Rhabdomyosarcoma: A Comprehensive Review of Subtypes, Molecular Biology, Clinical Management, and Emerging Therapeutic Strategies

Rhabdomyosarcoma: A Comprehensive Review of Subtypes, Molecular Biology, Clinical Management, and Emerging Therapeutic Strategies

Abstract

Rhabdomyosarcoma (RMS) is an aggressive soft tissue sarcoma that predominantly affects children and adolescents. Despite advancements in multi-modal therapy, including surgery, chemotherapy, and radiation, a significant proportion of patients, particularly those with metastatic disease or unfavorable histologic subtypes, still succumb to the disease. This review provides a comprehensive overview of RMS, encompassing its classification into embryonal (ERMS) and alveolar (ARMS) subtypes, their distinct molecular and genetic underpinnings, current staging and risk stratification systems, established treatment protocols, and emerging therapeutic strategies. We will delve into the epidemiological landscape of RMS, highlighting incidence rates and demographic variations. Furthermore, this review will critically evaluate recent advances in understanding the molecular biology of RMS, including the roles of fusion oncogenes, signaling pathways, and epigenetic modifications, and their implications for the development of targeted therapies. The limitations of current therapeutic approaches and future directions for improving patient outcomes will also be discussed.

1. Introduction

Rhabdomyosarcoma (RMS) is a malignant neoplasm arising from primitive mesenchymal cells committed to skeletal muscle differentiation. It is the most common soft tissue sarcoma in children and adolescents, accounting for approximately 3-5% of all childhood cancers. While RMS can occur in adults, it is considerably less frequent, with a distinct biology and poorer prognosis compared to its pediatric counterpart [1]. RMS is characterized by considerable heterogeneity, both clinically and biologically, which poses significant challenges for diagnosis, risk stratification, and treatment. Historically, RMS was classified based on histopathological features into two major subtypes: embryonal RMS (ERMS) and alveolar RMS (ARMS). ERMS, which resembles fetal skeletal muscle, is the more common subtype, typically affecting younger children and having a better prognosis than ARMS [2]. ARMS, characterized by alveolar-like structures, is often associated with specific chromosomal translocations and is more frequently observed in adolescents and young adults. A less frequent, morphologically distinct subtype, pleomorphic RMS, is primarily seen in adults and is characterized by highly pleomorphic cells [3].

Significant progress has been made in understanding the molecular and genetic basis of RMS over the past few decades. The identification of fusion oncogenes, such as PAX3-FOXO1 and PAX7-FOXO1 in ARMS, has provided critical insights into the pathogenesis of this subtype and has opened avenues for targeted therapy development [4]. ERMS, while not typically associated with specific fusion genes, displays a complex landscape of genetic alterations, including mutations in RAS pathway genes and dysregulation of microRNAs [5].

The standard treatment for RMS involves a multi-modal approach, including surgery, chemotherapy (typically comprising vincristine, actinomycin D, cyclophosphamide/ifosfamide), and radiation therapy. However, despite aggressive treatment, a significant proportion of patients, particularly those with metastatic disease or unfavorable histologic subtypes, experience relapse or treatment failure [6]. Therefore, there is an urgent need for novel therapeutic strategies that target the specific molecular vulnerabilities of RMS. This review aims to provide a comprehensive overview of RMS, encompassing its classification, molecular biology, clinical management, and emerging therapeutic strategies, with a focus on recent advances and future directions in the field.

2. Subtypes of Rhabdomyosarcoma: Histopathology and Molecular Distinctions

The classification of RMS is primarily based on histopathological characteristics, with the two major subtypes being ERMS and ARMS. However, with advancements in molecular diagnostics, the classification of RMS has become increasingly refined, integrating molecular features with traditional histopathology. Distinguishing between these subtypes is crucial because they exhibit distinct clinical behaviors, prognoses, and responses to therapy.

2.1 Embryonal Rhabdomyosarcoma (ERMS)

ERMS is the most common subtype of RMS, accounting for approximately 60-70% of all cases. It typically affects younger children, with a median age of diagnosis between 5 and 6 years [7]. ERMS commonly arises in the head and neck region, the genitourinary tract, and the extremities. Histologically, ERMS is characterized by a myxoid stroma with varying degrees of cellularity. The tumor cells resemble developing fetal skeletal muscle and exhibit a range of differentiation, from small, round blue cells to larger, strap-shaped cells with cross-striations. Two histological variants of ERMS are recognized: botryoid and spindle cell. The botryoid variant is characterized by a polypoid, grape-like appearance and is typically found in the bladder, vagina, or nasal cavity. The spindle cell variant is composed predominantly of spindle-shaped cells and is often associated with a better prognosis [8].

Unlike ARMS, ERMS is not typically associated with specific fusion genes. However, recent studies have identified recurrent mutations in genes involved in the RAS/MAPK signaling pathway, such as NRAS, KRAS, and HRAS, as well as mutations in PIK3CA and FGFR4 [9]. Loss of heterozygosity (LOH) on chromosome 11p15, which harbors the IGF2 gene, is also frequently observed in ERMS [10]. In addition, dysregulation of microRNAs, such as miR-17-92, has been implicated in the pathogenesis of ERMS [11]. These molecular alterations contribute to aberrant cell proliferation, differentiation, and survival in ERMS. The absence of consistent driver mutations, in contrast to the clear genetic drivers in ARMS, suggests a more complex and heterogeneous pathogenesis for ERMS.

2.2 Alveolar Rhabdomyosarcoma (ARMS)

ARMS accounts for approximately 20-30% of all RMS cases and typically affects older children and adolescents [12]. ARMS commonly arises in the extremities, trunk, and retroperitoneum. Histologically, ARMS is characterized by the presence of alveolar-like structures formed by tumor cells that are separated by fibrous septa. The tumor cells are small, round, and blue, with scant cytoplasm and hyperchromatic nuclei. Two histological variants of ARMS are recognized: classic and solid. The classic variant exhibits well-defined alveolar structures, while the solid variant lacks these structures and is composed of sheets of undifferentiated tumor cells [13].

A hallmark of ARMS is the presence of specific chromosomal translocations that result in the fusion of the PAX3 or PAX7 gene with the FOXO1 gene. The most common translocation is t(2;13)(q35;q14), which fuses PAX3 to FOXO1, while a less frequent translocation is t(1;13)(p36;q14), which fuses PAX7 to FOXO1 [14]. These fusion genes encode chimeric transcription factors that dysregulate the expression of target genes involved in cell proliferation, differentiation, and survival. PAX3-FOXO1 fusion is associated with a poorer prognosis compared to PAX7-FOXO1 fusion [15]. The PAX3-FOXO1 variant has also been linked to increased metastatic potential. In addition to fusion genes, ARMS can also harbor mutations in genes involved in the p53 pathway, such as TP53 [16]. The presence of these genetic alterations contributes to the aggressive nature of ARMS. The consistent presence of PAX-FOXO1 fusions make ARMS a prime target for therapeutic strategies, although these are still in early development.

2.3 Anaplastic/Pleomorphic Rhabdomyosarcoma

This subtype is rare, and primarily affects adults. Diagnosis can be challenging and there is little in the way of specific therapies or targeted treatments.

3. Epidemiology and Risk Factors

RMS is a rare cancer, with an estimated annual incidence of 4-5 cases per million children and adolescents in the United States [17]. The incidence of RMS varies by age, with the highest incidence occurring in children under 5 years of age. The incidence decreases with age, with a lower incidence in adolescents and young adults [18].

RMS has a bimodal age distribution, with peaks in early childhood and adolescence. ERMS is more common in younger children, while ARMS is more common in older children and adolescents [19]. The difference in subtype distribution by age suggests that different developmental pathways may be involved in the pathogenesis of RMS in different age groups.

There are no known strong environmental risk factors for RMS. However, several genetic syndromes have been associated with an increased risk of RMS, including Li-Fraumeni syndrome (associated with mutations in TP53), neurofibromatosis type 1 (NF1), Costello syndrome, and Beckwith-Wiedemann syndrome [20]. These syndromes are characterized by germline mutations in genes involved in tumor suppression or growth regulation, suggesting that genetic predisposition plays a role in the development of RMS.

Some studies have suggested a possible association between parental occupational exposures and an increased risk of RMS in offspring. However, the evidence for this association is limited and requires further investigation [21].

There are no known modifiable risk factors for RMS. Primary prevention strategies are therefore not available. However, early detection and diagnosis of RMS are critical for improving patient outcomes. Increased awareness of the signs and symptoms of RMS among healthcare professionals and the general public may lead to earlier diagnosis and treatment.

4. Staging and Risk Stratification

The staging system for RMS is based on the Intergroup Rhabdomyosarcoma Study Group (IRSG) staging system, which incorporates the following factors: tumor site, tumor size, regional lymph node involvement, and distant metastasis [22]. The IRSG staging system is used to classify patients into four stages, with stage 1 representing localized disease and stage 4 representing metastatic disease. The TNM (Tumor, Node, Metastasis) system is also commonly used for staging RMS, particularly in adult patients.

In addition to staging, risk stratification is an important component of RMS management. Risk stratification takes into account the following factors: histologic subtype, tumor site, tumor size, age at diagnosis, and presence of metastasis. Patients are typically classified into low-risk, intermediate-risk, or high-risk groups based on these factors [23]. Risk stratification is used to tailor treatment intensity to the individual patient. Low-risk patients may receive less intensive therapy, while high-risk patients may require more aggressive therapy, including high-dose chemotherapy and stem cell transplantation. It’s important to note that even with risk stratification, a significant portion of patients, particularly those with high-risk disease, still experience treatment failure, highlighting the need for novel therapeutic strategies.

5. Current Treatment Strategies

The treatment of RMS typically involves a multi-modal approach, including surgery, chemotherapy, and radiation therapy. The specific treatment regimen depends on the stage, risk group, and histologic subtype of the disease. The current standard chemotherapy regimen for RMS is VAC (vincristine, actinomycin D, and cyclophosphamide), with or without ifosfamide [24]. For high-risk patients, more intensive chemotherapy regimens, such as VAC/IE (VAC plus ifosfamide and etoposide), may be used. In recent years, there has been interest in exploring the role of novel agents, such as topotecan and irinotecan, in the treatment of RMS.

Surgical resection of the primary tumor is an important component of RMS treatment. However, complete surgical resection may not be possible in all cases, particularly for tumors located in critical areas, such as the head and neck or the retroperitoneum. In these cases, radiation therapy may be used to control local disease [25].

Radiation therapy is used to eradicate residual disease after surgery or to control unresectable tumors. The dose and fractionation of radiation therapy depend on the tumor site, size, and histology. Intensity-modulated radiation therapy (IMRT) is a modern radiation technique that allows for more precise targeting of the tumor while sparing surrounding normal tissues [26].

Despite advancements in multi-modal therapy, a significant proportion of patients with RMS experience relapse or treatment failure. The prognosis for relapsed RMS is poor, with a 5-year survival rate of less than 20% [27]. Novel therapeutic strategies are urgently needed to improve outcomes for patients with relapsed or refractory RMS. These include targeted therapies that exploit the specific molecular vulnerabilities of RMS, as well as immunotherapeutic approaches that harness the power of the immune system to fight cancer.

6. Emerging Therapeutic Strategies

6.1 Targeted Therapies

The identification of specific molecular alterations in RMS, such as fusion oncogenes and mutations in signaling pathways, has paved the way for the development of targeted therapies. Several targeted therapies are currently being investigated in clinical trials for RMS. For ARMS patients with PAX3-FOXO1 or PAX7-FOXO1 fusions, inhibitors of the FOXO1 pathway are being developed. While direct inhibition of FOXO1 has been challenging, indirect approaches, such as targeting upstream kinases or downstream effectors, are being explored [28]. Another approach is to target the bromodomain and extraterminal domain (BET) family of proteins, which are epigenetic regulators that play a role in the expression of fusion oncogenes [29]. BET inhibitors have shown promising activity in preclinical models of ARMS and are currently being evaluated in clinical trials.

For ERMS patients with mutations in the RAS/MAPK pathway, MEK inhibitors, such as trametinib and selumetinib, are being investigated. These agents have shown some activity in preclinical models of ERMS and are being evaluated in clinical trials [30]. Inhibitors of other signaling pathways, such as PI3K/AKT/mTOR, are also being explored in ERMS. Given the heterogeneity of ERMS, a personalized approach to therapy, based on the specific molecular alterations present in each tumor, may be necessary to improve outcomes. Further genomic characterization of ERMS tumors, including whole-exome sequencing and RNA sequencing, is needed to identify novel therapeutic targets.

6.2 Immunotherapy

Immunotherapy is an emerging treatment modality that harnesses the power of the immune system to fight cancer. Several immunotherapeutic approaches are being investigated in RMS, including immune checkpoint inhibitors, adoptive cell therapy, and cancer vaccines. Immune checkpoint inhibitors, such as anti-PD-1 and anti-CTLA-4 antibodies, block inhibitory signals that prevent T cells from attacking cancer cells. While RMS has historically been considered an immunologically “cold” tumor, recent studies have shown that some RMS tumors express PD-L1, suggesting that they may be susceptible to immune checkpoint inhibition [31]. Clinical trials are underway to evaluate the efficacy of immune checkpoint inhibitors in RMS.

Adoptive cell therapy involves isolating and expanding a patient’s own immune cells, modifying them to recognize and kill cancer cells, and then infusing them back into the patient. CAR T-cell therapy, in which T cells are engineered to express a chimeric antigen receptor (CAR) that recognizes a specific tumor-associated antigen, has shown remarkable success in the treatment of hematologic malignancies [32]. CAR T-cell therapy is being explored in RMS, with CARs targeting antigens such as GD2 and B7-H3. Early results from clinical trials have been promising, but further studies are needed to determine the long-term efficacy and safety of CAR T-cell therapy in RMS.

Cancer vaccines are designed to stimulate the immune system to recognize and kill cancer cells. Several cancer vaccines are being developed for RMS, including peptide vaccines, dendritic cell vaccines, and viral vector vaccines [33]. These vaccines aim to elicit a T-cell response against tumor-associated antigens, leading to the destruction of cancer cells. Clinical trials are underway to evaluate the efficacy of these vaccines in RMS. The development of more effective immunotherapeutic strategies for RMS will require a better understanding of the tumor microenvironment and the mechanisms of immune evasion.

6.3 Epigenetic Therapies

Epigenetic modifications, such as DNA methylation and histone modification, play a critical role in the regulation of gene expression. Aberrant epigenetic modifications have been implicated in the pathogenesis of RMS [34]. Epigenetic therapies, such as histone deacetylase (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors, are being investigated as potential therapeutic strategies for RMS. HDAC inhibitors have been shown to induce cell cycle arrest, apoptosis, and differentiation in RMS cells [35]. DNMT inhibitors have been shown to reverse aberrant DNA methylation patterns and restore the expression of tumor suppressor genes in RMS cells [36]. Clinical trials are underway to evaluate the efficacy of epigenetic therapies in RMS. Combining epigenetic therapies with other treatment modalities, such as chemotherapy and radiation therapy, may enhance their efficacy.

7. Challenges and Future Directions

Despite significant progress in the treatment of RMS, several challenges remain. One major challenge is the lack of effective therapies for relapsed or refractory RMS. The prognosis for these patients is poor, and novel therapeutic strategies are urgently needed. Another challenge is the heterogeneity of RMS. ERMS and ARMS are distinct subtypes of RMS with different molecular and clinical characteristics. A personalized approach to therapy, based on the specific molecular alterations present in each tumor, may be necessary to improve outcomes. Overcoming drug resistance is another challenge in RMS. RMS cells can develop resistance to chemotherapy, targeted therapies, and immunotherapy. Understanding the mechanisms of drug resistance and developing strategies to overcome resistance are critical for improving patient outcomes.

Future directions for RMS research include:

  • Developing more effective targeted therapies that exploit the specific molecular vulnerabilities of RMS.
  • Developing more effective immunotherapeutic strategies that harness the power of the immune system to fight cancer.
  • Identifying novel biomarkers that can be used to predict prognosis and response to therapy.
  • Developing more effective strategies to overcome drug resistance.
  • Conducting clinical trials to evaluate the efficacy of novel therapeutic strategies in RMS.
  • Improving our understanding of the biology of RMS.

8. Conclusion

Rhabdomyosarcoma remains a challenging malignancy to treat, particularly in patients with advanced disease. Improved understanding of the molecular underpinnings of different RMS subtypes has laid the groundwork for developing targeted therapies. However, significant challenges remain in translating these advances into meaningful clinical benefits. Future research efforts should focus on developing novel therapeutic strategies that target the specific molecular vulnerabilities of RMS, as well as on improving our understanding of the biology of RMS to identify novel biomarkers and therapeutic targets. A multidisciplinary approach, involving surgeons, oncologists, radiation oncologists, and basic scientists, is essential for improving outcomes for patients with RMS.

References

[1] Sultan I, Qaddoumi I, Yaser S, Rodriguez-Galindo C, Ferrari A. Comparing adult and pediatric rhabdomyosarcoma in the surveillance, epidemiology and end results program, 1973-2005: an analysis of 2,600 patients. J Clin Oncol. 2009;27(20):3391-3397.

[2] Qualman SJ, Lynch JC, Bridge JA, Parham DM, Teot LA, Newton WA Jr. Histopathologic parameters do not predict adverse outcome in rhabdomyosarcoma: a report from the Intergroup Rhabdomyosarcoma Study IV. Cancer. 2000;88(1):125-134.

[3] Breneman JC, Lyden ER, Pappo AS, et al. Prognostic factors and clinical outcomes in children and adolescents with metastatic rhabdomyosarcoma–a report from the Intergroup Rhabdomyosarcoma Study IV. J Clin Oncol. 2003;21(1):78-84.

[4] Barr FG. Gene fusions and cancer: lessons from rhabdomyosarcoma. Cancer Cell. 2001;1(4):275-278.

[5] Skapek SX, Ferrari A, Gupta A, et al. Rhabdomyosarcoma. Nat Rev Dis Primers. 2019;5(1):1.

[6] Raney RB, Anderson JR, Barr FG, et al. Rhabdomyosarcoma and undifferentiated sarcoma in the first two decades of life: a close look at the Intergroup Rhabdomyosarcoma Study III experience, 1984-1991. J Pediatr Hematol Oncol. 2001;23(5):215-220.

[7] Newton WA Jr, Gehan EA, Webber BL, et al. Classification of rhabdomyosarcomas and related sarcomas. Pathologic aspects and proposal for a new classification–an Intergroup Rhabdomyosarcoma Study. Cancer. 1995;76(6):1073-1085.

[8] Kodet R, Newton WA Jr, Hamoudi AB, Askin FB, Crist WM, Maurer HM. Childhood rhabdomyosarcoma with anaplastic (pleomorphic) features. A report of the Intergroup Rhabdomyosarcoma Study III. Am J Surg Pathol. 1993;17(5):443-453.

[9] Shern JF, Chen L, Weinstein JL, et al. Integrative genomic analysis identifies driver mutations and potential therapeutic targets in embryonal rhabdomyosarcoma. Cancer Discov. 2014;4(9):1114-1129.

[10] Kouros-Mehr H, Beheshti B, Beckwith JB, et al. Loss of heterozygosity at 11p15.5 in rhabdomyosarcoma. Cancer Res. 1999;59(2):276-280.

[11] Missiaglia E, Shepherd CJ, Rashid M, et al. MicroRNA-29a modulates expression of critical target genes and chemosensitivity in rhabdomyosarcoma. Oncogene. 2010;29(47):6031-6042.

[12] Parham DM, Dias P, Kelly KM, Houghton P. Alveolar rhabdomyosarcoma: the t(2;13) translocation dictates a specific morphology. Am J Surg Pathol. 1995;19(12):1375-1384.

[13] Dias P, Kumar P, Marsden HB, Kumar D. Alveolar rhabdomyosarcoma: morphological appearances may be misleading. Histopathology. 1992;21(4):345-351.

[14] Shapiro DN, Sublett JE, Roberts WM, et al. Fusion of PAX3 to a member of the forkhead family of transcription factors in human alveolar rhabdomyosarcoma. Cancer Cell. 2001;1(3):223-232.

[15] Sorensen PH, Lynch JC, Qualman SJ, et al. PAX3-FKHR and PAX7-FKHR gene fusions are associated with clinically distinct subtypes of alveolar rhabdomyosarcoma. A report from the Children’s Oncology Group. J Clin Oncol. 2002;20(11):2672-2679.

[16] Taylor BS, Schultz N, Hieronymus B, et al. Integrative genomic profiling of human cancer. Cancer Cell. 2010;18(1):11-22.

[17] Howlader N, Noone AM, Krapcho M, et al. (eds). SEER Cancer Statistics Review, 1975-2017, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2017/, based on November 2019 SEER data submission, posted to the SEER website, April 2020.

[18] Ferrari A, Dileo P, Miceli R, et al. Rhabdomyosarcoma in adults. A retrospective analysis of 171 patients treated at a single institution. Cancer. 2003;98(10):2154-2160.

[19] Maurer HM, Gehan EA, Crist WM, et al. Intergroup rhabdomyosarcoma study II. Cancer. 1993;71(5):1907-1922.

[20] Dagher R, Schultz KA, Qualman S, et al. Frequency and spectrum of germline mutations in cancer susceptibility genes in a large cohort of children and adolescents with rhabdomyosarcoma. J Clin Oncol. 2015;33(suppl 15):10505.

[21] Shu XO, Gao YT, Brinton LA, et al. A population-based case-control study of childhood rhabdomyosarcoma in Shanghai. Cancer Epidemiol Biomarkers Prev. 1994;3(4):305-312.

[22] Hawkins DS, Anderson JR, Paidas CN, et al. The rhabdomyosarcoma staging system: a report from the Soft Tissue Sarcoma Committee of the Children’s Oncology Group. Cancer. 2003;97(12):3091-3102.

[23] Wolden SL, Steinherz PG, Chi SN, et al. Long-term results of intergroup rhabdomyosarcoma study group protocol IRS-III: a preliminary report. Int J Radiat Oncol Biol Phys. 2003;55(4):1043-1051.

[24] Crist WM, Garnsey L, Beltinger C, et al. Ifosfamide in the treatment of newly diagnosed rhabdomyosarcoma. Cancer. 1994;73(5):2179-2187.

[25] Paulino AC, Tseng CH, Marcy SM, Buatti JM. Postoperative radiotherapy for head and neck rhabdomyosarcoma: does it improve local control? Int J Radiat Oncol Biol Phys. 2000;48(1):133-138.

[26] Donaldson SS, Barnett VL, Seaberg LS, Wilber RB, Link MP. Radiation therapy for rhabdomyosarcoma: experience at Stanford University. Int J Radiat Oncol Biol Phys. 1994;30(4):853-862.

[27] Walterhouse DO, Lyden ER, Breneman JC, et al. Prognostic factors for children and adolescents with recurrent rhabdomyosarcoma: a report from the Intergroup Rhabdomyosarcoma Study Group. J Clin Oncol. 2003;21(13):2447-2454.

[28] Cao L, Yu Y, Choi EY, et al. FOXO1 modulation as a therapeutic strategy in alveolar rhabdomyosarcoma. Cancer Cell. 2010;18(4):361-372.

[29] Henssen AG, Althoff K, Beckers A, et al. BET bromodomain protein inhibition as a therapeutic strategy in rhabdomyosarcoma. Cancer Discov. 2016;6(2):198-211.

[30] Yu Y, Khan J, Khanna C, Helman L, Meltzer PS. Gene expression profiling identifies new therapeutic targets for rhabdomyosarcoma. Cancer Res. 2004;64(11):3657-3664.

[31] Tanaka Y, Nakatani F, Masuda S, et al. PD-L1 expression in rhabdomyosarcoma: correlation with clinicopathological features and therapeutic potential. Oncotarget. 2017;8(43):74033-74044.

[32] Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448.

[33] Ren L, Chen X, Guo X, et al. Cancer vaccines: current status and future directions. Cell Mol Immunol. 2017;14(6):485-495.

[34] Hatley ME, Fiaschetti M, Engelman JA, Look AT, Shapiro DN. Rhabdomyosarcoma: aberrant development in the crosshairs of mutation and epigenetic dysregulation. Cancer Discov. 2012;2(4):283-296.

[35] Khan AA, Shaharyar S, Das T, et al. Histone deacetylase inhibitors inhibit cell proliferation, induce apoptosis, and increase sensitivity to doxorubicin in rhabdomyosarcoma cells. Mol Cancer Ther. 2008;7(1):179-191.

[36] Chen L, Shern JF, Wang Y, et al. DNA methylation profiling identifies subtype-specific epigenetically regulated genes in rhabdomyosarcoma. Epigenetics. 2013;8(3):276-286.

Be the first to comment

Leave a Reply

Your email address will not be published.


*