Next Article in Journal
High-Throughput CRISPR Screening in Hematological Neoplasms
Previous Article in Journal
Vessel-Sparing Lymphadenectomy Should Be Performed in Small Intestine Neuroendocrine Neoplasms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Precision Medicine Highlights Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric, Adolescents and Young Adult Sarcomas

by
Farinaz Barghi
1,2,
Harlan E. Shannon
2,
M. Reza Saadatzadeh
2,3,
Barbara J. Bailey
2,
Niknam Riyahi
2,4,
Khadijeh Bijangi-Vishehsaraei
2,3,
Marissa Just
3,
Michael J. Ferguson
3,
Pankita H. Pandya
2,3,* and
Karen E. Pollok
1,2,3,4,*
1
Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
2
Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
3
Department of Pediatrics, Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
4
Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(15), 3611; https://doi.org/10.3390/cancers14153611
Submission received: 6 June 2022 / Revised: 19 July 2022 / Accepted: 20 July 2022 / Published: 25 July 2022
(This article belongs to the Section Cancer Biomarkers)

Abstract

:

Simple Summary

This review provides an overview of clinical features and current therapies in children, adolescents, and young adults (AYA) with sarcoma. It highlights the basic and clinical findings on the cyclin-dependent kinases 4 and 6 (CDK4/6) cell cycle regulatory pathway in the context of the precision medicine-based molecular profiles of the three most common types of pediatric and AYA sarcomas—osteosarcoma (OS), rhabdomyosarcoma (RMS), and Ewing sarcoma (EWS).

Abstract

Despite improved therapeutic and clinical outcomes for patients with localized diseases, outcomes for pediatric and AYA sarcoma patients with high-grade or aggressive disease are still relatively poor. With advancements in next generation sequencing (NGS), precision medicine now provides a strategy to improve outcomes in patients with aggressive disease by identifying biomarkers of therapeutic sensitivity or resistance. The integration of NGS into clinical decision making not only increases the accuracy of diagnosis and prognosis, but also has the potential to identify effective and less toxic therapies for pediatric and AYA sarcomas. Genome and transcriptome profiling have detected dysregulation of the CDK4/6 cell cycle regulatory pathway in subpopulations of pediatric and AYA OS, RMS, and EWS. In these patients, the inhibition of CDK4/6 represents a promising precision medicine-guided therapy. There is a critical need, however, to identify novel and promising combination therapies to fight the development of resistance to CDK4/6 inhibition. In this review, we offer rationale and perspective on the promise and challenges of this therapeutic approach.

1. Introduction

Positive outcomes in the treatment of pediatric, as well as adolescent and young adult (AYA) cancers, have improved remarkably over the past several decades [1]. The first key to this success was the implementation of cooperative clinical research trial groups among academic centers that were focused on the treatment of pediatric and AYA cancers. This collaboration led to the generation of clinical data indicating patient response to treatment options from relatively large numbers of patients, which ultimately impacted therapeutic outcomes and increased long-term survival rates. Interdisciplinary multimodal therapy approaches such as polychemotherapy protocols and applying drug combinations sequentially was the second key to improving survival in pediatric patients [2]. This phenomenon is most evident in acute lymphoblastic leukemia (ALL), where survival in some subgroups of ALL is now up to 90% of patients [3]. Improvements in survival in solid tumors such as sarcoma have been slower, with the 5-year event-free survival (EFS) rate approaching 70%; however, 5-year survival for patients with metastatic disease still only remain between 20-30% [4]. This is compounded by the fact that sarcomas are quite rare and heterogeneous in children and AYA patients [5], making the testing of new therapies in adequately powered clinical trials extremely challenging [2]. Additionally, metastatic tumors harbor extremely complex genomic alterations—demonstrated by next-generation sequencing (NGS)—that can differ from the primary malignancy [6]. Moreover, these patients are at a very high risk of developing chronic health issues affecting long-term morbidity and mortality due to the intensity of front-line and salvage therapies [4]. Therefore, personalized treatment that focuses on specific actionable molecular alterations in sarcoma patients with high-risk signatures at diagnosis, or with metastatic and/or relapsed disease, have emerged as an opportunity to increase survival and improve quality of life [4]. While conventional chemotherapy can damage both cancer cells and normal tissues, treating sarcomas based on individual molecular profiles has the potential to specifically target the cancer cells and minimize the toxicity to normal tissues. Moreover, molecular signatures are increasingly used in diagnosis, as well as being used as biomarkers of therapeutic sensitivity and resistance. They also can be exploited to uncover additional intra-tumoral heterogeneity and, hence, redefine tumor subtype classifications [2]. By further refining classifications based on molecular signatures, tumor subtypes are being further subdivided into smaller categories, emphasizing the critical need to incorporate precision genomic approaches in guiding therapy selection [2].
Molecular analyses of pediatric and AYA sarcomas have led to the identification of cyclin-dependent kinases (CDK) CDK4 and CDK6 as a therapeutic targeting opportunity. Activation of the CDK4/6 pathway can be a powerful driver of sarcomagenesis [7]. It is a major cell cycle regulatory pathway in mammalian cells, regulating the progression of cells into the DNA synthesis (S phase) part of the cell cycle [7]. The objective of this review is two-fold. First, a brief overview of pediatric and AYA sarcoma clinical features and current therapies is provided. This is followed by an assessment of the basic and clinical findings that are already published in the literature on the CDK4/6 cell cycle regulatory pathway in the context of precision medicine-based molecular profiles in pediatric and AYA osteosarcoma (OS), Ewings Sarcoma (EWS), and rhabdomyosarcoma (RMS).

2. Pediatric and AYA Sarcomas

Sarcomas are rare among adult malignancies, representing only 8% of all the malignancies that are diagnosed in the United States each year [8,9,10]. Similarly, sarcoma is rare in children and adolescents, with OS, RMS, and EWS accounting for 1011 out of 16,850 diagnoses in this group in 2020 [10]. Though equally rare in childhood and adolescence, sarcomas are unfortunately the cause of 13% of cancer-related deaths in the US [4]. Additionally, it has been reported that approximately 1600 young adults are diagnosed with soft tissue sarcomas (STS) and bone sarcomas annually [11]. Sarcomas are of mesenchymal origin and can arise in bone or soft tissues. Among the bone sarcomas, OS and EWS are the most common types and represent 56% and 34% of bone sarcomas, respectively [1]. STS are further classified into rhabdomyosarcoma (RMS) and non-rhabdomyosarcoma [1]. Sarcomas are among the most “hard-to-cure malignancies” because of their aggressive biological behavior and occurrence at almost every anatomical site [9]. The epidemiology, clinical features, and cancer predisposition syndromes of the sarcomas have been reviewed elsewhere [11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39]. As mentioned above, this is also compounded by the rarity of these cancers, making it challenging to conduct clinical trials. Despite the administration of the optimized multimodal treatment, including the combination of surgery, chemotherapy, irradiation, immunotherapy, and/or targeted therapeutics, greater than 30% of sarcoma patients succumb to their disease [9]. For metastatic and relapsed sarcoma patients, the survival rates are abysmal, with less than 30% of patients alive several years after diagnosis [4]. Thus, the identification of actionable targets for these difficult-to-treat tumors is urgently needed. Technological advancements in NGS leading to precise molecular characterization of individual sarcomas has helped to identify and prioritize therapeutic targets and prognostic/predictive biomarkers for potential treatments [9]. The ultimate goal of precision medicine is to increase the validity of diagnosis and prognosis, as well as to identify the most effective and safe therapy for the treatment of pediatric and AYA sarcomas [2,9]. To achieve this goal, however, the molecular mechanisms that contribute to the development and pathogenesis of pediatric sarcomas require further investigation to identify targeted combination therapies and additional actionable therapeutic targets for future clinical use [4].

2.1. Osteosarcoma (OS)

Prognosis and the Standard of Care

OS is the most common primary bone cancer in children and AYA, with an overall incidence rate of 4.5 per million in patients under the age of 25 in the US [40]. Metastatic disease is found in up to 25% of OS patients, where the lungs are the most common site of disease spread [40]. The 5-year survival rates in localized and metastatic OS patients under the age of 25 are approximately 70% and 30%, respectively [13]. While metastasis is a major poor prognosis marker for OS, additional factors have been identified that contribute to poor prognosis in OS. These include large tumors (>10 cm), axial tumor locations, male gender, OS of the pelvis, and elevated alkaline phosphate levels at diagnosis [14,41]. The mutational landscape of OS is highly complex and differs remarkably between tumors. Unlike other pediatric sarcomas, OS lacks a specific chromosome translocation or recurrent driver mutations. Instead, it is characterized by a high level of chromosomal instabilities (CINs) such as copy number variations (CNVs) [42]. The genomic complexity of OS greatly complicates identifying therapeutic strategies for OS patients. Standard-of-care for OS patients is similar across most OS subtypes, which includes neoadjuvant chemotherapy, surgical resection of the primary tumor with wide margins, and adjuvant chemotherapy [12,14]. MAP (methotrexate, adriamycin, and cisplatin) chemotherapy is the most common initial treatment for OS [43]. Radiotherapy is not included in standard-of-care for OS patients, since these tumors are notoriously resistant to it [44]. The addition of chemotherapy regimens to surgical resection increased the 5-year overall survival from ~20% to up to 70% for patients with localized disease [14]. In the relapsed setting, there are no standardized second-line cytotoxic chemotherapies for OS. In terms of targeted agents, more investigations are needed to identify single and combinations of targeted agents for relapsed OS. The use of multi-receptor tyrosine kinase (RTK) inhibitors such as cabozantinib and regorafenib has recently exhibited some success in clinical trials [44,45].

2.2. Ewing Sarcoma (EWS)

Prognosis and the Standard of Care

EWS is the second most common primary bone malignancy in children and AYA patients and can be diagnosed as a primary soft tissue tumor with the incidence in children and adolescents of ~3 cases per 1,000,000 per year in the US. The anatomic site of tumor involvement, tumor size, and the effect of treatment on tumor necrosis are significant prognostic factors, but metastasis is the by far the most adverse prognostic factor in EWS patients [35]. In children and adolescents, patients with localized EWS have a 5-year overall survival of about 70%, but patients with metastases at diagnosis show a markedly lower overall survival (>30%) [33]. After both neoadjuvant and adjuvant chemotherapy, EWS 5-year overall survival may be lower in patients with pelvic tumors, large tumors (>8 cm), and incomplete tumor regression [46]. The main somatic driver mutation in EWS is the genetic translocation that fuses the EWSR1 gene on chromosome 22 with other genes, mainly the FLI1 gene, a member of the E26 transformation specific (ETS) gene family of transcription factors on chromosome 11 [47,48]. This fusion is found in 85–90% of EWS patients [49]. Therefore, the EWSR1/FLI1 fusion gene functions as the major oncogene by acting as an aberrant transcription factor in Ewing sarcoma [48]. For example, E2F3, a member of the E2F family of transcription factors, is directly activated by the EWSR1/FLI1 fusion protein. The EWSR1/FLI1-mediated activation of E2F3 leads to activation of the genes that are involved in cell cycle progression [50]. In 10% of EWS cases, the EWSR1 gene is also fused to the ERG gene, another member of the ETS gene family of transcription factors, on chromosome 22 which also results in an aberrant transcription factor [49]. Clinically, the suppression of fusion proteins has not been successfully achieved in EWS for more than 20 years; however, the promising inhibition of fusion proteins—especially EWS/FLI1—has been shown preclinically and is currently in clinical trials [51]. Standard-of-care for EWS patients involves multidisciplinary management with surgical resection; local radiation; chemotherapy with vincristine and doxorubicin; and cyclophosphamide with ifosfamide and etoposide [52].

2.3. Rhabdomyosarcoma (RMS)

Prognosis and the Standard of Care

RMS is the most common soft tissue sarcoma in children, with an overall incidence rate of approximately 4.5 patients per million in individuals younger than 20 years old in the US. Prognostic indicators in RMS, by which patient outcome is directly influenced, include patient age, histology subtype, tumor site, tumor size, and stage of the disease. It has been shown that patients younger than ten years who have localized tumors that measure <5 cm have a higher chance of survival [26,27,30]. Additionally, favorable prognosis is achieved when tumors occur at the following sites: head and neck, non-parameningeal orbital, paratesticular and genitourinary-non-bladder/prostate sites [27,30]. In RMS, 5-year overall survival is the worst for alveolar RMS (ARMS) patients, with approximately 25% and 30% 5-year survival, respectively. ERMS has the highest 5-year survival rate at approximately 70%. There are no significant differences in the survival between gender or race among all of the RMS subtypes [53].
The improvement in cure rates for localized RMS has increased from 25% to 70% and is largely due to multidisciplinary cooperative group trials and multimodal treatment protocols [30]. However, <30% of patients with the metastatic disease survive beyond 5 years, despite aggressive therapy [54]. RMS patients can also be classified genetically as fusion-positive or fusion-negative cases. Most ARMS cases (~80%) are fusion positive with chromosomal translocations of the PAX genes—PAX3 on chromosome 2 or PAX7 on chromosome 1, to FOXO1 on chromosome 13 [55]. By resulting in aberrant transcription factors, these gene fusions lead to poor patient prognosis, as they are correlated with tumor aggressiveness [54,55]. Since developing small molecule inhibitors that are selective for fusion transcription factors is challenging, indirect inhibition is more promising by targeting the genes that are controlled by the fusion proteins [56]. For example, the amplification of CDK4, MYCN, and MIR17HG genes has been found predominantly in PAX gene fusion-positive (PFP) tumors, which can be considered as therapeutic targets in PFP RMS patients [54]. A new gene fusion involving a FET-family gene with TFCP2 has also been recently identified in epithelioid- and spindle-cell RMS patients, revealing a new fusion-positive RMS subgroup of highly aggressive craniofacial intraosseous RMS [57,58]. In most intraosseous RMS cases, tumors usually destroy the bone cortex and invade the surrounding soft tissue. The most common bones affected are craniofacial bones [57]. Following a histological confirmation of RMS, almost all patients receive chemotherapy, including vincristine, actinomycin, and cyclophosphamide (VAC), before undergoing surgery and/or radiotherapy [59,60]. Neoadjuvant chemotherapy maximizes the chance of complete resection at surgery. However, complete resection is rarely possible, particularly in tumors with unfavorable sites, which highlights the importance of radiotherapy as standard of care in RMS [27]. Second-line treatment for relapsed RMS patients is not standardized but is based on institutional preference, which can include various combinations of cytotoxic drugs such as ifosfamide, doxorubicin, temsirolimus, and vincristine [60,61].

3. Precision Medicine and Targeted Therapies in Pediatric and AYA Sarcomas: Effects on Diagnosis and Prognosis

Overall, there is an unmet clinical need to develop more efficacious and well tolerated therapeutic strategies for all three common forms of sarcoma (OS, RMS, and EWS) in children and AYAs. The molecular profiling of tumor samples can be a valuable tool for identifying actionable targets and corresponding inhibitors that can be a component of the clinical treatment plan in pediatric and AYA sarcoma patients. The evolution of NGS approaches has revolutionized preclinical and clinical cancer research in recent years by providing a major step forward in advancing precision medicine in the clinic [62]. Actionable alterations that are potentially responsive to specific targeted therapies can be detected in patients with various types of tumors by using the most common NGS approaches, which include whole genome sequencing (WGS), whole exome sequencing (WES), cancer-specific gene panels, RNA-seq, and proteomics [63,64]. Identifying tumor-specific aberrations with the potential to offer rational personalized treatments has improved the precision medicine concept in the clinic [62]. This is exemplified by a growing interest in the establishment of multi-disciplinary precision medicine programs that are focused on pediatric cancers, including sarcomas [65,66,67]. However, precision-guided treatments are in their infancy, and much remains to be learned in this field regarding the translation of all knowledge to the clinic in pediatric and AYA sarcoma patients [4]. Recent studies have detected targetable alterations in sarcoma patients and have used the findings to guide treatment [9]. Dysregulation of the primary cell cycle regulators is one of the detected actionable alterations that are gained by multiple-precision genomics/transcriptomics programs in pediatric and AYA sarcoma patients, and they are discussed in detail in the next section of this article.

3.1. The CDK4/6 Cell Cycle Regulatory Pathway Dysregulation in Pediatric and AYA Sarcomas

Dysregulation of the Cyclin D–CDK4/6 axis through various mechanisms leads to uncontrolled cell proliferation, frequently shown in many types of cancer, which are thought to show greater dependency on CDK4/6 [68,69]. Cancer cells frequently bypass the RB-dependent restriction point in the G1 phase of the cell cycle, typically through alterations in cell cycle machinery genes that lead to constitutive Cyclin D–CDK4/6 activation, or through the loss of RB itself [70]. Loss of the p16INK4A function is also a common event leading to dysregulation of the Cyclin D–CDK4/6 axis in cancer cells, as it can predict strong CDK4/6 dependence in cancer cells [68]. Increased growth factor-mediated mitogenic signaling through key oncogenic pathways, such as hyperactive PI3K and MAPK pathways, can also assist cancer cells in bypassing the RB-dependent restriction point in the G1 phase of the cell cycle by promoting Cyclin D-CDK4/6 activity (Figure 1) [69]. Cyclin D–CDK4/6 axis dysregulation has been reported in a subset of pediatric and AYA sarcoma patients. Precision medicine programs indicating dysregulation of this major cell cycle regulatory pathway in pediatric and AYA sarcoma patients are discussed below.

3.1.1. Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric and AYA OS

Recent precision medicine trials (PMTs) using -omics approaches have demonstrated dysregulation of the CDK4/6 cell cycle regulatory pathway in OS patients (Table 1). The Individualized Therapy for Relapsed Malignancies in Childhood (INFORM) precision medicine study on children with high-risk relapsed/refractory malignancies, including OS, aimed to detect therapeutic targets individually. In this study, whole-exome, low-coverage whole-genome, and RNA sequencing, as well as methylation and expression microarray analyses, were used. The results showed that in two out of four OS patients ≤ 20 years of age, CDK4/6 pathway-associated therapeutic targets, including either G91A point mutation in CDKN2B (predicted but not proven to affect CDKN2B function) or CDK4 amplification, were detected. CDK4 inhibition was the matched targeted therapy for these genomic alterations in these patients [71]. The Precision in Pediatric Sequencing Program is a PMT in which the prospective clinical WES of patient-matched tumor-normal samples and the RNA-seq of tumors were conducted to identify potential targetable variants in patients who were initially diagnosed with pediatric malignancies, including OS, with the age range of 2 weeks to 26 years. In one out of the six OS patients who were enrolled in this study, CCNE1 over-expression was detected as a potential therapeutic biomarker for the use of a CDK4/6 inhibitor. RB splice mutation was also detected as a clinically impactful germline alteration in one out of the six OS patients in this study [72]. In the individualized cancer therapy (iCat) trial, the feasibility of identifying actionable alterations was assessed in patients aged ≤30 years with extracranial solid tumors, including OS patients. In this trial, mutations were detected by Sequenom assay or targeted NGS, and copy number was assessed by comparative genomic hybridization (aCGH). In addition, RNA seq was performed in the trial if sufficient tumor specimen was available. CCNE1 high copy number gain was identified as a potentially actionable in two out of the 11 OS patients who were enrolled in the trial [73]. Dinaciclib, which selectively inhibits cyclin-dependent kinases CDK1, CDK2, CDK5, and CDK9 [74], was recommended as a targeted agent to inhibit the CCNE1-mediated activation of CDK2 [73]. CCND1 high copy number gain was also detected in one of the OS patients. For this actionable alteration, CDK4/6 inhibitor ribociclib was recommended as the targeted therapy [73]. In the molecular biology tumor board (MBB) program at Institute Curie, tumor profiling was conducted using panel-based NGS and array-comparative genomic hybridization on patients with a poor prognosis or relapsed/refractory solid tumors, including OS. In this study, in one out of four OS patients under the age of 19, CDKN2A homozygous deletion was identified as potentially actionable alteration, for whom a CDK4/6 inhibitor was recommended [75]. In the TRICEPS study (the personalized targeted therapy in refractory or relapsed cancer in childhood study), molecular profiling of pediatric and adolescent patients at the age of ≤18 was achieved by the WES of a matched tumor to normal germline samples and RNA sequencing of the tumor. In two out of the seven OS who were patients enrolled in this study, CDKN2A exhibited alterations, including either CDKN2A copy loss or CDKN2A homozygous deletion; one OS patient had RB copy loss [76]. In a phase 1 clinical trial program at MD Anderson Cancer Center, genomic profiling results gained by NGS were analyzed on advanced sarcoma patients, including OS patients with the age range of 8–76 years. In one out of 11 OS patients, CDK4 amplification was one of the actionable alterations found [77]. Based on an extensive panel of NGS data that were obtained from 67 pediatric and adult OS patients enrolled in a PMT by Memorial Sloan-Kettering Cancer Center (MSKCC), CDK4 amplification in 13.4% of cases, and CDKN2A/B deletion/mutation in 26.9% of cases were identified as potentially actionable alterations for CDK4/6 inhibitors. No significant differences were identified between pediatric and adult OS patients in the frequency of the potentially actionable alterations [78]. In the Zero Childhood Cancer Program, 252 tumors from high-risk pediatric and AYA patients, including OS patients, were molecularly profiled using WGS and RNA-seq. Among the eight OS patients who were enrolled in this PMT, overexpression and CNV of CCNE1 were identified in one case. Moreover, in two OS patients, somatic CNV and SV were detected in RB [67]. CDK4/6 inhibition was among the recommended therapies in this study for these patients [67]. The St. Jude Children’s Research Hospital-Washington University Pediatric Cancer Genome Project characterized the genomic landscape of OS by performing WGS on 34 pediatric and AYA OS tumors and matched normal tissue samples. The data showed that RB underwent significant recurrent somatic alterations in 10 out of 34 cases. RB was mutated by both point mutations and structural variations (SVs) in these cases. In addition, in one patient, the SV of CCND3 was detected [79]. In another study, “CRB cancer des Hôpitaux de Toulouse; BB-0033-00014”, WGS was performed on tumor samples from seven high-grade OS patients who were aged ≤20 at diagnosis, who were matched with non-tumor tissues from the same patients. A stop-gain mutation in the RB gene was identified in one case. In this patient, a low number of secondary mutations in other genes were observed, compared to patients with the wild-type RB gene [80]. As part of another study, 59 tumor/normal pairs of pediatric OS samples were examined using WES, WGS, and RNA-seq. CDKN2A/B deletions were identified as potentially actionable alterations in 2 of 20 OS patients. These two patients’ tumor samples were RB-proficient, providing rationale for treatment with a CDK4/6 inhibitor [81]. In the Molecular Screening for Cancer Treatment Optimization (MOSCATO-01) trial on pediatric and AYA patients with recurrent or refractory solid tumors aged under 23 at diagnosis, four OS patients were among the patients who were enrolled in the trial. The tumor samples were analyzed by CGH array, NGS, WES, and RNA seq. RB deletion was detected in one OS patient [82].

3.1.2. Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric and AYA RMS

Recent PMTs have demonstrated dysregulation of the CDK4/6 cell cycle regulatory pathway in a population of RMS patients (Table 1). In the Zero Childhood Cancer Program, a PMT with 17 RMS patients (11 fusion-positive (FP) and six fusion-negative (FN) patients) out of 252 pediatric and AYA high-risk cancer patients; dysregulation of Cyclin D–CDK4/6 axis associated genes, including overexpression of CDK4, CDKN2A/B, CCND3 or CCNE1; and CNVs of CDK4, CDKN2A/B, CCND2, or CCND3 were identified as therapeutic response biomarkers for CDK4/6 inhibition in ~45% of FP patients and in ~30% of FN RMS patients. CDK4/6 inhibition was among the recommended therapies for cell-cycle associated dysregulation [67]. In the INFORM study, 33% (two out of six) of cases had either CDKN2A/B deletion or a CDK4 copy number gain in RMS patients who were 15 years or younger and diagnosed with either ERMS or ARMS. CDK4 inhibition as a matching targeted therapy was suggested for these patients [71]. For the iCat trial with patients aged 30 or younger who were diagnosed with extracranial solid tumors, including RMS tumors, CDKN2A/B deletion was observed as a potential biomarker of therapeutic response to CDK4/6 inhibitor therapy in one out of 11 ERMS patients. The CDK4/6 inhibitor, ribociclib, was recommended as targeted therapy for this patient [73]. In the MOSCATO-01 trial with 12 RMS patients, CDKN2A deletion was detected in three ERMS patients. CDK inhibition was recommended as a precision genomic guided therapy for the patients with this alteration [82]. ClinOmics Program, a clinical genomics study of children and AYAs with relapsed and refractory cancers aged 25 or younger, examined the feasibility of genome-guided precision therapy in the patients, including RMS, by performing a combination of WES, RNA-seq, and high-density single-nucleotide polymorphism array analysis of the tumor. In one out of three RMS patients who were enrolled in the study, somatic alterations of CDKN2A and RB were detected, where CDKN2A and RB underwent the loss of heterozygosity, leading to cell cycle dysregulation [83]. In the MBB trial, among four RMS patients aged under 19, CDK4 amplification was detected as a potentially actionable alteration in one patient who was diagnosed with metastatic ARMS. This patient received the CDK4/6 inhibitor palbociclib and showed partial remission. However, the therapy was stopped after two months because of disease progression, emphasizing the need for a combination therapy approach and raising the question of whether earlier intervention may be the optimal strategy in the future [75]. In the TRICEPS study, CDKN2A point mutation was detected in one out of four RMS patients at 18 years or younger who were enrolled in the study. RB point mutation and loss of heterozygosity were identified in another patient who was diagnosed with ARMS [76]. In a phase 1 clinical trial program at MD Anderson Cancer Center, CDK4 amplification was identified as an actionable gene alteration, defined as a gene alteration that could be targeted by CDK4/6 inhibitor palbociclib in 2 of 8 RMS cases. All the patients who were enrolled in this trial were diagnosed with advanced sarcoma (ages 8–76 years) [77]. A comparative genomic hybridization array analysis of the primary tumor from a 27-year-old patient with relapsed and chemotherapy-refractory ARMS identified the highest level of amplification in the 12q13–14 region—more specifically, CDK4 was highly amplified in this region. In this study, CDK4 inhibition was suggested as a potential therapeutic strategy for RMS [84]. In another study, array-CGH demonstrated copy number alteration of CDKN2A as a recurrent mutation in RMS patients, including pediatric and adult patients with FET-TFCP2 fusion. CDKN2A homozygous deletion was identified in 90% of these cases [57].

3.1.3. Dysregulation of the CDK4/CDK6 Cell Cycle Regulatory Pathway in Pediatric and AYA EWS

Recent PMTs have also demonstrated dysregulation of the CDK4/6 cell cycle regulatory pathway in EWS patients (Table 1). In the INFORM precision medicine study, in 45.5% of (5/11) EWS patients aged 28 or younger, one of the following genes associated with Cyclin D–CDK4/6 axis was dysregulated, including the amplification of CCND2, overexpression of CCND1, or CDKN2A/B deletion. In one patient, both the overexpression of CCND1 and CDKN2A/B deletion were identified. All these patients were EWS/ETS (either EWSR1/FLI1 or EWSR1/ERG) fusion positive. CDK inhibitors such as CDK4/6 inhibitors were suggested for these patients as precision genomics (PG)- directed therapy [71]. In the iCat trial with patients aged ≤30 who were diagnosed with extracranial solid tumors, including EWS tumors, either two copy losses of CDKN2A/B or a single copy number gain of CCND1 was identified in two out of 12 EWS patients. The iCat recommendation was CDK4/6 inhibitor ribociclib for these patients [73]. EWS patients were included in the ClinOmics Program for children and AYAs with relapsed and refractory cancers aged ≤25. In approximately 10% of EWS patients who were positive for EWSR1/FLI1 fusion, CDKN2A homozygous loss was identified as a potential biomarker of therapeutic response to CDK4/6 inhibitor therapy [83]. In the TRICEPS study, in one of two EWS patients aged ≤18, CDKN2A copy loss was identified as a potentially actionable somatic alteration. This patient was positive for EWSR1/FLI1 fusion [76]. In one of three EWSR1 fusion-positive EWS patients enrolled in the phase 1 clinical trial program at MD Anderson Cancer Center, CDK4 amplification was detected. CDK4 amplification was one of the most actionable alterations in this study, which was defined as a gene alteration that could be directly targeted by CDK4/6 inhibitor palbociclib. All patients with different advanced sarcomas who were enrolled in this study were 8–76 years old [77]. The Zero Childhood Cancer Program reported that data on 18 EWS patients (out of 252 pediatric and AYA high-risk cancer patients who were enrolled in the study) demonstrated the homozygous deletion of CDKN2A/B as a potential biomarker of therapeutic response to CDK4/6 inhibition in three of the EWS patients. One of these three patients was FP for EWSR1/FLI1. In this study, CDK4/6 inhibition was recommended as a targeted therapy for these patients with deletion of CDKN2A/B [67]. CDKN2A/B deletions were demonstrated in four out of five EWS patients aged under 23 who underwent copy-number analysis and were enrolled in the MOSCATO-01 trial. Two of these patients with CDKN2A/B deletions were EWS/FLI1 FP EWS patients. CDK4 inhibition therapy was recommended as targeted therapy for these patients [82]. In another PMT on pediatric and AYA EWS patients aged between 4 months and 21 years old, the molecular profiles of the tumors from the patients were obtained using WES, WGS, SNP array, and RNA-seq. EWS/ETS rearrangements—mostly EWSR1/FLI1—were identified in these patients. Focal deletions at chromosome 9p21.3, which was consistent with a loss of CDKN2A, occurred in 22% of the EWS tumors using a combination of SNP array and WES [85].
Altogether, these studies indicate that -omics analyses are critical in providing guidance on whether a CDK4/6 inhibitor should be considered as a potential targeted therapy in these three common types of pediatric and AYA sarcomas.

4. Inhibition of Cyclin-Dependent Kinases 4/6 (CDK4/6) as a Source of Therapeutic Targets in Pediatric and AYA Sarcomas Based on Precision Genomics/Transcriptomics Profiling

4.1. Cyclin D–CDK4/6 Axis Role in Cell Cycle Regulation

Dysregulation of the cell cycle is one of cancer’s hallmarks and provides numerous options for therapeutic intervention [68]. CDK4 and CDK6 are major cell cycle regulators in mammalian cells, where they induce the progression of cells into the DNA synthetic (S) phase of the cell cycle [70]. In the first gap phase (G1) of the cell cycle, the enzymatic activity of CDK4 and CDK6 are regulated positively by D-type cyclins (D1, D2, and D3), expressed in response to multiple extracellular signals such as stimulatory mitogens, cell–cell interaction, and differentiation inducers [86,87]. The three D-type cyclins are expressed, alone or together, in different cell lineages, which may be due to tissue-specific aspects of normal physiology underlying the differential expression of the D-type cyclins [86]. The cyclins form a holoenzyme complex, assembled with CDK4 or CDK6 (Cyclin D-CDK4/6 complex) [87]. The Cyclin D-CDK4/6 complex is not yet enzymatically fully activated, unless by CDK-activating kinase (CAK)-mediated activating phosphorylation of CDK4/6 [88]. CDK4 and CDK6 have mostly overlapping functions. Both can associate with all three D-type cyclins, as they share 71% amino acid identity [89]. However, it has been shown that CDK4 and CDK6 can differentially regulate cancer progression through various mechanisms in breast, prostate, and pancreatic cancer models, with the CDK4-dependent regulation of pro-metastatic inflammatory pathways, and the CDK6-dependent control of the genes that are involved in DNA replication and repair mechanisms. In these models, silencing CDK6 but not CDK4 led to the induction of DNA damage and defective DNA repair. In contrast, silencing CDK4 but not CDK6 enriched the TNF signaling pathway that is involved in the regulation of proinflammatory cytokines and chemokines [90]. It is currently not known if CDK4 and CDK6 differential regulation is operative in sarcomas, and it is an area that is worthy of investigation.
The Cyclin D-CDK4/6 complex regulates interactions between the RB protein and E2F transcription factors [69]. Among the seven E2F family members (E2F1-5, 2A7E, and E2F7), three of them, E2F1-3, are transcriptional activators [91]. Upon the direct interaction of E2F1-3 with RB, the transcriptional activity of E2Fs is inhibited, leading to a pause in cell cycle transition [91]. The expression level of E2F1-3 differs at various cell cycle phases; E2F3 expression is increased in early-G1 to mid-G1, while the increased expression of E2F1 and E2F2 occurs at the G1/S boundary [91]. E2F transcriptional activity is not only inhibited by the direct binding to RB, but also by the RB-mediated recruitment of histone deacetylase (HDAC) to the RB-E2F complex [92]. Initially, upon Cyclin D-CDK4/6-mediated RB phosphorylation, RB affinity to E2F and HDAC is reduced, promoting the transcription of genes such as Cyclin E2 [92]. Subsequently, the Cyclin E2-CDK2 complex hyperphosphorylates RB which induces full dissociation of the RB-E2F complex and the transcription of genes encoding proteins playing critical roles in the next phases of the cell cycle, such as Cyclin A2, Cyclin B1 and others involved in DNA replication and repair [92,93,94]. Cyclin E2-CDK2 exemplifies that the cell cycle is not only positively regulated by Cyclin D-CDK4/6, but also by other Cyclin-CDKs (Figure 1).
CDK4/6 can also regulate the cell cycle independently of RB. CDK4/6 initiates the phosphorylation of FOXM1 in G1, leading to the transcriptional activation of FOXM1, which is required for the expression of the genes that are involved in cell cycle progression [95]. CDK4 negatively regulates SMAD2/3, components of the TGFβ signaling cascade that exhibit anti-proliferative activity [96]. Moreover, the CDK4-mediated phosphorylation of SMAD3 leads to its inhibition and the release of G1 arrest. Therefore, diminishing SMAD3 activity by CDK4-mediated phosphorylation could contribute to tumorigenesis [97].
CDK/cyclin activity is negatively regulated by CDK inhibitors (CKIs) and is classified into two families of cell cycle inhibitors, including the CDK-interacting protein/kinase inhibitory protein (CIP/KIP) family comprising p21CIP1, p27KIP1 and p57KIP2, and the inhibitor of cyclin-dependent kinase 4 (INK4) family, including p16INK4A, p15INK4B, p18INK4C, and p19INK4D [98,99]. The INK4 family of proteins specifically inhibits Cyclin D-CDK4/6 activity by directly binding to CDK4/6 to inhibit the formation of CDK4/6-cyclin D1, D2, and D3 complexes [98]. In contrast to the INK family, the CIP/KIP proteins can bind to all CDKs driving the cell cycle, including CDK1, CDK2, and CDK4/6 [98]. For example, p21CIP1 (inhibitor of CDK1 and CDK2) can bind to Cyclin D-CDK4/6 complex and relieve CDK2, further increasing its binding to Cyclin E2 and cell cycle progression [100]. p16INK4A and p15INK4B, encoded by CDKN2A and CDKN2B, respectively, play significant roles as tumor suppressors in the INK4 family [101].
Dysregulation of the Cyclin D–CDK4/6 axis through various mechanisms, such as the gene amplification of positive regulators, or gene rearrangement; loss of negative regulators; epigenetic alterations; and point mutations in significant Cyclin D–CDK4/6 pathway components, leads to uncontrolled cell proliferation frequently shown in many types of cancer including sarcomas [69,102]. Therefore, because of the significant role of CDK4/6 activity in cancer cells, CDK4/6 inhibitors have emerged as well validated targeted therapy for cancer treatment [69]. As discussed later in this review, alterations leading to Cyclin D-CDK4/6 axis dysregulation could confer resistance or sensitivity to CDK4/6 inhibitors, an important point for the stratification of patients for CDK4/6 inhibitor therapy.

4.2. Selective CDK4/6 Inhibitors: Palbociclib, Abemaciclib, Ribociclib, Trilaciclib and Dalpiciclib

In addition to CDK4 and CDK6, other CDKs also play potential roles in cell cycle regulation. To date, 21 different CDKs have been identified in the human body that can interact with 29 different cyclins [103,104]. Therefore, the administration of pan-CDK inhibitors could cause significant toxicity, since multiple CDKs that are essential for the function of normal cells could also be inhibited [103,104]. The first-generation pan-CDK inhibitors such as flavopiridol and roscovitine lacked selectivity among the CDK family members [105]. The second generation including, dinaciclib, P276-00, AT7519, TG02, roniciclib, and RGB-286638 were developed based on the first-generation group but with increasing selectivity against CDK1 and CDK2 to reduce the off-target risks of CDK inhibitors [105,106]. These two generations of CDK inhibitors demonstrated limited efficacy and tolerable toxicity in clinical trials [86]. A third generation of CDK inhibitors with high selectivity for CDK4/6 was subsequently developed with the goal of reducing the risk of toxicity, while improving efficacy [107]. The high selectivity of CDK inhibitors to CDK4/6 is associated with reduced toxicity in normal cells. In addition, Santamaria et al. demonstrated that the genetic knockout of CDK4/6 in mice embryos did not interfere with normal mouse embryonic fibroblast cells and organogenesis, suggesting that the selective targeting of CDK4/6 should be tolerated. In these embryos, the compensatory effect of CDK1 was sufficient for the phosphorylation of RB and the expression of the genes that were regulated by E2F transcription factors [108]. To date, there are four U.S. Food and Drug Administration (FDA)-approved third-generation CDK4/6 inhibitors: palbociclib (Ibrance, PD0332991); ribociclib (Kisqali, LEE011); abemaciclib (Verzenio, LY2835219); and trilaciclib (Cosela, G1T28), all of which are employed in the clinic and have demonstrated some degree of clinical efficacy [106,109]. Palbociclib was the first orally bioavailable, highly selective CDK4/6 inhibitor and approved by the U.S. FDA in 2015 [110]. Subsequently, in 2017, ribociclib and abemaciclib were approved by the U.S. FDA [74,75]. Trilaciclib is the most recent CDK4/6 inhibitor to be approved by the U.S. FDA in 2021 [109]. As anticipated, the four selective CDK4/6 inhibitors suppress RB phosphorylation, thus causing G1 arrest [106,109]. There is also emerging evidence that CDK4/6 inhibition can block metabolic pathways, potentially leading to the control of tumor progression that is independent of RB status [111,112]. In addition, CDK4/6 inhibition is also very attractive, as it can influence the tumor microenvironment by impacting cell-mediated immunity and enhancing anti-tumor responses. For example, CDK4/6 inhibition increased IL-2 and CD4+ T-cell activation [113] and blocked the growth of immunosuppressive regulatory T cells [114]. A cellular assessment of the CDK4/6 inhibitors that measured biochemical potency and drug affinity demonstrated apparent differences among palbociclib, abemaciclib, and ribociclib. Palbociclib showed equivalent potency toward CDK4/Cyclin D3 and CDK6/Cyclin D1; however, both abemaciclib and ribociclib had significantly more CDK4/Cyclin D3 potency [115]. The biochemical profiling of trilaciclib demonstrated similar potency toward CDK4/Cyclin D1 and CDK6/Cyclin D3 [116]. FDA-approved palbociclib, abemaciclib, and ribociclib are currently used for the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-) advanced or metastatic breast cancer, in combination with endocrine therapy [107,117,118]. Among the three CDK4/6 inhibitors, abemaciclib is the only one that shows promising efficacy as a single agent to treat HR+ breast cancer [119]. In addition to clinical trials for breast cancer, the three CDK4/6 inhibitors underwent and are still undergoing testing in several clinical trials for other types of cancer, both as a single drug and in combination therapy [107]. Specifically, for pediatric sarcomas, there are several ongoing clinical trials to investigate the efficacy and safety of these CDK4/6 inhibitors (Table 2). Despite the similar mechanism of action for the first three CDK4/6 inhibitors, palbociclib, abemaciclib, and ribociclib, they exhibit different toxicity profiles and dose-delivery schedules. Since the half-lives of palbociclib and ribociclib are more than 24 h, they are dosed daily. Due to their myelosuppressive effects, daily dosing is non-continuous. Administration is for 21 days followed by a break of one week for neutrophil count recovery. In comparison, the half-life of abemaciclib is <24 h. It induces relatively less myelosuppression and is dosed twice daily [120] Palbociclib and ribociclib have minimal GI toxicities compared to abemaciclib, which has extensive GI toxicities [121]. However, abemaciclib-mediated bone marrow toxicity is less serious than palbociclib and ribociclib-mediated bone marrow toxicities [120]. The rate of hematological toxicity, mainly neutropenia, is higher for palbociclib and ribociclib [122]. Moreover, it has been shown that ribociclib can induce hepatic toxicity [123]. The most recently U.S. FDA-approved CDK4/6 inhibitor, trilaciclib, has been used to reduce the rate of chemotherapy-induced myelosuppression in patients with small-cell lung cancer. The administration of trilaciclib prior to chemotherapy could transiently induce G1 arrest of the cell cycle to prevent chemotherapy-induced myelosuppression [109]. Trilaciclib is well tolerated in the patients. Its administration before chemotherapy is not associated with a clinically relevant toxicity increase, and it significantly reduces chemotherapy-related toxicity [124]. To date, there are no clinical trials to investigate the efficacy and safety of trilaciclib in pediatric and AYA sarcomas. In addition to four U.S. FDA-approved CDK4/6 inhibitors, a novel CDK4/6 inhibitor, dalpiciclib (SHR6390), is approved by China’s National Medical Products Administration (NMPA) for the treatment of HR+ and HER2- recurrent or metastatic breast cancer with endocrine agents [125,126]. Dalpiciclib has a similar potency against CDK4 and CDK6 [126]. The oral administration of dalpiciclib every three weeks with a one-week break in patients with advanced breast cancer was well-tolerated [126].

4.2.1. Intrinsic and Acquired Mechanisms of Resistance to CDK4/6 Inhibitors

While the selective CDK4/6 inhibitors show promising targeted therapy for cancer treatment, the onset of therapeutic resistance to these agents through both intrinsic and acquired molecular mechanisms has been encountered in cancer treatment. The development of resistance to CDK4/6 inhibitors leads to their suboptimal cytostatic effects, highlighting a need for combination therapy to evoke the cell death response [127,128].

Markers of Intrinsic Resistance and Sensitivity to CDK4/6 Inhibitors

Intrinsic mechanisms of resistance and sensitivity to CDK4/6 inhibitors are associated with alterations in specific cell cycle genes, which can serve as biomarkers of therapeutic resistance or sensitivity. Lack of RB, the primary target of CDK4/6, is a crucial contributor to CDK4/6 inhibitors’ intrinsic resistance [129]. Tumor cells with loss, or a low expression of RB are generally more resistant to CDK4/6 inhibitors than RB-proficient cells [68]. As a result, the vast majority of studies on CDK4/6 inhibitors have focused on RB-proficient cancer cells. In addition, alterations in other genes such as CDK4, CCND (the gene that encodes Cyclin D), CDKN2A, E2F, and CDK2 can also contribute to CDK4/6 inhibitors’ intrinsic resistance [129]. The mentioned gene alterations are discussed in the specific studies below.
Konecny et al. studied the effect of palbociclib on 40 ovarian cancer cell lines and demonstrated that the copy number loss of CDKN2A/B and RB correlates with sensitivity and resistance to palbociclib, respectively [130]. A transcript microarray analysis of both palbociclib-sensitive and palbociclib-resistant human breast cancer cell lines identified 450 genes that were differentially expressed between sensitive cell lines (palbociclib IC50 < 150 nM) and resistant cell lines (palbociclib IC50 > 1000 nM). An increased expression of RB and CCND1 and a decreased expression of CDKN2A were identified in association with sensitivity to the effect of palbociclib [131]. A phase II trial of palbociclib in liposarcoma patients with CDK4 amplification and RB expression revealed that the sensitivity of these patients to palbociclib was associated with a favorable progression-free survival rate [132]. However, in FP RMS cell lines, resistance to ribociclib was evident in CDK4-amplified versus CDK4-wildtype cell lines. This differential responsiveness to ribociclib was also demonstrated in xenograft models of CDK4-amplified and wildtype FP RMS [133], implying that high levels of CDK4 may not be optimally inhibited. In addition, in FP RMS cell lines with a higher level of RB expression, increased sensitivity to ribociclib was shown, compared to FN RMS cell lines with a lower level of RB expression [133]. Consistent with the higher level of RB expression in FP RMS cell lines [133], CDK4 amplification has also been demonstrated predominantly in FP RMS tumors, compared to FN RMS tumors [134]. In a panel of melanoma cell lines, the most prevalent alterations were found in CDKN2A, including copy number loss, methylation, or mutation in this gene, which resulted in a decreased or absent CDKN2A protein and increased palbociclib sensitivity [135]. In a breast cancer cell line, E2F2 overexpression was sufficient to bypass sensitivity to palbociclib [136]. Therefore, if E2F levels are high, cells can evade cell cycle arrest that is mediated by CDK4/6 inhibition [136]. Moreover, another study found that glioblastoma (GBM) xenograft lines with CDKN2A expression and either RB mutation or CDK4 amplification were resistant to palbociclib. They suggested that CDK4 amplification leads to palbociclib resistance due to persistent RB phosphorylation by excessive CDK4. In addition, they demonstrated palbociclib sensitivity in an in vivo orthotopic GBM survival model. Palbociclib sensitivity was significant in this in vivo model with CDKN2A/B-deletion and CDK4 wild-type [137]. As part of another study, cell response to palbociclib was assessed on single-cell clones with high and low CDK2 activity sorted from triple-negative breast cancer cells. It was shown that the subpopulation of cells with high CDK2 activity bypassed the CDK4/6-dependency for cell cycle progression by the temporal deregulation of Cyclin E1 expression. However, the subpopulation of cells with low CDK2 activity could not overcome the inhibition of CDK4/6; therefore, they entered a quiescent state [138].
The underlying mechanisms that are associated with an acquired resistance to CDK4/6 inhibitors are discussed in the next sub section.

Acquired Mechanisms of Resistance to CDK4/6 Inhibitors and Possible Combination Therapies with CDK4/6 Inhibitors

Acquired resistance is the main reason for the therapeutic failure of CDK4/6 inhibitors in RB-proficient tumors, where tumor cells can acquire the ability to survive in the absence of CDK4/6 function [139]. Elucidation of the potential mechanisms of acquired resistance to CDK4/6 inhibitors may help to identify effective combination therapies to improve therapeutic responses to CDK4/6 inhibitors [87]. In contrast to cytotoxic drugs, CDK4/6 inhibitors mostly induce a cytostatic response by blocking CDK4/6-dependent cell cycle progression from the G1 to the S phase of the cell cycle [140]. The combination of CDK4/6 inhibitors with additional targeted therapies in RB-proficient tumors may change the cytostatic responses of CDK4/6 inhibitors to senescence and apoptosis [87]. Targeting major mitogenic signaling pathways such as the phosphatidylinositol 3’-kinase (PI3K) pathway or the mitogen-activated protein kinase (MAPK) pathway, along with CDK4/6 inhibition, may be a more promising combination for improving therapeutic efficacy (Figure 1) [87]. Moreover, both the PI3K and MAPK pathways are typically activated in cancer even before treatment. Therefore, it is possible that PI3K or MEK inhibitors, in combination with CDK4/6 inhibitors, are required to push the cells from cytostasis to cell death. For example, in estrogen receptor (ER)-positive breast cancer cell lines, upregulation of the PI3K pathway was identified as a consequence of chronic exposure to the CDK4/6 inhibitor palbociclib, which in turn upregulated Cyclin D1. In these cell lines, activated Cyclin D1 could drive cell cycle progression independently of CDK4/6 through the activation of CDK2; therefore, these cells could evade the cytostatic effect of the CDK4/6 inhibitor through noncanonical Cyclin D1-CDK2–mediated S-phase entry. In this study, the dual inhibition of the CDK4/6 and PI3K pathway prevented resistance to palbociclib both in vitro and in vivo [127]. In addition, it was shown that ribociclib, combined with PDK1 inhibitor GSK2334470 or the PI3K inhibitor alpelisib, decreased ER+ breast cancer xenograft tumor growth more efficaciously than either drug alone. In this study, primary in vitro studies showed the upregulation of phosphorylated and total PDK1 protein levels in ribociclib-resistant ER+ breast cancer cells, generated by chronic drug exposure [141]. PDK1 functions downstream of PI3K and activates AKT, where PDK1 at the plasma membrane binds to phosphatidylinositol-3,4,5-trisphosphate (PIP3), a product of PI3K. PIP3 phosphorylates and activates AKT [142]. The results of another study revealed that in palbociclib-resistant ER+ breast cancer cells, the Cyclin D-CDK4/6 pathway could be reactivated, but it remained sensitive to mTOR inhibitor vistusertib, suggesting that the inhibition of the PI3K pathway through the inhibition of mTOR may be a potential option for patients who have relapsed on therapy with CDK4/6 inhibitors. In this study, the combination of palbociclib with vistusertib resulted in more potent and durable regressions in breast cancer cell lines and xenografts [143]. Moreover, WES data on the biopsies from a melanoma patient showed that the oncogenic mutation PI3KCAE545K pre-existed in a rare tumor cell subpopulation prior to combination therapy of the CDK4/6 inhibitor ribociclib and the MEK inhibitor MEK162. In this patient, it was demonstrated that these rare tumor cell subpopulations were positively selected and expanded early after ribociclib + MEK162 combination therapy. PI3KCAE545K was identified as the only oncogenic mutation that confers resistance via increased activation of S6K1, a key downstream mTOR effector in the PI3K pathway. Therefore, an mTOR inhibitor could help to alleviate this resistance [144], possibly developed by CDK4/6 inhibitor ribociclib. As mentioned earlier, the MAPK signaling pathway can also contribute to CDK4/6 inhibitor resistance. In the following studies, the integration of an RNA sequencing analysis and phosphoproteomics profiling revealed that the MAPK pathway was hyperactivated in palbociclib-resistant prostate adenocarcinoma cell models. These palbociclib-resistant models were sensitized to MEK inhibitors, showing dependency on the active MAPK signaling pathway for tumor development when CDK4/6 was inhibited [145]. A study on bladder cancer also found that 995 genes were identified as significantly enriched after treatment with palbociclib in a bladder cancer cell line, using a genome-scale CRISPR-Cas9 activation screen. Significantly enriched related single-guide RNAs (sgRNAs) were randomly cloned and validated on both molecular and functional levels for mediating resistance to palbociclib. MAP3K20 gene, a member of the MAPK signaling pathway, was among the validated sgRNAs considered as resistance mediators to palbociclib. In addition, the dual inhibition of the CDK4/6 and MAPK pathway exhibited synergistic effects in vitro and in vivo [146]. In agreement with these observations, the dual inhibition of the CDK4/6 and MAPK pathway has revealed beneficial preclinical outcomes in melanoma [147] and colorectal cancer [148,149,150]. These findings suggest that the dual inhibition of the CDK4/6 and PI3K or MAPK pathways may be more efficacious at overcoming resistance to CDK4/6 inhibitors and increasing their antitumor activity in patients who are sensitive to these inhibitors in the clinic.
Our data also support the efficacy of the dual inhibition of the CDK4/6 and PI3K or MAPK pathways in models of aggressive OS (Barghi et al., unpublished observations). Based on our in vitro screening data, the combination therapy of CDK4/6 and PI3K/mTOR inhibition induced a synergistic cell-growth inhibition that correlated with increased cell death in a panel of human RB+ OS cell lines. Moreover, our in vivo data demonstrated that in a pediatric patient-derived xenograft (PDX) model of relapsed OS that exhibited CDK4/6 hyperactivation, the dual inhibition of the CDK4/6 and PI3K/mTOR pathways was well tolerated and tumor growth was significantly decreased, compared to single agents (Barghi et al., unpublished observations).

5. Conclusions and Future Directions

Precision medicine in pediatric and AYA cancers, including sarcomas, seeks to identify therapeutic biomarkers that can reliably provide rationale and prioritization of the best therapy for each individual patient. To this end, the time of the biopsy collection and analysis may be critical. One can envision that the identification of high-risk signatures prior to front-line therapy is a strategy to pursue. Additionally, at relapsed stages, it is important to note that precision-guided treatment could be different from that at early stages. Therefore, obtaining another biopsy is appropriate for verifying previous actionable targets and/or identifying additional therapeutic biomarkers at the relapse stage [151]. To improve outcomes in pediatric and AYA sarcoma patients harboring poor prognostic signatures, it is essential to continue to develop and refine in-vitro and in-vivo modeling approaches that recapitulate not only the molecular signature of high-risk sarcoma patients, but also the tumor microenvironments, which can vary greatly in patients with aggressive sarcomas. To ultimately improve patient outcomes in the clinic, linking a patient’s clinical history to preclinical safety and efficacy data obtained from their PDX model is critical for studying pathogenesis and validating alternative or combination therapies. The next goal of precision medicine in pediatric and AYA cancer patients is to understand in detail the tumor adaptive response and develop combination therapies that can mitigate inhibitor resistance, such as that discussed on CDK4/6 inhibition. The development of resistance to CDK4/6 inhibitors from both the pre-activation of the PI3K or MAPK pathway or compensatory activation is being explored extensively in breast cancer, and clearly warrants preclinical testing in pediatric and AYA sarcomas with CDK4/6 hyperactivation. Importantly, continued approaches to understand and minimize toxic side effects need to receive more attention, as future therapies will move towards combinations of small molecule inhibitors [152]. In this article, we reviewed multiple pediatric and AYA precision medicine trials to highlight the dysregulation of the CDK4/6 cell cycle regulatory pathway in the most common types of pediatric and AYA sarcomas, as well as to provide insight into the promise and challenges of targeting the CDK4/6 pathway. In conclusion, the identification of potential therapeutic biomarkers by precision medicine has great potential to accelerate and advance successful patient outcomes, as well as develop less toxic treatments for pediatric and AYA cancer sarcoma patients. Thus, the establishment of precision medicine programs in academic centers and related clinical trials on pediatric and AYA sarcomas is the key to identifying and refining targeted therapies for these rare diseases, which, once classified into each subtype, represent even smaller patient populations [153].

Author Contributions

Conceptualization, F.B., H.E.S., M.R.S., B.J.B., N.R., K.B.-V., M.J., M.J.F., P.H.P. and K.E.P.; methodology, F.B., P.H.P. and K.E.P.; formal analysis, F.B., P.H.P. and K.E.P.; resources, P.H.P. and K.E.P.; data curation, P.H.P. and K.E.P.; writing—original draft preparation, F.B., H.E.S., M.R.S., B.J.B., N.R., K.B.-V., M.J., M.J.F., P.H.P. and K.E.P.; writing—review and editing, F.B., H.E.S., M.R.S., B.J.B., N.R., K.B.-V., M.J., M.J.F., P.H.P. and K.E.P.; visualization, F.B., P.H.P. and K.E.P.; supervision, H.E.S., P.H.P. and K.E.P.; project administration, P.H.P. and K.E.P.; funding acquisition, P.H.P. and K.E.P. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by the Sarcoma Foundation of America, Cure Kids Cancer, NICHD/NIH Specialized Centers in Research in Pediatric Developmental Pharmacology (P50HD090215), NIH/NCI Cancer Center Support Grant (P30CA082709), The Tyler Trent Cancer Research Endowment for the Riley Hospital for Children IU-Health, The Grand Challenge-Precision Health Initiative-Pre-Sarcoma/Sarcoma Pillar, and the Riley Children’s Foundation.

Conflicts of Interest

H.E.S is a retiree and shareholder of Eli Lilly and Co. All other authors declare no conflict of interest.

References

  1. Ries, L.A.G. Cancer Incidence and Survival among Children and Adolescents: United States Seer Program, 1975–1995; National Cancer Institute: Bethesda, MD, USA, 1999. [Google Scholar]
  2. Burdach, S.E.G.; Westhoff, M.-A.; Steinhauser, M.F.; Debatin, K.-M. Precision medicine in pediatric oncology. Mol. Cell. Pediatr. 2018, 5, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hunger, S.P.; Mullighan, C.G. Acute Lymphoblastic Leukemia in Children. N. Engl. J. Med. 2015, 373, 1541–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hingorani, P.; Janeway, K.; Crompton, B.D.; Kadoch, C.; Mackall, C.L.; Khan, J.; Shern, J.F.; Schiffman, J.; Mirabello, L.; Savage, S.; et al. Current state of pediatric sarcoma biology and opportunities for future discovery: A report from the sarcoma translational research workshop. Cancer Genet. 2016, 209, 182–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Jacobs, A.J.; Lindholm, E.B.; Levy, C.F.; Fish, J.D.; Glick, R.D. Racial and ethnic disparities in treatment and survival of pediatric sarcoma. J. Surg. Res. 2017, 219, 43–49. [Google Scholar] [CrossRef] [PubMed]
  6. Kurzrock, R.; Giles, F.J. Precision oncology for patients with advanced cancer: The challenges of malignant snowflakes. Cell Cycle 2015, 14, 2219–2221. [Google Scholar] [CrossRef] [PubMed]
  7. Kohlmeyer, J.L.; Gordon, D.J.; Tanas, M.R.; Monga, V.; Dodd, R.D.; Quelle, D.E. CDKs in Sarcoma: Mediators of Disease and Emerging Therapeutic Targets. Int. J. Mol. Sci. 2020, 21, 3018. [Google Scholar] [CrossRef] [PubMed]
  8. Gage, M.M.; Nagarajan, N.; Ruck, J.M.; Canner, J.K.; Khan, S.; Giuliano, K.; Gani, F.; Wolfgang, C.; Johnston, F.M.; Ahuja, N. Sarcomas in the United States: Recent trends and a call for improved staging. Oncotarget 2019, 10, 2462–2474. [Google Scholar] [CrossRef] [Green Version]
  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. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
  11. Bleyer, A.; Montello, M.; Budd, T.; Saxman, S. National survival trends of young adults with sarcoma: Lack of progress is associated with lack of clinical trial participation. Cancer 2005, 103, 1891–1897. [Google Scholar] [CrossRef]
  12. Ballatori, S.E.; Hinds, P.W. Osteosarcoma: Prognosis plateau warrants retinoblastoma pathway targeted therapy. Signal Transduct. Target. Ther. 2016, 1, 16001. [Google Scholar] [CrossRef] [Green Version]
  13. Duchman, K.R.; Gao, Y.; Miller, B.J. Prognostic factors for survival in patients with high-grade osteosarcoma using the Surveillance, Epidemiology, and End Results (SEER) Program database. Cancer Epidemiol. 2015, 39, 593–599. [Google Scholar] [CrossRef]
  14. Gianferante, D.M.; Mirabello, L.; Savage, S.A. Germline and somatic genetics of osteosarcoma—Connecting aetiology, biology and therapy. Nat. Rev. Endocrinol. 2017, 13, 480–491. [Google Scholar] [CrossRef]
  15. Haynes, L.; Kaste, S.C.; Ness, K.K.; Wu, J.; Ortega-Laureano, L.; Bishop, M.; Neel, M.; Rao, B.; Fernandez-Pineda, I. Pathologic fracture in childhood and adolescent osteosarcoma: A single-institution experience. Pediatr. Blood Cancer 2016, 64, e26290. [Google Scholar] [CrossRef] [Green Version]
  16. Klein, M.J.; Siegal, G.P. Osteosarcoma. Am. J. Clin. Pathol. 2006, 125, 555–581. [Google Scholar] [CrossRef]
  17. Mirabello, L.; Troisi, R.J.; Savage, S.A. International osteosarcoma incidence patterns in children and adolescents, middle ages and elderly persons. Int. J. Cancer 2009, 125, 229–234. [Google Scholar] [CrossRef] [Green Version]
  18. Moore, D.D.; Luu, H.H. Osteosarcoma. Orthop. Oncol. 2014, 162, 65–92. [Google Scholar]
  19. Nie, Z.; Peng, H. Osteosarcoma in patients below 25 years of age: An observational study of incidence, metastasis, treatment and outcomes. Oncol. Lett. 2018, 16, 6502–6514. [Google Scholar] [CrossRef] [Green Version]
  20. Wang, T.; Liao, S.; Ding, X.; Anil, K.C.; Huang, Q.; Lin, C.; Mo, J.; Tang, H.; Liu, Y. Intraperitoneal extraosseous osteosarcoma: A case report and literatures review. BMC Musculoskelet. Disord. 2020, 21, 1–5. [Google Scholar] [CrossRef]
  21. Kong, E.; Hinds, P.W. The Retinoblastoma Protein in Osteosarcomagenesis. In Osteosarcoma; INTECH Open Access Publisher: Rijeka, Croatia, 2012. [Google Scholar]
  22. Mai, P.L.; Best, A.F.; Peters, J.A.; DeCastro, R.M.; Khincha, P.P.; Loud, J.T.; Bremer, R.C.; Rosenberg, P.S.; Savage, S.A. Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort. Cancer 2016, 122, 3673–3681. [Google Scholar] [CrossRef] [Green Version]
  23. Alaggio, R.; Zhang, L.; Sung, Y.S.; Huang, S.C.; Chen, C.L.; Bisogno, G.; Zin, A.; Agaram, N.P.; LaQuaglia, M.P.; Wexler, L.H.; et al. A Molecular Study of Pediatric Spindle and Sclerosing Rhabdomyosarcoma: Identification of Novel and Recurrent VGLL2-related Fusions in Infantile Cases. Am. J. Surg. Pathol. 2016, 40, 224–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Breitfeld, P.P.; Meyer, W.H. Rhabdomyosarcoma: New Windows of Opportunity. Oncologist 2005, 10, 518–527. [Google Scholar] [CrossRef] [PubMed]
  25. Huh, W.; Bejar, D.E. Rhabdomyosarcoma in adolescent and young adult patients: Current perspectives. Adolesc. Heal. Med. Ther. 2014, 5, 115–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Jawad, M.U.; Cheung, M.C.; Min, E.S.; Schneiderbauer, M.M.; Koniaris, L.G.; Scully, S.P. Ewing sarcoma demonstrates racial disparities in incidence-related and sex-related differences in outcome: An analysis of 1631 cases from the SEER database, 1973–2005. Cancer 2009, 115, 3526–3536. [Google Scholar] [CrossRef]
  27. Jawad, N.; McHugh, K. The clinical and radiologic features of paediatric rhabdomyosarcoma. Pediatr. Radiol. 2019, 49, 1516–1523. [Google Scholar] [CrossRef]
  28. Lychou, S.E.; Gustafsson, G.G.; Ljungman, G.E. Higher rates of metastatic disease may explain the declining trend in Swedish paediatric rhabdomyosarcoma survival rates. Acta Paediatr. 2016, 105, 74–81. [Google Scholar] [CrossRef]
  29. Ognjanovic, S.; Linabery, A.; Charbonneau, B.; Ross, J.A. Trends in childhood rhabdomyosarcoma incidence and survival in the United States, 1975–2005. Cancer 2009, 115, 4218–4226. [Google Scholar] [CrossRef] [Green Version]
  30. Perez, E.A.; Kassira, N.; Cheung, M.C.; Koniaris, L.G.; Neville, H.L.; Sola, J. Rhabdomyosarcoma in Children: A SEER Population Based Study. J. Surg. Res. 2011, 170, e243–e251. [Google Scholar] [CrossRef]
  31. Stiller, C.; Parkint, D.M. International variations in the incidence of childhood soft-tissue sarcomas. Paediatr. Périnat. Epidemiol. 1994, 8, 107–119. [Google Scholar] [CrossRef]
  32. Skapek, S.X.; Ferrari, A.; Gupta, A.A.; Lupo, P.J.; Butler, E.; Shipley, J.; Barr, F.G.; Hawkins, D.S. Rhabdomyosarcoma. Nat. Rev. Dis. Primers 2019, 5, 1. [Google Scholar] [CrossRef]
  33. Esiashvili, N.; Goodman, M.; Marcus, R.B., Jr. Changes in incidence and survival of Ewing sarcoma patients over the past 3 decades: Surveillance Epidemiology and End Results data. J. Pediatr. Hematol. Oncol. 2008, 30, 425–430. [Google Scholar] [CrossRef]
  34. Tavakkoli, M.; Mueller, L. Cutaneous Ewing Sarcoma and Ewing Sarcoma of the Bone: Distinct Diseases. Case Rep. Oncol. 2018, 11, 729–734. [Google Scholar] [CrossRef]
  35. Choi, E.Y.; Gardner, J.M.; Lucas, D.R.; McHugh, J.B.; Patel, R.M. Ewing sarcoma. Semin. Diagn. Pathol. 2014, 31, 39–47. [Google Scholar] [CrossRef]
  36. Grünewald, T.G.P.; Cidre-Aranaz, F.; Surdez, D.; Tomazou, E.M.; de Álava, E.; Kovar, H.; Sorensen, P.H.; Delattre, O.; Dirksen, U. Ewing sarcoma. Nat. Rev. Dis. Primers 2018, 4, 5. [Google Scholar] [CrossRef]
  37. Brohl, A.S.; Patidar, R.; Turner, C.E.; Wen, X.; Song, Y.K.; Wei, J.S.; Calzone, K.A.; Khan, J. Frequent inactivating germline mutations in DNA repair genes in patients with Ewing sarcoma. Genet. Med. 2017, 19, 955–958. [Google Scholar] [CrossRef] [Green Version]
  38. Farid, M.; Ngeow, J. Sarcomas Associated With Genetic Cancer Predisposition Syndromes: A Review. Oncologist 2016, 21, 1002–1013. [Google Scholar] [CrossRef] [Green Version]
  39. Kleinerman, R.A.; Schonfeld, S.J.; Tucker, M.A. Sarcomas in hereditary retinoblastoma. Clin. Sarcoma Res. 2012, 2, 15. [Google Scholar] [CrossRef] [Green Version]
  40. Marko, T.A.; Diessner, B.; Spector, L.G. Prevalence of Metastasis at Diagnosis of Osteosarcoma: An International Comparison. Pediatr. Blood Cancer 2016, 63, 1006–1011. [Google Scholar] [CrossRef] [Green Version]
  41. Isakoff, M.S.; Barkauskas, D.A.; Ebb, D.; Morris, C.; Letson, D.G. Poor Survival for Osteosarcoma of the Pelvis: A Report from the Children’s Oncology Group. Clin. Orthop. Relat. Res. 2012, 470, 2007–2013. [Google Scholar] [CrossRef] [Green Version]
  42. Martin, J.W.; Squire, J.; Zielenska, M. The Genetics of Osteosarcoma. Sarcoma 2012, 2012, 1–11. [Google Scholar] [CrossRef] [Green Version]
  43. Bielack, S.S.; Smeland, S.; Whelan, J.S.; Marina, N.; Jovic, G.; Hook, J.M.; Krailo, M.D.; Gebhardt, M.; Pápai, Z.; Meyer, J.; et al. Methotrexate, Doxorubicin, and Cisplatin (MAP) Plus Maintenance Pegylated Interferon Alfa-2b Versus MAP Alone in Patients With Resectable High-Grade Osteosarcoma and Good Histologic Response to Preoperative MAP: First Results of the EURAMOS-1 Good Response Randomized Controlled Trial. J. Clin. Oncol. 2015, 33, 2279–2287. [Google Scholar] [CrossRef] [PubMed]
  44. Zuch, D.; Giang, A.-H.; Shapovalov, Y.; Schwarz, E.; Rosier, R.; O’Keefe, R.; Eliseev, R.A. Targeting Radioresistant Osteosarcoma Cells With Parthenolide. J. Cell. Biochem. 2011, 113, 1282–1291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Duffaud, F.; Italiano, A.; Bompas, E.; Rios, M.; Penel, N.; Mir, O.; Piperno-Neumann, S.; Chevreau, C.; Delcambre, C.; Bertucci, F.; et al. Efficacy and safety of regorafenib in patients with metastatic or locally advanced chondrosarcoma: Results of a non-comparative, randomised, double-blind, placebo controlled, multicentre phase II study. Eur. J. Cancer 2021, 150, 108–118. [Google Scholar] [CrossRef] [PubMed]
  46. Gaspar, N.; Hawkins, D.S.; Dirksen, U.; Lewis, I.J.; Ferrari, S.; Le Deley, M.-C.; Kovar, H.; Grimer, R.; Whelan, J.; Claude, L.; et al. Ewing Sarcoma: Current Management and Future Approaches Through Collaboration. J. Clin. Oncol. 2015, 33, 3036–3046. [Google Scholar] [CrossRef]
  47. Li, Y.; Luo, H.; Liu, T.; Zacksenhaus, E.; Ben-David, Y. The ets transcription factor Fli-1 in development, cancer and disease. Oncogene 2014, 34, 2022–2031. [Google Scholar] [CrossRef] [Green Version]
  48. Sorensen, P.H.; Lessnick, S.L.; Lopez-Terrada, D.; Liu, X.F.; Triche, T.J.; Denny, C.T. A second Ewing’s sarcoma translocation, t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat. Genet. 1994, 6, 146–151. [Google Scholar] [CrossRef]
  49. Berger, M.; Dirksen, U.; Braeuninger, A.; Koehler, G.; Juergens, H.; Krumbholz, M.; Metzler, M. Genomic EWS-FLI1 Fusion Sequences in Ewing Sarcoma Resemble Breakpoint Characteristics of Immature Lymphoid Malignancies. PLoS ONE 2013, 8, e56408. [Google Scholar] [CrossRef] [Green Version]
  50. Bilke, S.; Schwentner, R.; Yang, F.; Kauer, M.; Jug, G.; Walker, R.L.; Davis, S.; Zhu, Y.J.; Pineda, M.; Meltzer, P.S.; et al. Oncogenic ETS fusions deregulate E2F3 target genes in Ewing sarcoma and prostate cancer. Genome Res. 2013, 23, 1797–1809. [Google Scholar] [CrossRef] [Green Version]
  51. Harlow, M.L.; Chasse, M.H.; Boguslawski, E.A.; Sorensen, K.M.; Gedminas, J.M.; Kitchen-Goosen, S.M.; Rothbart, S.B.; Taslim, C.; Lessnick, S.L.; Peck, A.S.; et al. Trabectedin Inhibits EWS-FLI1 and Evicts SWI/SNF from Chromatin in a Schedule-dependent Manner. Clin. Cancer Res. 2019, 25, 3417–3429. [Google Scholar] [CrossRef] [Green Version]
  52. Karosas, A.O. Ewing’s sarcoma. Am. J. Health Syst. Pharm. 2010, 67, 1599–1605. [Google Scholar] [CrossRef]
  53. Amer, K.M.; Thomson, J.E.; Congiusta, D.; Dobitsch, A.; Chaudhry, A.; Li, M.; Chaudhry, A.; Bozzo, A.; Siracuse, B.; Aytekin, M.N.; et al. Epidemiology, Incidence, and Survival of Rhabdomyosarcoma Subtypes: SEER and ICES Database Analysis. J. Orthop. Res. 2019, 37, 2226–2230. [Google Scholar] [CrossRef]
  54. Shern, J.F.; Chen, L.; Chmielecki, J.; Wei, J.S.; Patidar, R.; Rosenberg, M.; Ambrogio, L.; Auclair, D.; Wang, J.; Song, Y.K.; et al. Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov. 2014, 4, 216–231. [Google Scholar] [CrossRef] [Green Version]
  55. Sun, W.; Chatterjee, B.; Wang, Y.; Stevenson, H.S.; Edelman, D.C.; Meltzer, P.S.; Barr, F.G. Distinct methylation profiles characterize fusion-positive and fusion-negative rhabdomyosarcoma. Mod. Pathol. 2015, 28, 1214–1224. [Google Scholar] [CrossRef]
  56. Wachtel, M.; Schäfer, B.W. PAX3-FOXO1: Zooming in on an “undruggable” target. Semin. Cancer Biol. 2018, 50, 115–123. [Google Scholar] [CrossRef]
  57. Le Loarer, F.; Cleven, A.H.G.; Bouvier, C.; Castex, M.-P.; Romagosa, C.; Moreau, A.; Salas, S.; Bonhomme, B.; Gomez-Brouchet, A.; Laurent, C.; et al. A subset of epithelioid and spindle cell rhabdomyosarcomas is associated with TFCP2 fusions and common ALK upregulation. Mod. Pathol. 2019, 33, 404–419. [Google Scholar] [CrossRef]
  58. Watson, S.; Perrin, V.; Guillemot, D.; Reynaud, S.; Coindre, J.; Karanian, M.; Guinebretière, J.; Freneaux, P.; Le Loarer, F.; Bouvet, M.; et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J. Pathol. 2018, 245, 29–40. [Google Scholar] [CrossRef] [Green Version]
  59. Lautz, T.B.; Hayes-Jordan, A. Recent progress in pediatric soft tissue sarcoma therapy. Semin. Pediatr. Surg. 2019, 28, 150862. [Google Scholar] [CrossRef]
  60. Zloto, O.; Minard-Colin, V.; Boutroux, H.; Brisse, H.J.; Levy, C.; Kolb, F.; Bolle, S.; Carton, M.; Helfre, S.; Orbach, D. Second-line therapy in young patients with relapsed or refractory orbital rhabdomyosarcoma. Acta Ophthalmol. 2020, 99, 334–341. [Google Scholar] [CrossRef]
  61. Mascarenhas, L.; Chi, Y.-Y.; Hingorani, P.; Anderson, J.R.; Lyden, E.R.; Rodeberg, D.A.; Indelicato, D.J.; Kao, S.; Dasgupta, R.; Spunt, S.L.; et al. Randomized Phase II Trial of Bevacizumab or Temsirolimus in Combination With Chemotherapy for First Relapse Rhabdomyosarcoma: A Report From the Children’s Oncology Group. J. Clin. Oncol. 2019, 37, 2866–2874. [Google Scholar] [CrossRef]
  62. Morganti, S.; Tarantino, P.; Ferraro, E.; D’Amico, P.; Duso, B.A.; Curigliano, G. Next Generation Sequencing (NGS): A Revolutionary Technology in Pharmacogenomics and Personalized Medicine in Cancer. Adv. Exp. Med. Biol. 2019, 1168, 9–30. [Google Scholar]
  63. Kamps, R.; Brandão, R.D.; van den Bosch, B.J.; Paulussen, A.D.; Xanthoulea, S.; Blok, M.J.; Romano, A. Next-Generation Sequencing in Oncology: Genetic Diagnosis, Risk Prediction and Cancer Classification. Int. J. Mol. Sci. 2017, 18, 308. [Google Scholar] [CrossRef]
  64. Rodriguez, H.; Zenklusen, J.C.; Staudt, L.M.; Doroshow, J.H.; Lowy, D.R. The next horizon in precision oncology: Proteogenomics to inform cancer diagnosis and treatment. Cell 2021, 184, 1661–1670. [Google Scholar] [CrossRef]
  65. Jones, D.T.W.; Banito, A.; Grünewald, T.G.P.; Haber, M.; Jäger, N.; Kool, M.; Milde, T.; Molenaar, J.J.; Nabbi, A.; Pugh, T.J.; et al. Molecular characteristics and therapeutic vulnerabilities across paediatric solid tumours. Nat. Cancer 2019, 19, 420–438. [Google Scholar] [CrossRef]
  66. Seibel, N.L.; Janeway, K.; Allen, C.E.; Chi, S.N.; Cho, Y.-J.; Bender, J.L.G.; Kim, A.; Laetsch, T.W.; Irwin, M.S.; Takebe, N.; et al. Pediatric oncology enters an era of precision medicine. Curr. Probl. Cancer 2017, 41, 194–200. [Google Scholar] [CrossRef]
  67. Wong, M.; Mayoh, C.; Lau, L.M.S.; Khuong-Quang, D.-A.; Pinese, M.; Kumar, A.; Barahona, P.; Wilkie, E.E.; Sullivan, P.; Bowen-James, R.; et al. Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer. Nat. Med. 2020, 26, 1742–1753. [Google Scholar] [CrossRef]
  68. Goel, S.; DeCristo, M.J.; McAllister, S.S.; Zhao, J.J. CDK4/6 Inhibition in Cancer: Beyond Cell Cycle Arrest. Trends Cell Biol. 2018, 28, 911–925. [Google Scholar] [CrossRef]
  69. Hamilton, E.; Infante, J.R. Targeting CDK4/6 in patients with cancer. Cancer Treat. Rev. 2016, 45, 129–138. [Google Scholar] [CrossRef] [Green Version]
  70. VanArsdale, T.; Boshoff, C.; Arndt, K.T.; Abraham, R.T. Molecular Pathways: Targeting the Cyclin D-CDK4/6 Axis for Cancer Treatment. Clin. Cancer Res. 2015, 21, 2905–2910. [Google Scholar] [CrossRef] [Green Version]
  71. Worst, B.C.; van Tilburg, C.M.; Balasubramanian, G.P.; Fiesel, P.; Witt, R.; Freitag, A.; Boudalil, M.; Previti, C.; Wolf, S.; Schmidt, S.; et al. Next-generation personalised medicine for high-risk paediatric cancer patients – The INFORM pilot study. Eur. J. Cancer 2016, 65, 91–101. [Google Scholar] [CrossRef] [Green Version]
  72. Oberg, J.A.; Bender, J.G.; Sulis, M.L.; Pendrick, D.; Sireci, A.N.; Hsiao, S.J.; Turk, A.T.; Cruz, F.S.D.; Hibshoosh, H.; Remotti, H.; et al. Implementation of next generation sequencing into pediatric hematology-oncology practice: Moving beyond actionable alterations. Genome Med. 2016, 8, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Harris, M.H.; DuBois, S.G.; Bender, J.L.G.; Kim, A.; Crompton, B.D.; Parker, E.; Dumont, I.P.; Hong, A.L.; Guo, D.; Church, A.; et al. Multicenter Feasibility Study of Tumor Molecular Profiling to Inform Therapeutic Decisions in Advanced Pediatric Solid Tumors: The Individualized Cancer Therapy (iCat) Study. JAMA Oncol. 2016, 2, 608–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Criscitiello, C.; Viale, G.; Esposito, A.; Curigliano, G. Dinaciclib for the treatment of breast cancer. Expert Opin. Investig. Drugs 2014, 23, 1305–1312. [Google Scholar] [CrossRef] [PubMed]
  75. Pincez, T.; Clément, N.; Lapouble, E.; Pierron, G.; Kamal, M.; Bieche, I.; Bernard, V.; Fréneaux, P.; Michon, J.; Orbach, D.; et al. Feasibility and clinical integration of molecular profiling for target identification in pediatric solid tumors. Pediatr. Blood Cancer 2017, 64, e26365. [Google Scholar] [CrossRef]
  76. Khater, F.; Vairy, S.; Langlois, S.; Dumoucel, S.; Sontag, T.; St-Onge, P.; Bittencourt, H.; Soglio, D.D.; Champagne, J.; Duval, M.; et al. Molecular Profiling of Hard-to-Treat Childhood and Adolescent Cancers. JAMA Netw. Open 2019, 2, e192906. [Google Scholar] [CrossRef]
  77. Groisberg, R.; Hong, D.S.; Holla, V.; Janku, F.; Piha-Paul, S.; Ravi, V.; Benjamin, R.; Patel, S.K.; Somaiah, N.; Conley, A.; et al. Clinical genomic profiling to identify actionable alterations for investigational therapies in patients with diverse sarcomas. Oncotarget 2017, 8, 39254–39267. [Google Scholar] [CrossRef]
  78. Suehara, Y.; Alex, D.; Bowman, A.; Middha, S.; Zehir, A.; Chakravarty, D.; Wang, L.; Jour, G.; Nafa, K.; Hayashi, T.; et al. Clinical Genomic Sequencing of Pediatric and Adult Osteosarcoma Reveals Distinct Molecular Subsets with Potentially Targetable Alterations. Clin. Cancer Res. 2019, 25, 6346–6356. [Google Scholar] [CrossRef]
  79. Chen, X.; Bahrami, A.; Pappo, A.; Easton, J.; Dalton, J.; Hedlund, E.; Ellison, D.; Shurtleff, S.; Wu, G.; Wei, L.; et al. Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep. 2014, 7, 104–112. [Google Scholar] [CrossRef] [Green Version]
  80. Bousquet, M.; Noirot, C.; Accadbled, F.; de Gauzy, J.S.; Castex, M.; Brousset, P.; Gomez-Brouchet, A. Whole-exome sequencing in osteosarcoma reveals important heterogeneity of genetic alterations. Ann. Oncol. 2016, 27, 738–744. [Google Scholar] [CrossRef]
  81. Perry, J.A.; Kiezun, A.; Tonzi, P.; van Allen, E.M.; Carter, S.L.; Baca, S.C.; Cowley, G.S.; Bhatt, A.S.; Rheinbay, E.; Pedamallu, C.S.; et al. Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proc. Natl. Acad. Sci. USA 2014, 111, E5564–E5573. [Google Scholar] [CrossRef] [Green Version]
  82. Harttrampf, A.C.; Lacroix, L.; Deloger, M.; Deschamps, F.; Puget, S.; Auger, N.; Vielh, P.; Varlet, P.; Balogh, Z.; Abbou, S.; et al. Molecular Screening for Cancer Treatment Optimization (MOSCATO-01) in Pediatric Patients: A Single-Institutional Prospective Molecular Stratification Trial. Clin. Cancer Res. 2017, 23, 6101–6112. [Google Scholar] [CrossRef] [Green Version]
  83. Chang, W.; Brohl, A.S.; Patidar, R.; Sindiri, S.; Shern, J.F.; Wei, J.S.; Song, Y.K.; Yohe, M.E.; Gryder, B.; Zhang, S.; et al. MultiDimensional ClinOmics for Precision Therapy of Children and Adolescent Young Adults with Relapsed and Refractory Cancer: A Report from the Center for Cancer Research. Clin. Cancer Res. 2016, 22, 3810–3820. [Google Scholar] [CrossRef] [Green Version]
  84. Park, S.; Lee, J.; Do, I.G.; Jang, J.; Rho, K.; Ahn, S.; Maruja, L.; Kim, S.J.; Kim, K.M.; Mao, M.; et al. Aberrant CDK4 Amplification in Refractory Rhabdomyosarcoma as Identified by Genomic Profiling. Sci. Rep. 2014, 4, 3623. [Google Scholar] [CrossRef] [Green Version]
  85. Crompton, B.D.; Stewart, C.; Taylor-Weiner, A.; Alexe, G.; Kurek, K.C.; Calicchio, M.L.; Kiezun, A.; Carter, S.L.; Shukla, S.A.; Mehta, S.S.; et al. The genomic landscape of pediatric Ewing sarcoma. Cancer Discov. 2014, 4, 1326–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Asghar, U.; Witkiewicz, A.K.; Turner, N.C.; Knudsen, E.S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug Discov. 2015, 14, 130–146. [Google Scholar] [CrossRef] [Green Version]
  87. Sherr, C.J.; Beach, D.; Shapiro, G.I. Targeting CDK4 and CDK6: From Discovery to Therapy. Cancer Discov. 2016, 6, 353–367. [Google Scholar] [CrossRef] [Green Version]
  88. Lolli, G.; Johnson, L.N. CAK—Cyclin-Dependent Activating Kinase: A Key Kinase in Cell Cycle Control and a Target for Drugs? Cell Cycle 2005, 4, 565–570. [Google Scholar] [CrossRef] [Green Version]
  89. Grossel, M.J.; Hinds, P.W. From Cell Cycle to Differentiation: An Expanding Role for Cdk6. Cell Cycle 2006, 5, 266–270. [Google Scholar] [CrossRef] [Green Version]
  90. Dai, M.; Boudreault, J.; Wang, N.; Poulet, S.; Daliah, G.; Yan, G.; Moamer, A.; Burgos, S.A.; Sabri, S.; Ali, S.; et al. Differential Regulation of Cancer Progression by CDK4/6 Plays a Central Role in DNA Replication and Repair Pathways. Cancer Res. 2021, 81, 1332–1346. [Google Scholar] [CrossRef]
  91. Giacinti, C.; Giordano, A. RB and cell cycle progression. Oncogene 2006, 25, 5220–5227. [Google Scholar] [CrossRef] [Green Version]
  92. Taylor, L.J.; Bar-Sagi, D. The Role of Rac and Rho in Cell Cycle Progression. In Handbook of Cell Signaling; Academic Press: Amsterdam, The Netherlands, 2010; pp. 1781–1784. [Google Scholar]
  93. Braden, W.A.; McClendon, A.K.; Knudsen, E.S. Cyclin-dependent kinase 4/6 activity is a critical determinant of pre-replication complex assembly. Oncogene 2008, 27, 7083–7093. [Google Scholar] [CrossRef] [Green Version]
  94. Zhu, W.; Giangrande, P.H.; Nevins, J.R. E2Fs link the control of G1/S and G2/M transcription. Embo J. 2004, 23, 4615–4626. [Google Scholar] [CrossRef] [PubMed]
  95. Anders, L.; Ke, N.; Hydbring, P.; Choi, Y.J.; Widlund, H.R.; Chick, J.M.; Zhai, H.; Vidal, M.; Gygi, S.P.; Braun, P.; et al. A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells. Cancer Cell 2011, 20, 620–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Matsuura, I.; Denissova, N.G.; Wang, G.; He, D.; Long, J.; Liu, F. Cyclin-dependent kinases regulate the antiproliferative function of Smads. Nature 2004, 430, 226–231. [Google Scholar] [CrossRef] [PubMed]
  97. Zelivianski, S.; Cooley, A.; Kall, R.; Jeruss, J.S. Cyclin-Dependent Kinase 4–Mediated Phosphorylation Inhibits Smad3 Activity in Cyclin D–Overexpressing Breast Cancer Cells. Mol. Cancer Res. 2010, 8, 1375–1387. [Google Scholar] [CrossRef] [Green Version]
  98. Bury, M.; Le Calvé, B.; Ferbeyre, G.; Blank, V.; Lessard, F. New Insights into CDK Regulators: Novel Opportunities for Cancer Therapy. Trends Cell Biol. 2021, 31, 331–344. [Google Scholar] [CrossRef]
  99. Yuan, C.; Li, J.; Selby, T.L.; Byeon, I.-J.L.; Tsai, M.-D. Tumor suppressor INK4: Comparisons of conformational properties between p16INK4A and p18INK4C. J. Mol. Biol. 1999, 294, 201–211. [Google Scholar] [CrossRef] [Green Version]
  100. Pack, L.R.; Daigh, L.H.; Chung, M.; Meyer, T. Clinical CDK4/6 inhibitors induce selective and immediate dissociation of p21 from cyclin D-CDK4 to inhibit CDK2. Nat. Commun. 2021, 12, 3356. [Google Scholar] [CrossRef]
  101. Kong, Y.; Hsieh, C.-H.; Alonso, L.C. ANRIL: A lncRNA at the CDKN2A/B Locus With Roles in Cancer and Metabolic Disease. Front. Endocrinol. 2018, 9, 405. [Google Scholar] [CrossRef] [Green Version]
  102. 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. 2016, 35, 151–163. [Google Scholar] [CrossRef]
  103. Malumbres, M. Cyclin-dependent kinases. Genome Biol. 2014, 15, 122. [Google Scholar] [CrossRef] [Green Version]
  104. Malumbres, M.; Harlow, E.; Hunt, T.; Hunter, T.; Lahti, J.M.; Manning, G.; Morgan, D.; Tsai, L.-H.; Wolgemuth, D.J. Cyclin-dependent kinases: A family portrait. Nat. Cell Biol. 2009, 11, 1275–1276. [Google Scholar] [CrossRef] [Green Version]
  105. Zhang, M.; Zhang, L.; Hei, R.; Li, X.; Cai, H.; Wu, X.; Zheng, Q.; Cai, C. CDK inhibitors in cancer therapy, an overview of recent development. Am. J. Cancer Res. 2021, 11, 1913–1935. [Google Scholar]
  106. Poratti, M.; Marzaro, G. Third-generation CDK inhibitors: A review on the synthesis and binding modes of Palbociclib, Ribociclib and Abemaciclib. Eur. J. Med. Chem. 2019, 172, 143–153. [Google Scholar] [CrossRef]
  107. Yuan, K.; Wang, X.; Dong, H.; Min, W.; Hao, H.; Yang, P. Selective inhibition of CDK4/6: A safe and effective strategy for developing anticancer drugs. Acta Pharm. Sin. B 2021, 11, 30–54. [Google Scholar] [CrossRef]
  108. 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]
  109. Powell, K.; Prasad, V. Concerning FDA approval of trilaciclib (Cosela) in extensive-stage small-cell lung cancer. Transl. Oncol. 2021, 14, 101206. [Google Scholar] [CrossRef]
  110. McCain, J. First-in-Class CDK4/6 Inhibitor Palbociclib Could Usher in a New Wave of Combination Therapies for HR+, HER2- Breast Cancer. Pharm. Ther. 2015, 40, 511–520. [Google Scholar]
  111. Álvarez-Fernández, M.; Malumbres, M. Mechanisms of Sensitivity and Resistance to CDK4/6 Inhibition. Cancer Cell 2020, 37, 514–529. [Google Scholar] [CrossRef]
  112. Topacio, B.R.; Zatulovskiy, E.; Cristea, S.; Xie, S.; Tambo, C.S.; Rubin, S.M.; Sage, J.; Kõivomägi, M.; Skotheim, J.M. Cyclin D-Cdk4,6 Drives Cell-Cycle Progression via the Retinoblastoma Protein’s C-Terminal Helix. Mol. Cell 2019, 74, 758–770.e4. [Google Scholar] [CrossRef]
  113. Deng, J.; Wang, E.S.; Jenkins, R.W.; Li, S.; Dries, R.; Yates, K.; Chhabra, S.; Huang, W.; Liu, H.; Aref, A.R.; et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018, 8, 216–233. [Google Scholar] [CrossRef] [Green Version]
  114. Goel, S.; DeCristo, M.J.; Watt, A.C.; BrinJones, H.; Sceneay, J.; Li, B.B.; Khan, N.; Ubellacker, J.M.; Xie, S.; Metzger-Filho, O.; et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature 2017, 548, 471–475. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, P.; Lee, N.V.; Hu, W.; Xu, M.; Ferre, R.A.; Lam, H.; Bergqvist, S.; Solowiej, J.; Diehl, W.; He, Y.-A.; et al. Spectrum and Degree of CDK Drug Interactions Predicts Clinical Performance. Mol. Cancer Ther. 2016, 15, 2273–2281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Bisi, J.E.; Sorrentino, J.A.; Roberts, P.J.; Tavares, F.X.; Strum, J.C. Preclinical Characterization of G1T28: A Novel CDK4/6 Inhibitor for Reduction of Chemotherapy-Induced Myelosuppression. Mol. Cancer Ther. 2016, 15, 783–793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Kim, E.S. Abemaciclib: First Global Approval. Drugs 2017, 77, 2063–2070. [Google Scholar] [CrossRef] [PubMed]
  118. Shah, A.; Bloomquist, E.; Tang, S.; Fu, W.; Bi, Y.; Liu, Q.; Yu, J.; Zhao, P.; Palmby, T.R.; Goldberg, K.B.; et al. FDA Approval: Ribociclib for the Treatment of Postmenopausal Women with Hormone Receptor-Positive, HER2-Negative Advanced or Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2999–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Dickler, M.N.; Tolaney, S.M.; Rugo, H.S.; Cortés, J.; Diéras, V.; Patt, D.; Wildiers, H.; Hudis, C.A.; O’Shaughnessy, J.; Zamora, E.; et al. MONARCH 1, A Phase II Study of Abemaciclib, a CDK4 and CDK6 Inhibitor, as a Single Agent, in Patients with Refractory HR(+)/HER2(-) Metastatic Breast Cancer. Clin. Cancer Res. 2017, 23, 5218–5224. [Google Scholar] [CrossRef] [Green Version]
  120. George, M.A.; Qureshi, S.; Omene, C.; Toppmeyer, D.L.; Ganesan, S. Clinical and Pharmacologic Differences of CDK4/6 Inhibitors in Breast Cancer. Front. Oncol. 2021, 11, 693104. [Google Scholar] [CrossRef]
  121. Rugo, H.S.; Huober, J.; García-Sáenz, J.A.; Masuda, N.; Sohn, J.H.; Andre, V.A.M.; Barriga, S.; Cox, J.; Goetz, M. Management of Abemaciclib-Associated Adverse Events in Patients with Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: Safety Analysis of MONARCH 2 and MONARCH 3. Oncologist 2021, 26, e53–e65. [Google Scholar] [CrossRef]
  122. Costa, R.; Costa, R.B.; Talamantes, S.M.; Helenowski, I.; Peterson, J.; Kaplan, J.; Carneiro, B.A.; Giles, F.J.; Gradishar, W.J. Meta-analysis of selected toxicity endpoints of CDK4/6 inhibitors: Palbociclib and ribociclib. Breast 2017, 35, 1–7. [Google Scholar] [CrossRef]
  123. Onesti, C.E.; Jerusalem, G. CDK4/6 inhibitors in breast cancer: Differences in toxicity profiles and impact on agent choice. A systematic review and meta-analysis. Expert Rev. Anticancer Ther. 2021, 21, 283–298. [Google Scholar] [CrossRef]
  124. Weiss, J.; Goldschmidt, J.; Andric, Z.; Dragnev, K.H.; Gwaltney, C.; Skaltsa, K.; Pritchett, Y.; Antal, J.M.; Morris, S.R.; Daniel, D. Effects of Trilaciclib on Chemotherapy-Induced Myelosuppression and Patient-Reported Outcomes in Patients with Extensive-Stage Small Cell Lung Cancer: Pooled Results from Three Phase II Randomized, Double-Blind, Placebo-Controlled Studies. Clin. Lung Cancer 2021, 22, 449–460. [Google Scholar] [CrossRef]
  125. Fan, Y.; Xu, B. Current clinical trials on breast cancer in China: A systematic literature review. Cancer 2020, 126, 3811–3818. [Google Scholar] [CrossRef]
  126. Zhang, P.; Xu, B.; Gui, L.; Wang, W.; Xiu, M.; Zhang, X.; Sun, G.; Zhu, X.; Zou, J. A phase 1 study of dalpiciclib, a cyclin-dependent kinase 4/6 inhibitor in Chinese patients with advanced breast cancer. Biomark. Res. 2021, 9, 24. [Google Scholar] [CrossRef]
  127. Herrera-Abreu, M.T.; Palafox, M.; Asghar, U.; Rivas, M.A.; Cutts, R.J.; Garcia-Murillas, I.; Pearson, A.; Guzman, M.; Rodriguez, O.; Grueso, J.; et al. Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor-Positive Breast Cancer. Cancer Res. 2016, 76, 2301–2313. [Google Scholar] [CrossRef] [Green Version]
  128. McCartney, A.; Migliaccio, I.; Bonechi, M.; Biagioni, C.; Romagnoli, D.; de Luca, F.; Galardi, F.; Risi, E.; de Santo, I.; Benelli, M.; et al. Mechanisms of Resistance to CDK4/6 Inhibitors: Potential Implications and Biomarkers for Clinical Practice. Front. Oncol. 2019, 9, 666. [Google Scholar] [CrossRef]
  129. Xu, X.Q.; Pan, X.H.; Wang, T.T.; Wang, J.; Yang, B.; He, Q.J.; Ding, L. Intrinsic and acquired resistance to CDK4/6 inhibitors and potential overcoming strategies. Acta Pharmacol. Sin. 2021, 42, 171–178. [Google Scholar] [CrossRef]
  130. 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] [Green Version]
  131. Finn, R.S.; Dering, J.; Conklin, D.; Kalous, O.; Cohen, D.J.; Desai, A.J.; Ginther, C.; Atefi, M.; Chen, I.; Fowst, C.; et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 2009, 11, R77. [Google Scholar] [CrossRef] [Green Version]
  132. 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]
  133. 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] [Green Version]
  134. Gonzalez Curto, G.; der Vartanian, A.; Frarma, Y.E.; Manceau, L.; Baldi, L.; Prisco, S.; Elarouci, N.; Causeret, F.; Korenkov, D.; Rigolet, M.; et al. The PAX-FOXO1s trigger fast trans-differentiation of chick embryonic neural cells into alveolar rhabdomyosarcoma with tissue invasive properties limited by S phase entry inhibition. PLoS Genet. 2020, 16, e1009164. [Google Scholar] [CrossRef]
  135. Young, R.J.; Waldeck, K.; Martin, C.; Foo, J.H.; Cameron, D.P.; Kirby, L.; Do, H.; Mitchell, C.; Cullinane, C.; Liu, W.; et al. Loss of CDKN2A expression is a frequent event in primary invasive melanoma and correlates with sensitivity to the CDK4/6 inhibitor PD0332991 in melanoma cell lines. Pigment Cell Melanoma Res. 2014, 27, 590–600. [Google Scholar] [CrossRef]
  136. Dean, J.L.; Thangavel, C.; McClendon, A.K.; Reed, C.A.; Knudsen, E.S. Therapeutic CDK4/6 inhibition in breast cancer: Key mechanisms of response and failure. Oncogene 2010, 29, 4018–4032. [Google Scholar] [CrossRef] [Green Version]
  137. Cen, L.; Carlson, B.L.; Schroeder, M.A.; Ostrem, J.L.; Kitange, G.J.; Mladek, A.C.; Fink, S.R.; Decker, P.A.; Wu, W.; Kim, J.S.; et al. P16-Cdk4-Rb axis controls sensitivity to a cyclin-dependent kinase inhibitor PD0332991 in glioblastoma xenograft cells. Neuro Oncol. 2012, 14, 870–881. [Google Scholar] [CrossRef] [Green Version]
  138. Asghar, U.S.; Barr, A.R.; Cutts, R.; Beaney, M.; Babina, I.; Sampath, D.; Giltnane, J.; Lacap, J.A.; Crocker, L.; Young, A.; et al. Single-Cell Dynamics Determines Response to CDK4/6 Inhibition in Triple-Negative Breast Cancer. Clin. Cancer Res. 2017, 23, 5561–5572. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, B.; Li, R.; Wu, S.; Liu, X.; Ren, J.; Li, J.; Bi, K.; Wang, Y.; Jia, H. Breast Cancer Resistance to Cyclin-Dependent Kinases 4/6 Inhibitors: Intricacy of the Molecular Mechanisms. Front. Oncol. 2021, 11, 651541. [Google Scholar] [CrossRef]
  140. McClendon, A.K.; Dean, J.L.; Rivadeneira, D.B.; Yu, J.E.; Reed, C.A.; Gao, E.; Farber, J.L.; Force, T.; Koch, W.J.; Knudsen, E.S. CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy. Cell Cycle 2012, 11, 2747–2755. [Google Scholar] [CrossRef] [Green Version]
  141. Jansen, V.M.; Bhola, N.E.; Bauer, J.A.; Formisano, L.; Lee, K.M.; Hutchinson, K.E.; Witkiewicz, A.K.; Moore, P.D.; Estrada, M.V.; Sánchez, V.; et al. Kinome-Wide RNA Interference Screen Reveals a Role for PDK1 in Acquired Resistance to CDK4/6 Inhibition in ER-Positive Breast Cancer. Cancer Res. 2017, 77, 2488–2499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Mora, A.; Komander, D.; van Aalten, D.M.; Alessi, D.R. PDK1, the master regulator of AGC kinase signal transduction. Semin. Cell Dev. Biol. 2004, 15, 161–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Michaloglou, C.; Crafter, C.; Siersbaek, R.; Delpuech, O.; Curwen, J.O.; Carnevalli, L.S.; Staniszewska, A.D.; Polanska, U.M.; Cheraghchi-Bashi, A.; Lawson, M.; et al. Combined Inhibition of mTOR and CDK4/6 Is Required for Optimal Blockade of E2F Function and Long-term Growth Inhibition in Estrogen Receptor-positive Breast Cancer. Mol. Cancer Ther. 2018, 17, 908–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Romano, G.; Chen, P.L.; Song, P.; McQuade, J.L.; Liang, R.J.; Liu, M.; Roh, W.; Duose, D.Y.; Carapeto, F.C.L.; Li, J.; et al. A Preexisting Rare PIK3CA(E545K) Subpopulation Confers Clinical Resistance to MEK plus CDK4/6 Inhibition in NRAS Melanoma and Is Dependent on S6K1 Signaling. Cancer Discov. 2018, 8, 556–567. [Google Scholar] [CrossRef] [Green Version]
  145. De Leeuw, R.; McNair, C.; Schiewer, M.J.; Neupane, N.P.; Brand, L.J.; Augello, M.A.; Li, Z.; Cheng, L.C.; Yoshida, A.; Courtney, S.M.; et al. MAPK Reliance via Acquired CDK4/6 Inhibitor Resistance in Cancer. Clin. Cancer Res. 2018, 24, 4201–4214. [Google Scholar] [CrossRef] [Green Version]
  146. Tong, Z.; Sathe, A.; Ebner, B.; Qi, P.; Veltkamp, C.; Gschwend, J.E.; Holm, P.S.; Nawroth, R. Functional genomics identifies predictive markers and clinically actionable resistance mechanisms to CDK4/6 inhibition in bladder cancer. J. Exp. Clin. Cancer Res. 2019, 38, 322. [Google Scholar] [CrossRef]
  147. Teh, J.L.F.; Cheng, P.F.; Purwin, T.J.; Nikbakht, N.; Patel, P.; Chervoneva, I.; Ertel, A.; Fortina, P.M.; Kleiber, I.; HooKim, K.; et al. In Vivo E2F Reporting Reveals Efficacious Schedules of MEK1/2-CDK4/6 Targeting and mTOR-S6 Resistance Mechanisms. Cancer Discov. 2018, 8, 568–581. [Google Scholar] [CrossRef] [Green Version]
  148. Lee, M.S.; Helms, T.L.; Feng, N.; Gay, J.; Chang, Q.E.; Tian, F.; Wu, J.Y.; Toniatti, C.; Heffernan, T.P.; Powis, G.; et al. Efficacy of the combination of MEK and CDK4/6 inhibitors in vitro and in vivo in KRAS mutant colorectal cancer models. Oncotarget 2016, 7, 39595–39608. [Google Scholar] [CrossRef] [Green Version]
  149. Pek, M.; Yatim, S.; Chen, Y.; Li, J.; Gong, M.; Jiang, X.; Zhang, F.; Zheng, J.; Wu, X.; Yu, Q. Oncogenic KRAS-associated gene signature defines co-targeting of CDK4/6 and MEK as a viable therapeutic strategy in colorectal cancer. Oncogene 2017, 36, 4975–4986. [Google Scholar] [CrossRef]
  150. Ziemke, E.K.; Dosch, J.S.; Maust, J.D.; Shettigar, A.; Sen, A.; Welling, T.H.; Hardiman, K.M.; Sebolt-Leopold, J.S. Sensitivity of KRAS-Mutant Colorectal Cancers to Combination Therapy That Cotargets MEK and CDK4/6. Clin. Cancer Res. 2016, 22, 405–414. [Google Scholar] [CrossRef] [Green Version]
  151. Carter, J.; Cheng, L.; Zucker, J.; Marshall, M.; Pollok, K.; Murray, M.; Li, L.; Renbarger, J. Use of Precision Medicine Molecular Profiling of Baseline Tumor Specimen May Not Benefit Outcomes in Children With Relapsed or Refractory Pediatric Sarcomas. Clin. Pharmacol. Ther. 2017, 101, 328–330. [Google Scholar] [CrossRef] [Green Version]
  152. Vo, K.T.; Parsons, D.W.; Seibel, N.L. Precision Medicine in Pediatric Oncology. Surg. Oncol. Clin. N. Am. 2020, 29, 63–72. [Google Scholar] [CrossRef]
  153. Weiss, A.; Gill, J.; Goldberg, J.; Lagmay, J.; Spraker-Perlman, H.; Venkatramani, R.; Reed, D. Advances in therapy for pediatric sarcomas. Curr. Oncol. Rep. 2014, 16, 395. [Google Scholar] [CrossRef]
Figure 1. Targeting dysregulation of Cyclin D–CDK4/6 axis by CDK4/6 inhibition. Cyclin D-CDK4/6-mediated RB phosphorylation occurs upon activation of Cyclin D-CDK4/6 complex in response to extracellular signals such as stimulatory mitogens, leading to ensuing cell cycle progression. Initially, upon Cyclin D-CDK4/6-mediated RB phosphorylation, RB affinity to E2F is reduced, promoting transcription of genes such as Cyclin E2. Subsequently, the Cyclin E2-CDK2 complex hyperphosphorylates RB, which induces full dissociation of the RB-E2F complex and transcription of genes encoding proteins playing critical roles in the next phases of the cell cycle. The p16INK4A (p16) tumor suppressor encoded by CDKN2A directly binds to CDK4/6 and inhibits the formation of the Cyclin D-CDK4/6 complex and prevents Cyclin D-CDK4/6-mediated RB phosphorylation. A dysregulated Cyclin D–CDK4/6 axis, which leads to uncontrolled cell proliferation, can be inhibited by CDK4/6 inhibitors. Chronic inhibition of CDK4/6 can trigger the upregulation of PI3K and MAPK pathways to compensate for inhibitory effects of CDK4/6 inhibitors on cell cycle progression by activation of D type cyclins. Dual inhibition of CDK4/6 and PI3K/mTOR or MEK may be an efficient combination treatment to prevent the compensatory effect of the PI3K or MAPK pathways on the development of CDK4/6 inhibitor resistance.
Figure 1. Targeting dysregulation of Cyclin D–CDK4/6 axis by CDK4/6 inhibition. Cyclin D-CDK4/6-mediated RB phosphorylation occurs upon activation of Cyclin D-CDK4/6 complex in response to extracellular signals such as stimulatory mitogens, leading to ensuing cell cycle progression. Initially, upon Cyclin D-CDK4/6-mediated RB phosphorylation, RB affinity to E2F is reduced, promoting transcription of genes such as Cyclin E2. Subsequently, the Cyclin E2-CDK2 complex hyperphosphorylates RB, which induces full dissociation of the RB-E2F complex and transcription of genes encoding proteins playing critical roles in the next phases of the cell cycle. The p16INK4A (p16) tumor suppressor encoded by CDKN2A directly binds to CDK4/6 and inhibits the formation of the Cyclin D-CDK4/6 complex and prevents Cyclin D-CDK4/6-mediated RB phosphorylation. A dysregulated Cyclin D–CDK4/6 axis, which leads to uncontrolled cell proliferation, can be inhibited by CDK4/6 inhibitors. Chronic inhibition of CDK4/6 can trigger the upregulation of PI3K and MAPK pathways to compensate for inhibitory effects of CDK4/6 inhibitors on cell cycle progression by activation of D type cyclins. Dual inhibition of CDK4/6 and PI3K/mTOR or MEK may be an efficient combination treatment to prevent the compensatory effect of the PI3K or MAPK pathways on the development of CDK4/6 inhibitor resistance.
Cancers 14 03611 g001
Table 1. Dysregulation of the CDK4/6 cell cycle regulatory pathway in pediatric and AYA OS, RMS and EWS patients.
Table 1. Dysregulation of the CDK4/6 cell cycle regulatory pathway in pediatric and AYA OS, RMS and EWS patients.
Precision Medicine TrialsGenomic and Protein/RNA Biomarkers Associated with CDK4/6 Pathway
OSRMSEWS
INFORM [71]Point mutations in CDKN2B and CDK4 amplificationCDKN2A/B deletion or gain of CDK4 copy numberCCND2 amplification, CDKN2A/B deletion, CCND1 overexpression
The Precision in Pediatric Sequencing (PIPseq) [72]RB splice mutation CCNE1 over-expression
The individualized cancer therapy (iCat) [73]CCND1 and CCNE1 high copy number gainCDKN2A/B deletionCDKN2A homozygous loss or single copy number gain of CCND1
The molecular biology tumor board (MBB) [75]CDKN2A homozygous deletionCDK4 amplification
TRICEPS [76]CDKN2A copy loss/CDKN2A homozygous deletion and RB copy lossCDKN2A point mutation, RB point mutation and loss of heterozygosity
Phase 1 clinical trial program at MD Anderson Cancer Center [77]CDK4 amplificationCDK4 amplificationCDK4 amplification
MSKCC [78]CDK4 amplification and CDKN2A deletion/mutation
The Zero Childhood Cancer Program [67]CNV/SV of CCNE1 and RB and CCNE1 over-expressionCNVs of CDK4, CDKN2A/B, CCND2; or CCND3 overexpression of CDK4, CCND3, or CCNE1; and down-regulation of CDKN2A/BHomozygous deletion of CDKN2A/B
The St. Jude Children’s Research Hospital—Pediatric Cancer Genome Project [79]SV of CCND3 and RB point mutation/SV
CRB cancer des Hôpitaux de Toulouse; BB-0033-00014 [80]Stop-gained mutation in RB
Complementary genomic study of OS patients [81]CDKN2A/B deletions
MOSCATO-01 [82]RB deletionCDKN2A deletionCDKN2A/B deletions
Clin Omics Program [83] CDKN2A and RB loss of heterozygosityCDKN2A homozygous loss
RMS Case study [84] CDK4 amplification
Molecular profiling of FET-TFCP2 RMS patients [57] CDKN2A homozygous deletion
Molecular profiling of EWS patients [85] CDKN2A deletion
Table 2. Ongoing clinical trials on CDK4/6 inhibitors in pediatric and AYA sarcoma patients.
Table 2. Ongoing clinical trials on CDK4/6 inhibitors in pediatric and AYA sarcoma patients.
Disease TreatmentPhaseClinicalTrial.gov Identifier
Recurrent or refractory RB-positive solid tumors, including OS, EWS, and RMSCDK4/6 inhibitor (palbociclib)2NCT03526250
Recurrent or refractory solid tumors, including EWS and RMSCDK4/6 inhibitor (palbociclib) in combination with temozolomide and irinotecan, and/or with topotecan and cyclophosphamide.2NCT03709680
Recurrent or refractory RB-positive solid tumors, including OS, EWS, and RMSCDK4/6 inhibitor (palbociclib) and other targeted therapies 2NCT03155620
Soft tissue sarcomas, RMSCDK4/6 inhibitor (ribociclib) in combination with doxorubicin hydrochloride)1NCT03009201
Recurrent or refractory solid tumors, including OS, EWS, and RMSCDK4/6 inhibitor (abemaciclib)1NCT02644460
Soft tissue and bone sarcoma, including OSCDK4/6 inhibitor (abemaciclib)2NCT04040205
EWSCDK4/6 inhibitor (palbociclib) in combination with ganitumab2NCT04129151
Relapsed or refractory solid tumors, including OS, EWS, and RMSCDK4/6 inhibitor (abemaciclib) in combination with irinotecan and temozolomide1NCT04238819
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Barghi, F.; Shannon, H.E.; Saadatzadeh, M.R.; Bailey, B.J.; Riyahi, N.; Bijangi-Vishehsaraei, K.; Just, M.; Ferguson, M.J.; Pandya, P.H.; Pollok, K.E. Precision Medicine Highlights Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric, Adolescents and Young Adult Sarcomas. Cancers 2022, 14, 3611. https://doi.org/10.3390/cancers14153611

AMA Style

Barghi F, Shannon HE, Saadatzadeh MR, Bailey BJ, Riyahi N, Bijangi-Vishehsaraei K, Just M, Ferguson MJ, Pandya PH, Pollok KE. Precision Medicine Highlights Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric, Adolescents and Young Adult Sarcomas. Cancers. 2022; 14(15):3611. https://doi.org/10.3390/cancers14153611

Chicago/Turabian Style

Barghi, Farinaz, Harlan E. Shannon, M. Reza Saadatzadeh, Barbara J. Bailey, Niknam Riyahi, Khadijeh Bijangi-Vishehsaraei, Marissa Just, Michael J. Ferguson, Pankita H. Pandya, and Karen E. Pollok. 2022. "Precision Medicine Highlights Dysregulation of the CDK4/6 Cell Cycle Regulatory Pathway in Pediatric, Adolescents and Young Adult Sarcomas" Cancers 14, no. 15: 3611. https://doi.org/10.3390/cancers14153611

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