Cyclin L1 and L2 Interactions with CDK11 and Splicing Factors. J. Biol. Chem. 2008, 283, 7721–7732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Chi, Y.; Huang, S.; Wang, L.; Zhou, R.; Wang, L.; Xiao, X.; Li, D.; Cai, Y.; Zhou, X.; Wu, J. CDK11p58inhibits ERα-positive breast cancer invasion by targeting integrin β3 via the repression of ERα signaling. BMC Cancer 2014, 14, 577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zong, H.; Chi, Y.; Wang, Y.; Yang, Y.; Zhang, L.; Chen, H.; Jiang, J.; Li, Z.; Hong, Y.; Wang, H.; et al. Cyclin D3/CDK11p58 Complex Is Involved in the Repression of Androgen Receptor. Mol. Cell. Biol. 2007, 27, 7125–7142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kren, B.T.; Unger, G.M.; Abedin, J.; I Vogel, R.; Henzler, C.M.; Ahmed, K.; Trembley, J.H. Preclinical evaluation of cyclin dependent kinase 11 and casein kinase 2 survival kinases as RNA interference targets for triple negative breast cancer therapy. Breast Cancer Res. 2015, 17, 19. [Google Scholar] [CrossRef] [Green Version]
  85. Conyers, R.; Young, S.; Thomas, D.M. Liposarcoma: Molecular Genetics and Therapeutics. Sarcoma 2010, 2011, 483154. [Google Scholar] [CrossRef] [Green Version]
  86. Assi, T.; Kattan, J.; Rassy, E.; Nassereddine, H.; Farhat, F.; Honore, C.; Le Cesne, A.; Adam, J.; Mir, O. Targeting CDK4 (cyclin-dependent kinase) amplification in liposarcoma: A comprehensive review. Crit. Rev. Oncol. 2020, 153, 103029. [Google Scholar] [CrossRef]
  87. Crago, A.M.; Dickson, M.A. Liposarcoma. Surg. Oncol. Clin. N. Am. 2016, 25, 761–773. [Google Scholar] [CrossRef] [Green Version]
  88. Crago, A.M.; Singer, S. Clinical and molecular approaches to well differentiated and dedifferentiated liposarcoma. Curr. Opin. Oncol. 2011, 23, 373–378. [Google Scholar] [CrossRef] [Green Version]
  89. Crozat, A.; Åman, P.; Mandahl, N.; Ron, D. Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 1993, 363, 640–644. [Google Scholar] [CrossRef]
  90. Rabbitts, T.H.; Forster, A.; Larson, R.; Nathan, P. Fusion of the dominant negative transcription regulator CHOP with a novel gene FUS by translocation t(12;16) in malignant liposarcoma. Nat. Genet. 1993, 4, 175–180. [Google Scholar] [CrossRef]
  91. Barretina, J.; Taylor, B.S.; Banerji, S.; Ramos, A.H.; Lagos-Quintana, M.; DeCarolis, P.L.; Shah, K.; Socci, N.D.; Weir, B.A.; Ho, A.; et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat. Genet. 2010, 42, 715–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Sueekantaiah, C.; Karakousis, C.P.; Leong, S.P.L.; Sandberg, A.A. Cytogenetic findings in liposarcoma correlate with histopathologic subtypes. Cancer 1992, 69, 2484–2495. [Google Scholar] [CrossRef]
  93. Yen, C.-C.; Chen, T.W.-W. Next frontiers in systemic therapy for soft tissue sarcoma. Chin. Clin. Oncol. 2018, 7, 43. [Google Scholar] [CrossRef] [PubMed]
  94. Manji, G.A.; Schwartz, G.K. Managing Liposarcomas: Cutting Through the Fat. J. Oncol. Pr. 2016, 12, 221–227. [Google Scholar] [CrossRef]
  95. Hemming, M.L.; Bhola, P.; Loycano, M.A.; Anderson, J.A.; Taddei, M.L.; Doyle, L.A.; Lavrova, E.; Andersen, J.L.; Klega, K.S.; Benson, M.R.; et al. Preclinical Modeling of Leiomyosarcoma Identifies Susceptibility to Transcriptional CDK Inhibitors through Antagonism of E2F-Driven Oncogenic Gene Expression. Clin. Cancer Res. 2022, 28, 2397–2408. [Google Scholar] [CrossRef] [PubMed]
  96. Serrano, C.; George, S. Leiomyosarcoma. Hematol. Clin. N. Am. 2013, 27, 957–974. [Google Scholar] [CrossRef]
  97. Meza-Zepeda, L.A.; Kresse, S.H.; Barragan-Polania, A.H.; Bjerkehagen, B.; Ohnstad, H.O.; Namløs, H.M.; Wang, J.; Kristiansen, B.E.; Myklebost, O. Array Comparative Genomic Hybridization Reveals Distinct DNA Copy Number Differences between Gastrointestinal Stromal Tumors and Leiomyosarcomas. Cancer Res. 2006, 66, 8984–8993. [Google Scholar] [CrossRef] [Green Version]
  98. Dei Tos, A.P.; Maestro, R.; Doglioni, C.; Piccinin, S.; Libera, D.D.; Boiocchi, M.; Fletcher, C.D. Tumor suppressor genes and related molecules in leiomyosarcoma. Am. J. Pathol. 1996, 148, 1037–1045. [Google Scholar]
  99. Yang, J.; Du, X.; Chen, K.; Ylipää, A.; Lazar, A.J.; Trent, J.; Lev, D.; Pollock, R.; Hao, X.; Hunt, K.; et al. Genetic aberrations in soft tissue leiomyosarcoma. Cancer Lett. 2009, 275, 1–8. [Google Scholar] [CrossRef] [Green Version]
  100. Coley, H.M.; Shotton, C.F.; Thomas, H. Seliciclib (CYC202; r-roscovitine) in combination with cytotoxic agents in human uterine sarcoma cell lines. Anticancer Res. 2007, 27, 273–278. [Google Scholar]
  101. Coley, H.M.; Shotton, C.F.; Kokkinos, M.I.; Thomas, H. The effects of the CDK inhibitor seliciclib alone or in combination with cisplatin in human uterine sarcoma cell lines. Gynecol. Oncol. 2007, 105, 462–469. [Google Scholar] [CrossRef] [PubMed]
  102. Riva, P.; Dalprà, L.; Gualandri, V.; Volontè, M.; Miozzo, M.; Malgara, R.; Conti, A.F.; Larizza, L. 19p deletion in recurring leiomyosarcoma lesions from the same patient. Cancer Genet. Cytogenet. 2000, 119, 102–108. [Google Scholar] [CrossRef]
  103. Knösel, T.; Altendorf-Hofmann, A.; Lindner, L.; Issels, R.; Hermeking, H.; Schuebbe, G.; Gibis, S.; Siemens, H.; Kampmann, E.; Kirchner, T. Loss of p16(INK4a) is associated with reduced patient survival in soft tissue tumours, and indicates a senescence barrier. J. Clin. Pathol. 2014, 67, 592–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kawaguchi, K.-I.; Oda, Y.; Saito, T.; Yamamoto, H.; Tamiya, S.; Takahira, T.; Miyajima, K.; Iwamoto, Y.; Tsuneyoshi, M. Mechanisms of inactivation of thep16INK4a gene in leiomyosarcoma of soft tissue: Decreased p16 expression correlates with promoter methylation and poor prognosis. J. Pathol. 2003, 201, 487–495. [Google Scholar] [CrossRef] [PubMed]
  105. Carvalho, S.D.; Pissaloux, D.; Crombé, A.; Coindre, J.-M.; Le Loarer, F. Pleomorphic Sarcomas. Surg. Pathol. Clin. 2019, 12, 63–105. [Google Scholar] [CrossRef] [PubMed]
  106. Goertz, O.; Pieper, A.; von der Lohe, L.; Stricker, I.; Dadras, M.; Behr, B.; Lehnhardt, M.; Harati, K. The Impact of Surgical Margins and Adjuvant Radiotherapy in Patients with Undifferentiated Pleomorphic Sarcomas of the Extremities: A Single-Institutional Analysis of 192 Patients. Cancers 2020, 12, 362. [Google Scholar] [CrossRef] [Green Version]
  107. Taylor, B.S.; Barretina, J.; Maki, R.G.; Antonescu, C.R.; Singer, S.; Ladanyi, M. Advances in sarcoma genomics and new therapeutic targets. Nat. Cancer 2011, 11, 541–557. [Google Scholar] [CrossRef] [Green Version]
  108. Guillou, L.; Aurias, A. Soft tissue sarcomas with complex genomic profiles. Virchows Arch. 2009, 456, 201–217. [Google Scholar] [CrossRef] [Green Version]
  109. Reid, A.H.; Tsai, M.M.; Venzon, D.J.; Wright, C.F.; Lack, E.E.; Oʼleary, T.J. MDM2 Amplification, P53 Mutation, and Accumulation of the P53 Gene Product in Malignant Fibrous Histiocytoma. Diagn. Mol. Pathol. 1996, 5, 65–73. [Google Scholar] [CrossRef]
  110. Bieging, K.T.; Mello, S.S.; Attardi, L.D. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 2014, 14, 359–370. [Google Scholar] [CrossRef] [Green Version]
  111. Pérot, G.; Chibon, F.; Montero, A.; Lagarde, P.; de Thé, H.; Terrier, P.; Guillou, L.; Ranchère, D.; Coindre, J.-M.; Aurias, A. Constant p53 Pathway Inactivation in a Large Series of Soft Tissue Sarcomas with Complex Genetics. Am. J. Pathol. 2010, 177, 2080–2090. [Google Scholar] [CrossRef] [PubMed]
  112. Li, G.Z.; Okada, T.; Kim, Y.-M.; Agaram, N.P.; Sanchez-Vega, F.; Shen, Y.; Tsubokawa, N.; Rios, J.; Martin, A.S.; Dickson, M.A.; et al. Rb and p53-Deficient Myxofibrosarcoma and Undifferentiated Pleomorphic Sarcoma Require Skp2 for Survival. Cancer Res. 2020, 80, 2461–2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Dodd, L.G.; Ware, P.; Duncan, D.; Hertel, J. Doing more with less: New markers for sarcoma diagnosis and their applicability to cytology specimens. Diagn. Cytopathol. 2016, 44, 351–360. [Google Scholar] [CrossRef]
  114. Dodd, R.D. Emerging targets in sarcoma: Rising to the challenge of RAS signaling in undifferentiated pleomorphic sarcoma. Cancer 2015, 122, 17–19. [Google Scholar] [CrossRef]
  115. Dodd, R.D.; Mito, J.K.; Eward, W.C.; Chitalia, R.; Sachdeva, M.; Ma, Y.; Barretina, J.; Dodd, L.; Kirsch, D.G. NF1 Deletion Generates Multiple Subtypes of Soft-Tissue Sarcoma That Respond to MEK Inhibition. Mol. Cancer Ther. 2013, 12, 1906–1917. [Google Scholar] [CrossRef] [Green Version]
  116. van der Horst, C.A.J.; Bongers, S.L.M.; Versleijen-Jonkers, Y.M.H.; Ho, V.K.Y.; Braam, P.M.; Flucke, U.E.; de Wilt, J.H.W.; Desar, I.M.E. Overall Survival of Patients with Myxofibrosarcomas: An Epidemiological Study. Cancers 2022, 14, 1102. [Google Scholar] [CrossRef] [PubMed]
  117. Cao, S.; Li, J.; Zhang, J.; Li, H. Development and validation of a prognostic nomogram for predicting the overall survival of myxofibrosarcoma patients: A large population-based study. Transl. Cancer Res. 2021, 10, 923–937. [Google Scholar] [CrossRef]
  118. Radaelli, S.; Pasquali, S.; Colombo, C.; Callegaro, D.; Sanfilippo, R.; Stacchiotti, S.; Provenzano, S.; Sangalli, C.; Morosi, C.; Barisella, M.; et al. Treatment strategies and outcomes of primary Myxofibrosarcomas in a large patients cohort. Eur. J. Surg. Oncol. 2022. [Google Scholar] [CrossRef]
  119. Ogura, K.; Hosoda, F.; Arai, Y.; Nakamura, H.; Hama, N.; Totoki, Y.; Yoshida, A.; Nagai, M.; Kato, M.; Arakawa, E.; et al. Integrated genetic and epigenetic analysis of myxofibrosarcoma. Nat. Commun. 2018, 9, 2765. [Google Scholar] [CrossRef] [Green Version]
  120. Ratner, N.; Miller, S.J. A RASopathy gene commonly mutated in cancer: The neurofibromatosis type 1 tumour suppressor. Nat. Cancer 2015, 15, 290–301. [Google Scholar] [CrossRef]
  121. Tsai, J.-W.; Li, C.-F.; Kao, Y.-C.; Wang, J.-W.; Fang, F.-M.; Wang, Y.-H.; Wu, W.-R.; Wu, L.-C.; Hsing, C.-H.; Li, S.-H.; et al. Recurrent Amplification at 7q21.2 Targets CDK6 Gene in Primary Myxofibrosarcomas and Identifies CDK6 Overexpression as an Independent Adverse Prognosticator. Ann. Surg. Oncol. 2012, 19, 2716–2725. [Google Scholar] [CrossRef] [PubMed]
  122. Li, C.-F.; Fang, F.-M.; Lan, J.; Wang, J.-W.; Kung, H.-J.; Chen, L.-T.; Chen, T.-J.; Li, S.-H.; Wang, Y.-H.; Tai, H.-C.; et al. AMACR Amplification in Myxofibrosarcomas: A Mechanism of Overexpression That Promotes Cell Proliferation with Therapeutic Relevance. Clin. Cancer Res. 2014, 20, 6141–6152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Li, C.-F.; Huang, H.-Y.; Wu, W.-R.; Liang, S.-S.; Chen, Y.-L.; Chen, L.-R.; Peng, Y.-T.; Lee, H.-C.; Shiue, Y.-L. Clinical Aggressiveness of Myxofibrosarcomas Associates with Down-Regulation of p12CDK2AP1: Prognostic Implication of a Putative Tumor Suppressor that Induces Cell Cycle Arrest and Apoptosis Via Mitochondrial Pathway. Ann. Surg. Oncol. 2014, 21, 711–720. [Google Scholar] [CrossRef] [PubMed]
  124. Kohlmeyer, J.L.; Gordon, D.J.; Tanas, M.R.; Dodd, R.D.; Monga, V.; Darbro, B.W.; Quelle, D.E. Combination therapies for MPNSTs targeting RABL6A-RB1 signaling. Oncotarget 2021, 12, 10–14. [Google Scholar] [CrossRef] [PubMed]
  125. Magallón-Lorenz, M.; Fernández-Rodríguez, J.; Terribas, E.; Creus-Batchiller, E.; Romagosa, C.; Estival, A.; Sidelnikova, D.P.; Salvador, H.; Villanueva, A.; Blanco, I.; et al. Chromosomal translocations inactivating CDKN2A support a single path for malignant peripheral nerve sheath tumor initiation. Qual. Life Res. 2021, 140, 1241–1252. [Google Scholar] [CrossRef]
  126. Kohlmeyer, J.L.; Kaemmer, C.A.; Pulliam, C.; Maharjan, C.K.; Samayoa, A.M.; Major, H.J.; Cornick, K.E.; Knepper-Adrian, V.; Khanna, R.; Sieren, J.C.; et al. RABL6A Is an Essential Driver of MPNSTs that Negatively Regulates the RB1 Pathway and Sensitizes Tumor Cells to CDK4/6 Inhibitors. Clin. Cancer Res. 2020, 26, 2997–3011. [Google Scholar] [CrossRef] [Green Version]
  127. Berner, J.; Sørlie, T.; Mertens, F.; Henriksen, J.; Sæter, G.; Mandahl, N.; Brøgger, A.; Myklebost, O.; Lothe, R.A. Chromosome band 9p21 is frequently altered in malignant peripheral nerve sheath tumors: Studies of CDKN2A and other genes of the pRB pathway. Genes, Chromosom. Cancer 1999, 26, 151–160. [Google Scholar] [CrossRef]
  128. Bhalla, A.D.; Landers, S.M.; Singh, A.K.; Landry, J.P.; Yeagley, M.G.; Myerson, G.S.B.; Delgado-Baez, C.B.; Dunnand, S.; Nguyen, T.; Ma, X.; et al. Experimental models of undifferentiated pleomorphic sarcoma and malignant peripheral nerve sheath tumor. Lab. Investig. 2022, 102, 658–666. [Google Scholar] [CrossRef]
  129. Kohlmeyer, J.; Kaemmer, C.; Umesalma, S.; Gourronc, F.; Klingelhutz, A.; Quelle, D. RABL6A Regulates Schwann Cell Senescence in an RB1-Dependent Manner. Int. J. Mol. Sci. 2021, 22, 5367. [Google Scholar] [CrossRef]
  130. Miettinen, M.M.; Antonescu, C.R.; Fletcher, C.D.; Kim, A.; Lazar, A.; Quezado, M.M.; Reilly, K.M.; Stemmer-Rachamimov, A.; Stewart, D.R.; Viskochil, D.; et al. Histopathologic evaluation of atypical neurofibromatous tumors and their transformation into malignant peripheral nerve sheath tumor in patients with neurofibromatosis 1—A consensus overview. Hum. Pathol. 2017, 67, 1–10. [Google Scholar] [CrossRef]
  131. Korfhage, J.; Lombard, D.B. Malignant Peripheral Nerve Sheath Tumors: From Epigenome to Bedside. Mol. Cancer Res. 2019, 17, 1417–1428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Herz, H.-M.; Shilatifard, A. The JARID2–PRC2 duality: Figure 1. Genes Dev. 2010, 24, 857–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Lee, W.; Teckie, S.; Wiesner, T.; Ran, L.; Granada, C.N.P.; Lin, M.; Zhu, S.; Cao, Z.; Liang, Y.; Sboner, A.; et al. PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors. Nat. Genet. 2014, 46, 1227–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Cichowski, K.; Shih, T.S.; Schmitt, E.; Santiago, S.; Reilly, K.; McLaughlin, M.E.; Bronson, R.T.; Jacks, T. Mouse Models of Tumor Development in Neurofibromatosis Type 1. Science 1999, 286, 2172–2176. [Google Scholar] [CrossRef]
  135. Keng, V.W.; Rahrmann, E.P.; Watson, A.L.; Tschida, B.R.; Moertel, C.L.; Jessen, W.J.; Rizvi, T.A.; Collins, M.H.; Ratner, N.; Largaespada, D.A. PTEN and NF1 Inactivation in Schwann Cells Produces a Severe Phenotype in the Peripheral Nervous System That Promotes the Development and Malignant Progression of Peripheral Nerve Sheath Tumors. Cancer Res. 2012, 72, 3405–3413. [Google Scholar] [CrossRef] [Green Version]
  136. Vogel, K.S.; Klesse, L.J.; Velasco-Miguel, S.; Meyers, K.; Rushing, E.J.; Parada, L.F. Mouse Tumor Model for Neurofibromatosis Type 1. Science 1999, 286, 2176–2179. [Google Scholar] [CrossRef]
  137. Kim, W.Y.; Sharpless, N.E. The Regulation of INK4/ARF in Cancer and Aging. Cell 2006, 127, 265–275. [Google Scholar] [CrossRef] [Green Version]
  138. Kim, A.; Stewart, D.R.; Reilly, K.M.; Viskochil, D.; Miettinen, M.M.; Widemann, B.C. Malignant Peripheral Nerve Sheath Tumors State of the Science: Leveraging Clinical and Biological Insights into Effective Therapies. Sarcoma 2017, 2017, 7429697. [Google Scholar] [CrossRef]
  139. Chaney, K.E.; Perrino, M.R.; Kershner, L.J.; Patel, A.V.; Wu, J.; Choi, K.; Rizvi, T.A.; Dombi, E.; Szabo, S.; Largaespada, D.A.; et al. Cdkn2a Loss in a Model of Neurofibroma Demonstrates Stepwise Tumor Progression to Atypical Neurofibroma and MPNST. Cancer Res. 2020, 80, 4720–4730. [Google Scholar] [CrossRef]
  140. Rhodes, S.D.; He, Y.; Smith, A.; Jiang, L.; Lu, Q.; Mund, J.; Li, X.; Bessler, W.; Qian, S.; Dyer, W.; et al. Cdkn2a (Arf) loss drives NF1-associated atypical neurofibroma and malignant transformation. Hum. Mol. Genet. 2019, 28, 2752–2762. [Google Scholar] [CrossRef]
  141. Pemov, A.; Hansen, N.F.; Sindiri, S.; Patidar, R.; Higham, C.S.; Dombi, E.; Miettinen, M.M.; Fetsch, P.; Brems, H.; Chandrasekharappa, S.C.; et al. Low mutation burden and frequent loss of CDKN2A/B and SMARCA2, but not PRC2, define premalignant neurofibromatosis type 1–associated atypical neurofibromas. Neuro-Oncology 2019, 21, 981–992. [Google Scholar] [CrossRef] [PubMed]
  142. Pemov, A.; Li, H.; Presley, W.; Wallace, M.R.; Miller, D.T. Genetics of human malignant peripheral nerve sheath tumors. Neuro-Oncol. Adv. 2019, 2, i50–i61. [Google Scholar] [CrossRef] [PubMed]
  143. Röhrich, M.; Koelsche, C.; Schrimpf, D.; Capper, D.; Sahm, F.; Kratz, A.; Reuss, J.; Hovestadt, V.; Jones, D.T.W.; Bewerunge-Hudler, M.; et al. Methylation-based classification of benign and malignant peripheral nerve sheath tumors. Acta Neuropathol. 2016, 131, 877–887. [Google Scholar] [CrossRef] [PubMed]
  144. Brohl, A.S.; Kahen, E.; Yoder, S.J.; Teer, J.K.; Reed, D. The genomic landscape of malignant peripheral nerve sheath tumors: Diverse drivers of Ras pathway activation. Sci. Rep. 2017, 7, 14992. [Google Scholar] [CrossRef] [Green Version]
  145. Zhang, X.; Hagen, J.; Muniz, V.P.; Smith, T.; Coombs, G.S.; Eischen, C.M.; Mackie, D.I.; Roman, D.L.; Van Rheeden, R.; Darbro, B.; et al. RABL6A, a Novel RAB-Like Protein, Controls Centrosome Amplification and Chromosome Instability in Primary Fibroblasts. PLoS ONE 2013, 8, e80228. [Google Scholar] [CrossRef] [Green Version]
  146. Muniz, V.P.; Askeland, R.W.; Zhang, X.; Reed, S.M.; Tompkins, V.S.; Hagen, J.; McDowell, B.D.; Button, A.; Smith, B.J.; Weydert, J.A.; et al. RABL6A Promotes Oxaliplatin Resistance in Tumor Cells and Is a New Marker of Survival for Resected Pancreatic Ductal Adenocarcinoma Patients. Genes Cancer 2013, 4, 273–284. [Google Scholar] [CrossRef] [Green Version]
  147. Maharjan, C.K.; Umesalma, S.; Kaemmer, C.A.; Muniz, V.P.; Bauchle, C.; Mott, S.L.; Zamba, K.D.; Breheny, P.; Leidinger, M.R.; Darbro, B.W.; et al. RABL6A Promotes Pancreatic Neuroendocrine Tumor Angiogenesis and Progression In Vivo. Biomedicines 2021, 9, 633. [Google Scholar] [CrossRef]
  148. Umesalma, S.; Kaemmer, C.A.; Kohlmeyer, J.; Letney, B.L.; Schab, A.M.; Reilly, J.A.; Sheehy, R.; Hagen, J.; Tiwari, N.; Zhan, F.; et al. RABL6A inhibits tumor-suppressive PP2A/AKT signaling to drive pancreatic neuroendocrine tumor growth. J. Clin. Investig. 2019, 129, 1641–1653. [Google Scholar] [CrossRef]
  149. Lin, D.I.; Hemmerich, A.; Edgerly, C.; Duncan, D.; Severson, E.A.; Huang, R.S.; Ramkissoon, S.H.; Connor, Y.D.; Shea, M.; Hecht, J.L.; et al. Genomic profiling of BCOR-rearranged uterine sarcomas reveals novel gene fusion partners, frequent CDK4 amplification and CDKN2A loss. Gynecol. Oncol. 2020, 157, 357–366. [Google Scholar] [CrossRef]
  150. Baggott, J.E.; Vaughn, W.H.; Juliana, M.M.; Eto, I.; Krumdieck, C.L.; Grubbs, C.J. Effects of Folate Deficiency and Supplementation on Methylnitrosourea-Induced Rat Mammary Tumors. JNCI J. Natl. Cancer Inst. 1992, 84, 1740–1744. [Google Scholar] [CrossRef]
  151. Palumbo, A.; Lau, G.; Saraceni, M. Abemaciclib: The Newest CDK4/6 Inhibitor for the Treatment of Breast Cancer. Ann. Pharmacother. 2018, 53, 178–185. [Google Scholar] [CrossRef]
  152. Dhillon, S. Palbociclib: First Global Approval. Drugs 2015, 75, 543–551. [Google Scholar] [CrossRef] [PubMed]
  153. Burki, T.K. Ribociclib in HR-positive, HER2-negative breast cancer. Lancet Oncol. 2016, 17, e482. [Google Scholar] [CrossRef]
  154. Dhillon, S. Trilaciclib: First Approval. Drugs 2021, 81, 867–874. [Google Scholar] [CrossRef] [PubMed]
  155. Dickson, M.A.; Schwartz, G.K.; Keohan, M.L.; D’Angelo, S.P.; Gounder, M.M.; Chi, P.; Antonescu, C.R.; Landa, J.; Qin, L.-X.; Crago, A.M.; et al. Progression-Free Survival Among Patients With Well-Differentiated or Dedifferentiated Liposarcoma Treated With CDK4 Inhibitor Palbociclib. JAMA Oncol. 2016, 2, 937–940. [Google Scholar] [CrossRef] [Green Version]
  156. Dickson, M.A.; Tap, W.D.; Keohan, M.L.; D’Angelo, S.P.; Gounder, M.M.; Antonescu, C.R.; Landa, J.; Qin, L.-X.; Rathbone, D.D.; Condy, M.M.; et al. Phase II Trial of the CDK4 Inhibitor PD0332991 in Patients With Advanced CDK4-Amplified Well-Differentiated or Dedifferentiated Liposarcoma. J. Clin. Oncol. 2013, 31, 2024–2028. [Google Scholar] [CrossRef] [Green Version]
  157. Dickson, M.A. Molecular Pathways: CDK4 Inhibitors for Cancer Therapy. Clin. Cancer Res. 2014, 20, 3379–3383. [Google Scholar] [CrossRef] [Green Version]
  158. Hsu, J.Y.; Seligson, N.D.; Hays, J.L.; Miles, W.O.; Chen, J.L. Clinical Utility of CDK4/6 Inhibitors in Sarcoma: Successes and Future Challenges. JCO Precis. Oncol. 2022, 6, e2100211. [Google Scholar] [CrossRef]
  159. Penel, N.; Van Glabbeke, M.; Marreaud, S.; Ouali, M.; Blay, J.Y.; Hohenberger, P. Testing new regimens in patients with advanced soft tissue sarcoma: Analysis of publications from the last 10 years. Ann. Oncol. 2011, 22, 1266–1272. [Google Scholar] [CrossRef]
  160. Finn, R.S.; Crown, J.P.; Lang, I.; Boer, K.; Bondarenko, I.M.; Kulyk, S.O.; Ettl, J.; Patel, R.; Pinter, T.; Schmidt, M.; et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): A randomised phase 2 study. Lancet Oncol. 2015, 16, 25–35. [Google Scholar] [CrossRef]
  161. Mangini, N.S.; Wesolowski, R.; Ramaswamy, B.; Lustberg, M.B.; Berger, M.J. Palbociclib. Ann. Pharmacother. 2015, 49, 1252–1260. [Google Scholar] [CrossRef] [PubMed]
  162. Lu, J. Palbociclib: A first-in-class CDK4/CDK6 inhibitor for the treatment of hormone-receptor positive advanced breast cancer. J. Hematol. Oncol. 2015, 8, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Dickson, M.A.; Koff, A.; D’Angelo, S.P.; Gounder, M.M.; Keohan, M.L.; Kelly, C.M.; Chi, P.; Antonescu, C.R.; Landa, J.; Qin, L.-X.; et al. Phase 2 study of the CDK4 inhibitor abemaciclib in dedifferentiated liposarcoma. J. Clin. Oncol. 2019, 37, 11004. [Google Scholar] [CrossRef]
  164. Hientz, K.; Mohr, A.; Bhakta-Guha, D.; Efferth, T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2016, 8, 8921–8946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Knudsen, E.S.; Kumarasamy, V.; Nambiar, R.; Pearson, J.D.; Vail, P.; Rosenheck, H.; Wang, J.; Eng, K.; Bremner, R.; Schramek, D.; et al. CDK/cyclin dependencies define extreme cancer cell-cycle heterogeneity and collateral vulnerabilities. Cell Rep. 2022, 38, 110448. [Google Scholar] [CrossRef] [PubMed]
  166. Chawla, S.P.; Blay, J.Y.; Italiano, A.; Gutierrez, M.; Le Cesne, A.; Gomez-Roca, C.A.; Gouw, L.G.; von Mehren, M.; Wagner, A.; Maki, R.G.; et al. Phase Ib study of RG7112 with doxorubicin (D) in advanced soft tissue sarcoma (ASTS). J. Clin. Oncol. 2013, 10, 10514. [Google Scholar] [CrossRef]
  167. Elvin, J.A.; Gay, L.M.; Ort, R.; Shuluk, J.; Long, J.; Shelley, L.; Lee, R.; Chalmers, Z.R.; Frampton, G.M.; Ali, S.M.; et al. Clinical Benefit in Response to Palbociclib Treatment in Refractory Uterine Leiomyosarcomas with a Common CDKN2A Alteration. Oncol. 2017, 22, 416–421. [Google Scholar] [CrossRef] [Green Version]
  168. Boddu, S.; Walko, C.M.; Bienasz, S.; Bui, M.M.; Henderson-Jackson, E.; Naghavi, A.O.; Mullinax, J.E.; Joyce, D.M.; Binitie, O.; Letson, G.D.; et al. Clinical Utility of Genomic Profiling in the Treatment of Advanced Sarcomas: A Single-Center Experience. JCO Precis. Oncol. 2018, 2, 1–8. [Google Scholar] [CrossRef]
  169. Peguero, J.; Sohal, D.P.S.; O’Neil, B.H.; Safran, H.; Kelly, K.; Grilley-Olson, J.E.; Subbiah, V.; Nadauld, L.; Purkayastha, D.; Stealey, E.; et al. Tissue/Site-Agnostic Study of Ribociclib for Tumors With Cyclin D–CDK4/6 Pathway Genomic Alterations: A Phase II, Open-Label, Single-Arm Basket Study. JCO Precis. Oncol. 2019, 3, 1–10. [Google Scholar] [CrossRef]
  170. Morris, D.G.; Bramwell, V.H.C.; Turcotte, R.; Figueredo, A.T.; Blackstein, M.E.; Verma, S.; Matthews, S.; Eisenhauer, E.A. A Phase II Study of Flavopiridol in Patients With Previously Untreated Advanced Soft Tissue Sarcoma. Sarcoma 2006, 2006, 64374. [Google Scholar] [CrossRef]
  171. Carnero, A. Targeting the cell cycle for cancer therapy. Br. J. Cancer 2002, 87, 129–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Tramontana, T.F.; Marshall, M.S.; Helvie, A.E.; Schmitt, M.R.; Ivanovich, J.; Carter, J.L.; Renbarger, J.L.; Ferguson, M.J. Sustained Complete Response to Palbociclib in a Refractory Pediatric Sarcoma With BCOR-CCNB3 Fusion and Germline CDKN2B Variant. JCO Precis. Oncol. 2020, 4, 466–471. [Google Scholar] [CrossRef] [PubMed]
  173. Andrikopoulou, A.; Shalit, A.; Zografos, E.; Koutsoukos, K.; Korakiti, A.-M.; Liontos, M.; Dimopoulos, M.-A.; Zagouri, F. MicroRNAs as Potential Predictors of Response to CDK4/6 Inhibitor Treatment. Cancers 2021, 13, 4114. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cell cycle and a simplified illustration of interactions with selected kinases and cyclins. G1 is the cell cycle phase; in this phase, the cell increases in volume, but mitosis has not yet taken place. It is the first part of the interphase and transition into the S phase, the cell cycle’s replication phase. DNA replication takes place in this phase. The S phase (2nd phase) is usually constant in time (about 7 to 8 h) and lies between the G1 and G2 phases. Third, the G2 phase of the cell cycle is the second part of the interphase. It follows the S phase and enters the prophase of mitosis, in which the cell’s chromatin condenses into chromosomes. The following M phase describes the division of the cell. The nucleus splits (mitosis), and the entire cell divides (cytokinesis). Finally, the facultative G0 phase is the cell-cycle stage in which dormant or differentiated cells (e.g., nerve and muscle cells) are found [18].
Figure 1. Cell cycle and a simplified illustration of interactions with selected kinases and cyclins. G1 is the cell cycle phase; in this phase, the cell increases in volume, but mitosis has not yet taken place. It is the first part of the interphase and transition into the S phase, the cell cycle’s replication phase. DNA replication takes place in this phase. The S phase (2nd phase) is usually constant in time (about 7 to 8 h) and lies between the G1 and G2 phases. Third, the G2 phase of the cell cycle is the second part of the interphase. It follows the S phase and enters the prophase of mitosis, in which the cell’s chromatin condenses into chromosomes. The following M phase describes the division of the cell. The nucleus splits (mitosis), and the entire cell divides (cytokinesis). Finally, the facultative G0 phase is the cell-cycle stage in which dormant or differentiated cells (e.g., nerve and muscle cells) are found [18].
Cancers 14 03380 g001
Table 1. Registered ongoing and completed clinical trials with CDK-targeted therapy in STSs (www.clinicaltrials.gov (accessed on 30 June 2022)).
Table 1. Registered ongoing and completed clinical trials with CDK-targeted therapy in STSs (www.clinicaltrials.gov (accessed on 30 June 2022)).
StatusStudy TypeSTS TypeCDK TargetDrug(s)Estimated EnrollmentsIdentifier
RecruitingSingle-arm, single-institution, open-label, prospective phase II trialLPSCDK4/6Ribociclib30 participantsNCT03096912
UnknownSingle-arm, single-institution, open-label, prospective phase II trialAll STSs, LPS excludedCDK4/6Ribociclib45 participantsNCT04040205
RecruitingPhase I/II studyKaposi sarcomaCDK4/6Abemaciclib43 participantsNCT04941274
UnknownSingle-arm, single-institution, open-label, prospective phase II trialLPSCDK4/6Ribociclib30 participantsNCT02571829
RecruitingPhase III studyAll STSs and othersCDK4/6 with multiple othersNilotinib, ceritinib, capmatinib, Palbociclib (and 8 more.)960 participantsNCT03784014
RecruitingPhase II studyLPSCDK4/6 + anti-PD1Palbociclib, INCMGA0001242 participantsNCT04438824
RecruitingMulticenter, open-label, dose-escalation phase I trialMultiple sarcoma subtypesCDK9PRT252730 participantsNCT05159518
RecruitingPhase I, open-label, multicenter, nonrandomized, multiple-dose, safety, tolerability, pharmaco-kinetic, and pharmaco-dynamic studyLPSCDK4 + chemotherapyPF-07220060, letrozole, fulvestrant118 participantsNCT04557449
CompletedPhase Ib/II, open-label, multicenter studyLPSCDK4/6 + MDM2Siremadlin, ribociclib74 participantsNCT02343172
RecruitingNon-randomized, phase I/II studyAll STSs and othersCDK2 + chemotherapyBLU-222, carboplatin, ribociclib, fulvestrant366 participantsNCT05252416
RecruitingPhase Ib dose-escalation studyAll STSs and othersCDK4/6 + chemotherapyAbemaciclib, irinotecan, temozo-lomide60 participantsNCT04238819
RecruitingNon-randomized, open, two-cohort, phase II, multicenter national clinical trial. Twenty sites in Spain.STSs and othersCDK4/6Palbociclib40 participantsNCT03242382
RecruitingPhase I, open-label, dose-escalation, safety, pharmacokinetic, and pharmacodynamic studySTSsCDK9TP-128770 participantsNCT03604783
Active, not recruitungTwo-center, two-arm, phase II studyLPSCDK4/6Ribociclib, Everolimus50 participantsNCT03114527
CompletedPhase II studyLPSCDK4/6Palbociclib90 participantsNCT01209598
Active, not recruitungPhase II studyDDLPSCDK4/6Abemaciclib33 participantsNCT02846987
RecruitingPhase III, multicenter,
randomized double-blind study
DDLPSCDK4/6Abemaciclib vs. placebo108 participantsNCT04967521
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thiel, J.T.; Daigeler, A.; Kolbenschlag, J.; Rachunek, K.; Hoffmann, S. The Role of CDK Pathway Dysregulation and Its Therapeutic Potential in Soft Tissue Sarcoma. Cancers 2022, 14, 3380. https://doi.org/10.3390/cancers14143380

AMA Style

Thiel JT, Daigeler A, Kolbenschlag J, Rachunek K, Hoffmann S. The Role of CDK Pathway Dysregulation and Its Therapeutic Potential in Soft Tissue Sarcoma. Cancers. 2022; 14(14):3380. https://doi.org/10.3390/cancers14143380

Chicago/Turabian Style

Thiel, Johannes Tobias, Adrien Daigeler, Jonas Kolbenschlag, Katarzyna Rachunek, and Sebastian Hoffmann. 2022. "The Role of CDK Pathway Dysregulation and Its Therapeutic Potential in Soft Tissue Sarcoma" Cancers 14, no. 14: 3380. https://doi.org/10.3390/cancers14143380

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