Vol 19, No 4 (2023)
Review paper
Published online: 2023-01-27

open access

Page views 1293
Article views/downloads 1140
Get Citation

Connect on Social Media

Connect on Social Media

Diagnosis and treatment of rhabdomyosarcomas

Michał Łomiak12, Tomasz Świtaj1, Mateusz Spałek1, Joanna Radzikowska3, Marzanna Chojnacka4, Sławomir Falkowski1, Michał Wągrodzki5, Wojciech Kukwa3, Anna Szumera-Ciećkiewicz5, Piotr Rutkowski1, Anna M. Czarnecka1
Oncol Clin Pract 2023;19(4):250-279.

Abstract

Rhabdomyosarcoma (RMS) is a soft tissue sarcoma. The primary tumor is most commonly localized in the head and neck, the urogenital system, or the limbs. Classification by the World Health Organization has distinguished four histopathological RMS subtypes: embryonal, alveolar, pleomorphic, and spindle cell/sclerosing. Differential diagnosis of RMS includes melanoma, malignant neoplasm of peripheral nerve sheaths, liposarcoma, and PEComa. Among typical cytogenetic changes in RMS are chromosomal translocations t(2;13)(q35;q14) and t(1;13) (p36;q14). They lead to the formation of fusion genes that have a prognostic value. In the course of RMS, changes may also be present in signaling pathways, including RAS-PI3K, Wnt/b-catenin, receptor tyrosine kinase pathways, and myogenesis regulation. In 30% of patients at the time of diagnosis of RMS, distant metastases are present, most commonly to lungs, lymph nodes, bones, and bone marrow. Treatment of patients with RMS requires a multidisciplinary approach, and steadily perfected diagnostic techniques contribute to the individualization of therapeutic strategies. Optimal treatment of localized RMS is based on surgery combined with radiotherapy and chemotherapy. If distant metastases are present, the basic therapeutic method is multidrug chemotherapy, most frequently based on vincristine, dactinomycin, ifosfamide/cyclophosphamide, and etoposide. Despite intensive treatment, the 5-year survival index for RMS is not greater than 50%. There are still no unequivocal guidelines concerning the treatment in patients with local or distant recurrences.

REVIEW ARTICLE

Translation: Prof. Ewa Bartnik

Oncology in Clinical Practice

DOI: 10.5603/OCP.2022.0034

Copyright © 2023 Via Medica

ISSN 2450–1654

e-ISSN 2450–6478

Diagnosis and treatment of rhabdomyosarcomas

Michał Łomiak12*Tomasz Świtaj1*Mateusz Spałek1*Joanna Radzikowska3Marzanna Chojnacka4Sławomir Falkowski1Michał Wągrodzki5Wojciech Kukwa3Anna Szumera-Ciećkiewicz5Piotr Rutkowski1Anna M. Czarnecka1
1Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
2Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
3Department of Otorhinolaryngology, Faculty of Dental Medicine, Medical University of Warsaw, Warsaw, Poland
4Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
5Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
(*) equal participation

Address for correspondence:

Prof. Anna M. Czarnecka

Department of Soft Tissue/Bone Sarcoma

and Melanoma Maria Sklodowska-Curie

National Research Institute of Oncology,

ul. Roentgena 5, 02–781 Warsaw, Poland

tel.: +48 22 5462027

e-mail: am.czarnecka@coi.pl

Received: 28.09.2021 Accepted: 29.09.2021 Early publication date: 27.01.2023

ABSTRACT

Rhabdomyosarcoma (RMS) is a soft tissue sarcoma. The primary tumor is most commonly localized in the head and neck, the urogenital system, or the limbs. Classification by the World Health Organization has distinguished four histopathological RMS subtypes: embryonal, alveolar, pleomorphic, and spindle cell/sclerosing. Differential diagnosis of RMS includes melanoma, malignant neoplasm of peripheral nerve sheaths, liposarcoma, and PEComa. Among typical cytogenetic changes in RMS are chromosomal translocations t(2;13)(q35;q14) and t(1;13)(p36;q14). They lead to the formation of fusion genes that have a prognostic value. In the course of RMS, changes may also be present in signaling pathways, including RAS-PI3K, Wnt/b-catenin, receptor tyrosine kinase pathways, and myogenesis regulation. In 30% of patients at the time of diagnosis of RMS, distant metastases are present, most commonly to lungs, lymph nodes, bones, and bone marrow. Treatment of patients with RMS requires a multidisciplinary approach, and steadily perfected diagnostic techniques contribute to the individualization of therapeutic strategies. Optimal treatment of localized RMS is based on surgery combined with radiotherapy and chemotherapy. If distant metastases are present, the basic therapeutic method is multidrug chemotherapy, most frequently based on vincristine, dactinomycin, ifosfamide/cyclophosphamide, and etoposide. Despite intensive treatment, the 5-year survival index for RMS is not greater than 50%. There are still no unequivocal guidelines concerning the treatment in patients with local or distant recurrences.

Key words: rhabdomyosarcoma, sarcoma, soft tissue sarcomas, RMS, RAS, translocation

Oncol Clin Pract 2023; 19, 4: 250279

Introduction

Sarcomas are a heterogeneous group of malignant neoplasms derived from mesenchymal tissue. The usual classification includes sarcomas derived from bone and soft tissue sarcomas [1]. Rhabdomyosarcoma (RMS) is a soft tissue sarcoma, whose cells differentiate in the direction of striated muscles, which is proved by the expression of skeletal muscle markers [2]. The current World Health Organization (WHO) clas­sification divides RMS into four histological types: alveolar rhabdomyosarcoma (ARMS), embryonic rhabdomyosarcoma (EMRS), pleomorphic rhabdo­myosarcoma (PRMS) and spindle cell/sclerosing rhabdomyosarcoma (SCRMS) [3]. In the last several decades, a small improvement in the survival of patients with RMS has been observed. This is due to the development of diagnostic methods allowing correct diagnosis of the disease and improving access to multidisciplinary treatment, including modern radiotherapy (RT) techniques. Establishing uniform standards of the procedure that included pediatric treatment protocols in the therapy of adult patients has led to the increase of the 5-year survival rate from 36% to 54% in comparison to treatment with other protocols [4]. Nevertheless, the diversity of anatomical localization of primary RMS tumors, limited methods of treating metastatic disease, and the current lack of targeted therapies make for an unfavorable prognosis in this group of patients. In retrospect, the median overall survival (OS) for RMS patients with distant metastases is from 7 to 22 months [5–7]. Direct toxicity (e.g. cardiotoxicity) and other undesirable effects (e.g. neutropenia) of cytotoxic drugs used in RMS treatment, both in the localized stage and in the disseminated one, remain a large limitation in the choice of optimal treatment [8].

Table 1. Genetic syndromes predisposing to rhabdomyosarcoma

Genetic syndromes

Responsible genes

References

Beckwith-Wiedemann

IGF2, CDKN1C, H19, and KCNQ1OT1

[23]

Costello

HRAS

[24]

DICER1

DICER1

[25]

Type 1 neurofibromatosis

NF1

[19, 26, 27]

Li-Fraumeni

TP53

[15]

Noonan

BRAF, KRAS, NRAS, PTPN11, RAF1 and SOS1

[16]

CMMRD

MLH1, MSH2, MSH6, PMS2

[28]

Rubenstein-Taybi

CREBBP

[29]

Hereditary retinoblastoma

RB1

[30]

Gorlin

PTCH1, PTCH2, and SUFU

[31, 32]

Cardiofaciocutaneous

BRAF, MAP2K1, MAP2K2, KRAS

[33]

Epidemiology

The incidence of soft tissue sarcomas in Poland is determined to be 45 cases per 100 000 persons, which is about 1000 new patients per year [9]. Simultaneously they constitute not more than one percent of all malignant neoplasms in adults [9–11]. Rhabdomyosarcoma is very rare in adults and is responsible for about 3% of soft tissue sarcomas [12]. Epidemiological analysis of 2600 patients with RMS indicated that slightly over 40% of all RMS cases are in adults [13]. These sarcomas occur four times more frequently in the white than in the black population The largest cohort of adults with RMS described so far includes 1071 persons [13]. Moreover, there is a small number of publications from reference centers in Europe, the United States, and Asia, which includes groups from several dozen to several hundred patients. The median age of adults with RMS is very differentiated and varies from 26 to 71.5 years [7, 14].

Risk factors

Based on the available literature two main groups of risk factors for RMS can be distinguished, that is genetic and environmental factors. Persons with some heritable genetic syndromes including Li-Fraumeni [15] or Noonan syndrome [16] are at an increased risk of RMS (Tab. 1). Most patients with RMS do not have any first-degree relatives with neoplastic diseases in their medical history; however, neoplasms among first-degree relatives under 30 years of age are more frequent in RMS patients than in the healthy population. Congenital RMS cases have also been described [17, 18]. Among other factors which increase the risk of RMS are congenital defects [19], prenatal exposure to ionizing radiation [20], cocaine and cannabinoid use by the mothers and fathers [21], or pre-term birth [22].

Table 2. Changes in intracellular signal pathways in rhabdomyosarcoma

Signal pathway

Altered genes

Remarks

References

RAS-PI3K

NRAS, KRAS, HRAS PTPN11, NF1, BRAF,

PIK3CA

Over 80% of RMS tumors show PI3K pathway activation.

In 1/3 of FNRMS tumors, RAS pathway perturbations are present.

Mutations of the PI3K/AKT signal pathway define an ERMS subgroup with an unfavorable clinical course

[57, 58, 72–75]

RTK

FGFR2, FGFR4, IGF1R, ERBB2, EPHA3, EFNA1, PDGFRA

FGFR4 mutations occur in about 7% of FNRMS

IGFR1 overexpression is observed in FPRMS

PDGFRA gene overexpression is characteristic of FPRMS tumors

[46, 76–80]

Oncogenesis suppression pathways

PTEN, TP53, MDM2, CDKN2A, CDKN1C

PTEN gene mutation occurs in FNRMS tumors

TP53 gene mutation occurs in about 12% of FNRMS tumors

[36, 81–83]

Wnt/b-catenin

CTNNB1

CTNNB1 gene mutation (encoding b-catenin) is common in FNRMS tumors

[83]

Sonic Hedgehog signal pathway

GLI1

In ERMS an excess of genetic material from the 12q13 may be present, where the transcription factor GLI1, which is frequently overexpressed, is located

[57]

Pathways of regulating epigenetics and myogenesis

MYOD1, BCOR, ARID1A

MYOD1 mutations are characteristic of a particularly aggressive form of FNRMS

[57, 64, 84]

Pathogenesis

Even though RMS cells differentiate in the direction of myoblasts, it is not clear if they develop from the same cell lines from which striated muscle differentiates. Considering the anatomical variety of this neoplasm and the range of its oncogenic lesions, it can be assumed that they attain the myoblast phenotype by induction of the expression of genes characteristic of skeletal muscle [34]. Moreover, it has been shown that RMS can form as a result of oncogene expression both in skeletal myoblast cell lines [35, 36] and in non-myogenic cell lines [37]. Rhabdomyosarcomas may be derived from tissues such as skin, fat, or nerves [38]. The pathogenesis of RMS is based both on genetic material mutations (see subchapter 5) and the resulting perturbations of signal transduction pathways regulating cell function (Tab. 2). A high tumor mutational burden defined as the sum of somatic mutations of the genetic material of the tumor correlates with a poorer prognosis for RMS patients [39].

Cytogenetic aberrations

Alveolar RMS

The most characteristic chromosomal translocations present in ARMS are t(2;13)(q35;q14) and t(1;13)(p36;q14), leading to the formation of fusion genes PAX3-FOXO1 and PAX7-FOXO1 [40]. The former of these translocations are more common (55% vs. 23%). Their presence is associated with a poorer clinical prognosis [41]. The protein products of these genes are transcription factors. Their expression is stronger in comparison to the products of corresponding genes which did not undergo fusion (wild-type) [42, 43]. Additionally, the products of fusion genes are stabilized at the post-translational stage by the phosphorylation of the chimeric protein, which decreases their intracellular degradation [44]. Transcription factor PAX3-FOXO1 increases the expression of the following genes: ALK receptor tyrosine kinase gene, encoding the anaplastic lymphoma kinase, FGFR4, fibroblast growth factor receptor 4 gene, protooncogene MYCN, MYOD1 myogenic differentiation 1 gene, and MYOG (myogenin gene) [34]. In about 80% of ARMS, strong cytoplasmic ALK expression has been observed, most commonly associated with the amplification of this gene. In single cases, the presence of mutations has also been observed (substitution or the loss of a whole exon) [45]. Moreover, PAX3-FOXO1 interacts with proteins participating in modulating chromatin activity including BRD4, (bromodomain-containing protein 4) and CHD4 (chromodomain helix DNA-binding protein 4) [46, 47]. A role in the development and invasiveness of RMS is also played by excessive activation of the MET protooncogene, which takes place probably as a result of the activity of the fusion protein [48]. Amplification of 13q31 with the MIR17HG region (encoding miR-17-92, which also undergoes amplification in other neoplasms), occurring mainly in ARMS with the PAX7-FOXO1 fusion, is probably associated with a poorer clinical course of the disease [49]. The 12q13-14, amplification with the CDK4 locus characteristic almost exclusively for ARMS with the PAX3-FOXO1 fusion, has a similar prognostic value [50]. In about 20% of ARMS, the fusion of FOXO1(FKHR) or PAX3 is not present. These tumors, both in the molecular and clinical aspect, resemble ERMS (embryonal RMS) more closely, which indicates the key role of genetic diagnosis and creates a natural division of ARMS into fusion-positive (FPRMS) and fusion-negative (FNRMS) types [38]. For this reason, molecular investigations aimed at identifying PAX/FOXO1 fusions are recommended in all cases of alveolar and embryonal RMS [51] (Tab. 3).

Table 3. Molecular analyses to identify the PAX/FOXO1 fusion

Alveolar rhabdo- myosarcoma

PAX3-FOXO1

PAX7-FOXO1

PAX3-FOXO4

PAX3-NCOA1

PAX3-NCOA2

FOXO1-FGFR1

t(2;13)(Q35;Q14)

t(1;13)(p36;q14)

t(X;2)(q13;q36)

t(2;2)(p23;q36)

t(2;8)(q36;q13)

t(8;13;9)(p11;q14;q32)

Embryonal ERMS

In as many as 2550% of ERMS tumors, chromosomal number aberrations are present [52]. They generally concern additional copies of chromosome pairs 2, 7, 8 (even in 70% of cases), 11, 12, 13, and 20 [53]. The loss of chromosome pairs 9 and 10 and 15 is described in 30% of ERMS cases. If gene amplification occurs, it is detected in chromosome regions 12q13-q15, whereas in region11p15.5. homo- or heterozygous deletions are common. Moreover, in this region, the phenomenon of uniparental disomy and gene imprinting may occur [54–56]. Mutations in oncogenes and suppressor genes are more characteristic for ERMS than for ARMS. In both subtypes, changes in cellular signaling associated with receptors for growth factors RAS/PI3K are common through somatic mutations (more common in ERMS) or changes in the expression of key genes for this pathway (through specific fusions in ARMS) (Tab. 2, 4) [57].

Pleomorphic RMS

RMS pathogenesis at the molecular level is poorly characterized. A complex karyotype is most commonly present in PRMS with numerous structural and numerical aberrations but without specific changes, which has been confirmed by molecular analyses [53, 58]. A common gene mutation in PRMS is probably a mutation of the TP53 gene, especially in tumors appearing at a young age [59].

Spindle cell/sclerosing RMS

Spindle cell/sclerosing rhabdomyosarcoma is the most recent RMS to be distinguished in the histopathological classification. Within this subtype successive ones are distinguished with characteristic molecular changes and clinical appearance [60]. The first group are variants with rearrangement of the VGLL2/NCOA2 genes, which occur in children under the age of 5 years or as congenital neoplasms [61] (Tab. 5).

Table 4. Histopathological differential diagnosis of Rhabdomyosarcoma (RMS)

Neoplasm

Morphological characteristics

Immunohistochemical markers

Other information

Keratin

Desmin

Protein S100

Myogen /MyoD1

HMB-45/Melan A

SMA

ERMS

Ovoid and star-shaped cells loosely placed in myxoid stroma, less commonly morphology of small, round cells, presence of cells with the character of immature rhabdomyoblasts

+/–

+

–/+

+

+/–

Lack of characteristic cytogenetic marker, in most cases loss of heterozygosity at locus 11p15 [136]

ARMS

Small, round, and monomorphic myoblasts separated by empty oval or elongated spaces (similar to lung tissue structure)

+/–

+

–/+

+

+/–

7090% of cases show translocations: t(2;13)(q35;q14) or t(1;13)(p36;q14) [137], FISH with set of FOXO1 (FKHR) and PAX3 probes used in diagnosis

Ewing sarcoma

Visible areas composed of small monomorphic cells; Homer-Wright rosettes present

+/–

–/+

Positive staining for FLI-1 in about 80% of cases [138], Positive staining for CD99

Melanoma

Pleomorphic, epithelial, or fusiform cells with poor cohesion, melanin, presence of distinct nucleoli

+

+

BRAF mutations in approx. 50% of patients

MPNST

Various numbers of cells within neoplastic lesions, cells in bundles, coils, or “herringbone pattern”

+/–

In over 80% of cases neurofibromin 1 (NF1) gene mutations present [139], loss of nuclear expression of H3K27me3/INI1

PEComa

Perivascular proliferation of epithelial and fusiform cells with light, acidophilic

cytoplasm with granulocytes; nucleoli visible

+/–

+

+

In about 80% of cases deletions and/or loss of heterozygosity (LOH) in the 16p13.3 region [140, 141]

DFSP

Large density of cell arrangement in histological appearance with poorly visible borders of neoplastic lesions, radial arrangement of cells with fusiform morphology

Characteristic translocation t(17;22)(q22;q13) [142], CD34 expression [143]

Table 5. Variants with rearrangement of the VGLL2/NCOA2 genes occurring in children under the age of 5 years or as congenital neoplasms

Spindle cell/sclerosing rhabdomyosarcoma congenital/infant

SRF-NCOA2

TEAD1-NCOA2

VGLL2-NCOA2

VGLL2-CITED2

t(6;8)(p21;q13)

t(8;11)(q13;p15)

t(6;8)(q22;q13)

t(6;6)(q22;q24)

They are characterized by a good prognosis with a tendency to local recurrence [62]. Other possible gene fusions occurring in RMS with this clinical presentation are SRF/NCOA2, TEAD1/NCOA2, VGLL2/CITED2 [60]. A separate group is spindle cell/sclerosing RMS with a somatic activating mutation of the MYOD1 gene at position Lys122, occurring both in children and adults [63]. The mutation may be homo- or heterozygous and the mutated gene interacts with the MYC oncogene [64]. Tumors of this type are characterized by a poor prognosis, especially in children and adolescents [63]. In adolescents the mutation MyoD1 p.Leu122Arg associated with a very poor prognosis is characteristic. The third group are patients with spindle cell RMS without molecular changes and the fourth patients with a diagnosis of spindle cell RMS developing in bones with EWSR1/FUS-TFCP2 or MEIS1-NCOA2 translocations. In adults the presence of bone tumors is correlated with very poor prognosis and is characteristic for spindle cell/sclerosing RMS with the fusion of MEIS1/NOAC2 or EWSR1/ /FUS-TFCP2 genes, and so far it is known only from the description of several dozen cases [65–70]. The remaining cases of spindle cell RMS, which do not have the alterations described above, occur most commonly around the area of the testes or within the abdominal cavity [71].

Figure 1. Diagnostic algorithm procedure for rhabdomyosarcoma (RMS). Based on [51]; IHC immunohistochemistry staining; MRI magnetic resonance imaging; PET positron emission tomography; PET-CT positron emission tomography-computed tomography; PMR spinocerebral fluid

Clinical picture

Rhabdomyosarcoma may occur in almost any anatomical localization, most commonly in the extremities (approx. 25%), head and neck region (approx. 20%), and urogenital tract (approx. 20%) [13]. Localization within the head and neck also encompasses the area of the eye socket [85] and the parameningeal area including the nasal cavity [86], sinuses [87–89], the nasopharyngeal cavity [90], and the subtemporal fossa. It may develop within the parotid glands [91], the thyroid [92], and the oral cavity [93]. RMS within the urogenital tract may occur, among others, in the bladder [94], prostate [95], urethra [96], uterus [97, 98], vulva [99], or scrotum [100]. In adult RMS patients it more commonly develops in an unfavorable anatomical localization, i.e. other than the head and neck (except for parameningeal areas), urogenital tract (except for the bladder and prostate), and the biliary pathways [13, 101, 102]. A few cases of RMS are described in more rare localizations such as the liver [103], breast [104, 105], mediastinum [106], bronchi and lung [107, 108], cardiac muscle [109], pericardium [110], diaphragm [111], retroperitoneal space [112, 113], esophagus [114], stomach [115], or ileum [116].

Primary RMS is, in general, characterized by rapid and aggressive growth with the formation of a pseudobursa. The multiplicity of possible localizations is associated with a differentiated clinical picture. In the initial stages, the course of the disease may be asymptomatic [38]. The symptoms of focal damage of the nervous system appear in the case of RMS of the perimeningeal area. Within the eye socket, it may cause exophthalmos, perturbations of eyeball mobility, or vision perturbations [117–119]. Localized within the head and neck it may give symptoms of chronic or acute sinusitis, purulent or bloody discharge from the nasal cavity or ear canal, their obturation, or swallowing difficulti es [55, 120]. Because of aggressive growth in a limited anatomical space, cranial nerve paralysis may occur [121]. Rhabdomyosarcoma localized within the urogenital pathways, pelvis minor, or the abdominal cavity may give various symptoms such as chronic abdominal pain [122], bleeding from the birth canal [97], dysuria [123], jaundice [124], intussusception [125], or intestinal obstruction [126]. Edema, often painless, appearing in the case of RMS of the extremities or in the vicinity of the genital organs, may be ascribed to mechanical damage, which delays appropriate diagnosis. There are also descriptions of cases of disseminated RMS, where the first symptoms were perturbations of muscle strength of the extremities or limb paralysis [98]. Metastases may disseminate both through the lymphatic system and blood vessels [127, 128]. The most common localizations of metastases encompass the lungs, lymph nodes, and bone marrow. However, RMS may give metastases to almost all organs. In the literature, there are descriptions of metastatic RMS foci in the breast, peritoneum, pleura, central nervous system, and skin [14, 129, 130]. Bone metastases may manifest as bone pain and hypercalcemia and massive occupation of the bone marrow may cause symptoms typical for leukemia (cytopenia, bleeding, and infections) [45, 131–133]. Epidemiological analysis including 1017 adults with RMS indicated that over 28% of distant metastases were present at the moment of diagnosis, whereas regional dissemination (occupation of regional lymph nodes or primary lesions crossing the boundaries of the primary organ) was present in over 25% of the patients [5].

Diagnostic procedure

Preliminary diagnosis

If RMS is suspected, it is indispensable to carefully plan the whole diagnostic procedure (Fig. 1). The procedure is initiated by coarse needle biopsy with a pathological diagnosis in a sarcoma treatment reference center. Before performing the biopsy, it is recommended to evaluate the progress of the disease by visual imaging [computed tomography (CT) or magnetic resonance imaging (MRI)] of the primary focus and the regional lymph flow to optimally plan the biopsy and eventual further surgical treatment. A significant element of primary diagnosis is also a clinical and radiological evaluation of not only regional but also distant lymph nodes, whose occupation constitutes a generalized neoplastic disease. The criteria of occupation of lymph nodes in RMS have so far not been standardized, and, generally, a node larger than 1 cm in diameter is considered suspicious, regardless of its appearance in radiological imaging [56].

Figure 2. Alveolar rhabdomyosarcoma; A. Typical architecture with pseudoalveolar spaces (H&E, 40×); B. Neoplastic cells with acidophilic cytoplasm and eccentric cell nucleus differentiation in the direction of rhabdomyoblasts (H&E, 100×); C. In general, strong and diffuse myogenin expression (IHC clone F5D, Dako, 200×)
Histopathological diagnosis

Histopathological diagnosis of RMS is difficult, which is confirmed by the statistics of the international Intergroup Rhabdomyosarcoma Study Group (IRSG), according to which every fifth RMS diagnosis was incorrect [101]. In a light or electron microscope, cells can be seen that show differentiation in the direction of skeletal muscle cells “myoblast-like cells”. The next step is to perform immunohistochemical staining for the expression of proteins characteristic for muscle, which include muscle-specific actin and myosin, desmin, myoglobin, the MyoD1 protein, and myogenin [134]. The last two proteins are considered the most important markers of rhabdomyoblastic neoplasm differentiation [52]. Morphologically, RMS myoblasts can have different forms: they may be poorly differentiated (spherical, oval), fusiform, or fully differentiated [52]. Highly differentiated rhabdomyoblasts present as spherical or oval cells containing an acidophilic grainy cytoplasm with an eccentric or central spherical, single or double nucleus [52]. It should be, however, kept in mind that demonstrating rhabdomyoblastic differentiation among the cells of a neoplasm does not by itself determine an RMS diagnosis, as other neoplasms such as mesenchymal chondrosarcomas or sarcomatoid cancers are also characterized by the presence of these cells [135]. Rhabdomyosarcoma embryonal and alveolar belongs to the group of small round blue cell tumors, which are characterized by a low grade of differentiation and morphological similarity (small cell with a large, spherical strongly hyperchromatic nucleus, staining navy blue with hematoxylin) [55]. Other neoplasms that must be considered in the differential diagnosis of RMS are malignant peripheral nerve sheath tumor (MPNST), dermatofibrosarcoma protuberans (DFSP), perivascular epithelioid cell tumors (PEC-oma), melanoma, Ewing sarcoma, ectomesenchymoma, sarcoma-like cancer, including skin and salivary gland cancer, melanoma, liposarcoma, malignant teratoma, anaplastic thyroid cancer and neoplasms derived from nervous tissue. Differential diagnosis is presented in Table 4.

Figure 3. Embryonal Rhabdomyosarcoma of the vaginal wall of a 5-year-old patient (botryoid alveolar subtype); A. Primitive, small and ovoid neoplastic cells with poorly expressed differentiation in the direction of rhabdomyoblasts, placed loosely in a myxoid stroma. Supra-epithelial densification of cells visible the cambial layer H&E, 100×); B. Myogenin expression is generally visible only focally (IHC clone F5D, Dako, 200×)

During histopathological diagnosis, the RMS subtype must be established.

Figure 4. Pleomorphic rhabdomyosarcoma; A. Atypical polygonal and fusiform neoplastic cells typical for pleomorphic sarcomas; B. Desmin expression is generally multifocal or diffuse, but this marker does not allow for reliable differentiation of RMS from pleomorphic leiomyosarcoma (IHC clone D33, Dako, 40×); C. Myogenin is a highly specific marker for RMS, but in pleomorphic RMS, its expression may be poor (IHC F5D, Dako, 200×)
Alveolar RMS

Alveolar RMS constitutes about 25% of all RMS diagnoses in adults [13, 144]. In the adult population, it is most common in 1025 years old but may occur at any age. It is often localized in the soft tissues of the extremities, head and neck, retroperitoneal space, the urinary bladder, or the reproductive organ [145]. It is made up of tightly placed, small, circular, and monomorphic cells separated by empty oval or elongated spaces, which resemble lung alveoli in the histopathological picture. Cell aggregates may also be separated by connective tissue. The cells show a high nucleo-cytoplasmic ratio and are characterized by a high mitotic index. Some ARMS do not have the characteristic segmented location of cells, and their cells are uniformly clustered (the so-called solid form of ARMS), which makes the differential diagnosis with ERMS difficult [146]. Molecular methods including FISH with the FOXO1A set of probes are useful in diagnosing tumors with a solid structure, less characteristic for ARMS (Fig. 2).

Embryonal RMS

According to the available data including the largest group of adult patients with RMS, the frequency of occurrence of this subtype is 2030% of all histopathological RMS diagnoses [144, 147]. A frequent site for this neoplasm is the head and neck, in particular the eye socket, tissues associated with the meninges, middle ear, nasopharyngeal cavity, and the urogenital system, soft tissues of the extremities, the pelvis, and the retroperitoneal space [148]. It is built of acidophilic primitive ovoid cells, less commonly of round cells resembling immature rhabdomyoblasts. They are loosely disseminated in myxoid stromal tissue [149]. The cells are not distributed in an alveolar fashion characteristic for ARMS [149]. Cellular composition of ERMS reflects embryonal striated muscle development as very poorly differentiated cells up to fully differentiated cells may appear [52]. Anaplastic cells with a hyperchromatic enlarged nucleus are present in about 313% ERMS, and their presence may correlate with a poorer prognosis for the patients [150, 151]. In the botryoid form of ERMS (botryoid RMS), neoplastic cells form a layer called the cambial layer. This ERMS subtype with a good prognosis is characterized by a linear placement of neoplastic cells and occurs, in general, in the vicinity of mucous membranes, e.g. in the bladder [60]. Botryoid ERMS also often occupies the vagina, biliary pathways, the nasopharynx, and the nasal cavity [152]. In the anaplastic form of ERMS, which is a subtype with a poorer prognosis, the cells have an atypical multiform morphology. Immunohistochemical staining for myogenin indicates a heterogeneous and punctate expression of this protein in ERMS cells, but expression can also be uniform [153] (Fig. 3).

Figure 5. Spindle cell/sclerosing rhabdomyosarcoma; A. Fusiform morphology of cells arranged in long bundles (H&E, 100×); B. Another part of the same tumor with a more sclerosing and homogeneous stroma and spherical cells sclerosing variant of RMS (H&E, 100×). Spindle cell/sclerosing rhabdomyosarcoma is the morphological spectrum of the same neoplasm (both photos: H&E, 100×); C. Nuclear myogenin expression does not differentiate between particular RMS subtypes (IHC clone F5D, Dako, 100×); D. Strong and diffuse MyoD1 expression, often stronger than myogenin expression, is characteristic of SCRMS and is generally a result of the MYOD1 gene mutation at position L122R (IHC clone 5.8A, Dako, 100×)
Pleomorphic RMS

Pleomorphic RMS (PRMS)occurs almost exclusively in adults, in particular, after the sixth decade of life, and constitutes even up to 43% of RMS in adults [154, 155]. Its localization is most commonly in soft tissues of the lower extremities (especially the thighs), retroperitoneal space, abdominal cavity, chest, spermatid cord, and the vicinity of the testes [156]. The histopathological picture shows a very low degree of differentiation in the direction of rhabdomyoblasts and requires careful differentiation with undifferentiated pleomorphic sarcoma [38]. Cells that show signs of atypia are pleomorphic; they have shapes from small epithelial to large cells with segmented nuclei and distinct nucleoli. They may be placed in groups, be linear, or be irregularly disseminated [38]. Cellular pleomorphism is diffuse in contrast to disseminated anaplastic cells in ERMS. PRMS is also characterized by poor myogenin expression, however, the largest study including 38 cases indicated that each of the tumors showed positive staining for at least one skeletal muscle marker [60] (Fig. 4).

Spindle cell/sclerosing RMS

In 2013, SCRMS was distinguished as a distinct variant of RMS on the basis of its genetic profile, whereas previously it had been identified as an ERMS subtype [3, 157]. According to WHO, it is the rarest RMS subtype [158]. In the pediatric population, SCRMS is associated with a better prognosis, but in the few descriptions of cases with this neoplasm in adult patients, this no longer holds [159, 160]. Spindle cell/sclerosing rhabdomyosarcoma with the presence of the NCOA2 and VGLL2 translocation is correlated with a better prognosis in infants [62]. A poorer clinical prognosis is characteristic of tumors in the parameningeal area and associated with MYOD1 [62, 161]. In adults, the most common SCRMS localization is the head and neck, less commonly, the extremities and the retroperitoneal space [160]. Histopathologically, the tumor tissue is composed of fusiform cells arranged in bundles or placed in a swirl. The cells have an elongated and spoke-like nucleus, small nucleoli, and eosinophilic cytoplasm and present varied nuclear atypia, mitotic activity, and pleomorphism [162]. These tumors often contain a rich collagen stroma with disseminated small neoplastic cells, hence the description sclerosing of this RMS variant [163]. Some cases show properties of sclerosing with gaps simulating vessels [158]. Immunohistochemical studies indicate strong positive staining for desmin and MyoD1, local or disseminated myogenin, and no or local immunoreactivity to cytokeratins [164, 165] (Fig. 5).

Extended diagnostic evaluation

After confirming RMS, the next step is to evaluate disease progression and qualification for treatment. The following analyses are recommended: blood morphology with a smear, extended biochemical blood analysis, urine analysis, chest, abdominal and pelvic CT, bone scintigraphy or PET-CT (detecting metastases to lymph nodes and bones), trepanobiopsy of the bone marrow, brain and spine MRI, and cerebrospinal fluid puncture (primary focus within the meninges and if the occupation of meninges is suspected), and sometimes a diagnostic biopsy of suspected lymph nodes [45]. In the case of extremity and trunk RMS, where the percentage of metastases to regional lymph nodes is particularly high, mapping of the lymph system and evaluation of the sentinel lymph node is possible to consider [166, 167].

Table 6. Evaluation of the degree of RMS progression according to the classification of the Intergroup Rhabdomyosarcoma Study Group TNM

Stage

Localization

T

Size

N

M

1

Favorable

T1/T2

a/b

Any

M0

2

Unfavorable

T1/T2

a

N0/Nx

M0

3

Unfavorable

T1/T2

a

N1

M0

b

Any N

M0

4

Any

T1/T2

a/b

Any

M1

T: locally advanced

N: lymph nodes

M: distant metastases

T1: locally limited, not infiltrating

T2: locally advanced, infiltrating

Diameter:

a 5 cm

b > 5 cm

N0: not occupied

N1: occupied regional lymph nodes (> 1 cm in CT/MRI/18F-FDG)

Nx: unknown status

M0: distant metastases absent

M1: distant metastases present,

spinal cord and the presence of metastatic

tumor in the pleura or peritoneum]metastases in

extra-regional lymph nodes as well

the presence of free tumor cells

in the pleural, peritoneal, and cerebral fluid

Good localizations

Eye socket, head, and neck (except for the parameningeal area), urogenital system (except for bladder and prostate)

Poor localizations

Parameningeal area, limbs, retroperitoneal space, bladder, prostate, biliary pathways*, other

Table 7. Prognostic RMS evaluation according to the Intergroup Rhabdomyosarcoma Study Group classification

Prognosis (EFS, event-free survival)

Stage acc. to IRSG TNM

Clinical group

Localization

Size

FOX01 rearrangement (fusion)

M

N

Excellent

(> 85%)

Low-risk subgroup A

1

I

Favorable

a/b

Negative

M0

N0

1

II

Favorable

a/b

Negative

M0

N0

1

III

Eye socket

a/b

Negative

M0

N0

2

I

Unfavorable

a

Negative

M0

N0/Nx

1

II

Favorable

a/b

Negative

M0

N1

Very good

(7085%)

Low-risk subgroup B

1

III

Eye socket

a/b

Negative

M0

N1

1

III

Favorable, except for eye socket

a/b

Negative

M0

N0/N1/Nx

2

II

Unfavorable

a

Negative

M0

N0/Nx

3

I/II

Unfavorable

a

Negative

M0

N1

3

I/II

Unfavorable

b

Negative

M0

N0/N1/Nx

Good

(5070%)

Moderate

risk subgroup

2

III

Unfavorable

a

Negative

M0

No/Nx

3

III

Unfavorable

a

Negative

M0

N1

3

III

Unfavorable

b

Negative

M0

N0/N1/Nx

1/2/3

I/II/III

Any

a/b

Positive

M0

N0/N1/Nx

4

IV

Any

a/b

Negative

M1

N0/N1/Nx

Poor (< 30%)

High-risk subgroup

4

IV

Any

a/b

Negative

M1

N0/N1/Nx

4

IV

Any

a/b

Positive

M1

N0/N1/Nx

Bone scintigraphy or PET-CT and two-sided aspirational bone marrow biopsy allow the evaluation of the possible occupation of the bone system and/or bone marrow. In selected cases (tumor < 5 cm, FN-RMS, no evidence of lymph node occupation), it is possible not to evaluate the diagnosis of RMS metastases to bones [51]. Imaging studies allow determining the risk group, which is the basic criterium determining prognosis and treatment intensity (Tab. 6, 7). The TNM RMS system was elaborated by the Intergroup Rhabdomyosarcoma Study Group (IRSG) in 2001 for the pediatric population and young adults [167]. For adult patients both the above-mentioned TNM IRSG system and the TNM classification, according to the American Joint Committee on Cancer for soft tissue sarcomas, can be used [168].

Table 8. Classification of patients to clinical groups taking into consideration the extent of the surgery and the progression of the disease according to the Intergroup Rhabdomyosarcoma Study Group

Clinical group* (CG)

Stage plus result of surgery

I

A localized disease, no infiltration of surrounding structures and spaces, microscopically radical resection

B localized disease, infiltration of surrounding structures and spaces, microscopically radical resection

II

A localized disease, no infiltration of surrounding structures and spaces, resection microscopically non-radical, macroscopically radical

B regional lymph nodes occupied, microscopically radical resection

C regional lymph nodes occupied, resection microscopically non-radical, macroscopically radical

III

A tumor regardless of local stage and regional lymph node occupation, exclusively biopsy

B tumor regardless of local stage and regional lymph node occupation, surgery macroscopically non-radical, more than 50% of tumor volume

IV

Any local stage, any surgery result, presence of distant metastases

General principles of treating localized disease

Treatment sequence

Rhabdomyosarcomas should be treated in a multidisciplinary fashion in reference centers for pediatric and adult sarcomas. The treatment regimens are based on resection of the primary tumor and eventual metastases to lymph nodes with perioperative radiotherapy or radical radiotherapy when surgical treatment is not possible. Methods of local treatment should be combined with multidrug chemotherapy based on cyclophosphamide or ifosfamide in combination with anthracycline, or dactinomycin and vincristine, or dacarbazine.

Surgery

Surgery is the basic therapeutic option for RMS patients, regardless of the risk group to which they belong. Local treatment must be considered first after diagnosis with the intent of complete resection of the tumor and obtaining microscopically radical surgical margins. Currently, surgery is frequently preceded by chemotherapy and/or radiotherapy [170]. In some cases, there are indications for radical regional lymphadenectomy. If metastases to regional lymph nodes are present, radical RT is used. Taking into consideration the complications of radiotherapy, histological examination of the lymph nodes should be performed to exclude non-neoplastic reactive lymphadenopathy [51]. For localizations in the extremities sparing treatments are preferred [144]. If radical surgery cannot be performed because of considerable local disease progression, localization, or other contraindications for surgery, qualifying the patient for radical RT should be considered. In the case of ARMS, performing non-radical surgery, decompression treatments, or aggressive mutilating palliative surgery does not improve the prognosis but only delays the moment of initiating systemic treatment (an exception are ERMS metastases to the retroperitoneal space). The scope of the used surgical treatment is important for further planning of radiotherapy. In clinical practice, the classification into clinical groups (CG) according to IRSG is used (Tab. 8).

Radiotherapy

Supplementary radiotherapy is indicated in all patients with RMS stages 13 according to IRSG TNM and CG IIII except for ARMS without the FOX01 rearrangement (then a decision should be taken based on risk and benefit analysis). Typically, treatment should be started after the fourth course of chemotherapy. Experience so far suggests that even if cranial nerves are affected or the tumor infiltrates the base of the skull, RT can only be started in week 12 of treatment if it was preceded by a rapid start of neoadjuvant chemotherapy [171]. Radiotherapy should be initiated urgently, regardless of the number of received chemotherapy courses in a situation of vision loss or spinal compression.

Depending on the localization and clinical group (supplementary RT after surgery or radical RT) the used total doses vary in the range of 5065 Gy. Because of complications after irradiation (particularly visible in children), the aim is to reduce the total dose or to use other methods or RT techniques T [172, 173]. The results of recent IRSG studies indicate that in the pediatric population simultaneously treated with chemotherapy, the RT doses can be decreased to 3650.4 Gy (28 fractions of 1.8 Gy) without affecting the treatment results. There are no data concerning optimal RT regimens in the adult population. The target volume should be the volume of the primary tumor before chemotherapy and surgery and regional lymph nodes (clinically suspected or in imaging studies). Using modern techniques of RT, proton radiotherapy, or brachytherapy may be associated with the protection of critical organs and contribute to decreasing the percentage of distant treatment complications. RT use in CG II improves local effectiveness from 65% to 83%. The comparison of the results of European (MMT) and American (IRS) studies suggests that in localized forms (low and intermediate-risk group) early use of local treatment using RT may be associated with better local control and OS (84% in IRS-IV studies vs. 71% in MMT89 studies, where local treatment, mainly by surgery, was used after obtaining a response to chemotherapy). Also, the percentages of 5-year progression-free survivals were higher in the IRS-IV study, 78% vs. 57% in the MMT89 study. A higher number of complications of surgical treatment was, however, observed.

In clinical practice, preferred regimens of irradiation depend on CG:

  • CG I: 41.4 Gy in 23 fractions of 1.8 Gy;
  • CG II: 45 Gy in 25 fractions of 1.8 Gy;
  • CG III in localization other than the eye socket with residual tumor < 5 cm: 50.4 Gy in 25 fractions of 1.8 Gy;
  • CG III in localization other than the eye socket with residual tumor > 5 cm: 50.4 Gy in 25 fractions of 1.8 Gy, simultaneous increase of total dose on the residual tumor to 56 Gy in fractions of 2 Gy;
  • CG III in eye socket localization: 45 Gy in 25 fractions of 1.8 Gy with simultaneous chemotherapy based on a regimen containing cyclophosphamide;
  • Parameningeal localization (the nasal cavity, nasopharynx, paranasal sinuses, middle ear, mastoid process, subtemporal fossa, pterygopalatine fossa): the elective volume is the primary tumor, adjacent meninges, and the intracranial area, preferred fractionation scheme is 5459.4 Gy in 30-33 fractions of 1.8 Gy.
Figure 6. The treatment plan for a 5-year-old boy with a diagnosis of embryonal RMS of the right eye socket (superior medial wall) after chemotherapy. Plan performed by the VMAT SIB technique assumed giving a 45 Gy dose on the area of the primary tumor with the margin (eye socket), with an increase to 50 Gy in the area of the residual tumor in 25 fractionated doses of 1.8 Gy and 2 Gy, respectively

The described regimens apply to the pediatric patient population. For decisions on adult patients concerning fractionation, regimens should be individually taken for each case (Fig. 6, 7).

Figure 7. Radiotherapy plans of an adult female patient with pleomorphic rhabdomyosarcoma with a limited number of metastases to the lungs (oligometastatic disease) diagnosed during pregnancy. The patient received chemotherapy based on doxorubicin up to the moment of birth. After the birth of a healthy child, she was qualified for hypofractionated preoperative radiotherapy 5 × 5 Gy (A), resection, postoperative chemotherapy, and stereotactic radiotherapy 10 × 4 Gy on the volume of two lung metastases (B and C)
Chemotherapy

Adding neo- and adjuvant chemotherapy to the treatment of patients without metastases allowed to obtain 6090% percent of 5-year survival. In patients over 16 years of age and adults, both in multicenter studies and in retrospective analyses, the results of treatment are worse, and the percentage of 5-year survival is in the range of 3040%. The intensity (2- or 3-drug treatments) and the duration of treatment (6, 12, or 24 months) depend on the risk group (Tab. 6, 7).

In the trials patients both in the disseminated and the localized stage were treated with various chemotherapy regimens, among the most common were doxorubicin monotherapy, doxorubicin plus ifosfamide, doxorubicin plus ifosfamide plus dacarbazine. Some of the patients received regimens in agreement with pediatric standards of RMS treatment, most commonly: ifosfamide plus vincristine plus actinomycin D, ifosfamide plus vincristine plus doxorubicin plus dacarbazine, and ifosfamide plus vincristine plus actinomycin D plus doxorubicin. In over 30% of treated patients, local recurrence of the neoplastic disease was observed, and another 40% developed distant metastases, but both using radiotherapy (p = 0.011) and chemotherapy according to pediatric protocols (p = 0.003) were associated with statistically better overall survival (OS) in multifactorial analysis. Moreover, using pediatric chemotherapy regimens in treating localized RMS in adults was described in the research of Kojima et al. [174]. This included the following protocols: 1) vincristine 1.5 mg/m2 (days 1, 8 and 15) plus cyclophosphamide 2.2 mg/ m2 (day 1) plus actinomycin D 1.5 mg/m2 (day 1); 2) vincristine plus dactinomycin plus another component chosen among: ifosfamide, etoposide, or doxorubicin. Not only the pediatric chemotherapy regimens but also the whole therapeutic procedure for the adult patient with localized RMS according to pediatric recommendations for the treatment of this disease affects OS and increases the percentage of patients with 5-year local recurrence-free survival (LRFS) [144].

Basic treatment regimens for ERMS and ARMS are presented in Table 8. VAC and VAI/IVA regimens appear to be equivalent. Adding other active drugs to the basic regimen (VAC), such as doxorubicin, etoposide, cisplatin, carboplatin, ifosfamide, or melphalan, according to trial results published so far, did not have a statistically significant effect on OS in patients with RMS in clinical groups III and IV (Tab. 8). Evaluation of the combination of standard therapy VAC/VAI with irinotecan or topotecan is the subject of the ongoing trial IRS-V. The results of the European trial with random selection of patients European Paediatric Soft Tissue Sarcoma Study Group (EpSSG) RMS2005 (population < 18 years old) have not been published yet, its aim, among others, was to evaluate in a subgroup of patients with ARMS with the N1 characteristic the effect of adding doxorubicin to standard IVA chemotherapy and the effectiveness of supportive care with vinorelbine and cyclophosphamide (altogether 50 weeks). Based on available data, it is difficult to unequivocally determine the duration of systemic treatment and indications for supportive treatment in adult patients. In adult patients (with ARMS high-risk group), treatment should last up to 4852 weeks. In the case of RMS treated in the AI regimen (doxorubicin, ifosfamide plus mesna) or MAID (doxorubicin, ifosfamide plus mesna, dacarbazine), systemic treatment is generally performed until the maximum dose of doxorubicin has been used (if the progression of the disease is not noted previously during the treatment) [14, 175–178]. No clinical trials have evaluated specific chemotherapy regimens of patients with pleomorphic RMS. Most patients receive adjuvant chemotherapy (anthracyclines and alkylating cytostatics) with local tumor treatment by surgery and/or radiotherapy. Because of the differences in the biology and phenotype of this RMS subtype in comparison with ERMS and ARMS, multicomponent pediatric chemotherapy regimens may not be applicable in such cases. On the contrary, in the case of adult patients with RMS other than pleomorphic, the use of regimens described in pediatric guidelines for the treatment of this neoplasm is recommended, and the criteria of age for the inclusion in clinical trials evaluating the effects of treatment with pediatric regimens are often extended from the pediatric population to adults [51, 144] (Tab. 9, Fig. 8).

Table 9. Chemotherapy regimens used in rhabdomyosarcoma treatment

Regimen name

Administered drugs

References

Most common regimens of RMS treatment

VA

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Dactinomycin 0.15 mg/kg/d. (max. 0.5 mg/d.), day 1.–5.

[179]

VAC

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Dactinomycin 0.15 mg/kg/d. (max. 0.5 mg/d.), day 1.–5.

Cyclophosphamide 2.2 g/m2 plus mesna, day 1.

[179]

VAC

Vincristine 1.4 mg/m2 (maximum dose 2 mg), day 1, 8, 15

Actinomycin D 1.25 mg/m2 (maximum dose 2mg), day 1.

Cyclophosphamide 1200 mg/m2, day 1.

[180]

VAC/IE

Vincristine 1.6 mg/m2, day 1.

Actinomycin D 0.45 mg/kg, day 1.

Cyclophosphamide 1200 mg/m2, day 1.

Ifosfamide 1800 mg/m2 plus mesna days 21.–25.

Etoposide 100 mg/m2, day 21.

Second line of treatment: cisplatin and etoposide

[88]

VAI

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Dactinomycin 0.15 mg/kg/d. (max. 0.5 mg/d.), day 1.–5.

Ifosfamide 1.8 g/m2/d. plus mesna, day 1.–5.

[179]

VIE

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Ifosfamide 1.8 g/m2/d. plus mesna, day 1.–5.

Etoposide 100 mg/m2/d., day 1.–5.

[179]

IVA

Ifosfamide 3 g/m2/d. plus mesna, day 1.–3.

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Dactinomycin 1.5 mg/m2 (max. 2 mg/d.), day 1.

[179]

IVADo

Ifosfamide 3 g/m2/d. plus mesna, day 1.–2.

Vincristine 1.5 mg/m2 (max. 2 mg), day 1.

Dactinomycin 1.5 mg/m2 (max. 2 mg/d.), day 1.

Doxorubicin 30 mg/m2, 4-hour infusion, day 1.–2.

[179]

VDC

Vincristine 1.5 mg/m2 (max. 2 mg) Dactinomycin 75 mg/m2

Dexrazoxane

[51]

IE

Ifosfamide 9 g/m2

Etoposide 500 mg/m2

[51]

VI

Vincristine 1.5 mg/m2 (max. 2 mg)

Irinotecan 50 mg/m2

[51]

VA

Vincristine 1.5 mg/m2 (max. 2 mg)

Dactinomycin 0.045 mg/kg (max. 2.5 mg)

[51]

VAC

Vincristine 1.5 mg/m2 (max. 2 mg)

Dactinomycin 0.045 mg/kg (max. 2.5 mg)

Cyclophosphamide 1200 mg/m2

[51]

Less common regimens of RMS treatment

Vincristine 2 mg

Doxorubicin 7590 mg/m2 72 h infusion plus dexamethasone (cardioprotection)

Ifosfamide 10 g/m2 divided in boluses for 45 days

[38]

First line of treatment:

Doxorubicin 60 mg/m2

Cyclophosphamide 600 mg/m2

Second line of treatment:

Cisplatin 75 mg/m2

Taxol 200 mg/m2

Next line of treatment:

Gemcitabine 1000 mg/m2

Carboplatin AUC 5

[98]

Preoperative chemotherapy:

Doxorubicin (50 mg/m2)

Dacarbazine (1000 mg/m2)

Vincristine (1.4 mg/m2)

Cyclophosphamide (700 mg/m2)

Postoperative:

Methotrexate (2 g/m2)

[181]

Cyclophosphamide 1200 mg/m2 day 1.

Vinorelbine 25 mg/m2 day 1 and 8

Temsirolimus 15 mg/m2 day 1, 8, and 15

(maximum 12 cycles of treatment).

[182]

Cyclophosphamide 250 mg/m2 day 1.5.

Topotecan 0.75 mg/m2 day 5.

[183]

Cyclophosphamide 25 mg/m2 each day of the cycle

Vinorelbine 25 mg/m2 on days 1, 8, 15, and 28 of the cycle

[184]

Dactinomycin 25 mg/m2 day 1.3.

Ifosfamide 2500 mg/m2 day 1.4.

[185]

Dactinomycin 75 mg/m2

day 1 (every 21 days up to 6 cycles)

[185]

Vinorelbine in monotherapy

Vinorelbine 30 mg/m2 on days 1 and 8 of the cycle every 3 weeks OR

Vinorelbine 33.75 mg/m2 every week for 6 weeks, then 2 weeks without the drug

[186, 187]

Vincristine 1.5 mg/m2, day 1 and weeks 1, 2, 4 and 5

Irinotecan 50 mg/m2 for 5 days in week 1 and 4

[188]

GD

Gemcitabine 900 mg/m2 day 1 and 8

Docetaxel 100 mg/m2 day 8

In a 21-day cycle

[189]

Observation after treatment

After completion of the treatment, the patient should be observed carefully. The recommended procedure includes the physical examination and imaging studies in the form of CT or MRI of the primary localization and CT of the chest, abdominal cavity, and pelvis using a contrast agent. Medical visits should take place every three months for the first two years, then every six months for the next three years, and subsequently once a year.

General principles of treating disease with distant metastases

In a high percentage of patients, distant metastases are found at the moment of diagnosis, which is linked with a poor prognosis. Treating the patient with distant metastases will include each of the three methods used for localized disease, that is surgery, RT, and chemotherapy. Some authors recommend limiting chemotherapy to VAC or VAC/VI regimens taking into consideration the patient’s quality of life and the poor prognosis in this group [51]. Surgery and/or RT of the primary tumor are used for RMS with a limited number of metastases to limit the risk of failure of subsequent therapy. In the case of multiple metastases, the priority is obtaining control of the disease, and if this is successful, local treatment can be considered. In most patients with RMS with numerous metastases, therapeutic procedures are palliative in character. In a retrospective multicenter analysis of RMS patients in stage 4 according to IRSG TNM, among 13 patients included in the trial two underwent resection of the primary tumor, six received palliative RT, and seven palliative chemotherapy [7]. Median OS was 7.1 months. The most common chemotherapy regimens were doxorubicin monotherapy, ifosfamide with doxorubicin, and multicomponent chemotherapy vincristine plus doxorubicin plus cyclophosphamide. Among 14 patients receiving chemotherapy admitted to hospitals with primary disseminated neoplastic disease and progressing to metastatic disease, only in 7 patients a clinical benefit was observed in response to chemotherapy [PR (partial response) or SD (stable disease)]. The only responses after administration of successive lines of chemotherapy in patients with a partial response or stable disease after the first line were observed in patients receiving chemotherapy according to the VAC protocol (vincristine, doxorubicin, cyclophosphamide). Median progression-free survival (PFS) was 2.3 months. Another retrospective study included 4 patients with RMS M1 and palliative chemotherapy was initiated in all patients, and in one of them, treatment was supplemented by palliative radiotherapy [6]. The median overall survival of the treated patients was 21.7 months. In an observational study by Bompas et al. [147] among 46 patients with stage M1 RMS, 19 received surgery, 26 radiotherapy, and 29 received palliative chemotherapy (doxorubicin ± ifosfamide or multidrug therapy based on ifosfamide, vincristine, actinomycin with or without supporting chemotherapy with cyclophosphamide). Complete remission in this group was obtained in only 13 patients (28%). Five-year survival of patients with metastatic disease was 5% (median: 13 months). The results of the clinical trial VIT-0910 indicated that adding temozolomide to the vincristine and irinotecan regimen improves the survival of patients with recurring or resistant RMS [190]. The results of studies in centers treating RMS indicate that the most common chemotherapy regimens used in such patients are multicomponent regimens using combinations such as vincristine, doxorubicin, and cyclophosphamide (VAC), or ifosfamide with doxorubicin or doxorubicin, ifosfamide, dacarbazine, and mesna (MAID) [38]. There are single descriptions of treating RMS patients with a small molecule tyrosine kinase inhibitor pazopanib which suggest that this drug could find application in patients previously treated with standard chemotherapy regimens [191, 192]. Unfortunately, another small-molecule tyrosine kinase inhibitor (crizotinib), which, among others, inhibits ALK kinase, did not have clinically significant activity in monotherapy of ARMS patients [193].

Figure 8. Proposed rhabdomyosarcoma (RMS) treatment regimens in children and young adults. Based on [51]; IE ifosfamide and etoposide; RTX radiotherapy; VA vincristine, actinomycin; VAC vincristine, actinomycin, cyclophosphamide; VDC vincristine, doxorubicin, deksrazoksan; VI vincristine and irinotecan

Procedure in the case of disease progression during or after treatment

In at least one-third of patients with RMS local or general recurrence will occur [51]. In patients in whom the disease has progressed during the first line of treatment, the prognosis is particularly poor. Patients who completed RMS treatment often do not obtain a full radiological response in layered imaging studies, despite normalization of the PET scan picture, which is probably due to the scarring of the primary tumor site or the differentiation of that tissue. For that reason, the biopsy of the tumor bed after removal of the primary tumor is not recommended except for situations where the primary tumor increases in size or pain occurs [194]. Patients whose PET scan indicates an enhanced signal in the site of the primary tumor pose a particular challenge in respect to the choice of further therapy. Indubitably, they belong to the group of patients with an increased risk of local recurrence and development of distant metastases, and the decision on performing a biopsy of the primary site of the tumor or further surgical resection should be taken after stratification of both risks and potential benefits [195]. A definite suspicion of RMS recurrence requires taking tissue material using a biopsy and histopathological confirmation. Surgical resection of the tumor may be considered if access to the site of recurrence allows this. Radiotherapy is quite often used for the treatment of the primary tumor (if it had not been treated previously) and metastases to the bones and lungs if this is doable. Radiotherapy may be delayed, especially in respect to neoplastic metastases, to evaluate the response to chemotherapy and to avoid myelosuppressive complications due to systemic cytotoxic treatment. Currently, it is particularly important to include patients with RMS recurrence or progression into clinical trials and to use chemotherapeutics with proven activity against RMS. There are no data permitting comparison of the effectiveness of treating with specific chemotherapy regimens, thus the decision about the choice of a given type of therapy in patients with RMS recurrence depends on many factors, including among others the first-line treatment protocol, the patient’s general status, and the tolerance of earlier therapy. As a rule, second-line chemotherapy is used containing previously mentioned active drugs (platinum derivatives, camptothecin, etoposide, doxorubicin, vinorelbine). In patients with recurrence or primarily generalized RMS, attempts at high-dose chemotherapy have also been made. Phase III trials with a random selection of patients were conducted, in which standard chemotherapy was compared with myeloablative treatment. There are insufficient data to determine the optimum length of chemotherapy duration in the case of recurrence/progression. Patients, in general, receive at least 8 cycles of chemotherapy, if a complete response to therapy and acceptable tolerance of therapy occurs [51]. In the MMT89 and MMT91 trials, a group of 52 patients from a high-risk group who had undergone myeloablative therapy after standard induction chemotherapy were nonrandomly compared with 44 patients treated only with standard inductive and supplementary chemotherapy. The percentages of progression-free survivals were 30% for myeloablative chemotherapy and 19% for standard treatment. In this group of patients, a more significant prognostic factor associated with treatment turned out to be the response to initial inductive chemotherapy. The percentage of OS in patients who were in complete remission after surgery and chemotherapy up to week 18 was 41% in comparison with 14% in patients in whom a complete response had not been obtained (p = 0.0001). The available data do not allow a valid evaluation of myeloablative treatment in young adults and suggest the resistance of RMS cells to mega chemotherapy, as most recurrences after treatment occurred in sites previously occupied by the neoplasm [196, 197]. The effectiveness of chemotherapeutics commonly used in treating other types of soft tissue sarcomas in adults such as gemcitabine, docetaxel, or pazopanib, has so far not been sufficiently evaluated in RMS. Moreover, few published data are indicating a good effect of using tyrosine kinase inhibitors in the therapy of patients with RMS. Also, the role of the increasingly popular immunotherapy, including checkpoint inhibitors (antibodies directed against PD-1, PD-L1, or CTLA-1), immunomodulating drugs, or CAR-T cell therapy, has not yet been verified in the context of RMS treatment, but there are single cases of complete response to treatment with drugs from these groups [198].

The algorithm of the procedure to follow if recurrence or progression of rhabdomyosarcoma is suspected is presented in Figure 9 [51].

Figure 9. Algorithm of procedure for suspicion of rhabdomyosarcoma recurrence or progression. After [51]; CTX chemotherapy; RTX radiotherapy

RMS in adult patients selected aspects

Due to the rare occurrence of RMS in the adult population, there are no unequivocal guidelines concerning the treatment of this neoplasm. There are no published results of clinical trials with randomization which could be the basis of uniform principles of care for such patients. So far experience in treating adult patients with RMS is based on small groups collected in retrospective analyses [144, 147, 178]. The used treatment regimens differ considerably depending on the center in which the patients are treated and the therapy standards in that center.

In the largest meta-analysis published so far including 533 adult patients with RMS in a localized stage, a large variety of treatment protocols chosen by specialists for patients with RMS was presented [5]. Currently, three main methods are used for the treatment of localized RMS, which include oncological surgery, radiotherapy, and chemotherapy. In the mentioned meta-analysis, the most used methods were surgery plus chemotherapy (27.5%), a combination of the 3 methods (25.1%), and surgery alone (19.0%). The combination of surgery and radiotherapy (1.2%) and radiotherapy and chemotherapy (13.2%) were slightly less common. Monotherapy, that is chemotherapy or radiotherapy, was the least common (3.8%). The median radiation dose used in the case of radiotherapy and surgery was 54 Gy (from 14 to 110 Gy), while when radiotherapy was the only method used the median was slightly higher, namely 56.5 Gy (from 36 to 110 Gy). The most common chemotherapy was as an adjuvant treatment (42.9%) or as primary therapy (15.3%). The protocols were mainly based on cyclophosphamide (22.5%), a combination of cyclophosphamide with anthracycline (21.2%), and a combination of ifosfamide with anthracycline (13.9%). Another study which included 82 patients with locoregionally advanced RMS, had a 5-year overall survival index of 44% [155]. The treatment was most commonly a combination of surgery, radiotherapy, and chemotherapy (30 persons), a combination of radiotherapy and chemotherapy (28 persons), and a combination of radiotherapy and surgery (15 persons). Radiotherapy in the case of a preoperative procedure included irradiation with a median of 50 Gy, whereas postoperative radiotherapy and radiotherapy without surgery had a median of 60 Gy. Chemotherapy was given to 58 patients and included administering doxorubicin or actinomycin D in combination with vincristine or cyclophosphamide. Disease recurrence occurred in 47 patients (57%), most commonly in the form of distant metastases (22 persons), less frequently as a local (11 persons) or loco-regional recurrence (11 persons). In the retrospective study of Noujaim et al. [7] 32 patients with localized RMS were described in whom in 26 cases radical surgery removal of the primary tumor was performed. Frequently in as many as 15 patients, postoperative radiotherapy was applied, whereas only 3 persons received chemotherapy or preoperative radiotherapy. Local and distant recurrence was present in 4 and 10 persons, respectively. In a study from another center, including 16 patients with RMS in a localized stage, the most common procedure was radiotherapeutic treatment combined with chemotherapy (11 persons) and chemotherapy alone (2 persons) [6]. Three persons were treated by primary surgery supplemented by chemotherapy or/and radiotherapy. The most frequently chosen chemotherapy regimen (regardless of the stage of the disease) was vincristine, actinomycin D, cyclophosphamide (VAC) alternating with ifosfamide and etoposide (IE). During radiotherapy in 10 patients, simultaneous chemotherapy was used in the vincristine and cyclophosphamide (VC) regimen. Among patients treated with non-palliative radiotherapy, the median radiation dose was 56 Gy. Among 16 persons treated for primary localized RMS, 4 had local recurrence, whereas metastases or regional dissemination were observed in 6 persons. Treatment with radiotherapy (p = 0.009) and chemotherapy lasting longer than 19 weeks (p = 0.009), as well as adding a simultaneous regimen of VC chemotherapy to radiotherapy (p = 0.01), was associated with a longer OS. In the next group of patients including 111 adults with localized RMS, surgery of the primary tumor was performed in 80% (89 persons), radiotherapy in 73% (81 persons), and chemotherapy was administered to 75% of patients (83 persons) [147]. CT, MRI, and PET-CT can be used to evaluate the periodic effectiveness of the treatment.

Table 10. Studies describing the treatment of adult patients with diagnosed rhabdomyosarcoma

Refe-rences

Number of patients

Patients’ age (median)

mOS

5-OS

Median PFS/DFS/RFS/EFS

M(–)

M(+)

M(–)

M(+)

M(–)

M(+)

[7]

45

71.5 (28.492.8)

12.8

7.1

29%

7.3 (RFS)

2.3 (PFS)

[6]

20

34 (1979)

53.2

21.7

20% (3year)

19.8 (DFS)

20.4 (DFS)

[147]

157

37 (1886)

40.0

13.0

43%

5%

9.3 (RFS)

[147]

292

55 (1899)

40.0 (whole cohort)

40.0 (whole cohort)

[178]

84

31 (1676)

35.0

15.0

50%

22%

[200]

36

29 (2172)

22.4 (PFS)

13.3 (PFS)

[207]

59

56 (3872)

11.0

9.0

[14]

39

26 (1682)

44%

0%

[155]

82

(M–)

27 (1781)

38

44%

6.5 (PFS)

[144]

171

27 (1983)

45.7%

4.3%

[13]

1071

> 19

47%

[4]

138

28 (1686)

45%

approx. 18%

[180]

8

24 (1860)

27.3

17.0 (PFS)

[203]

66

28 (1871)

30.0

11.0

36%

11%

17.0 (EFS)

[208]

239

19 (10102)

45.6

16.8

44.1%

18%

22.8 (RFS)

10.8 (PFS)

[203]

66

28 (1871)

30.0

11.0

35%

11%

Because RMS is rare in adults there is a limited number of papers reporting the results of treating this neoplasm, and their main limitation is their retrospective character (Tab. 10). The lack of unequivocal guidelines concerning the treatment of metastatic disease leads to a large variety of chemotherapy regimens in studies from large centers, which, moreover, differ in their standards of oncological care. Survival of adults with RMS is still much lower than the results obtained in pediatric populations, where 5-year overall survival is OS = 7787% in children and OS = 2040% in adults [5, 144, 167, 178]. This fact can probably be explained by several aspects. First, age was shown to be an independent prognostic factor for patients with RMS [200]. A significant difference is the use of lower doses of supportive chemotherapy components in adults in comparison to children, due to the high frequency of serious complications, including bone marrow suppression, infections, and neurotoxic effects, among others [174]. Additionally, the more common histopathological RMS subtypes in adults are pleomorphic and alveolar RMS, they are associated with a poorer prognosis [13]. Five-year overall survival in RMS patients is in the range of 4050% for localized disease and is from zero to 30% in the case of metastatic disease based on available analyses from large centers [7, 14, 147, 178]. Nevertheless, retrospective studies have shown that initiation of pediatric protocols of localized disease treatment is associated with a better prognosis and obtaining 5-year survivals of patients at the level of 61.5% [144, 147]. The use of radiotherapy was also associated with better survival of the patients both in the local disease stage, as well as for disseminated disease [147, 200, 201]. Including chemotherapy was found to be an independent prognostic factor causing lower mortality due to the progress of the neoplastic disease according to the analysis of patients from a center in Thailand [202]. There are, however, also reports on the lack of improvement of survival of patients subjected to chemotherapy in comparison to a control group [201]. Moreover, tumors smaller than 5 centimeters in size correlate with longer patient survival regardless of other factors [155, 178]. Favorable tumor localization has been distinguished in some elaborations of retrospective studies as an independent factor affecting survival [4]; however, there are also opposite conclusions that multifactorial analysis indicates that this aspect is not statistically significant [5, 203]. In the case of tumors of the urinary tract, localization within the prostate gives a better prognosis as compared to the urinary bladder or kidney RMS [204]. It is worth underlining that obtaining negative surgical margins after tumor resection was distinguished as a prognostic factor in the context of disease recurrence and progression [147, 178]. Surgical resection of the primary tumor remains the standard in localized disease, but it has been shown that this procedure improves survival in disseminated disease in adults [147, 205]. After treatment careful control of recurrences is necessary. Medical visits should take place every 3 months for the first year, every 46 months during the second and third year, and subsequently once a year. Among analyses performed during control visits are interview and physical- examination, peripheral blood morphology and biochemistry (parameters of liver and kidney function), imaging studies CT every 36 months for the first 2 years, subsequently once a year for the next 3 years, bone scintigraphy (every 6 months for the first 2 years, subsequently once a year for the next 3 years), the remaining examinations (ultrasound/CT/MRI) of the area of the primary tumor and PET-CT depending on the decision of the multi-specialist team [206].

Table 11. Stratification to rhabdomyosarcoma (RMS) risk groups according to Children’s Oncology Group-Soft-Tissue Sarcoma. Based on [131, 213, 214]

Risk

Histology

Grade

Clinical group

Low

ERMS

1

I, II, III

ERMS

2, 3

I, II

Standard

ERMS

2, 3

III

ARMS

1, 2, 3

I, II, III

High

ERMS

4

IV

ARMS

4

IV

Table 12. Stratification to risk groups for the localized form of rhabdomyosarcoma (RMS) according to the European Paediatric Soft Tissue Sarcoma Group. Based on [131, 213, 214]

Risk

Histology

Clinical stage

Localization

N Status

Tumor size and patient’s age

Low

ERMS

I

All

N0

5 cm and 10 years

Standard

ERMS

I

All

N0

> 5 cm or > 10 years

ERMS

II, III

Favorable

N0

all

ERMS

II, III

Unfavorable

N0

5 cm and 10 years

High

ERMS

II, III

Unfavorable

N0

> 5 cm or > 10 years

ERMS

II, III

All

N1

All

ARMS

I, II, III

All

N0

All

Very high

ARMS

II, III

All

N1

All

Table 13. Modified risk stratification system including the PAX3/7-FOXO1 fusion [219]

Risk

IRSG TNM stage

Clinical group

Age

Rearrangement FOXO1 (fusion)

Low

1

I, II, III (only eye socket)

Any

FOXO1–

2

I, II

Standard

1

III (without eye socket)

Any

FOXO1–

1, 2, 3

I, II, III

FOXO1+

2, 3

III

FOXO1–

3

I, II

FOXO1–

4

IV

< 10 years

FOXO1–

High

4

IV

≥ 10 years

FOXO1–

Any

FOXO1+

Selected aspects of pediatric RMS

Rhabdomyosarcoma is the most diagnosed soft tissue sarcoma in children. It constitutes about 57% of all pediatric neoplasms and 60% of soft tissue sarcomas. Over one-half of the cases appear in small children aged 26 years. In the group of pediatric patients, ERMS (5570%) and ARMS (2530%) are the most common [13, 209–211]. The most frequent localization of the disease in children is the head and neck area (eye socket, parameningeal area, soft tissues of the face and neck; about 36%). These are generally cases of ERMS, diagnosed before the age of eight years, rarely metastasizing to regional lymph nodes [166]. The urogenital tract is also a frequent site of RMS occurrence in children (approx. 23%). In respect to prognosis, this localization can be divided into the area of the bladder and prostate, and the area without the bladder and prostate (testes, epididymis, the peritesticular area, penis, vulva, vagina, ovary, uterus) [212]. Vaginal RMS deserves particular attention in this group; it occurs, in general, in small girls and has a very characteristic clinical presentation of botryoid masses “falling out” of the vaginal vestibule, causing bleeding and/ or discharge [124, 203]. Uterine tumors are generally oligosymptomatic and are thus diagnosed in advanced stages [166, 212]. Extremities are a less common localization in children (approx. 20%). The neoplasm, in general, develops in the form of a painless tumor, often giving metastases to regional lymph nodes (50%) [166]. In about 15% of RMS cases in children, at the moment of diagnosis generalized disease is found (stage four of clinical progression according to TNM for RMS) with metastases to the lungs (50%), bone marrow (3040%), bones (10%) and/or lymph nodes (depending on the localization 550%) [166, 212].

Treatment of soft tissue sarcomas in children is based on international protocols of the Cooperative Weichteilsarkom Studiengruppe (CWS) and the European Paediatric Soft Tissue Sarcoma Group (EpSSG), recommended by the Polish Pediatric Solid Tumor Group. The basis for the treatment strategy is appropriate stratification to risk groups based on the following prognostic factors: the disease stage according to the IRSG classification, histological type, age of the patient, size, and localization of the tumor. The risk stratification system is periodically updated. Taking into consideration the most recent data on the prognostic value of the genetic status of ARMS, it should be kept in mind that the current system of stratification will need to be verified in the near future. Currently, this system assumes a division into risk groups. The assignment to a given risk group determines the choice of a specified therapeutic regimen. The American system of risk stratification elaborated by the Children’s Oncology Group-Soft-Tissue Sarcoma (COG-STS), differs slightly from the European system created by EpSSG. A detailed description of both systems of risk stratification is presented in Tables 11 and 12.

Recent studies have confirmed the importance of PAX3/7-FOXO1 fusion status as a critical prognostic biomarker following M status [215–217]. Other molecular factors of potential prognostic significance, which have not yet been used in RMS risk stratification, are under investigation. The INternational Soft Tissue SaRcoma ConsorTium will supervise the coordination of further research work and combining clinical and molecular data from different research studies from various medical centers. A modification of the current risk stratification system by including the PAX3/7-FOXO1 fusion status is a subject of several prospective clinical trials [COG (ARST1431) and EpSSG Frontline and Relapsed-Rhabdo-MyoSarcoma (FaR-RMS)] (Tab. 13) [218].

Unfavorable localization encompasses limbs, the parameningeal area, the bladder, and the prostate. The alveolar type of sarcoma, the patient’s age > 10 years, and tumor size > 5 cm are also associated with a poorer prognosis [13, 170, 209–211].

Currently, in about 70% of children with locally advanced disease, a permanent cure is obtained after using combined treatment [209, 210]. The optimal time for initiating local therapy is controversial. European protocols recommend surgery and/or radiotherapy after the 3rd cycle, i.e. in week 13 from starting chemotherapy, and for metastatic disease from week 22 [209, 210]. The modality of surgical treatment of RMS is due to the possibility of infiltration of various sites by the tumor, and the course of the disease may be different depending on the tumor localization. Radical surgery is an important prognostic factor, but because of the localization and size of the tumor, it is generally difficult to perform. However, only in 10% of the patients at the moment of diagnosis, the extent of neoplastic disease allows radical surgical resection [212]. In the remaining patients, the surgical intervention is limited to a biopsy. Analysis of survival did not indicate the superiority of debulking surgery as compared to a biopsy [56]. After performing non-radical tumor resection, a second evaluation by the surgeon is recommended to determine the possibility of radicalization and to consider performing such a procedure before initiating systemic therapy (PRE, pre-treatment re-excision). The premise of the surgical protocol is complete (macroscopic and microscopic) removal of the neoplastic tumor with the margin of the surrounding tissues, without a significant cosmetic effect nor perturbation of function [56, 212, 214]. The surgery before initiating systemic treatment affects risk stratification, allowing classification of the patient to a better group compared to the classification of the primary surgery [220]. The subject of safe tissue margins during primary resection in children remains controversial. Most frequently obtaining a margin of about 0.5 cm is recommended [56]. In the case of locally advanced disease, surgical treatment is only considered after completing induction chemotherapy (DPE, delayed primary excision) when imaging studies show a residual tumor qualifying for radical resection.

Histopathological verification of regional lymph nodes is recommended in patients with the suspected occupation of lymph nodes in a clinical investigation or imaging study and primary RMS localization within the limbs and the peritesticular area (≥ 10 years). This procedure is also recommended in children with ARMS with the PAX/FOXO1 translocation [56]. The recommended method is a biopsy of the sentinel node [56]. Confirmation of regional lymph node involvement is an indication for radiotherapy, as radical lymphadenectomy was not found to improve survival [56].

Rhabdomyosarcoma is a neoplasm with high sensitivity to chemotherapy, therefore, current regimens are based on neoadjuvant chemotherapy. Despite many studies on the intensification of this treatment, the standard in Europe is still the regimen using three drugs: ifosfamide, vincristine, and actinomycin D [209–211]. In turn, COG recommends the VAC regimen composed of vincristine, actinomycin D, and cyclophosphamide. The European group explains the substitution of cyclophosphamide by ifosfamide by a decreased risk of toxicity to the gonads. The treatment of patients qualified to the low-risk group is shorter, and a reduction of the dose of cyclophosphamide is also possible without affecting overall survival. In patients from the moderate risk group, a reduction of the cyclophosphamide dose requires adding the next drug (e.g. irinotecan) to the basic treatment regimen. The greatest challenge is the treatment of patients from the high-risk group and patients with disease recurrence, as for years no improvement of survival indices has been observed. In these groups, attempts are made to introduce new drugs into the treatment regimens now in force and to introduce new treatment methods.

Radiotherapy, except for cases from the low-risk group, is standard supplementary treatment after surgery. However, there are premises for including this treatment before surgery: an easier and more precise definition of the target for irradiation, limiting the volume of normal tissues receiving a high dose, decreasing the risk of secondary tumors (most of the irradiated tissues will be removed), and the radiobiological advantage of irradiating tissues which are better oxygenated. So far little data have been published on this subject. In a group of 17 children with diagnosed RMS of the urogenital tract, Seitz et al. [221] obtained a 5-year EFS of 82%, which is a very promising result. However, the basic advantage of such a treatment sequence is the decrease in the risk of late toxicity. Radiotherapy in the pelvic or parameningeal area, especially in children under 3 years of age, is a treatment associated with a high risk of hindering the development of irradiated tissues and serious toxicity depending on the dose administered to normal organs. The fear of initiating such aggressive treatment in neonates probably contributes to the poorer survival indices in this age group [214]. Hence modern treatment methods utilizing volumetric modulated arc therapy (VMAT) or proton therapy, with a greater possibility of protecting healthy tissues, are currently recommended as the treatment of choice [170, 221, 222]. To further improve the conformality of the distribution and to decrease the dose outside the target volume, the technique of simultaneous irradiation is used, in which different doses are given in different areas of the target volume simultaneously. For a selected group of patients, especially with RMS localized in the organs of the pelvis minor, brachytherapy may be used as part of the procedure together with sparing surgery. This is, however, a form of treatment performed only in a few reference centers [210].

Table 14. Current clinical trials of adult patients with rhabdomyosarcoma (RMS)

Clinical phase

Intervention

Primary endpoints of the clinical trial

Age of patients recruited for the trial

I/II

AMG479 antibody (Ganitumab) against IGF-1R receptor combined with Src family kinase inhibitor (Dasatinib)

Phase I

Determining a safe dose of dasatinib combined with ganitumab in patients with recurrent RMS or resistant-to-treatment embryonal or alveolar RMS

Phase II

Number of patients with ORR (CR or PR)

> 2 years

III

VAC alternating with vincristine and irinotecan (VI) vs. VAC/VI plus temsirolimus

EFS

< 40 years

III

VAC vs. VAC alternating with vincristine and irinotecan (VI)

EFS

RR

OS

< 49 years

II

Cabozantinib-s-malate (XL184) small-molecule tyrosine kinase inhibitor (c-Met, VEGFR2, AXL, RET)

ORR (CR and PR)

230 years

I

NK cells from donors without compatibility in HLA system combined with ALT803 (IL-15 analog increasing NK cell cytotoxicity)

Establishing maximum tolerated dose of NK cells in combination with ALT803 administration

18100 years

II

Vemurafenib (BRAF inhibitor)

ORR (CR and PR)

121 years

II

Nab-paclitaxel combined with gemcitabine

RR

PFS

1230 years

I

Vorinostat, vincristine, irinotecan, temozolomide

Establishing maximum tolerated dose of vorinostat (combined with other chemotherapeutics)

130 years

I/II

Eribulin and irinotecan

Phase I:

Establishing maximum tolerated dose of eribulin combined with irinotecan and establishing the appropriate dose of a combination of drugs for phase II of a clinical trial

Phase II:

ORR (CR and PR)

5 months–25 years

I

Mocetinostat combined with vinorelbine

Determining the toxicity of the drug combination

Establishing maximum tolerated dose of drug combination

> 13 years

I

Immunotherapy using B7H3 CAR-T cells and B7H3 x CD19 CAR-T cells

Establishing maximum tolerated dose of CAR-T cells

Evaluation of immunotherapy toxicity

Evaluation of the technology of preparing

bispecific B7H3 x CD19 CAR-T cells

< 26 years

II

Regorafenib

PFS

> 5 years

I

Palbociclib combined with temozolomide and irinotecan

Evaluation of toxicity and adverse effects of drug combination

RR

220 years

I

Lyso-thermosensitive liposomal doxorubicin

Establishing maximum tolerated dose of the drug

Evaluation of drug toxicity

Evaluation of drug pharmacokinetics

< 30 years

I

Immunotherapy using EGFR806 CAR-T cells

Maximum tolerated dose

Evaluation of drug toxicity and adverse effects

Evaluation of the yield of the process of obtaining CAR-T cells

< 26 years

I

CLR131

Evaluation of drug toxicity

221 years

I/II

Prexasertib combined with irinotecan

Establishing recommended dose of prexasertib combined with irinotecan for phase II trial

Evaluation of response to treatment among

> 1 year

I

Olaparib combined with temozolomide or olaparib in combination with temozolomide and irinotecan

Maximum tolerated dose of drug combinations

> 16 years

I/II

Infusion of haploidentical activated NK cells

RR

< 80 years

II

Vincristine and irinotecan or vincristine, irinotecan and temozolomide

PR or CR

6 months–50 years

In irradiation of children with an RMS diagnosis a broad range of doses from 36 to 54 Gy is used and, in the case of monotherapy, even up to 59.4 Gy. This depends on the localization and histological type of the sarcoma and, above all, on the extent of the residual disease [209–211]. Research on the use of doses escalated to 59.4 Gy in all patients with tumors exceeding 5 cm in size is ongoing [223].

There are many contradictory data on procedures in metastatic disease. In choosing the optimal type of treatment criteria, identifying 4 prognostic factors may be helpful: age, localization of the primary focus, occupation of the bone marrow, and metastases to at least 3 localizations. Patients with the presence of only one factor attain a 3-year EFS of 44% in comparison to 14% of patients with 24 factors [209]. There are no unequivocal guidelines as to the role of surgery in treating generalized neoplastic disease. The recommended procedure is a biopsy, performed to confirm the presence of a metastatic focus, and surgical resection of the persisting focus after chemotherapy [56, 224].

Despite significant improvements in treating children diagnosed with RMS from the low and standard risk group (3-year EFS > 70%), the effects of treating more advanced diseases are still unsatisfactory, especially in the presence of factors with adverse effects on the prognosis [209–211]. In 2030% of pediatric patients with localized RMS and 70% with metastatic disease, a recurrence of the disease will occur [225, 226]. Despite gradual improvement in the treatment of such patients, indices of 5-year post-relapse survival (PROS) in this group do not exceed 30% [227].

There are still many questions regarding, among others, the optimal time for introducing radiotherapy, the benefit of escalating the dose, or the role of radiotherapy in persistent disease, especially with a poor prognosis. The possibilities of conventional treatment intensification are limited by complications, and the low survival indices in the group of patients with recurrence and generalized disease indicate that new safer methods of targeted therapy must be sought. Among substances with proven activity against RMS, which have been tested in vitro and in vivo, are monoclonal antibodies against IGF-1R (cixutumumab and robatumumab), IGF-1R inhibitor (BMS-754807), VEGFR inhibitor (cediranib), RTK inhibitor (sunitinib), AAK inhibitor (alisertib), and mTOR kinase inhibitor (rapamycin). Cixutumumab (IMC-A12), temsirolimus (mTOR kinase inhibitor), and bevacizumab (monoclonal antibody against VEGF) are being tested in clinical trials in combination with chemotherapy in patients with RMS recurrence and generalized neoplastic disease [228–230]. New reports on the efficacy and good tolerance of the combination of vinorelbine with the histone deacetylase inhibitor mocetinostat in RMS is interesting [231].

Conclusion

Most of the data concerning survival and prognostic factors of adults with RMS come from retrospective, single-center analyses including several dozen to several hundred patients (Tab. 10). Despite the development of diagnostic techniques and new technologies in radiotherapeutic treatment, the spectacular improvement in survival attained in the pediatric population has not been possible for adults. This fact indicates that the course of the disease is considerably different in children and teenagers in comparison with adults. In care for the patient with local tumor development, the greatest role is played by oncological surgery combined with radiotherapy and chemotherapy. The evaluation of the effectiveness of these combinations of methods in patients with disseminated RMS is crucial in the context of elaborating an optimal sequence of treatment and chemotherapeutic and radiotherapeutic regimens in adults. The standardization and verification of procedures used in treating patients with RMS metastases are particularly important to prolong and improve their quality of life. Taking into consideration the rarity and the complexity of this disease, patients with RMS should be treated in highly specialized hospital wards with long-term practice in care for persons with soft tissue sarcomas. Cooperation within a multidisciplinary team is crucial. That team should be composed of an oncological surgeon, a clinical oncologist, a radiotherapist, and, depending on the need, physicians of other specializations (e.g. a gynecologist or an ear and throat specialist). Multidirectional treatment and the experience of oncological teams in large specialist centers allow us to obtain the best results of treatment. The need to include adult patients into multicenter clinical trials has been repeatedly stressed, as their results may be the basis for elaborating uniform standards of care for patients with RMS.

Current clinical trials for adult patients with RMS are presented in Table 14.

Article Information and Declarations

Conflict of interest

Authors declare no conflict of interest.

Financing

Prepared within the remit and scientific plan of the NIO-PIB.

References

  1. Ceyssens S, Stroobants S. Sarcoma. Methods Mol Biol. 2011; 727: 191203, doi: 10.1007/978-1-61779-062-1_11, indexed in Pubmed: 21331935.
  2. Agaram NP. Update on Myogenic Sarcomas. Surg Pathol Clin. 2019; 12(1): 5162, doi: 10.1016/j.path.2018.10.003, indexed in Pubmed: 30709448.
  3. Rudzinski ER, Anderson JR, Hawkins DS, et al. The World Health Organization Classification of Skeletal Muscle Tumors in Pediatric Rhabdomyosarcoma: A Report From the Children’s Oncology Group. Arch Pathol Lab Med. 2015; 139(10): 12811287, doi: 10.5858/arpa.2014-0475-OA, indexed in Pubmed: 25989287.
  4. Gerber NK, Wexler LH, Singer S, et al. Adult rhabdomyosarcoma survival improved with treatment on multimodality protocols. Int J Radiat Oncol Biol Phys. 2013; 86(1): 5863, doi: 10.1016/j.ijrobp.2012.12.016, indexed in Pubmed: 23414767.
  5. Elsebaie MAT, Amgad M, Elkashash A, et al. Management of Low and Intermediate Risk Adult Rhabdomyosarcoma: A Pooled Survival Analysis of 553 Patients. Sci Rep. 2018; 8(1): 9337, doi: 10.1038/s41598-018-27556-1, indexed in Pubmed: 29921891.
  6. Liu YT, Wang CW, Hong RL, et al. Prognostic Factors and Treatment Outcomes of Adult Patients With Rhabdomyosarcoma After Multimodality Treatment. Anticancer Res. 2019; 39(3): 13551364, doi: 10.21873/anticanres.13249, indexed in Pubmed: 30842169.
  7. Noujaim J, et al. Adult Pleomorphic Rhabdomyosarcoma: A Multicentre Retrospective Study. Anticancer Res. 2015; 35(11): 62136217.
  8. Hortobágyi GN. Anthracyclines in the treatment of cancer. An overview. Drugs. 1997; 54(Suppl 4): 17.
  9. Wojciechowska U. A.U.J.D.. Cancer in Poland in 2012. Cancer in Poland in 2012. 2013; 63(3): 197216.
  10. ESMO / European Sarcoma Network Working Group. Soft tissue and visceral sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2012; 23 Suppl 7: vii92vii99, doi: 10.1093/annonc/mds253, indexed in Pubmed: 22997462.
  11. Stiller CA, et al. Descriptive epidemiology of sarcomas in Europe: report from the RARECARE project. Eur J Cancer. 2013; 49(3): 684695.
  12. Alkhormi AM, et al. Primary duodenal embryonal rhabdomyosarcoma in adults: a case report. AME Case Rep. 2019; 3: 29.
  13. Sultan I, Qaddoumi I, Yaser S, et al. Comparing adult and pediatric rhabdomyosarcoma in the surveillance, epidemiology and end results program, 1973 to 2005: an analysis of 2,600 patients. J Clin Oncol. 2009; 27(20): 33913397, doi: 10.1200/JCO.2008.19.7483, indexed in Pubmed: 19398574.
  14. Esnaola N, Rubin B, Baldini E, et al. Response to Chemotherapy and Predictors of Survival in Adult Rhabdomyosarcoma. Ann Surg. 2001; 234(2): 215223, doi: 10.1097/00000658-200108000-00012.
  15. Diller L, Sexsmith E, Gottlieb A, et al. Germline p53 mutations are frequently detected in young children with rhabdomyosarcoma. J Clin Invest. 1995; 95(4): 16061611, doi: 10.1172/jci117834.
  16. Kratz C, Rapisuwon S, Reed H, et al. Cancer in Noonan, Costello, cardiofaciocutaneous and LEOPARD syndromes. Am J Med Genet C Semin Med Genet. 2011; 157(2): 8389, doi: 10.1002/ajmg.c.30300.
  17. Russo I, Paolo VDi, Gurnari C, et al. Congenital Rhabdomyosarcoma: a different clinical presentation in two cases. BMC Pediatrics. 2018; 18(1), doi: 10.1186/s12887-018-1128-5.
  18. Lee YC. Congenital Eyelid Rhabdomyosarcoma. Ophthalmic Plast Reconstr Surg. 2016; 32(5): e104e106.
  19. Yang P, Grufferman S, Khoury MJ, et al. Association of childhood rhabdomyosarcoma with neurofibromatosis type i and birth defects. Genetic Epidemiology. 2005; 12(5): 467474, doi: 10.1002/gepi.1370120504.
  20. Grufferman S, Ruymann F, Ognjanovic S, et al. Prenatal X-ray exposure and rhabdomyosarcoma in children: a report from the children’s oncology group. Cancer Epidemiol Biomarkers Prev. 2009; 18(4): 12711276, doi: 10.1158/1055-9965.EPI-08-0775, indexed in Pubmed: 19293315.
  21. Grufferman S, et al. Parents’ use of cocaine and marijuana and increased risk of rhabdomyosarcoma in their children. Cancer Causes Control. 1993; 4(3): 217224.
  22. Lupo PJ, Danysh HE, Skapek SX, et al. Maternal and birth characteristics and childhood rhabdomyosarcoma: a report from the Children’s Oncology Group. Cancer Causes Control. 2014; 25(7): 905913, doi: 10.1007/s10552-014-0390-6, indexed in Pubmed: 24831857.
  23. Weksberg R, Shuman C, Beckwith JB. Beckwith-Wiedemann syndrome. Eur J Hum Genet. 2010; 18(1): 814, doi: 10.1038/ejhg.2009.106, indexed in Pubmed: 19550435.
  24. Estep AL, Tidyman WE, Teitell MA, et al. HRAS mutations in Costello syndrome: detection of constitutional activating mutations in codon 12 and 13 and loss of wild-type allele in malignancy. Am J Med Genet A. 2006; 140(1): 816, doi: 10.1002/ajmg.a.31078, indexed in Pubmed: 16372351.
  25. Doros L, Yang J, Dehner L, et al. DICER1 mutations in embryonal rhabdomyosarcomas from children with and without familial PPB-tumor predisposition syndrome. Pediatr Blood Cancer. 2012; 59(3): 558560, doi: 10.1002/pbc.24020, indexed in Pubmed: 22180160.
  26. Hartley AL. Neurofibromatosis in children with soft tissue sarcoma. Pediatric hematology and oncology. 1988; 5(1): 716.
  27. Choi JS, Choi JS, Kim EJ. Primary pulmonary rhabdomyosarcoma in an adult with neurofibromatosis-1. Ann Thorac Surg. 2009; 88(4): 13561358, doi: 10.1016/j.athoracsur.2009.02.083, indexed in Pubmed: 19766846.
  28. Wimmer K, Rosenbaum T, Messiaen L. Connections between constitutional mismatch repair deficiency syndrome and neurofibromatosis type 1. Clin Genet. 2017; 91(4): 507519, doi: 10.1111/cge.12904, indexed in Pubmed: 27779754.
  29. Boot MV, van Belzen MJ, Overbeek LI, et al. Benign and malignant tumors in Rubinstein-Taybi syndrome. Am J Med Genet A. 2018; 176(3): 597608, doi: 10.1002/ajmg.a.38603, indexed in Pubmed: 29359884.
  30. Kleinerman RA, Tucker MA, Abramson DH, et al. Risk of soft tissue sarcomas by individual subtype in survivors of hereditary retinoblastoma. J Natl Cancer Inst. 2007; 99(1): 2431, doi: 10.1093/jnci/djk002, indexed in Pubmed: 17202110.
  31. Cajaiba M, Bale A, Alvarez-Franco M, et al. Rhabdomyosarcoma, Wilms tumor, and deletion of the patched gene in Gorlin syndrome. Nature Clinical Practice Oncology. 2006; 3(10): 575580, doi: 10.1038/ncponc0608.
  32. Hahn H, Wojnowski L, Zimmer A, et al. Rhabdomyosarcomas and radiation hypersensitivity in a mouse model of Gorlin syndrome. Nature Medicine. 1998; 4(5): 619622, doi: 10.1038/nm0598-619.
  33. Innes A, Chudley A. Rhabdomyosarcoma in a Patient With CardioFacioCutaneous Syndrome. J Pediatr Hematol Oncol. 2000; 22(6): 546, doi: 10.1097/00043426-200011000-00017.
  34. Skapek SX, et al. Rhabdomyosarcoma. Nat Rev Dis Primers. 2019; 5(1): 1.
  35. Linardic CM, Naini S, Herndon JE, et al. The PAX3-FKHR fusion gene of rhabdomyosarcoma cooperates with loss of p16INK4A to promote bypass of cellular senescence. Cancer Res. 2007; 67(14): 66916699, doi: 10.1158/0008-5472.CAN-06-3210, indexed in Pubmed: 17638879.
  36. Keller C, Arenkiel BR, Coffin CM, et al. Alveolar rhabdomyosarcomas in conditional Pax3:Fkhr mice: cooperativity of Ink4a/ARF and Trp53 loss of function. Genes Dev. 2004; 18(21): 26142626, doi: 10.1101/gad.1244004, indexed in Pubmed: 15489287.
  37. Drummond CJ, et al. Hedgehog Pathway Drives Fusion-Negative Rhabdomyosarcoma Initiated From Non-myogenic Endothelial Progenitors. Cancer Cell. 2018; 33(1): 108124.e5.
  38. Ruiz-Mesa C, Goldberg J, Munoz AC, et al. Rhabdomyosarcoma in Adults: New Perspectives on Therapy. Curr Treat Options Oncol. 2015; 16(6), doi: 10.1007/s11864-015-0342-8.
  39. Casey DL, et al. Genomic Determinants of Clinical Outcomes in Rhabdomyosarcoma. Clin Cancer Res. 2020; 26(5): 11351140.
  40. Sorensen PHB, Lynch JC, Qualman SJ, et al. PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children’s oncology group. J Clin Oncol. 2002; 20(11): 26722679, doi: 10.1200/JCO.2002.03.137, indexed in Pubmed: 12039929.
  41. Davicioni E, Anderson JR, Buckley JD, et al. Gene expression profiling for survival prediction in pediatric rhabdomyosarcomas: a report from the children’s oncology group. J Clin Oncol. 2010; 28(7): 12401246, doi: 10.1200/JCO.2008.21.1268, indexed in Pubmed: 20124188.
  42. Bennicelli JL, Edwards RH, Barr FG. Mechanism for transcriptional gain of function resulting from chromosomal translocation in alveolar rhabdomyosarcoma. Proc Natl Acad Sci U S A. 1996; 93(11): 54555459, doi: 10.1073/pnas.93.11.5455, indexed in Pubmed: 8643596.
  43. Davis RJ, Barr FG. Fusion genes resulting from alternative chromosomal translocations are overexpressed by gene-specific mechanisms in alveolar rhabdomyosarcoma. Proc Natl Acad Sci U S A. 1997; 94(15): 80478051, doi: 10.1073/pnas.94.15.8047, indexed in Pubmed: 9223312.
  44. Thalhammer V, Lopez-Garcia LA, Herrero-Martin D, et al. PLK1 phosphorylates PAX3-FOXO1, the inhibition of which triggers regression of alveolar Rhabdomyosarcoma. Cancer Res. 2015; 75(1): 98110, doi: 10.1158/0008-5472.CAN-14-1246, indexed in Pubmed: 25398439.
  45. Paulino AC, Okcu MF. Rhabdomyosarcoma. Curr Probl Cancer. 2008; 32(1): 734, doi: 10.1016/j.currproblcancer.2007.11.001, indexed in Pubmed: 18206520.
  46. Gryder BE, Yohe ME, Chou HC, et al. PAX3-FOXO1 Establishes Myogenic Super Enhancers and Confers BET Bromodomain Vulnerability. Cancer Discov. 2017; 7(8): 884899, doi: 10.1158/2159-8290.CD-16-1297, indexed in Pubmed: 28446439.
  47. Böhm M, Wachtel M, Marques JG, et al. Helicase CHD4 is an epigenetic coregulator of PAX3-FOXO1 in alveolar rhabdomyosarcoma. J Clin Invest. 2016; 126(11): 42374249, doi: 10.1172/JCI85057, indexed in Pubmed: 27760049.
  48. Taulli R, et al. Validation of met as a therapeutic target in alveolar and embryonal rhabdomyosarcoma. Cancer Res. 2006; 66(9): 47424749.
  49. Reichek JL, Duan F, Smith LM, et al. Genomic and clinical analysis of amplification of the 13q31 chromosomal region in alveolar rhabdomyosarcoma: a report from the Children’s Oncology Group. Clin Cancer Res. 2011; 17(6): 14631473, doi: 10.1158/1078-0432.CCR-10-0091, indexed in Pubmed: 21220470.
  50. Barr FG, Duan F, Smith LM, et al. Genomic and clinical analyses of 2p24 and 12q13-q14 amplification in alveolar rhabdomyosarcoma: a report from the Children’s Oncology Group. Genes Chromosomes Cancer. 2009; 48(8): 661672, doi: 10.1002/gcc.20673, indexed in Pubmed: 19422036.
  51. Borinstein SC. Consensus and controversies regarding the treatment of rhabdomyosarcoma. Pediatr Blood Cancer. 2018; 65(2).
  52. Dziuba I, Kurzawa P, Dopierała M, et al. Rhabdomyosarcoma in children - current pathologic and molecular classification. Pol J Pathol. 2018; 69(1): 2032, doi: 10.5114/pjp.2018.75333, indexed in Pubmed: 29895123.
  53. Mitelman F, Mertens F. Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer. 2015. http://cgap.nci.nih.gov/Chromosomes/Mitelman.
  54. Parham DM, Barr FG. Sceletal Muscle Tumours. In: Fletcher CDM, Unni KK, Mertens F. ed. World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of Soft Tissue and Bone. IARC Press, Lyon 2002.
  55. Radzikowska J. Rhabdomyosarcoma of the head and neck in children. Contemp Oncol (Pozn). 2015; 19(2): 98107.
  56. Dasgupta R, Fuchs J, Rodeberg D. Rhabdomyosarcoma. Semin Pediatr Surg. 2016; 25(5): 276283, doi: 10.1053/j.sempedsurg.2016.09.011, indexed in Pubmed: 27955730.
  57. Shern JF, 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(2): 216231.
  58. Gordon A, McManus A, Anderson J, et al. Chromosomal imbalances in pleomorphic rhabdomyosarcomas and identification of the alveolar rhabdomyosarcoma-associated PAX3-FOXO1A fusion gene in one case. Cancer Genetics and Cytogenetics. 2003; 140(1): 7377, doi: 10.1016/s0165-4608(02)00631-3.
  59. Hettmer S, Archer N, Somers G, et al. Anaplastic rhabdomyosarcoma inTP53germline mutation carriers. Cancer. 2013; 120(7): 10681075, doi: 10.1002/cncr.28507.
  60. Leiner J, Le Loarer F. The current landscape of rhabdomyosarcomas: an update. Virchows Arch. 2020; 476(1): 97108, doi: 10.1007/s00428-019-02676-9, indexed in Pubmed: 31696361.
  61. Watson S, Perrin V, Guillemot D, et al. Transcriptomic definition of molecular subgroups of small round cell sarcomas. J Pathol. 2018; 245(1): 2940, doi: 10.1002/path.5053.
  62. Alaggio R, 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(2): 224235.
  63. Agaram N, LaQuaglia M, Alaggio R, et al. MYOD1-mutant spindle cell and sclerosing rhabdomyosarcoma: an aggressive subtype irrespective of age. A reappraisal for molecular classification and risk stratification. Modern Pathology. 2018; 32(1): 2736, doi: 10.1038/s41379-018-0120-9.
  64. Kohsaka S, Shukla N, Ameur N, et al. A recurrent neomorphic mutation in MYOD1 defines a clinically aggressive subset of embryonal rhabdomyosarcoma associated with PI3K-AKT pathway mutations. Nat Genet. 2014; 46(6): 595600, doi: 10.1038/ng.2969, indexed in Pubmed: 24793135.
  65. Agaram NP. Expanding the Spectrum of Intraosseous Rhabdomyosarcoma: Correlation Between 2 Distinct Gene Fusions and Phenotype. Am J Surg Pathol. 2019; 43(5): 695702.
  66. Chrisinger JSA, Wehrli B, Dickson BC, et al. Epithelioid and spindle cell rhabdomyosarcoma with FUS-TFCP2 or EWSR1-TFCP2 fusion: report of two cases. Virchows Arch. 2020; 477(5): 725732, doi: 10.1007/s00428-020-02870-0, indexed in Pubmed: 32556562.
  67. Dashti NK, Wehrs RN, Thomas BC, et al. Spindle cell rhabdomyosarcoma of bone with FUS-TFCP2 fusion: confirmation of a very recently described rhabdomyosarcoma subtype. Histopathology. 2018; 73(3): 514520, doi: 10.1111/his.13649, indexed in Pubmed: 29758589.
  68. Wong DD, van Vliet C, Gaman A, et al. Rhabdomyosarcoma with FUS re-arrangement: additional case in support of a novel subtype. Pathology. 2019; 51(1): 116120, doi: 10.1016/j.pathol.2018.09.056, indexed in Pubmed: 30477883.
  69. Tagami Y, et al. Spindle cell rhabdomyosarcoma in a lumbar vertebra with FUS-TFCP2 fusion. Pathol Res Pract. 2019; 215(8): 152399.
  70. Le Loarer F, Cleven AHG, Bouvier C, et al. A subset of epithelioid and spindle cell rhabdomyosarcomas is associated with TFCP2 fusions and common ALK upregulation. Mod Pathol. 2020; 33(3): 404419, doi: 10.1038/s41379-019-0323-8, indexed in Pubmed: 31383960.
  71. Leuschner I, et al. Spindle Cell Variants of Embryonal Rhabdomyosarcoma in the Paratesticular Region. Am J Surg Pathol. 1993; 17(8): 858, doi: 10.1097/00000478-199308000-00019.
  72. Shukla N, Ameur N, Yilmaz I, et al. Oncogene mutation profiling of pediatric solid tumors reveals significant subsets of embryonal rhabdomyosarcoma and neuroblastoma with mutated genes in growth signaling pathways. Clin Cancer Res. 2012; 18(3): 748757, doi: 10.1158/1078-0432.CCR-11-2056, indexed in Pubmed: 22142829.
  73. Stewart E, et al. Identification of Therapeutic Targets in Rhabdomyosarcoma through Integrated Genomic, Epigenomic, and Proteomic Analyses. Cancer Cell. 2018; 34(3): 411426.e19.
  74. Martinelli S, McDowell H, Vigne S, et al. RAS signaling dysregulation in human embryonal Rhabdomyosarcoma. Genes, Chromosomes and Cancer. 2009; 48(11): 975982, doi: 10.1002/gcc.20702.
  75. Seki M, Nishimura R, Yoshida K, et al. Abstract 482: Integrated genetic and epigenetic analysis defines novel molecular clusters in rhabdomyosarcoma. Tumor Biology. 2015, doi: 10.1158/1538-7445.am2015-482.
  76. Cao L, Yu Y, Bilke S, et al. Genome-wide identification of PAX3-FKHR binding sites in rhabdomyosarcoma reveals candidate target genes important for development and cancer. Cancer Res. 2010; 70(16): 64976508, doi: 10.1158/0008-5472.CAN-10-0582, indexed in Pubmed: 20663909.
  77. Bridge JA, Liu J, Qualman SJ, et al. Genomic gains and losses are similar in genetic and histologic subsets of rhabdomyosarcoma, whereas amplification predominates in embryonal with anaplasia and alveolar subtypes. Genes Chromosomes Cancer. 2002; 33(3): 310321, doi: 10.1002/gcc.10026, indexed in Pubmed: 11807989.
  78. Ganti R, Skapek SX, Zhang J, et al. Expression and genomic status of EGFR and ErbB-2 in alveolar and embryonal rhabdomyosarcoma. Mod Pathol. 2006; 19(9): 12131220, doi: 10.1038/modpathol.3800636, indexed in Pubmed: 16729016.
  79. Mark HF, Brown S, Sun CL, et al. Fluorescent in situ hybridization detection of HER-2/neu gene amplification in rhabdomyosarcoma. Pathobiology. 1998; 66(2): 5963, doi: 10.1159/000027997, indexed in Pubmed: 9645628.
  80. Taniguchi E, et al. PDGFR-A is a therapeutic target in alveolar rhabdomyosarcoma. Oncogene. 2008; 27(51): 65506560.
  81. Sharma P, Lioutas A, Fernandez-Fuentes N, et al. Arginine citrullination at the C-terminal domain controls RNA polymerase II transcription. Mol Cell. 2019; 73(1): 8496.e7, doi: 10.1016/j.molcel.2018.10.016.
  82. Naini S, et al. Defining the cooperative genetic changes that temporally drive alveolar rhabdomyosarcoma. Cancer Res. 2008; 68(23): 95839588.
  83. Hayes MN, Langenau DM. Discovering novel oncogenic pathways and new therapies using zebrafish models of sarcoma. Methods Cell Biol. 2017; 138: 525561, doi: 10.1016/bs.mcb.2016.11.011, indexed in Pubmed: 28129857.
  84. Huynh K, Fischle W, Verdin E, et al. BCoR, a novel corepressor involved in BCL-6 repression. Genes Dev. 2000; 14(14): 18101823, doi: 10.1101/gad.14.14.1810.
  85. Cortes Barrantes P, Jakobiec FA, Dryja TP. A Review of the Role of Cytogenetics in the Diagnosis of Orbital Rhabdomyosarcoma. Semin Ophthalmol. 2019; 34(4): 243251, doi: 10.1080/08820538.2019.1620802, indexed in Pubmed: 31146616.
  86. Huang HJ, Li XO, Zhong DR. Nasal cavity and paranasal sinus rhabdomyosarcoma:a clinicopathological and immunohistochemistry characteristics study of fifteen cases. Zhonghua Bing Li Xue Za Zhi. 2019; 48(11): 884886, doi: 10.3760/cma.j.issn.0529-5807.2019.11.010, indexed in Pubmed: 31775439.
  87. Torres-Peña JL, Ramos Castrillo AI, Mencía-Gutiérrez E, et al. Nasal Cavity or Alveolar Paranasal Sinus Rhabdomyosarcoma with Orbital Extension in Adults: 2 Cases. Plast Reconstr Surg Glob Open. 2015; 3(6): e414, doi: 10.1097/GOX.0000000000000366, indexed in Pubmed: 26180715.
  88. Purkayastha A, Sarin A, Bhatnagar S, et al. Adult Alveolar Paranasal Rhabdomyosarcoma; A Rare Aggressive Disease with Pulmonary, Brain and Skeletal Metastasis: Review of an Institutional Experience. Review of an Institutional Experience. SAJ Cancer Sci 5. 2018; 101.
  89. Campo M, Flamarique S, Asin G, et al. Multidisciplinary approach of a locally advanced adult alveolar rhabdomyosarcoma of paranasal sinuses: a case report and literature review. Rhinology Online. 2018; 1(1): 104107, doi: 10.4193/rhinol/18.034.
  90. Kanagalingam J, Medcalf M, Courtauld E, et al. Rhabdomyosarcoma of the Adult Nasopharynx. ORL. 2002; 64(3): 233236, doi: 10.1159/000058032.
  91. Warner BM, Griffith CC, Taylor WD, et al. Sclerosing rhabdomyosarcoma: presentation of a rare sarcoma mimicking myoepithelial carcinoma of the parotid gland and review of the literature. Head Neck Pathol. 2015; 9(1): 147152, doi: 10.1007/s12105-014-0540-x, indexed in Pubmed: 24710732.
  92. Febrero B, Oviedo I, Ríos A, et al. Primary rhabdomyosarcoma of the thyroid in an adult with auricular thrombosis. Eur Ann Otorhinolaryngol Head Neck Dis. 2017; 134(1): 4951, doi: 10.1016/j.anorl.2016.01.014, indexed in Pubmed: 27595524.
  93. McInturff M, Adamson A, Donaldson C, et al. Embryonal Rhabdomyosarcoma of the Oral Cavity. Head Neck Pathol. 2016; 11(3): 385388, doi: 10.1007/s12105-016-0761-2.
  94. Aggarwal A, Singh V, Pandey S, et al. Embryonal rhabdomyosarcoma of urinary bladder in an adult patient: an unusual manifestation. BMJ Case Rep. 2018; 2018: bcr2018224255, doi: 10.1136/bcr-2018-224255, indexed in Pubmed: 29654103.
  95. Schildhaus HU, Lokka S, Fenner W, et al. Spindle cell embryonal rhabdomyosarcoma of the prostate in an adult patient - case report and review of clinicopathological features. Diagn Pathol. 2016; 11(1): 56, doi: 10.1186/s13000-016-0507-1, indexed in Pubmed: 27357857.
  96. Townsend MF. Ureteral rhabdomyosarcoma. Urology. 1999; 54(3): 561.
  97. Alavi S, Eckes L, Kratschell R, et al. Pleomorphic Rhabdomyosarcoma of the Uterus - Case Report and a Systematic Review of the Literature. Anticancer Res. 2017; 37(5): 25092514, doi: 10.21873/anticanres.11592, indexed in Pubmed: 28476820.
  98. Alkhaledi A, Hanafi I, Alsabe H, et al. Rhabdomyosarcoma of the uterus with multiple metastases in a post-menopausal woman. Oxf Med Case Reports. 2019; 2019(3): omz017, doi: 10.1093/omcr/omz017, indexed in Pubmed: 30949355.
  99. Issam L, Sana L, Ismail E, et al. Vaginal embryonal rhabdomyosarcoma in young woman: A case report and literature review. Archives of Cancer Science and Therapy. 2020; 4(1): 034037, doi: 10.29328/journal.acst.1001020.
  100. Gong W, Gao Q, Xu Z, et al. Giant intrascrotal embryonal rhabdomyosarcoma in an adult: a case report and review of the literature. J Med Case Rep. 2018; 12(1): 149, doi: 10.1186/s13256-018-1607-1, indexed in Pubmed: 29804543.
  101. Breitfeld PP, Meyer WH. Rhabdomyosarcoma: new windows of opportunity. Oncologist. 2005; 10(7): 518527, doi: 10.1634/theoncologist.10-7-518, indexed in Pubmed: 16079319.
  102. Lawrence Jr W, Anderson JR, Gehan EA. Pretreatment TNM staging of childhood rhabdomyosarcoma: a report of the Intergroup Rhabdomyosarcoma Study Group. Children’s Cancer Study Group. Pediatric Oncology Group. Cancer. 1997; 80(6): 11651170, indexed in Pubmed: 9305719.
  103. Yin J, Liu Z, Yang K. Pleomorphic rhabdomyosarcoma of the liver with a hepatic cyst in an adult: Case report and literature review. Medicine (Baltimore). 2018; 97(29): e11335, doi: 10.1097/MD.0000000000011335, indexed in Pubmed: 30024506.
  104. Kim DoY, Seol YMi, Kim H, et al. Primary rhabdomyosarcoma of the breast in a 17-year-old girl: Case report. Medicine (Baltimore). 2017; 96(49): e9076, doi: 10.1097/MD.0000000000009076, indexed in Pubmed: 29245322.
  105. Kallianpur AA, Shukla NK, Deo SVS, et al. Primary mammary rhabdomyosarcoma in a nineteen year old female: A case report and review of literature. Indian J Cancer. 2015; 52(3): 295296, doi: 10.4103/0019-509X.176702, indexed in Pubmed: 26905115.
  106. Motola-Kuba D, M.-N. I, Fernandez-Ferreira R. Primary mediastinal embryonal rhabdomyosarcoma in adult: literature review and a case report. Journal of Cancer Biology & Research. 2018.
  107. Ammar-Boukhris A, Kamoun-Sellami N, Chtourou A, et al. Endobronchial pulmonary rhabdomyosarcoma. A case report. Rev Pneumol Clin. 2002; 58(5 Pt 1): 286289, indexed in Pubmed: 12486379.
  108. Ji Gy, Mao H. Primary pulmonary rhabdomyosarcoma in an adult: a case report and review of the literature. J Zhejiang Univ Sci B. 2013; 14(9): 859865, doi: 10.1631/jzus.B1200248, indexed in Pubmed: 24009208.
  109. Suda H, Koga N, Ohteki H. A case report of primary rhabdomyosarcoma of the heart treated with mitral valve replacement. Kyobu Geka. 1992; 45(13): 11831186, indexed in Pubmed: 1474694.
  110. DeLuca WM, Soderberg Jr CH, Riley RS, et al. Soliditary rhabdomyosarcoma of the pericardium: a case report and pathologic discussion. R I Med J. 1980; 63(3): 7983, indexed in Pubmed: 6929073.
  111. Medeiros CW, Kondo W, Baptista I, et al. Primary rhabdomyosarcoma of the diaphragm: case report and literature review. Rev Hosp Clin Fac Med Sao Paulo. 2002; 57(2): 6772, doi: 10.1590/s0041-87812002000200004, indexed in Pubmed: 11981587.
  112. Kuwabara T, Morioka H, Maki A, et al. The retroperitoneal rhabdomyosarcoma: a case report. Hinyokika Kiyo. 1990; 36(4): 433436, indexed in Pubmed: 2198788.
  113. Yu L, Yang SJ. Spindle cell rhabdomyosarcoma of the retroperitoneum: an unusual case developed in a pregnant woman but obscured by pregnancy. Int J Clin Exp Pathol. 2014; 7(8): 49044912.
  114. Shah R, Sabanathan S, Okereke CD, et al. Rhabdomyosarcoma of the oesophagus. A case report. J Cardiovasc Surg (Torino). 1995; 36(1): 99100, indexed in Pubmed: 7721934.
  115. Palermo M, Mastronardi LM, García RH, et al. Primary gastric rhabdomyosarcoma. Case report. Acta Gastroenterol Latinoam. 2012; 42(2): 131134, indexed in Pubmed: 22876716.
  116. Damiani S, Nappi O, Eusebi V. Primary rhabdomyosarcoma of the ileum in an adult. Arch Pathol Lab Me. 1991; 115(3): 235238, indexed in Pubmed: 2001160.
  117. Karcioglu Z, Hadjistilianou D, Rozans M, et al. Orbital Rhabdomyosarcoma. Cancer Control. 2017; 11(5): 328333, doi: 10.1177/107327480401100507.
  118. Sharada S. Adult Rhabdomyosarcoma: A Rare Case Report and the Associated Challenges. Journal of Clinical and Experimental Ophthalmology. 2020; 11(3), doi: 10.35248/2155-9570.20.11.838.
  119. Ahmad TY, Al Houri HN, Al Houri AN, et al. Aggressive orbital rhabdomyosarcoma in adulthood: A case report in a public hospital in Damascus, Syria. Avicenna J Med. 2018; 8(3): 110113, doi: 10.4103/ajm.AJM_62_18, indexed in Pubmed: 30090751.
  120. Dilger AE, S AL, Cramer J, et al. Rhabdomyosarcoma of the Paranasal Sinuses Initially Diagnosed as Acute Sinusitis. Sinusitis. 2017.
  121. Kariya S, Cureoglu S, Schachern P, et al. Histopathological temporal bone study of the metastatic rhabdomyosarcoma. Auris Nasus Larynx. 2009; 36(2): 221223, doi: 10.1016/j.anl.2008.05.008.
  122. Ormeci T, Durmus O, Saral I, et al. Persistent abdominal pain after exercise: an unexpected diagnosis. Acta Reumatol Port. 2015; 40(2): 195197, indexed in Pubmed: 26219974.
  123. Kam J, Yuminaga Y, Maclean F, et al. Rapidly growing massive pleomorphic rhabdomyosarcoma of the bladder presenting with bladder outlet obstruction. ANZ J Surg. 2018; 88(3): E208E209, doi: 10.1111/ans.13319, indexed in Pubmed: 26381078.
  124. Aldabagh SM, Shibata CS, Taxy JB. Rhabdomyosarcoma of the common bile duct in an adult. Arch Pathol Lab Med. 1986; 110(6): 547550, indexed in Pubmed: 3518654.
  125. Xi S, Tong W. Pleomorphic rhabdomyosarcoma metastasis to small intestine causing intussusception: A case report. Medicine (Baltimore). 2018; 97(51): e13648, doi: 10.1097/MD.0000000000013648, indexed in Pubmed: 30572480.
  126. Yadav SK, Sinha DK, Ahmed A, et al. Primary Intra-Abdominal Rhabdomyosarcoma in an Adult: an Unusual Presentation and Review of Literature. Indian J Surg Oncol. 2015; 6(2): 119122, doi: 10.1007/s13193-015-0376-1, indexed in Pubmed: 26405418.
  127. Oberlin O, Rey A, Lyden E, et al. Prognostic factors in metastatic rhabdomyosarcomas: results of a pooled analysis from United States and European cooperative groups. J Clin Oncol. 2008; 26(14): 23842389, doi: 10.1200/JCO.2007.14.7207, indexed in Pubmed: 18467730.
  128. Tallroth K. Lymphatic dissemination of bone and soft tissue sarcomas: a lymphographic investigation. Acta Radiol Suppl. 1976; 349: 184, indexed in Pubmed: 206099.
  129. Saboo S, Krajewski K, Zukotynski K, et al. Imaging Features of Primary and Secondary Adult Rhabdomyosarcoma. American Journal of Roentgenology. 2012; 199(6): W694W703, doi: 10.2214/ajr.11.8213.
  130. Luporsi M, Cassou-Mounat T, Amiot HM, et al. Rhabdomyosarcoma Revealed by a Breast Metastasis. Clinical Nuclear Medicine. 2018; 43(3): e98e100, doi: 10.1097/rlu.0000000000001971.
  131. Dasgupta R, Rodeberg DA. Update on rhabdomyosarcoma. Semin Pediatr Surg. 2012; 21(1): 6878, doi: 10.1053/j.sempedsurg.2011.10.007, indexed in Pubmed: 22248972.
  132. Newton W, Gehan E, Webber B, et al. Classification of rhabdomyosarcomas and related sarcomas. Pathologic aspects and proposal for a new classification-an intergroup rhabdomyosarcoma study. Cancer. 1995; 76(6): 10731085, doi: 10.1002/1097-0142(19950915)76:6<1073::aid-cncr2820760624>3.0.co;2-l.
  133. Hawkins DS, Gupta AA, Rudzinski ER. What is new in the biology and treatment of pediatric rhabdomyosarcoma? Curr Opin Pediatr. 2014; 26(1): 5056, doi: 10.1097/MOP.0000000000000041, indexed in Pubmed: 24326270.
  134. Rekhi B, Gupta C, Chinnaswamy G, et al. Clinicopathologic features of 300 rhabdomyosarcomas with emphasis upon differential expression of skeletal muscle specific markers in the various subtypes: A single institutional experience. Ann Diagn Pathol. 2018; 36: 5060, doi: 10.1016/j.anndiagpath.2018.07.002, indexed in Pubmed: 30098515.
  135. Folpe AL, Graham RP, Martinez A, et al. Mesenchymal chondrosarcomas showing immunohistochemical evidence of rhabdomyoblastic differentiation: a potential diagnostic pitfall. Hum Pathol. 2018; 77: 2834, doi: 10.1016/j.humpath.2018.03.012, indexed in Pubmed: 29559236.
  136. Xia SJ, Pressey JG, Barr FG. Molecular pathogenesis of rhabdomyosarcoma. Cancer Biol Ther. 2002; 1(2): 97104, doi: 10.4161/cbt.51, indexed in Pubmed: 12170781.
  137. Sumegi J, Streblow R, Frayer R, et al. Recurrent t(2;2) and t(2;8) translocations in rhabdomyosarcoma without the canonicalPAX-FOXO1fusePAX3to members of the nuclear receptor transcriptional coactivator family. Genes, Chromosomes and Cancer. 2009; 49(3): 224236, doi: 10.1002/gcc.20731.
  138. Folpe AL, Hill CE, Parham DM, et al. Immunohistochemical detection of FLI-1 protein expression: a study of 132 round cell tumors with emphasis on CD99-positive mimics of Ewing’s sarcoma/primitive neuroectodermal tumor. Am J Surg Pathol. 2000; 24(12): 16571662, doi: 10.1097/00000478-200012000-00010, indexed in Pubmed: 11117787.
  139. Brohl AS, Kahen E, Yoder SJ, et al. The genomic landscape of malignant peripheral nerve sheath tumors: diverse drivers of Ras pathway activation. Sci Rep. 2017; 7(1): 14992, doi: 10.1038/s41598-017-15183-1, indexed in Pubmed: 29118384.
  140. Agaram N, Sung YS, Zhang L, et al. Dichotomy of Genetic Abnormalities in PEComas With Therapeutic Implications. American Journal of Surgical Pathology. 2015; 39(6): 813825, doi: 10.1097/pas.0000000000000389.
  141. Sangiorgio V, Daniele L, Gallo T, et al. Ultrasound-guided fine needle aspiration cytology in the diagnosis of hepatic and pancreatic perivascular epithelioid cell tumors: A case series. Diagn Cytopathol. 2019; 47(4): 315319, doi: 10.1002/dc.24111, indexed in Pubmed: 30417984.
  142. Thway K, Noujaim J, Jones RL, et al. Dermatofibrosarcoma protuberans: pathology, genetics, and potential therapeutic strategies. Ann Diagn Pathol. 2016; 25: 6471, doi: 10.1016/j.anndiagpath.2016.09.013, indexed in Pubmed: 27806849.
  143. Larbcharoensub N, Kayankarnnavee J, Sanpaphant S, et al. Clinicopathological features of dermatofibrosarcoma protuberans. Oncol Lett. 2016; 11(1): 661667, doi: 10.3892/ol.2015.3966, indexed in Pubmed: 26870263.
  144. Ferrari A, Dileo P, Casanova M, et al. Rhabdomyosarcoma in adults. A retrospective analysis of 171 patients treated at a single institution. Cancer. 2003; 98(3): 571580, doi: 10.1002/cncr.11550, indexed in Pubmed: 12879475.
  145. Parham DM, B FG, Fletcher CDM, et al. Alveolar rhabdomyosarcoma WHO Classification of Tumours of Soft Tissue and Bone. IARC, Lyon 2013.
  146. Parham DM, Barr FG. Classification of rhabdomyosarcoma and its molecular basis. Adv Anat Pathol. 2013; 20(6): 387397, doi: 10.1097/PAP.0b013e3182a92d0d, indexed in Pubmed: 24113309.
  147. Bompas E, Campion L, Italiano A, et al. Outcome of 449 adult patients with rhabdomyosarcoma: an observational ambispective nationwide study. Cancer Med. 2018; 7(8): 40234035, doi: 10.1002/cam4.1374, indexed in Pubmed: 29956493.
  148. Parham DM, B FG, Fletcher CDM, et al. Embryonal rhabdomyosarcoma. WHO Classification of Tumours of Soft Tissue and Bone. IARC, Lyon 2013.
  149. Rudzinski E, Teot L, Anderson J, et al. Dense Pattern of Embryonal Rhabdomyosarcoma, a Lesion Easily Confused With Alveolar Rhabdomyosarcoma. American Journal of Clinical Pathology. 2013; 140(1): 8290, doi: 10.1309/ajcpa1wn7arpcmkq.
  150. Qualman S, et al. revalence and clinical impact of anaplasia in childhood rhabdomyosarcoma : a report from the Soft Tissue Sarcoma Committee of the Children’s Oncology Group. Cancer. 2008; 113(11): 32423247.
  151. Kodet R, Newton Jr WA, Hamoudi AB, et al. Childhood rhabdomyosarcoma with anaplastic (pleomorphic) features. A report of the Intergroup Rhabdomyosarcoma Study. Am J Surg Pathol. 1993; 17(5): 443453, doi: 10.1097/00000478-199305000-00002, indexed in Pubmed: 8470759.
  152. Goldblum JR, F AL, Weiss SW. Enzinger and Weiss’s Soft Tissue Tumors. Elsevier Saunders, Philadelphia 2014: 601638.
  153. Dias P, Chen B, Dilday B, et al. Strong Immunostaining for Myogenin in Rhabdomyosarcoma Is Significantly Associated with Tumors of the Alveolar Subclass. Am J Pathol. 2000; 156(2): 399408, doi: 10.1016/s0002-9440(10)64743-8.
  154. Furlong MA, Mentzel T, Fanburg-Smith JC. Pleomorphic rhabdomyosarcoma in adults: a clinicopathologic study of 38 cases with emphasis on morphologic variants and recent skeletal muscle-specific markers. Mod Pathol. 2001; 14(6): 595603, doi: 10.1038/modpathol.3880357, indexed in Pubmed: 11406662.
  155. Little DJ, et al. Adult rhabdomyosarcoma: outcome following multimodality treatment. Cancer. 2002; 95(2): 377388.
  156. Montgomery EA, B FG, Fletcher CDM, et al. Pleomorphic rhabdomyosarcoma. WHO Classification of Tumours of Soft Tissue and Bone. IARC, Lyon 2013.
  157. Doyle LA. Sarcoma classification: an update based on the 2013 World Health Organization Classification of Tumors of Soft Tissue and Bone. Cancer. 2014; 120(12): 17631774, doi: 10.1002/cncr.28657, indexed in Pubmed: 24648013.
  158. Nascimento AF, B FG, Fletcher CDM, et al. Spindle cell/sclerosing rhabdomyosarcoma. WHO Classification of Tumours of Soft Tissue and Bone. IARC, Lyon 2013.
  159. Cavazzana AO, Schmidt D, Ninfo V, et al. Spindle cell rhabdomyosarcoma. A prognostically favorable variant of rhabdomyosarcoma. Am J Surg Pathol. 1992; 16(3): 229235, indexed in Pubmed: 1599014.
  160. Val-Bernal J, Fernández N, Gómez-Román J. Spindle cell rhabdomyosarcoma in adults. A case report and literature review. Pathology - Research and Practice. 2000; 196(1): 6772, doi: 10.1016/s0344-0338(00)80024-2.
  161. Agaram N, Chen CL, Zhang L, et al. RecurrentMYOD1mutations in pediatric and adult sclerosing and spindle cell rhabdomyosarcomas: Evidence for a common pathogenesis. Genes, Chromosomes and Cancer. 2014; 53(9): 779787, doi: 10.1002/gcc.22187.
  162. Zhao Z, Yin Y, Zhang J, et al. Spindle cell/sclerosing rhabdomyosarcoma: case series from a single institution emphasizing morphology, immunohistochemistry and follow-up. Int J Clin Exp Pathol. 2015; 8(11): 1381413820, indexed in Pubmed: 26823695.
  163. Folpe A, McKenney J, Bridge J, et al. Sclerosing Rhabdomyosarcoma in Adults. Am J Surg Pathol. 2002; 26(9): 11751183, doi: 10.1097/00000478-200209000-00008.
  164. Carroll SJ, Nodit L. Spindle cell rhabdomyosarcoma: a brief diagnostic review and differential diagnosis. Arch Pathol Lab Med. 2013; 137(8): 11551158, doi: 10.5858/arpa.2012-0465-RS, indexed in Pubmed: 23899074.
  165. Rekhi B, Singhvi T. Histopathological, immunohistochemical and molecular cytogenetic analysis of 21 spindle cell/sclerosing rhabdomyosarcomas. APMIS. 2014; 122(11): 11441152, doi: 10.1111/apm.12272, indexed in Pubmed: 24730567.
  166. Hayes-Jordan A, Andrassy R. Rhabdomyosarcoma in children. Curr Opin Pediatr. 2009; 21(3): 373378, doi: 10.1097/MOP.0b013e32832b4171, indexed in Pubmed: 19448544.
  167. Crist WM, Anderson JR, Meza JL, et al. Intergroup rhabdomyosarcoma study-IV: results for patients with nonmetastatic disease. J Clin Oncol. 2001; 19(12): 30913102, doi: 10.1200/JCO.2001.19.12.3091, indexed in Pubmed: 11408506.
  168. Tanaka K, Ozaki T. New TNM classification (AJCC eighth edition) of bone and soft tissue sarcomas: JCOG Bone and Soft Tissue Tumor Study Group. Jpn J Clin Oncol. 2019; 49(2): 103107, doi: 10.1093/jjco/hyy157, indexed in Pubmed: 30423153.
  169. Crane JN, Xue W, Qumseya A, et al. Clinical group and modified TNM stage for rhabdomyosarcoma: A review from the Children’s Oncology Group. Pediatr Blood Cancer. 2022; 69(6): e29644, doi: 10.1002/pbc.29644, indexed in Pubmed: 35253352.
  170. Meza JL, Anderson J, Pappo AS, et al. Children’s Oncology Group. Analysis of prognostic factors in patients with nonmetastatic rhabdomyosarcoma treated on intergroup rhabdomyosarcoma studies III and IV: the Children’s Oncology Group. J Clin Oncol. 2006; 24(24): 38443851, doi: 10.1200/JCO.2005.05.3801, indexed in Pubmed: 16921036.
  171. Spalding AC, Hawkins DS, Donaldson SS, et al. The effect of radiation timing on patients with high-risk features of parameningeal rhabdomyosarcoma: an analysis of IRS-IV and D9803. Int J Radiat Oncol Biol Phys. 2013; 87(3): 512516, doi: 10.1016/j.ijrobp.2013.07.003, indexed in Pubmed: 24074925.
  172. Ladra MM, Szymonifka JD, Mahajan A, et al. Preliminary results of a phase II trial of proton radiotherapy for pediatric rhabdomyosarcoma. J Clin Oncol. 2014; 32(33): 37623770, doi: 10.1200/JCO.2014.56.1548, indexed in Pubmed: 25332253.
  173. Benkhaled S, Mané M, Jungels C, et al. Successful treatment of synchronous chemoresistant pulmonary metastasis from pleomorphic rhabdomyosarcoma with stereotaxic body radiation therapy: A case report and a review of the literature. Cancer Treat Res Commun. 2021; 26: 100282, doi: 10.1016/j.ctarc.2020.100282, indexed in Pubmed: 33360328.
  174. Kojima Y, Hashimoto K, Ando M, et al. Comparison of dose intensity of vincristine, d-actinomycin, and cyclophosphamide chemotherapy for child and adult rhabdomyosarcoma: a retrospective analysis. Cancer Chemother Pharmacol. 2012; 70(3): 391397, doi: 10.1007/s00280-012-1920-0, indexed in Pubmed: 22806306.
  175. Childhood rhabdomyosarcoma treatment for health professionals. . Nat. Cancer Institute. http://www.cancer.gov.
  176. Raney RB, Maurer HM, Anderson JR, et al. The Intergroup Rhabdomyosarcoma Study Group (IRSG): Major Lessons From the IRS-I Through IRS-IV Studies as Background for the Current IRS-V Treatment Protocols. Sarcoma. 2001; 5(1): 915, doi: 10.1080/13577140120048890, indexed in Pubmed: 18521303.
  177. Simon JH, Paulino AC, Ritchie JM, et al. Presentation, prognostic factors and patterns of failure in adult rhabdomyosarcoma. Sarcoma. 2003; 7(1): 17, doi: 10.1080/1357714031000114147, indexed in Pubmed: 18521362.
  178. Hawkins WG, Hoos A, Antonescu CR, et al. Clinicopathologic analysis of patients with adult rhabdomyosarcoma. Cancer. 2001; 91(4): 794803, indexed in Pubmed: 11241248.
  179. Jeziorski A, Rutkowski P. Biblioteka Chirurga Onkologa. Tom 3. Mięsaki tkanek miękkich. Via Medica, Gdańsk 2015.
  180. Keskin S, Ekenel M, Basaran M, et al. Clinicopathological characteristics and treatment outcomes of adult patients with paratesticular rhabdomyosarcoma (PRMS): A 10-year single-centre experience. Can Urol Assoc J. 2012; 6(1): 4245, doi: 10.5489/cuaj.11121, indexed in Pubmed: 22396368.
  181. Wu TH, Huang JS, Wang HM, et al. Long-term survivors of adult rhabdomyosarcoma of maxillary sinus following multimodal therapy: case reports and literature reviews. Chang Gung Med J. 2010; 33(4): 466471, indexed in Pubmed: 20804675.
  182. Mascarenhas L, Chi YY, Hingorani P, 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. Journal of Clinical Oncology. 2019; 37(31): 28662874, doi: 10.1200/jco.19.00576.
  183. Saylors RL, Stine KC, Sullivan J, et al. Pediatric Oncology Group. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J Clin Oncol. 2001; 19(15): 34633469, doi: 10.1200/JCO.2001.19.15.3463, indexed in Pubmed: 11481351.
  184. Minard-Colin V, Ichante JL, Nguyen L, et al. Phase II study of vinorelbine and continuous low doses cyclophosphamide in children and young adults with a relapsed or refractory malignant solid tumour: good tolerance profile and efficacy in rhabdomyosarcoma-a report from the Société Française des Cancers et leucémies de l’Enfant et de l’adolescent (SFCE). Eur J Cancer. 2012; 48(15): 24092416, doi: 10.1016/j.ejca.2012.04.012, indexed in Pubmed: 22633624.
  185. Casanova M, Ferrari A, Spreafico F, et al. Vinorelbine in previously treated advanced childhood sarcomas: evidence of activity in rhabdomyosarcoma. Cancer. 2002; 94(12): 32633268, doi: 10.1002/cncr.10600, indexed in Pubmed: 12115359.
  186. Kuttesch JF, Krailo MD, Madden T, et al. Children’s Oncology Group. Phase II evaluation of intravenous vinorelbine (Navelbine) in recurrent or refractory pediatric malignancies: a Children’s Oncology Group study. Pediatr Blood Cancer. 2009; 53(4): 590593, doi: 10.1002/pbc.22133, indexed in Pubmed: 19533657.
  187. Mascarenhas L, Lyden ER, Breitfeld PP, et al. Randomized phase II window trial of two schedules of irinotecan with vincristine in patients with first relapse or progression of rhabdomyosarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2010; 28(30): 46584663, doi: 10.1200/JCO.2010.29.7390, indexed in Pubmed: 20837952.
  188. Bay JO, Ray-Coquard I, Fayette J, et al. Groupe Sarcome Français. Docetaxel and gemcitabine combination in 133 advanced soft-tissue sarcomas: a retrospective analysis. Int J Cancer. 2006; 119(3): 706711, doi: 10.1002/ijc.21867, indexed in Pubmed: 16496406.
  189. Judson I, Verweij J, Gelderblom H, et al. Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: a randomised controlled phase 3 trial. Lancet Oncol. 2014; 15(4): 415423, doi: 10.1016/s1470-2045(14)70063-4, indexed in Pubmed: 24618336.
  190. Defachelles A, Bogart E, Casanova M, et al. Randomized phase 2 trial of the combination of vincristine and irinotecan with or without temozolomide, in children and adults with refractory or relapsed rhabdomyosarcoma (RMS). Journal of Clinical Oncology. 2019; 37(15_suppl): 1000010000, doi: 10.1200/jco.2019.37.15_suppl.10000.
  191. Seto T, Song MN, Trieu M, et al. Real-World Experiences with Pazopanib in Patients with Advanced Soft Tissue and Bone Sarcoma in Northern California. Med Sci (Basel). 2019; 7(3), doi: 10.3390/medsci7030048, indexed in Pubmed: 30889920.
  192. Hashimoto A, Takada K, Takimoto R, et al. Effective treatment of metastatic rhabdomyosarcoma with pazopanib. Gan To Kagaku Ryoho. 2014; 41(8): 10411044, indexed in Pubmed: 25132042.
  193. Schöffski P, Wozniak A, Leahy MG, et al. The tyrosine kinase inhibitor crizotinib does not have clinically meaningful activity in heavily pre-treated patients with advanced alveolar rhabdomyosarcoma with FOXO rearrangement: European Organisation for Research and Treatment of Cancer phase 2 trial 90101 ‘CREATE’. Eur J Cancer. 2018; 94: 156167, doi: 10.1016/j.ejca.2018.02.011, indexed in Pubmed: 29567632.
  194. Rodeberg DA, Stoner JA, Hayes-Jordan A, et al. Prognostic significance of tumor response at the end of therapy in group III rhabdomyosarcoma: a report from the children’s oncology group. J Clin Oncol. 2009; 27(22): 37053711, doi: 10.1200/JCO.2008.19.5933, indexed in Pubmed: 19470937.
  195. Casey DL, Wexler LH, Fox JJ, et al. Predicting outcome in patients with rhabdomyosarcoma: role of [(18)f]fluorodeoxyglucose positron emission tomography. Int J Radiat Oncol Biol Phys. 2014; 90(5): 11361142, doi: 10.1016/j.ijrobp.2014.08.005, indexed in Pubmed: 25539372.
  196. Admiraal R, van der Paardt M, Kobes J, et al. High-dose chemotherapy for children and young adults with stage IV rhabdomyosarcoma. Cochrane Database Syst Rev. 2010(12): CD006669, doi: 10.1002/14651858.CD006669.pub2, indexed in Pubmed: 21154373.
  197. Womer RB, Pressey JG. Rhabdomyosarcoma and soft tissue sarcoma in childhood. Curr Opin Oncol. 2000; 12(4): 337344, doi: 10.1097/00001622-200007000-00010, indexed in Pubmed: 10888419.
  198. Tlemsani C, Leroy K, Gimenez-Roqueplo AP, et al. Chemoresistant pleomorphic rhabdomyosarcoma: whole exome sequencing reveals underlying cancer predisposition and therapeutic options. J Med Genet. 2020; 57(2): 104108, doi: 10.1136/jmedgenet-2018-105594, indexed in Pubmed: 30352869.
  199. Quaglia MLa, Heller G, Ghavimi F, et al. The effect of age at diagnosis on outcome in rhabdomyosarcoma. Cancer. 1994; 73(1): 109117, doi: 10.1002/1097-0142(19940101)73:1<109::aid-cncr2820730120>3.0.co;2-s.
  200. Kojima Y, Hashimoto K, Ando M, et al. Clinical outcomes of adult and childhood rhabdomyosarcoma treated with vincristine, d-actinomycin, and cyclophosphamide chemotherapy. J Cancer Res Clin Oncol. 2012; 138(7): 12491257, doi: 10.1007/s00432-012-1199-x, indexed in Pubmed: 22441933.
  201. Khosla D, Sapkota S, Kapoor R, et al. Adult rhabdomyosarcoma: Clinical presentation, treatment, and outcome. J Cancer Res Ther. 2015; 11(4): 830834, indexed in Pubmed: 26881526.
  202. Sookprasert A, Ungareewittaya P, Manotepitipongse A, et al. Treatment Outcome and Predictors of Survival in Thai Adult Rhabdomyosarcoma Cases. Asian Pac J Cancer Prev. 2016; 17(3): 14491452, doi: 10.7314/apjcp.2016.17.3.1449.
  203. Drabbe C, Benson C, Younger E, et al. Embryonal and Alveolar Rhabdomyosarcoma in Adults: Real-Life Data From a Tertiary Sarcoma Centre. Clin Oncol. 2020; 32(1): e27e35, doi: 10.1016/j.clon.2019.07.007.
  204. Patel SR, Hensel CP, He J, et al. Epidemiology and survival outcome of adult kidney, bladder, and prostate rhabdomyosarcoma: A SEER database analysis. Rare Tumors. 2020; 12: 2036361320977401, doi: 10.1177/2036361320977401, indexed in Pubmed: 33329884.
  205. Ben Arush M, Minard-Colin V, Mosseri V, et al. Does aggressive local treatment have an impact on survival in children with metastatic rhabdomyosarcoma? Eur J Cancer. 2015; 51(2): 193201, doi: 10.1016/j.ejca.2014.11.009, indexed in Pubmed: 25471261.
  206. Egas-Bejar D, Huh WW. Rhabdomyosarcoma in adolescent and young adult patients: current perspectives. Adolesc Health Med Ther. 2014; 5: 115125, doi: 10.2147/AHMT.S44582, indexed in Pubmed: 24966711.
  207. Kashtan M, Jayakrishnan T, Rajeev R, et al. Age-based disparities in treatment and outcomes of retroperitoneal rhabdomyosarcoma. Int J Clin Oncol. 2015; 21(3): 602608, doi: 10.1007/s10147-015-0918-0.
  208. Dumont SN, Araujo DM, Munsell MF, et al. Management and outcome of 239 adolescent and adult rhabdomyosarcoma patients. Cancer Med. 2013; 2(4): 553563, doi: 10.1002/cam4.92, indexed in Pubmed: 24156028.
  209. Mandeville HC. Radiotherapy in the Management of Childhood Rhabdomyosarcoma. Clin Oncol (R Coll Radiol). 2019; 31(7): 462470, doi: 10.1016/j.clon.2019.03.047, indexed in Pubmed: 30992168.
  210. Wasti A, Mandeville H, Gatz S, et al. Rhabdomyosarcoma. Paediatrics and Child Health. 2018; 28(4): 157163, doi: 10.1016/j.paed.2018.03.001.
  211. Kazanowska B, G J. Mięsaki tkanek miękkich. In: Chybicka A, Sawicz-Birkowska K. ed. Onkologia i Hematologia Dziecięca. PZWL, Warszawa 2008.
  212. Kapoor G, Das K. Soft tissue sarcomas in children. Indian J Pediatr. 2012; 79(7): 936942, doi: 10.1007/s12098-011-0560-4, indexed in Pubmed: 21935710.
  213. Arndt CAS. Risk stratification of rhabdomyosarcoma: a moving target. Am Soc Clin Oncol Educ Book. 2013: 415419, doi: 10.14694/EdBook_AM.2013.33.415, indexed in Pubmed: 23714563.
  214. Gosiengfiao Y, Reichek J, Walterhouse D. What is new in rhabdomyosarcoma management in children? Paediatr Drugs. 2012; 14(6): 389400, doi: 10.2165/11599440-000000000-00000, indexed in Pubmed: 23050743.
  215. Skapek SX, Anderson J, Barr FG, et al. PAX-FOXO1 fusion status drives unfavorable outcome for children with rhabdomyosarcoma: a children’s oncology group report. Pediatr Blood Cancer. 2013; 60(9): 14111417, doi: 10.1002/pbc.24532, indexed in Pubmed: 23526739.
  216. Missiaglia E, Williamson D, Chisholm J, et al. PAX3/FOXO1 fusion gene status is the key prognostic molecular marker in rhabdomyosarcoma and significantly improves current risk stratification. J Clin Oncol. 2012; 30(14): 16701677, doi: 10.1200/JCO.2011.38.5591, indexed in Pubmed: 22454413.
  217. Hibbitts E, Chi YY, Hawkins DS, et al. Refinement of risk stratification for childhood rhabdomyosarcoma using FOXO1 fusion status in addition to established clinical outcome predictors: A report from the Children’s Oncology Group. Cancer Med. 2019; 8(14): 64376448, doi: 10.1002/cam4.2504, indexed in Pubmed: 31456361.
  218. Hettmer S, Linardic CM, Kelsey A, et al. Molecular testing of rhabdomyosarcoma in clinical trials to improve risk stratification and outcome: A consensus view from European paediatric Soft tissue sarcoma Study Group, Children’s Oncology Group and Cooperative Weichteilsarkom-Studiengruppe. Eur J Cancer. 2022 [Epub ahead of print]; 172: 367386, doi: 10.1016/j.ejca.2022.05.036, indexed in Pubmed: 35839732.
  219. Haduong JH, Heske CM, Allen-Rhoades W, et al. An update on rhabdomyosarcoma risk stratification and the rationale for current and future Children’s Oncology Group clinical trials. Pediatr Blood Cancer. 2022; 69(4): e29511, doi: 10.1002/pbc.29511, indexed in Pubmed: 35129294.
  220. Hays D, Lawrence W, Wharam M, et al. Primary reexcision for patients with ‘microscopic residual’ tumor following initial excision of sarcomas of trunk and extremity sites. J Pediatr Surg. 1989; 24(1): 510, doi: 10.1016/s0022-3468(89)80290-8.
  221. Seitz G, Dantonello TM, Int-Veen C, et al. CWS-96 Study Group. Treatment efficiency, outcome and surgical treatment problems in patients suffering from localized embryonal bladder/prostate rhabdomyosarcoma: a report from the Cooperative Soft Tissue Sarcoma trial CWS-96. Pediatr Blood Cancer. 2011; 56(5): 718724, doi: 10.1002/pbc.22950, indexed in Pubmed: 21370402.
  222. Greenberger BA, Yock TI. The role of proton therapy in pediatric malignancies: Recent advances and future directions. Semin Oncol. 2020; 47(1): 822, doi: 10.1053/j.seminoncol.2020.02.002, indexed in Pubmed: 32139101.
  223. Wolden SL, Lyden ER, Arndt CA, et al. Local Control for Intermediate-Risk Rhabdomyosarcoma: Results From D9803 According to Histology, Group, Site, and Size: A Report From the Children’s Oncology Group. Int J Radiat Oncol Biol Phys. 2015; 93(5): 10711076, doi: 10.1016/j.ijrobp.2015.08.040, indexed in Pubmed: 26581144.
  224. Dantonello TM, Winkler P, Boelling T, et al. CWS Study Group. Embryonal rhabdomyosarcoma with metastases confined to the lungs: report from the CWS Study Group. Pediatr Blood Cancer. 2011; 56(5): 725732, doi: 10.1002/pbc.22862, indexed in Pubmed: 21370403.
  225. Hibbitts E, Chi YY, Hawkins DS, et al. Refinement of risk stratification for childhood rhabdomyosarcoma using FOXO1 fusion status in addition to established clinical outcome predictors: A report from the Children’s Oncology Group. Cancer Med. 2019; 8(14): 64376448, doi: 10.1002/cam4.2504, indexed in Pubmed: 31456361.
  226. Heske CM, Mascarenhas L. Relapsed Rhabdomyosarcoma. J Clin Med. 2021; 10(4), doi: 10.3390/jcm10040804, indexed in Pubmed: 33671214.
  227. Dantonello T, Int-Veen C, Schuck A, et al. Survival following disease recurrence of primary localized alveolar rhabdomyosarcoma. Pediatric Blood & Cancer. 2013; 60(8): 12671273, doi: 10.1002/pbc.24488.
  228. Wagner LM, Fouladi M, Ahmed A, et al. Phase II study of cixutumumab in combination with temsirolimus in pediatric patients and young adults with recurrent or refractory sarcoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2015; 62(3): 440444, doi: 10.1002/pbc.25334, indexed in Pubmed: 25446280.
  229. Weigel B, Malempati S, Reid JM, et al. Phase 2 trial of cixutumumab in children, adolescents, and young adults with refractory solid tumors: a report from the Children’s Oncology Group. Pediatr Blood Cancer. 2014; 61(3): 452456, doi: 10.1002/pbc.24605, indexed in Pubmed: 23956055.
  230. Okada K, Yamasaki K, Tanaka C, et al. Phase I study of bevacizumab plus irinotecan in pediatric patients with recurrent/refractory solid tumors. Jpn J Clin Oncol. 2013; 43(11): 10731079, doi: 10.1093/jjco/hyt124, indexed in Pubmed: 24002900.
  231. Federman N, Crane J, Gonzales A, et al. A phase 1 dose-escalation/ /expansion clinical trial of mocetinostat in combination with vinorelbine in adolescents and young adults with refractory and/or recurrent rhabdomyosarcoma: Interim results. Journal of Clinical Oncology. 2022; 40(16_suppl): 1155311553, doi: 10.1200/jco.2022.40.16_suppl.11553.