Open access peer-reviewed chapter

Thoracic Follicular Dendritic Cell Sarcoma

Written By

Vinay V. and Priya Sharma

Submitted: 02 September 2023 Reviewed: 11 October 2023 Published: 21 March 2024

DOI: 10.5772/intechopen.1003838

From the Edited Volume

Soft Tissue Sarcoma and Leiomyoma - Diagnosis, Management, and New Perspectives

Gamal Abdul Hamid

Chapter metrics overview

19 Chapter Downloads

View Full Metrics

Abstract

Follicular dendritic cell sarcoma (FDCS), which makes up 0.4% of all soft tissue sarcomas, is an uncommon low-grade malignant tumor that develops from follicular dendritic cells in germinal centres of lymphoid tissue. The pathophysiology of FDCS is unknown. It can arise in both nodal and extra-nodal areas harboring FDCs and is commonly diagnosed in middle-aged individual who are in their fifth decade of life. The extranodal lymph node locations include the liver, spleen, and GI tract, where FDCS occurs most frequently (79.4%). It is quite uncommon to develop a pulmonary follicular dendritic cell sarcoma. At the microscopic scale, the FDCS displays a wide range of architectural patterns, including fascicular, storiform, whorled, and diffuse patterns. It is frequently not considered a differential diagnosis for a spindle cell neoplasm because of its rarity, misdiagnosis, and diagnostic difficulties. There is no effective treatment for this uncommon tumor, and the value of adjuvant therapy is still debatable. The median survival period following surgery for thoracic FDCS is approximately 4.41 years, with a 5-year recurrence-free survival rate of 47%. Patients diagnosed with localized disease exhibit a 5-year overall survival rate of 55%, whereas those with metastatic disease have a lower rate of 38%.

Keywords

  • follicular dendritic cell
  • follicular dendritic cell sarcoma
  • extranodal spread
  • pulmonary involvement
  • immunohistochemistry
  • outcome

1. Introduction

Tumors related to macrophage/monocyte and dendritic cell morphologies are collectively referred to as histiocytic neoplasms. They include both benign (e.g., Erdheim-Chester illness, Langerhans cell histiocytosis/LCH) and neoplastic (interdigitating dendritic cell sarcoma, histiocytic sarcomas, indeterminate cell sarcomas, FDCS, Langerhans cell sarcoma) conditions [1, 2]. FDCS, a unique and distinctive low-grade sarcoma showing spindle-shaped cells grouped in a whorled pattern, usually manifests as a slowly growing asymptomatic mass. Numerous examples have been reported to date since Monda and Rosai first identified primary malignant neoplasms (PMN) that had traits of follicular dendritic cells (FDC) in 1986 [3]. Perez-Ordonez and Rosai’s 1998 publication was the first in-depth analysis of FDC tumors [4]. Among the lymph node groups, the most affected ones include cervical, axillary, mediastinal, and intra-abdominal sites, whereas the lung, spleen, and liver are the most affected extranodal sites [5, 6]. Although once thought to be a low-grade malignancy, FDCS shows a significant malignant potential. At least, 40% of cases have local recurrence and 25% of cases have metastatic spread. Because of the short follow-up durations in numerous researches in the literature, even these numbers are an understatement [7]. However, due to their rarity compared to spindle cell sarcoma or carcinoma, spindle cell tumors do not necessarily raise concerns for FDCS when they appear as extranodal neoplasms. Moreover, the extensive variety of morphological features in FDCS poses a greater challenge in its diagnosis. It is worth noting that the reported misdiagnosis rate for FDCS in extranodal sites can be as high as 58%, affirming the idea that cases of FDCS have likely remained undetected [8].

Advertisement

2. Epidemiology

Follicular dendritic cell sarcoma (FDCS) is an exceptionally uncommon tumor, and its occurrence within the thoracic region is even rarer. Nevertheless, the precise incidence remains undetermined [9]. Literature consists of a few cases, less than 50 (Table 1). FDCS comprise <0.4% of soft tissue sarcomas. In a meta-analysis of 462 FDCS cases, the median age (range = 8–90 years) of occurrence was fifth decade of life with no gender predilection [6]. FDCS’s unusual and distinctive inflammatory pseudotumor-like variant tends to affect women more than other FDCS variants [25]. The fact that several cases have been documented from East Asia raises the idea that FDCS may be more common among some ethnic groups or regions [6].

Sl noAuthorsType of ReportSite of FDCsAge (years)GenderClinical PresentationTreatment givenOutcome
1Fassina et al. [10]Case reportMediastinal mass48MaleChest painNANA
2Shah et al. [11]Case reportLeft lung mass33MaleDry coughNANA
3Leipsic et al. [12]Case reportMiddle Mediastinum43MaleChest PainNANA
4Dening et al. [13]Case reportRight lower lobe nodule64FemaleCough and breathlessnessNANA
5Bushan et al. [14]Case reportAnterosuperior mediastinal mass34MaleFever and malaiseSurgery followed by chemoradiationNo recurrence at 6 months Follow up.
6Lee et al. [15]Case reportPosterior mediastinum63MaleDry cough, breathlessnessSurgeryNA
7Butler et al. [16]Case reportRight main bronchus lesion19FemaleCough, fever, dyspnoeaSurgery with lymphadenectomyNo recurrence at 2 years Follow up
8Wang et al. [17]Case reportLung parenchyma76FemaleDyspnoeaSurgeryExpired
9Miyoshi et al. [18]Case reportPosterior Mediastinum16FemaleIncidental findingSurgeryNo recurrence at 2 years Follow up
10Ulises et al. [19]Case reportPosterior mediastinum59MaleOral ulcers/ pemphigusComplete excision with 1 cycle of Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone.No recurrence at 2 months Follow up
11Xu et al. [20]Case reportRight lateral chest wall44MaleChest wall painTumor resection with lymphadenectomy of level I axillary nodes.No recurrence at 18 months Follow up.
12Ilonen et al. [21]Case reportPosterior Mediastinum57MaleIncidental findingChemotherapy (Doxorubicin, Ifosfamide, Mesna), Surgery, RadiotherapyNo recurrence at 2 years Follow up
13He et al. [22]Case reportRight lower lobe mass64FemaleIncidental findingSurgeryNo recurrence at 6 months follow-up
14Li et al. [23]Case reportRight hila of Lung29MaleCough, expectorationSurgery
Tacrolimus
Cyclosporine
Symptomatic improvement at 1 month
15Vinay et al. [24]Case reportLeft upper lobe mass34MaleChest PainSurgeryNA

Table 1.

Summary of extranodal follicular dendritic cell sarcomas reported in the thorax (according to year of publication).

Advertisement

3. Pathogenesis

3.1 Origin and function of FDCs

Follicular dendritic cells, also known as dendritic reticulum cells, is a vital component in B-cell follicles. Their primary function is to gather, hold, and deliver antigens to nearby B cells. They are present in lymphoid follicles both primary and secondary [26], and their origin is mesenchymal [27]. Follicular dendritic cells were initially described as ‘embryonal non-phagocytic reticulum cell’ in Maximow’s 1927 illustration of B follicles in human lymph nodes [28]. Under electron microscopy, FDCs exhibited interdigitations and extension of their plasma membranes, closely interacting with other germinal centre cells. This shows their potential significance in the germinal centre response, which has now been proven by a wide range of experiments [29]. Within the immune system, histiocytic and dendritic cells play a vital role by aiding in phagocytosis, processing antigens, and presenting these antigens to B and T cells. FDCs are nonlymphoid, nonphagocytic immune system accessory cells that are crucial for antigen presentation and controlling germinal centre reactivity [7, 30, 31, 32]. They create a dense network inside lymphoid follicles.

A key function of the FDC is luring B and T cells to B follicles through the secretion of CXCL13 (C-X-C chemokine motif ligand 13) and the interaction with B cells through integrins and their appropriate receptors. Immunocomplexes that are ‘exposed’ to germinal centre B-cells border the FDC cell membrane. By releasing numerous B-cell growth factors, mainly B activating factor (BAFF), FDC helps B cells survive and mature. During the germinal centre reaction, FDC plays a role in the apoptosis and phagocytosis of B cells with low-affinity B-cell receptors, a process mediated by Mfge8 produced by FDC [33]. FDCs can impact the adaptive immune response by recognizing environmental innate stimuli, such as microbial lipopolysaccharides, through Toll-like receptor 4 (TLR4). This recognition triggers TLR signaling, which in turn, promotes the synthesis of proteins such as CXCL13, TGF-1, and BAFF that contribute to B cell survival, recruitment, and class-switch recombination (Figure 1) [34].

Figure 1.

By secreting CXCL13 (C-X-C motif chemokine ligand 13) and connecting with B cells via cognate receptors and integrins, FDC plays a critical role in attracting T and B cells to B follicles. Immunocomplexes that are ‘exposed’ to germinal Centre B cells border the FDC cell membrane. FDC promotes B-cell survival and maturation by releasing a number of B-cell growth factors, including BAFF (B-activating factor). The germinal Centre response, which is mediated by Mfge8 generated by FDC, results in the apoptosis and phagocytization of B-cells with low-affinity B-cell receptors. FDC can influence the acquired response by detecting, through TLR-4 (toll-like receptor 4), ambient innate stimuli (such as microbial lipopolysaccharides), which leads to TLR signaling that stimulates the synthesis of proteins (CXCL13, TGF-1, and BAFF) and promote B cell survival, recruitment, and class-switch recombination.

Inflammatory pseudotumor-like fibroblastic/follicular dendritic cell neoplasm is an uncommon neoplasm that arises from dendritic cells. Within the World Health Organization (WHO) classification of hematological and lymphoid organ cancers, sarcoma is recognized as a unique entity. There are unique clinical characteristics of this tumor, including a predilection for young to middle-aged women, an occurrence in the liver or spleen, and the presence of Epstein-Barr virus (EBV) positive neoplastic cells [35]. This could be explained by EBV entering these cells due to FDCs expressing CD21, which functions as a receptor for the virus. Additionally, FDCs have been linked to myasthenia gravis, paraneoplastic pemphigus, and Castleman disease. Epidermal growth factor receptor is expressed in FDCS and non-neoplastic FDCs of Castleman disease, which may encourage the persistence of FDCs and allow for mutations that could lead to FDCS [6, 36].

3.2 Morphology of FDC

Long dendritiform processes and an abundance of poorly defined eosinophilic cytoplasm are characteristics of FDC. The nuclear membrane is clearly defined, the chromatin is finely scattered, and the nucleus is big, ovoid to spherical in shape with a tiny eosinophilic nucleolus. Typically, bi- or multinucleated FDC exhibit a ‘kissing’ pattern of nuclear molding and overlap [37]; these characteristics make it easier to identify FDC in cytological smears. On electron microscopy, the FDC’s cytoplasmic extensions are coated in an amorphous substance that resembles immunocomplexes and form a complex network connected by desmosomes [38]. Various FDC markers, including CD21 (C3d receptor), CD23, CD35 (C3b receptor), R4/23, DRC-1 (R4/25), CNA.42, Ki-M4p, Ki-M4, and Ki-FDC1p, effectively highlight the dendritic characteristics of FDCs on both frozen and paraffin sections. Compared to other malignant histiocytic tumors, FDCS exhibits a unique immunophenotype.

The mechanism of acquiring neoplastic potential by these FDCs is still not well studied, and more research in this is needed. As such, no universal driver translocation or mutation has been identified by comprehensive genomic investigation. A wide range of chromosomal instability, dysregulation of cell cycle progression, activation of nuclear factor kappa beta (NF-kB), mitogen-activated protein kinase (MAPK), and broad immunological evasion are also related to FDCS [39, 40]. BRAF V600E mutations, present in many histiocytic diseases and are affecting 20% of people with FDCS, might be more prevalent in the inflammatory form [41]. Epstein-Barr virus is regularly linked to the inflammatory version [42], while human herpesvirus 8 (HHV-8) is not known to be involved [43].

Association with mutations in TP53, PTEN, and NF-kB pathways has been noted in the literature [40, 44]. The expression of PD-L1 and PD-L2 and copy number increases at 9p24 implying immune surveillance escape may be involved in tumor development [40]. EZH2 was found to be overexpressed in FDCS tumors in 67% of cases, whereas p-ERK1/2 (phospho-extracellular signal-related protein kinases 1 and 2) were robustly expressed in 80% of cases [45]. Notably, nearly all FDCS cases have elevated levels of the epidermal growth factor receptor (EGFR), a hallmark of epithelial malignancies. The survival and expansion of FDCS cells may depend on EGFR signaling, which is triggered by ligands in the microenvironment [46].

Advertisement

4. Clinical presentation

FDCS does not have a gender predilection. It primarily occurs in adults, typically appearing at a median age of 49 years ranging from 8 to 90 years. There have been very few reported instances of this condition in pediatric patients [6]. FDCS has the potential to affect various anatomical regions. A higher frequency of involvement is seen in extranodal sites (79.4%), particularly in the liver, spleen, gastrointestinal tract, tonsil/adenoid, mediastinum, and lungs compared to nodal involvement (15%) [38]. FDCS frequently appears as a gradually developing mass that can either be symptom-free or accompanied by discomfort. The variant resembling an inflammatory pseudotumor (IPT) displays distinctive clinical attributes. Females are more likely to be impacted, as is the existence of EBV infection inside the tumor cells with the involvement of the liver or spleen and frequent systemic symptoms such as fever, malaise, and weight loss [22, 42, 47, 48, 49, 50, 51]. FDCS is found to be associated with many conditions such as paraneoplastic pemphigus with or without myasthenia gravis [52, 53, 54, 55, 56], lymphoproliferative conditions such as Castleman Disease-Hyaline vascular type (HV-CD) [57] and Sjogren’s syndrome [58] manifesting predominantly in extranodal sites. FDCS is also observed in conjunction with neoplasms such as lymphomas, leukemias, and malignant neoplasms of epithelial or melanocytic origin [38].

Advertisement

5. Differential diagnosis

Because of histopathological diversity and the potential to manifest at various locations, FDCS can be mistaken for various types of tumors and even inflammatory processes. The reported rates of misidentification range from 30 to 58% especially when it arises from extranodal sites [59, 60]. It might be mistaken with interdigitating dendritic cell sarcoma (IDCS), Langerhans cell histiocytosis (LCH), Kaposi sarcoma, spindle cell thymoma, metastatic melanoma, metastatic carcinoma, inflammatory myofibroblastic tumor, pleomorphic sarcoma, solitary fibrous tumor, and gastrointestinal stromal tumor (GIST).

IDCSs are uncommon tumors believed to originate from interdigitating dendritic cells within the nodal paracortex. They are typically found in lymph nodes or the spleen and are associated with an aggressive nature. Histologically indistinguishable from FDCS, IDCS generally displays a higher level of atypical features and lacks the bundled structures and spiral growth pattern commonly observed in most cases of FDCS that necessitates immunohistochemical studies for a definite diagnosis. IDCS is positive for S100, negative or focally positive for clusterin. LCH comprises oval-shaped Langerhans cells, characterized by their nuclei exhibiting grooves, folds, or indentations. These nuclei have fine chromatin, inconspicuous nucleoli, and thin nuclear membranes. The cells possess moderately abundant cytoplasm that appears slightly eosinophilic. Langerhans cells are typically positive for CD1a and langerin. In the case of Kaposi’s sarcoma, the condition is characterized by atypical spindle-shaped cells that form slit-like vascular spaces containing red blood cells. Immunohistochemistry (IHC) reveals the presence of positive cells for HHV8.

Spindle cell thymoma is commonly located in the anterior mediastinum. It is characterized by the presence of spindle-shaped cells exhibiting a growth pattern that ranges from fascicular to whorls. These spindle cells test positive for keratin (AE1/AE3). In metastatic carcinoma, the epithelioid cells are found in sheets, clusters, or as single cell with pronounced nuclear atypia. These cells also show positivity for keratin (AE1/AE3). Metastatic melanoma exhibits varying cytology, which may include epithelioid or spindle-shaped cells displaying nuclear atypia and prominent nucleoli. Additionally, the cytoplasm of these cells may contain brown pigment (melanin). IHC in metastatic melanoma shows positivity for S100 and MelanA. Inflammatory myofibroblastic tumor (IMT) comprises spindled myofibroblasts and fibroblasts with bland nuclei. IMT cells are positive for anaplastic lymphoma kinase-1 (ALK1). Pleomorphic sarcoma is composed of pleomorphic cells that exhibit a diverse range of cytological characteristics, which can encompass spindled, plasmacytoid, epithelioid, or multinucleated forms. Notably, this type of sarcoma does not demonstrate positivity for clusterin, CXCL13, CD21, CD23, and CD35. Positivity for DOG1 and STAT6 are seen in solitary fibrous tumors that are usually negative in FDCS [61, 62]. In FDCS, tumor markers CD34 and CD117, typically used to identify gastrointestinal stromal tumors (GIST) tend to show negative results [38, 62].

Advertisement

6. Diagnosis

6.1 Histopathology

Macroscopically, FDCS appears as a gray-yellow firm mass with a well-circumscribed surface. Necrosis can be visible in few areas. This tumor has occasionally been confused with lymphoma, mesenchymal tumors such as gastrointestinal stromal tumors or solitary fibrous tumors, or poorly differentiated carcinoma, depending on the site of its presentation. The diagnosis of FDCS is consistently confirmed through immunohistochemistry, and it often requires the use of multiple FDC markers because the loss of antigens is a common occurrence [37]. Limited studies have described the EBV-associated FDCS. Based on the number and growth pattern of EBV, the degree of lymphoplasmacytic infiltration, and characteristics of the intratumoral blood vessels, the tumors were classified into the following groups: classic type (53.8%), lymphoma-like subtype (38.5%), and hemangioma-like subtype (7.7%) [63].

Most FDCSs are considered low-grade sarcomas. Histological sections reveal a spindle cell proliferation exhibiting diverse architectural arrangements with the most common being storiform or whorled (reminiscent of meningiomas) bundles (Figure 2A and B), fascicles (Figure 2C and D), trabecular formations, or widespread sheets. Multiple growth patterns are frequently observable within the same tumor. Abundant lymphocytes are often dispersed throughout the tumor, occupying the spaces between tumor cells and the areas around blood vessels. Some cases exhibit a predominance of B-cell lymphocytes, whereas others feature a preponderance of T-cell lymphocytes. Occasional multinucleated cells, albeit in small quantities, may be present, resembling Warthin-Finkeldey giant cells. These cells typically exhibit indistinct boundaries, resulting in a syncytial appearance. Their cytoplasm is moderately abundant, with a pale eosinophilic hue and the possibility of having a fibrillary texture. Tumor cells typically feature elongated or ovoid nuclei characterized by a thin nuclear membrane, vesicular or granular chromatin, and small nucleoli (Figure 2E and F). Nuclear pseudoinclusions are sporadically observed, and their frequency may increase following radiation therapy. The mitotic rate is generally low, ranging from 0 to 10 mitoses per 10 high-power fields, and necrosis is infrequently detected. The proliferation index, typically represented by Ki-67, typically falls within the range of 1–25% (Figure 3D). This index can be elevated in cases demonstrating clear atypia [63]. As lymphocytes, including both B and T cells as well as plasma cells, are often commingled with tumor cells in IPT-like variants, recognizing tumor cells can be challenging [38]. The histological features of this disorder may include prominent nuclear atypia, including irregular nuclear membranes and hyperchromatic nuclei, and high mitotic counts ranging from 11 to 35 mitoses per 10 high-power field. Additionally, extensive necrosis may be present. These high-grade features are typically linked to lesions located in deeper tissue layers and are indicative of a higher likelihood of recurrence or metastasis [62].

Figure 2.

Histological sections display a cellular arrangement featuring elongated to spindle-shaped cells with eosinophilic nuclei, interspersed among lymphocytes, eosinophils, and plasma cells (Image 2A & B). The spindle cells are arranged in irregular bundles, and the cytoplasm appears pale or eosinophilic, with nuclei exhibiting moderate cytoplasmic cells organized in fascicles [40× (Image 2C & 2D), 100× (Image 2E & 2F)].

Figure 3.

FDCS cells showing IHC positivity to CD 21 (Image 3A & 3B), podoplanin (Image 3C), and Ki-67 (Image 3D). Negative for CD 34 (Image 3E) and CD 45 (Image 3F).

6.2 Immunophenotype

Diagnosing FDCS typically necessitates support from immunohistochemistry, and the utilization of multiple FDC markers is often imperative due to the frequent occurrence of antigen loss. Tumor cells exhibit varying degrees of positivity for CD21 (as shown in Figure 3A and B) and CD23, as well as CD35. In contrast, clusterin, podoplanin (Figure 3C), and CXCL13 consistently display higher expression levels, demonstrating high specificity in distinguishing them from tumors that may resemble FDCS. As a marker for FDCS, podoplanin (D2-40) exhibits high sensitivity and strong membrane staining [62]. EGFR expression is frequently detected in FDCS, although its specificity is relatively limited. Reports indicate that FDCS may exhibit positive staining for epithelial membrane antigen, CD68, S-100 protein, and, albeit rarely, cytokeratin and CD20 [37]. On occasion, reports have indicated reactivity in FDCS for S100, smooth muscle actin, CD68, and EMA. FDCS usually yields negative results in tests for CD1a, langerin, CD34 (Figure 3E), CD45 (Figure 3F), lysozyme, CD163, myeloperoxidase, CD3, CD79a, cytokeratin, MART1, and HMB45 [38]. In recent advancements, FDC-secreted protein (FDCSP), SSTR2A, and serglycin (SRGN) have surfaced as valuable markers for the purpose of identification [64].

6.3 Electron microscopy

In the realm of electron microscopy, characteristic diagnostic elements in FDCS consists of elongated and intricately interlocking cytoplasmic extensions, occasionally forming a maze-like configuration, as well as desmosome-like connections, whereas cytoplasm typically exhibits limited organelles, notably lacking Birbeck granules and lysosomes [62].

Advertisement

7. Genetics and molecular findings

Vermi and colleagues in their FDCS series examined the prevalent genetic variations in EGFR (specifically substitutions in exons 18 and 21, as well as deletion of exon 19), KRAS (exon 1), NRAS (exons 1 and 2), and PI3KCA (exons 9 and 20). Notably, no mutations in these genes were detected in any of the cases studied. In an FDCS cell line, the EGFR pathway was activated, showing that local cognate ligands are involved, rather than oncogenic mutations [46]. Genomic alterations found in FDCS includes single nucleotide variations, loss/deletions, amplification of genes, copy number gains, and fusion rearrangements [39]. Five unique translocations leading to fusion proteins were identified in four cases: MAP3K1-GCOM1, NTRK1-PDIA3, TYK2-ATPAF2, HDGRFP3-SHC4, and BPTF-WDR72 [38]. FDCS is distinguished by genetic modifications affecting genes within the NF-κB regulatory pathway, encompassing copy number loss or missense mutations in TRAF3, SOCS3, TNFAIP3, and NFKBIA [65]. Additionally, mutated cell-cycle regulators leading to recurrent homozygous deletions of tumor suppressor genes CDKN2A, RB1, and CYLD and BIRC3 has also been observed [39]. Go et al. employed direct Sanger sequencing and peptide nucleic acid clamp quantitative polymerase chain reaction to ascertain the presence of mutations and detected BRAFV600E mutation in 18.5% (5 out of 27) of FDCS cases [41]. PDGFRB N666S mutation occurring in HV-CD was found in FDCS patients with history of Castleman disease. While PD-L1 expression is frequently observed in FDCS, a substantial number of cases within a comprehensive dataset exhibited a low mutational burden, measuring less than six mutations per mega base [65]. This discovery raises doubts about the potential effectiveness of immunotherapy as a treatment approach for this neoplasm [38].

Advertisement

8. Treatment

Various approaches have been addressed such as surgery, chemotherapy, tyrosine kinase inhibitors, and radiotherapy because FDCS lacks specific treatment guidelines. Surgery remains the mainstay of treatment in FDCS patients with localized disease. Systemic therapies used in the metastatic setting include doxorubicin, ifosfamide, or gemcitabine-based regimens, which provided prolonged stable disease [66]. Numerous regimens incorporating gemcitabine have been experimented. The rate of complete responders was 42% for those undergoing gemcitabine combined with taxotere therapy, as reported by Jain et al. [67]. In contrast, gemcitabine and taxane therapy achieved an impressive 80% complete response rate, with a notable median response duration of 13.4 months in docetaxel combination therapy, as highlighted by Conry et al. [68]. Various systemic treatment protocols have been explored, encompassing doxorubicin and ifosfamide; vincristine, doxorubicin, and cyclophosphamide (VAC); ifosfamide and etoposide with or without carboplatin (ICE/IE); cyclophosphamide, doxorubicin, vincristine, and prednisone; as well as gemcitabine, either alone or in combination with docetaxel, carboplatin, and irinotecan. A number of patients received tyrosine kinase inhibitors, including sorafenib, sirolimus, ridaforolimus, sunitinib, brivanib, imatinib, cabozantinib, and pazopanib, which resulted in extended positive outcomes, both when administered as a first-line treatment or in subsequent therapy. Research indicates that the maturation and activation of dendritic cells were impeded by vascular endothelial growth factor (VEGF). However, this inhibitory effect was counteracted using VEGF inhibitors, including bevacizumab, sorafenib, and sunitinib [69]. Enhanced comprehension of the molecular characteristics and causative factors behind this tumor could pave the way for innovative therapeutic approaches.

Advertisement

9. Outcome

The pooled analysis of published case reports indicated that patients who were administered adjuvant radiotherapy (RT; 51 in total) did not experience improved overall survival (OS) when compared to those who underwent surgery alone (78 in total) [6]. In FDCS, pleomorphism or prominent nuclear atypia, a size of 6 cm or more, five or more mitoses per 10 high-power fields, and coagulative necrosis on microscopy are associated with an unfavorable prognosis [70]. In the study conducted by Saygin et al., they documented 2-year survival rates of 82% for early-stage cases, 80% for locally infiltrated tumors, and 42% for cases with distant metastasis [6]. In the study conducted by Gounder M et al., it was observed that the median overall survival (OS) exhibited significance difference among patients with localized disease (9.8 years), in contrast to individuals with metastatic or recurrent disease (2.69 years). Median survival in thoracic FDCS was 4.41 years, and the 5-year overall survival was 0.47. Local recurrence or metastasis has been observed in one-third of the diagnosed FDCS cases. The 5-year survival rate for individuals diagnosed with localized disease is 55%, while it drops to 38% for those with metastatic disease [66].

Advertisement

10. Conclusions

The thoracic extranodal FDCS will probably continue to be an underappreciated tumor. Thoracic FDCs contains several characteristic histological features that should serve as an indication to include the tumor in the differential diagnosis and help avoid misdiagnosis. Recognition needs a high index of suspicion, although thoracic FDC sarcoma has a high number of pathological features. The detection of FDC sarcomas should be aided by a greater understanding of their morphologic spectrum and the proper immunostains (D2-40 and clusterin) for differentiation as it is extremely similar to various cancers with histiocytic or dendritic origin. Given this neoplasm’s substantial potential for recurrence and metastatic spread, accurate characterization is essential. For management aspect, at present, there are no clear indications or guidelines for surgical treatment alone, adjuvant chemotherapy, or radiotherapy in the case of thoracic FDCS. It is challenging for any institute to amass sufficient knowledge on the proper therapy of these patients because of the low prevalence of this condition. Future efforts at their identification, a more thorough characterization of their clinical behavior; potential causal linkages should benefit from a greater understanding of their morphologic spectrum and the appropriate application of FDC markers for any unusual-appearing tumors.

Acknowledgments

Dr. Maria Paul (Senior Resident, Department of Anaesthesiology and Critical Care, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi) sketched the image showing how follicular dendritic cells function (Figure 1), and I would like to extend my deepest gratitude to her.

Conflict of interest

None of the authors have any conflict of interest.

Abbreviation

FDC

follicular dendritic cell

FDCS

follicular dendritic cell sarcoma

LCH

langerhans cell histiocytosis

PMN

primary malignant neoplasm

EBV

Epstein-Barr virus

HHV-8

human herpesvirus 8

CXCL13

C-X-C motif chemokine ligand 13

BAFF

B activating factor

TLR4

toll-like receptor 4

NF-kB

nuclear factor kappa beta

MAPK

mitogen-activated protein kinase

EGFR

epidermal growth factor receptor

IPT

inflammatory pseudotumor

HV-CD

hyaline-vascular subtype of Castleman disease

IDCS

Interdigitating Dendritic Cell Sarcomas

GIST

gastrointestinal stromal tumor

IMT

inflammatory myofibroblastic tumor

ALK1

anaplastic lymphoma kinase-1

FDCSP

FDC secreted protein

SRGN

serglycin

VAC

vincristine, doxorubicin, and cyclophosphamide

ICE/IE

ifosfamide and etoposide+/−carboplatin

VEGF

vascular endothelial growth factor

OS

overall survival

References

  1. 1. Swerdlow SH, Campo E, Pileri SA, Lee Harris N, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127(20):2375-2390
  2. 2. Emile JF, Abla O, Fraitag S, Horne A, Haroche J, Donadieu J, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 2016;127(22):2672
  3. 3. Monda L, Warnke R, Rosai J. A primary lymph node malignancy with features suggestive of dendritic reticulum cell differentiation. A report of 4 cases. The American Journal of Pathology. 1986;122(3):562
  4. 4. Perez-Ordonez B, Rosai J. Follicular dendritic cell tumor: Review of the entity. Seminars in Diagnostic Pathology. 1998;15
  5. 5. Pileri SA, Grogan TM, Harris NL, Banks P, Campo E, Chan JKC, et al. Tumours of histiocytes and accessory dendritic cells: An immunohistochemical approach to classification from the International Lymphoma Study Group based on 61 cases. Histopathology. 2002;41
  6. 6. Saygin C, Uzunaslan D, Ozguroglu M, Senocak M, Tuzuner N. Dendritic cell sarcoma: A pooled analysis including 462 cases with presentation of our case series. Critical Reviews in Oncology/Hematology. 2013;88(2):253-271
  7. 7. Chan JKC. Proliferative lesions of follicular dendritic (Cells: An overview, including a detailed account of follicular dendritic cell sarcoma): A neoplasm with many faces and uncommon etiologic associations. Advances in Anatomic Pathology. 1997;4(6)
  8. 8. Duan GJ, Wu YL, Sun H, Lang L, Chen ZW, Yan XC. Primary follicular dendritic cell sarcoma of the urinary bladder: The first case report and potential diagnostic pitfalls. Diagnostic Pathology. 2017;12(1)
  9. 9. Perkins SM, Shinohara ET. Interdigitating and follicular dendritic cell sarcomas: A SEER analysis. American Journal of Clinical Oncology: Cancer Clinical Trials. 2013;36
  10. 10. Fassina A, Marino F, Poletti A, Rea F, Pennelli N, Ninfo V. Follicular dendritic cell tumor of the mediastinum. Annals of Diagnostic Pathology. 2001;5(6):361-367
  11. 11. Shah RN, Ozden O, Yeldandi A, Peterson L, Rao S, Laskin WB. Follicular dendritic cell tumor presenting in the lung: A case report. Human Pathology. 2001;32(7):745-749
  12. 12. Leipsic JA, McAdams HP, Sporn TA. Follicular dendritic cell sarcoma of the mediastinum. American Journal of Roentgenology-New Series. 2007;188(6):1694
  13. 13. Denning KL, Olson PR, Maley Richard HJ, Flati VR, Myers JL, Silverman JF. Primary pulmonary follicular dendritic cell neoplasm: A case report and review of the literature. Archives of Pathology & Laboratory Medicine. 2009;133(4):643-647
  14. 14. Bushan K. Follicular dendritic cell sarcoma mediastinum-a case report. Indian Journal of Surgical Oncology. 2014;5(4):290-292
  15. 15. Lee BE, Korst RJ, Taskin M. Right pneumonectomy for resection of a posterior mediastinal follicular dendritic cell sarcoma arising from Castleman’s disease. The Annals of Thoracic Surgery. 2014;97(4):e101-e103
  16. 16. Butler J, Yessayan L, Simoff M. Endobronchial follicular dendritic cell sarcoma. Chest. 2015;148(4):539A
  17. 17. Wang R, Han W, Qi L, Shan L, Wang Z, Wang L. Extranodal follicular dendritic cell sarcoma: A clinicopathological report of four cases and a literature review. Oncology Letters. 2015;9(1):391-398
  18. 18. Miyoshi R, Sonobe M, Miyamoto E, Date H. Completely resected follicular dendritic cell sarcoma of the posterior mediastinum: Report of a case. Surgery Case Report. 2016;2(1)
  19. 19. Sifuentes FUP, Teran SM, Contreras E, Carrera-Ceron R, Bolanos F. Follicular dendritic cell sarcoma in the posterior mediastinum. Chest. 2018;154(4):36A
  20. 20. Xu H, Chen B, Jiang C, Yang Z, Wang K. Follicular dendritic cell sarcoma in the right chest wall: A case report. Medicine. 2020;99(35)
  21. 21. Ilonen IK, Meltzer AJ, Ellozy S, Bains MS, Huang J. Follicular dendritic cell sarcoma of the chest. The Annals of Thoracic Surgery. 2022;113(4):e263-e266
  22. 22. He H, Xue Q , Tan F, Yang L, Wang X, Gao Y, et al. A rare case of primary pulmonary inflammatory pseudotumor-like follicular dendritic cell sarcoma successfully treated by lobectomy. Annals of Translational Medicine. 2021;9(1)
  23. 23. Li SH, Dong B, Wu CL, Li ZH, Qi Y. Primary lung follicular dendritic cell sarcoma with rare paraneoplastic pemphigus: A case report. 2022. DOI: 10.21203/rs.3.rs-1780663/v1. Available from: https://www.researchsquare.com/article/rs-1780663/latest
  24. 24. Vinay V, Bansal S, Purwar P, Dewan RK, Verma P, Mullick S, et al. Thoracic follicular dendritic cell sarcoma – An outlandish presentation of a rare tumour with review of literature. Monaldi Archives for Chest Disease. 2023;93(2)
  25. 25. Chan JKC. WHO Classification of Tumors of Haematopoietic and Lymphoid Tissues. Vol. 2. IARC Publications; 2008. p. 285
  26. 26. Muppidi JR, Lu E, Cyster JG. The G protein-coupled receptor P2RY8 and follicular dendritic cells promote germinal center confinement of B cells, whereas S1PR3 can contribute to their dissemination. The Journal of Experimental Medicine. 2015;212(13):2213-2222
  27. 27. Krautler NJ, Kana V, Kranich J, Tian Y, Perera D, Lemm D, et al. Follicular dendritic cells emerge from ubiquitous perivascular precursors. Cell. 2012;150(1)
  28. 28. Maximow A. Bindegewebe und blutbildende Gewebe. Springer; 1927. pp. 232-583. Available from: https://link.springer.com/chapter/10.1007/978-3-642-66441-0_2
  29. 29. Mitchell J, Abbot A. Ultrastructure of the antigen-retaining reticulum of lymph node follicles as shown by high-resolution autoradiography. Nature. 1965;208
  30. 30. Foucar K, Foucar E. The mononuclear phagocyte and immunoregulatory effector (M-PIRE) system: Evolving concepts. Seminars in Diagnostic Pathology. 1990;7
  31. 31. Tew JG, Kosco MH, Burton GF, Szakal AK. Follicular dendritic cells as accessory cells. Immunological Reviews. 1990;117(1):185-211
  32. 32. Kosco MH, Gray D. Signals involved in germinal center reactions. Immunological Reviews. 1992;126(1):63-76
  33. 33. Wang X, Rodda LB, Bannard O, Cyster JG. Integrin-mediated interactions between B cells and follicular dendritic cells influence germinal center B cell fitness. The Journal of Immunology. 2014;192(10)
  34. 34. Heesters BA, Myers RC, Carroll MC. Follicular dendritic cells: Dynamic antigen libraries. Nature Reviews. Immunology. 2014;14
  35. 35. Bharadwaj H, Freeman J. EBV+ inflammatory pseudotumor-like follicular/fibroblastic dendritic cell sarcoma: An uncommon and challenging tumor. American Journal of Clinical Pathology. 2020;154(Supplement_1)
  36. 36. Lee IJ, Kim SC, Kim HS, Bang D, Yang WI, Jung WH, et al. Paraneoplastic pemphigus associated with follicular dendritic cell sarcoma arising from Castleman’s tumor. Journal of the American Academy of Dermatology. 1999;40 (2 II Suppl.)
  37. 37. Facchetti F, Lorenzi L. Follicular dendritic cells and related sarcoma. Seminars in Diagnostic Pathology. 2016;33(5):262-276
  38. 38. Facchetti F, Simbeni M, Lorenzi L. Follicular dendritic cell sarcoma. Pathologica. 2021;113(5):316-329
  39. 39. Andersen EF, Paxton CN, O’Malley DP, Louissaint A, Hornick JL, Griffin GK, et al. Genomic analysis of follicular dendritic cell sarcoma by molecular inversion probe array reveals tumor suppressor-driven biology. Modern Pathology. 2017;30(9):1321-1334
  40. 40. Griffin GK, Sholl LM, Lindeman NI, Fletcher CDM, Hornick JL. Targeted genomic sequencing of follicular dendritic cell sarcoma reveals recurrent alterations in NF-κB regulatory genes. Modern Pathology. 2016;29(1)
  41. 41. Go H, Jeon YK, Huh J, Choi SJ, Choi YD, Cha HJ, et al. Frequent detection of BRAFV600E mutations in histiocytic and dendritic cell neoplasms. Histopathology. 2014;65(2):261-272
  42. 42. Cheuk W, Chan JKC, Shek TWH, Chang JH, Tsou MH, Yuen NWF, et al. Inflammatory pseudotumor-like follicular dendritic cell tumor: A distinctive low-grade malignant intra-abdominal neoplasm with consistent Epstein-Barr virus association. The American Journal of Surgical Pathology. 2001;25(6):721-731
  43. 43. Nayler SJ, Taylor L, Cooper K. HHV-8 is not associated with follicular dendritic cell tumours. Journal of Clinical Pathology – Molecular Pathology. 1998;51(3)
  44. 44. Starr JS, Attia S, Joseph RW, Menke D, Casler J, Smallridge RC. Follicular dendritic cell sarcoma presenting as a thyroid mass. Journal of Clinical Oncology. 2015;33(17)
  45. 45. Tian X, Xu J, Fletcher C, Hornick JL, Dorfman DM. Expression of enhancer of zeste homolog 2 (EZH2) protein in histiocytic and dendritic cell neoplasms with evidence for p-ERK1/2-related, but not MYC-or p-STAT3-related cell signaling. Modern Pathology. 2018;31(4)
  46. 46. Vermi W, Giurisato E, Lonardi S, Balzarini P, Rossi E, Medicina D, et al. Ligand-dependent activation of EGFR in follicular dendritic cells sarcoma is sustained by local production of cognate ligands. Clinical Cancer Research. 2013;19(18):5027-5038
  47. 47. Li XQ , Cheuk W, Lam PWY, Wang Z, Loong F, Yeong ML, et al. Inflammatory pseudotumor-like follicular dendritic cell tumor of liver and spleen: Granulomatous and eosinophil-rich variants mimicking inflammatory or infective lesions. The American Journal of Surgical Pathology. 2014;38(5):646-653
  48. 48. Gong S, Auer I, Duggal R, Pittaluga S, Raffeld M, Jaffe ES. Epstein-Barr virus–associated inflammatory pseudotumor presenting as a colonic mass. Human Pathology. 2015;46(12):1956-1961
  49. 49. Choe JY, Go H, Jeon YK, Yun JY, Kim YA, Kim HJ, et al. Inflammatory pseudotumor-like follicular dendritic cell sarcoma of the spleen: A report of six cases with increased IgG4-positive plasma cells. Pathology International. 2013;63(5):245-251
  50. 50. Pan ST, Cheng CY, Lee NS, Liang PI, Chuang SS. Follicular dendritic cell sarcoma of the inflammatory pseudotumor-like variant presenting as a colonic polyp. Korean Journal of Pathology. 2014;48(2):140-145
  51. 51. Chen Y, Shi H, Li H, Zhen T, Han A. Clinicopathological features of inflammatory pseudotumour-like follicular dendritic cell tumour of the abdomen. Histopathology. 2016;68(6):858-865
  52. 52. Liu KL, Shen JL, Yang CS, Chen YJ. Paraneoplastic pemphigus as the first manifestation of follicular dendritic cell sarcoma. Journal der Deutschen Dermatologischen Gesellschaft. 2014;12(1):68-71
  53. 53. Lu T, Song B, Pu H, Li X, Chen Q , Yang C. Paraneoplastic pemphigus and myasthenia gravis as the first manifestations of a rare case of pancreatic follicular dendritic cell sarcoma: CT findings and review of literature. BMC Gastroenterology. 2019;19(1)
  54. 54. Chow SCY, Yeung ECL, Ng CSH, Wong RHL, Fai To K, Wan IYP. Mediastinal follicular dendritic cell sarcoma with paraneoplastic pemphigus. Asian Cardiovascular & Thoracic Annals. 2015;23(6):732-734
  55. 55. Garza-Chapa JI, Ocampo- Garza J, Vázquez-Herrera NE, Miranda-Maldonado IC, Rendón-Ramírez E, González-Chávez JM, et al. Paraneoplastic pemphigus associated with primary pulmonar follicular dendritic cell sarcoma showing good response to treatment. Journal of the European Academy of Dermatology and Venereology. 2016;30(3):465-467
  56. 56. Zhuang JY, Zhang FF, Li QW, Chen YF. Intra-abdominal inflammatory pseudotumor-like follicular dendritic cell sarcoma associated with paraneoplastic pemphigus: A case report and review of the literature. World Journal of Clinical Cases. 2020;8(14):3097-3107
  57. 57. Lin O, Frizzera G. Angiomyoid and follicular dendritic cell proliferative lesions in Castleman’s disease of hyaline-vascular type: A study of 10 cases. The American Journal of Surgical Pathology. 1997;21(11):1295-1306
  58. 58. Phrathep DD, Healey KD, Anthony S, Fives KR, Boshkos MC, Galani R. Mediastinal follicular dendritic cell sarcoma with underlying Sjogren’s syndrome. Cureus. 2023;4:1
  59. 59. Shia J, Chen W, Tang LH, Carlson DL, Qin J, Guillem JG, et al. Extranodal follicular dendritic cell sarcoma: Clinical, pathologic, and histogenetic characteristics of an underrecognized disease entity. Virchows Archiv. 2006;449(2):148-158
  60. 60. Duan GJ, Wu F, Zhu J, Guo DY, Zhang R, Shen LL, et al. Extranodal follicular dendritic cell sarcoma of the pharyngeal region: A potential diagnostic pitfall, with literature review. American Journal of Clinical Pathology. 2010;133(1):49-58
  61. 61. Pathology Outlines. Follicular dendritic cell sarcoma (FDCS). Available from: https://www.pathologyoutlines.com/topic/lymphnodesfdcsarcoma.html [Accessed: Aug 18, 2023]
  62. 62. Wu A, Pullarkat S. Follicular dendritic cell sarcoma. Archives of Pathology & Laboratory Medicine. 2016;140(2):186-190
  63. 63. Li Y, Yang X, Tao L, Zeng W, Zuo M, Li S, et al. Challenges in the diagnosis of Epstein-Barr virus-positive inflammatory follicular dendritic cell sarcoma: Extremely wide morphologic spectrum and immunophenotype. The American Journal of Surgical Pathology. 2023;47(4):476-489
  64. 64. Lorenzi L, Döring C, Rausch T, Benes V, Lonardi S, Bugatti M, et al. Identification of novel follicular dendritic cell sarcoma markers, FDCSP and SRGN, by whole transcriptome sequencing. Oncotarget. 2017;8(10):16463
  65. 65. Massoth LR, Hung YP, Ferry JA, Hasserjian RP, Nardi V, Nielsen GP, et al. Histiocytic and dendritic cell sarcomas of hematopoietic origin share targetable genomic alterations distinct from follicular dendritic cell sarcoma. The Oncologist. 2021;26(7):e1263-e1272
  66. 66. Gounder M, Desai V, Kuk D, Agaram N, Arcila M, Durham B, et al. Impact of surgery, radiation and systemic therapy on the outcomes of patients with dendritic cell and histiocytic sarcomas. European Journal of Cancer. 2015;51(16):2413-2422
  67. 67. Jain P, Milgrom SA, Patel KP, Nastoupil L, Fayad L, Wang M, et al. Characteristics, management, and outcomes of patients with follicular dendritic cell sarcoma. British Journal of Haematology. 2017;178(3):403-412
  68. 68. Conry RM. Response of follicular dendritic cell sarcoma to gemcitabine and docetaxel: Report of two cases and literature review. Clin Sarcoma Res. 2014;4(1)
  69. 69. Alfaro C, Suarez N, Gonzalez A, Solano S, Erro L, Dubrot J, et al. Influence of bevacizumab, sunitinib and sorafenib as single agents or in combination on the inhibitory effects of VEGF on human dendritic cell differentiation from monocytes. British Journal of Cancer. 2009;100(7):1111-1119
  70. 70. Chen T, Gopal P. Follicular dendritic cell sarcoma. Archives of Pathology & Laboratory Medicine. 2017;141(4):596-599

Written By

Vinay V. and Priya Sharma

Submitted: 02 September 2023 Reviewed: 11 October 2023 Published: 21 March 2024