Context.—

A wide spectrum of mesenchymal tumors harboring ALK gene rearrangements has been identified outside the archetypal example of ALK-positive inflammatory myofibroblastic tumors.

Objective.—

To evaluate the molecular pathology of unusual ALK-positive mesenchymal tumors and their response to ALK-targeted treatments.

Design.—

Seven patients with ALK-positive mesenchymal tumors, including inflammatory epithelioid cell sarcoma, undifferentiated sarcoma, histiocytic neoplasm, smooth muscle tumor of uncertain malignant potential (STUMP), and atypical fibrohistiocytic tumor, were included on the basis of aberrant ALK immunoexpression. Patients with inflammatory myofibroblastic tumors were excluded from the study. ALK gene rearrangement was investigated either by fluorescence in situ hybridization or next-generation sequencing.

Results.—

ALK was immunolabeled in all patients, diffusely (≥50%) in 6 patients and partially (10%–50%) in 1 patient. ALK gene rearrangement was discovered in 5 of the 6 available patients. The 3′-partners of ALK fusion were identified in 3 of 4 investigated patients as follows: PRKAR1A-ALK (ALK-positive histiocytic neoplasm), TNS1-ALK (STUMP), and KIF5B-ALK (ALK-positive atypical fibrohistiocytic tumor). We failed to discover ALK translocation in 1 patient with ALK-positive inflammatory epithelioid cell sarcoma. However, transcriptomic investigation showed that this tumor was significantly enriched with ALK-related pathways, which suggested activation of ALK through a nontranslocation pathway, as a constitutive oncogenic mark in this tumor. ALK-targeted inhibitors, which were administered to 3 patients with metastatic diseases, achieved partial remission in 1 patient with ALK-positive inflammatory epithelioid cell sarcoma and stable disease in patients with ALK-positive undifferentiated sarcoma and STUMP.

Conclusions.—

Molecular investigation of ALK-positive mesenchymal neoplasms could allow for an accurate diagnosis and personalized treatment.

The ALK gene, located on chromosome band 2p23.2–2p23.1, encodes the receptor tyrosine kinase family protein ALK. Since this gene was first described in anaplastic large cell lymphoma in 1994,1  translocation and activation of ALK have been demonstrated in diverse neoplasms of epithelial, melanocytic, hematolymphoid, and mesenchymal origin.211  In addition to the archetypal example of ALK-positive inflammatory myofibroblastic tumor (IMT) and the rarer ALK-positive epithelioid inflammatory myofibroblastic sarcoma (EIMS), it is intriguing that a wide spectrum of mesenchymal tumors, including undifferentiated pleomorphic sarcoma, smooth muscle tumor of uncertain malignant potential (STUMP), leiomyosarcoma, histiocytosis, and fibrohistiocytic tumor, can harbor ALK gene rearrangements.10,1216 ALK-translocated neoplasms have predilections for certain fusion partners. For example, TPM3/4-ALK and CLTC-ALK are frequent in IMT, whereas RANBP2-ALK is frequent in EIMS.17,18  Next-generation sequencing (NGS) is revealing an increasing complexity of ALK alterations as the list of ALK translocations grow.5,11,12,18  Moreover, the recent advent of ALK-targeted tyrosine kinase inhibitors in ALK-positive mesenchymal tumors creates an urgent need to better understand the fundamental molecular pathology associated with ALK gene alterations so that these new regimens can be applied appropriately.10,12,1922  However, the molecular characteristics of ALK-positive mesenchymal tumors outside of IMT are not fully understood because of their rarity. Immunohistochemical and genetic analyses of ALK-driven mesenchymal neoplasms may allow for a molecularly driven diagnosis and personalized treatment. In this study, we investigated the clinical, histopathologic, and molecular characteristics of ALK-positive mesenchymal neoplasms. Aberrant expression and translocation of ALK were identified in a small cohort of rare and in some cases, difficult-to-classify mesenchymal tumors in which ALK inhibitors demonstrated promising results.

Discovery of ALK-Positive Mesenchymal Tumors

First, we retrieved tumors that were immunostained for ALK from the pathologic depository of Seoul National University Hospital (SNUH; Seoul, Republic of Korea) between 2012 and 2018. After eliminating nonmesenchymal tumors, including carcinoma, lymphoma, melanoma, glioma, germ cell tumor, and neuroblastoma, 2 pathologists (M.J. and C.L.) reviewed every diagnosis, ALK expression, and ALK gene translocation, using glass slides, fluorescence in situ hybridization (FISH) images, and NGS data. To exclusively examine unusual mesenchymal tumors with aberrant ALK immunoexpression, or “ALK-positive mesenchymal tumors,” we screened out the classical examples of IMT as based on the latest 2020 World Health Organization classification.23  Tumors showing patchy (<10%) expression of ALK were also excluded. Clinical information was obtained from the medical records. Oncologic response was assessed according to the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria.24  The institutional review board of SNUH approved this study (institutional review board No. H-2102-036-1195).

Immunohistochemical Investigation

A series of immunohistochemical staining, including ALK (1:50; clone 5A4, Leica, Wetzlar, Germany), was carried out with 4-μm-thick formalin-fixed, paraffin-embedded (FFPE) tissue (Supplemental Table 1, see supplemental digital content at https://meridian.allenpress.com/aplm in the December 2022 table of contents). ALK immunohistochemical staining was conducted with a BenchMark autostainer (Ventana, Basel, Switzerland). The expression of ALK was measured and determined to be partial (10%–50%) or diffuse (≥50%). The expression of other proteins was semiquantified as negative (0%), low expression (1%–30%), or high expression (>30%). The Ki-67 proliferative index was digitally analyzed within the highest area by using a QuPath viewer.25 

Fluorescence In Situ Hybridization

Interphase FISH was performed, and the results were assessed under conditions that have been previously described.26  Briefly, a Vysis ALK dual-color break-apart probe kit (Abbott Molecular, Abbott Park, Illinois) was hybridized on available 3-μm-thick FFPE slides. The slides were examined with an Allegro Plus with a Solo Touch Workstation (BioView Ltd, Rehovot, Israel) and were reviewed manually under an Olympus BX51TRF microscope (Olympus, Tokyo, Japan) equipped with the appropriate filters. Translocation of ALK was interpreted as recommended: break-apart green and red signals or isolated red signals were considered positive for ALK translocation.27 

Next-Generation Sequencing

We further investigated ALK gene fusion and potential druggable targets in 4 of the 5 available patients (80%) through targeted gene sequencing (TGS) using in-house hybrid capture–based panels of SNUH (FIRST-Cancer Panel and/or FIRST-Lung Cancer Panel) that cover both the exonic and intronic regions of ALK (Table 1). Briefly, nucleotides were extracted from FFPE tumor tissue by using a QIAamp DNA FFPE Tissue Kit (Qiagen, Valencia, California) and then fragmented by using adaptive focused acoustics (Covaris, Woburn, Massachusetts) following the manufacturer's protocols. The libraries were prepared by using the SureSelect XT Reagent Kit (Agilent, Santa Clara, California).

Table 1

Clinical Details and Treatment Outcomes of Patients With ALK-Positive Mesenchymal Tumors

Clinical Details and Treatment Outcomes of Patients With ALK-Positive Mesenchymal Tumors
Clinical Details and Treatment Outcomes of Patients With ALK-Positive Mesenchymal Tumors

In addition, mRNA sequencing (RNA-seq) was used to confirm the ALK fusion status and the functional significance of ALK overexpression in patient 3 by comparing the transcriptional profiles of tumor versus normal tissue (Macrogen, Seoul, Republic of Korea). To this end, total RNA was extracted from FFPE specimens, followed by selective enrichment of mRNA with a poly-A tail and preparation of a cDNA library using the TruSeq RNA Access Library Prep Kit (Illumina, San Diego, California). Paired-end sequencing was performed on a HiSeq2500 platform (Illumina). Differentially expressed genes were identified with an absolute fold change of 2 or greater as a cutoff. Enrichment of gene sets was investigated from the Kyoto Encyclopedia of Genes and Genomes (KEGG) database28  and the MsigDB gene sets.29  The detailed methods have been previously published.30 

Patient Characteristics

Patient data from a total of 7 individuals with ALK-positive mesenchymal tumors were analyzed (Table 1). The ages at diagnosis varied and ranged from 13 to 63 years. Three patients with malignancies (patients 1, 3, and 5) initially had distant metastasis. Patient 4 developed lung metastasis 19 months after diagnosis. Patient 2 had an ALK-positive inflammatory sarcoma confined to the lung. In contrast, patient 6 was diagnosed with STUMP, which recurred with multiple metastases. Patient 7, who was diagnosed with an ALK-positive atypical fibrohistiocytic tumor, had localized disease in the nipple, which was suspected to have borderline malignant potential.

Clinical Course

Table 1 summarizes the clinical course and treatment response of the patients. Among the 5 patients with systemic diseases (patients 1, 3, 4, 5, and 6), 3 (60%; patients 1, 4, and 5) received cytotoxic chemotherapy and 3 (60%; patients 1, 4, and 6) received ALK-targeted tyrosine kinase inhibitors. Patient 5 initially underwent wide excision, followed by systemic treatment, after which he received a second excision for the chemotherapy-resistant lesion. Patient 3 received localized treatments for both primary and metastatic lesions. The other patients (2 and 7) with localized disease underwent complete surgical excision. After treatments, patients 1 and 4 died of the disease, patient 6 was alive with the disease for 7 years, and the other patients were free of disease.

ALK Aberration and Diagnostic Considerations

The histopathologic characteristics, including immunostaining for ALK and ALK gene rearrangement, are described in Tables 1 and 2. All patients (7 of 7, 100%) had aberrant ALK immunoreaction, which was diffuse (≥50%) in 6 patients (86%) and partial (<50%) in 1 patient (14%) in the cytoplasm and other subcellular locations, including the cytoplasmic membranes (patients 3 and 4), the nuclear membrane (patient 4), and the nucleus (patient 7) (Table 2). Rearrangement of ALK was identified in 5 of the 6 investigated patients (83%) with either FISH or NGS (TGS or RNA-seq) (Table 1). Patient 2, a consultation case from an outside hospital, had no material for the molecular assay.

Table 2

Pathologic Characteristics of Patients With ALK-Positive Mesenchymal Tumors

Pathologic Characteristics of Patients With ALK-Positive Mesenchymal Tumors
Pathologic Characteristics of Patients With ALK-Positive Mesenchymal Tumors

Diffuse immunoexpression and genetic aberrations of ALK, together with histopathologic findings, indicated ALK-positive inflammatory epithelioid cell sarcoma with features of EIMS as a diagnosis for patients 1 through 3. Among these patients, patients 1 and 2 had initially been diagnosed otherwise, with malignant perivascular epithelioid cell tumor and intimal sarcoma, respectively, at outside hospitals. In patients 1 (Figure 1, A and B) and 2 (Figure 1, C and D), the tumor cells were spindled to epithelioid, arranged in a fascicle or in a solid growth pattern, and infiltrated by numerous inflammatory cells, including lymphocytes, plasma cells, and/or eosinophils. Along with diffuse ALK immunolabeling, ALK translocation was confirmed in patient 1 (Figure 1, B, inset). Similarly, patient 3 demonstrated round to polygonal tumor cells with diffuse ALK immunoexpression that were admixed with lymphocytes and plasma cells (Figure 1, E and F). Although structural variation in the ALK gene was undetected by FISH (Figure 1, F, inset), TGS, and RNA-seq (Table 1), gene expression profiling identified ALK-driven molecular pathways, as described below, which suggested ALK-positive inflammatory epithelioid cell sarcoma in this patient.

Figure 1

A through F, Inflammatory epithelioid cell sarcoma. A, C, and E, Epithelioid tumor cells admixed with lymphocytes and/or plasma cells (patients 1 through 3). B, D, and F, Diffuse ALK expression and ALK translocation (patients 1 through 3). G and H, Undifferentiated sarcoma (patient 4). G, Pleomorphic tumor cells without inflammatory cells. H, Diffuse ALK expression and ALK translocation (hematoxylin-eosin, original magnification, ×200 [A, C, E, and G]; ALK, original magnification ×200 [B, D, F, and H]; fluorescence in situ hybridization, original magnification ×1000 [insets B, F, and H]).

Figure 1

A through F, Inflammatory epithelioid cell sarcoma. A, C, and E, Epithelioid tumor cells admixed with lymphocytes and/or plasma cells (patients 1 through 3). B, D, and F, Diffuse ALK expression and ALK translocation (patients 1 through 3). G and H, Undifferentiated sarcoma (patient 4). G, Pleomorphic tumor cells without inflammatory cells. H, Diffuse ALK expression and ALK translocation (hematoxylin-eosin, original magnification, ×200 [A, C, E, and G]; ALK, original magnification ×200 [B, D, F, and H]; fluorescence in situ hybridization, original magnification ×1000 [insets B, F, and H]).

Close modal

Given that patient 4 had highly pleomorphic tumor cells and no intermixing inflammatory cells, ALK-positive undifferentiated sarcoma was diagnosed despite diffuse ALK expression and ALK translocation (Figure 1, G and H). Patient 5 had a 7.0-cm hypermetabolic mass in the right infrapatellar fat pad with involvement of the adjacent bone (Figure 2, A and B). A positron emission tomography scan also revealed other lesions in the left knee and right inguinal/external iliac lymph nodes, which raised the possibility of bilateral malignancy with lymph node metastasis (Figure 2, B). Excisional biopsy of the primary mass revealed discohesive, round to polygonal cells containing round nuclei, prominent nucleoli, and vacuolated cytoplasm, which frequently showed hemophagocytosis (Figure 2, C). Immunohistochemical staining for ALK was diffuse-positive, and FISH analysis revealed ALK translocation (Figure 2, D). On the basis of positivity for histiocytic markers, including CD68, CD163, and lysozyme (Table 2), this patient was diagnosed with an ALK-positive histiocytic neoplasm. Patient 6, who had been treated for STUMP, had locoregional recurrence and bony metastases. Recurred tumors also construed as STUMP showed brisk mitoses without coagulative necrosis and myxoid changes (Figure 2, E; Table 2). Although ALK was weakly and partially (<50%) expressed in tumor cells, rearrangement of ALK was demonstrated by FISH and TGS (Figure 2, F). Finally, patient 7 had a tumor in the nipple involving the epidermis to deep dermis that showed solid or fascicular proliferation of spindle cells with mild cytologic atypia and lymphoplasma cell-rich stroma (Figure 2, G and H). Assessment of ALK revealed diffuse immunoreaction and ALK translocation (Figure 2, I). Further studies showed histiocytic differentiation, with positivity for CD68 and CD163 but negativity for smooth muscle–related antigens, including smooth muscle actin (SMA), desmin, calponin, and caldesmon (Table 2). The dermal-centered location and absence of smooth muscle differentiation suggested an ALK-positive atypical fibrohistiocytic tumor.

Figure 2

A through D, ALK-positive histiocytic neoplasm (patient 5). A, Right knee mass. B, Main mass (arrow) and other lesions/lymph nodes (arrowheads). C, Hemophagocytosis in the primary tumor. D, Diffuse ALK expression and ALK translocation. E and F, Smooth muscle tumor of uncertain malignant potential (patient 6). E, Mitoses (circle). F, Partial ALK expression and ALK rearrangement. G through I, ALK-positive atypical fibrohistiocytic tumor (patient 7). G and H, Spindle cells with lymphocytes in the epidermis-dermis. I, Diffuse ALK expression and ALK translocation (hematoxylin-eosin, original magnifications ×200 [C and E], ×150 [G], and ×400 [H]; ALK, original magnifications ×200 [D and I] and ×400 [F]; fluorescence in situ hybridization, original magnification ×1000 [insets D, F, and I]).

Figure 2

A through D, ALK-positive histiocytic neoplasm (patient 5). A, Right knee mass. B, Main mass (arrow) and other lesions/lymph nodes (arrowheads). C, Hemophagocytosis in the primary tumor. D, Diffuse ALK expression and ALK translocation. E and F, Smooth muscle tumor of uncertain malignant potential (patient 6). E, Mitoses (circle). F, Partial ALK expression and ALK rearrangement. G through I, ALK-positive atypical fibrohistiocytic tumor (patient 7). G and H, Spindle cells with lymphocytes in the epidermis-dermis. I, Diffuse ALK expression and ALK translocation (hematoxylin-eosin, original magnifications ×200 [C and E], ×150 [G], and ×400 [H]; ALK, original magnifications ×200 [D and I] and ×400 [F]; fluorescence in situ hybridization, original magnification ×1000 [insets D, F, and I]).

Close modal

Characterization of Genetic Alterations of ALK

NGS studies were conducted to further characterize ALK gene alterations in 4 patients (3, 5 through 7), including patient 3, in whom we failed to identify ALK gene translocations by FISH. The results indicated the following fusion of the 3′ domains of ALK to 5′ partners (Figure 3, A): PRKAR1A-ALK (patient 5; Figure 3, B), TNS1-ALK (patient 6; Figure 3, C), and KIF5B-ALK (patient 7; Figure 3, D). Such chimeric proteins were expected to encompass the tyrosine kinase motifs of ALK. The same TNS1-ALK fusion was also discovered in the FoundationOne Heme panel (Foundation Medicine, Cambridge, Massachusetts) in patient 6.

Figure 3

A, ALK gene fusions identified in patients 5 through 7. B through D, Illustrations showing each fusion transcript and protein. These data are based on Genome Browser (https://genome.ucsc.edu) and UniProt (https://www.uniprot.org) without reflecting the actual scale.

Figure 3

A, ALK gene fusions identified in patients 5 through 7. B through D, Illustrations showing each fusion transcript and protein. These data are based on Genome Browser (https://genome.ucsc.edu) and UniProt (https://www.uniprot.org) without reflecting the actual scale.

Close modal

Although ALK translocation was absent in patient 3 by FISH, TGS, and RNA-seq, significant overexpression of the ALK gene was verified in the tumor compared to adjacent normal tissue (Figure 4, A). Transcriptomic data suggested that tumor cells were enriched with downstream cascades of ALK activation, including the phosphatidylinositol 3-kinase (PI3K)–Akt and mitogen-activated protein kinase (MAPK) pathways, which were identified among the top 5 significantly enriched KEGG pathways (Figure 4, B). In addition, gene sets associated with E2F targets (Figure 4, C) and the G2/M checkpoint (Figure 4, D) were enriched in the tumor, but the Akt (Figure 4, E) and ALK (Figure 4, F) pathways were downregulated in normal tissue. These results collectively suggested that ALK gene activation might be a driver oncogene in patient 3 through a nontranslocation pathway.

Figure 4

Transcriptomic analysis in patient 3. A, ALK gene overexpression in the tumor compared to adjacent normal tissue. B, Top 5 significant KEGG pathways represented by DEGs between the tumor and adjacent normal tissue. DEGs were compared against the KEGG molecular pathways. C through F, Gene set enrichment analysis calculates enrichment scores on the basis of distribution of the predetermined gene sets either at the top (upregulated in tumor, positive score) or at the bottom (downregulated in tumor, negative score) of the DEGs. Abbreviations: DEGs, differentially expressed genes; FDR, false discovery rate; KEGG, Kyoto Encyclopedia of Genes and Genomes; NES, normalized enrichment score.

Figure 4

Transcriptomic analysis in patient 3. A, ALK gene overexpression in the tumor compared to adjacent normal tissue. B, Top 5 significant KEGG pathways represented by DEGs between the tumor and adjacent normal tissue. DEGs were compared against the KEGG molecular pathways. C through F, Gene set enrichment analysis calculates enrichment scores on the basis of distribution of the predetermined gene sets either at the top (upregulated in tumor, positive score) or at the bottom (downregulated in tumor, negative score) of the DEGs. Abbreviations: DEGs, differentially expressed genes; FDR, false discovery rate; KEGG, Kyoto Encyclopedia of Genes and Genomes; NES, normalized enrichment score.

Close modal

Response to ALK-Targeted Treatments

ALK inhibitors were administered to patients 1, 4, and 6, who achieved partial remission (patient 1) or stable disease (patients 4 and 6) (Table 1). Patient 1, who was diagnosed with ALK-translocated inflammatory epithelioid cell sarcoma, was enrolled in clinical trials for the ALK inhibitors crizotinib (NCT01121588) and ceritinib (NCT01283516) after worsening of liver metastases despite cytotoxic chemotherapy. Both crizotinib, as a third-line treatment, and subsequently ceritinib, as a fourth-line treatment, successfully alleviated the tumor burden for 10 and 7 months, respectively (Figure 5, A). Patient 4, who was diagnosed with ALK-positive undifferentiated sarcoma, developed pulmonary metastases shortly after doxorubicin-based chemotherapy, had stable disease after 4 months of ceritinib treatment, and showed a slight decrease in metastatic nodules (Figure 5, B). Later, this patient developed a brain metastasis, and he died of the disease 9 months after diagnosis. Patient 6, who was diagnosed with STUMP, experienced a leiomyosarcoma-like clinical course with aggravating metastases in the spine and ileum. These bony lesions exhibited metabolic responses to alectinib for 6 months and showed a 30% or greater reduction in standardized uptake value (Figure 5, C). She had been living with the disease for 7 years.

Figure 5

Responses to ALK inhibitors. A, Initial partial remission, followed by disease progression after crizotinib and ceritinib treatment in inflammatory epithelioid cell sarcoma (patient 1). B, Response to ceritinib in metastatic undifferentiated sarcoma (patient 4). C, A metabolic response to alectinib in metastatic STUMP (patient 6) in both the spine (upper) and iliac bone (lower). Bold lines denote the duration of ALK-targeted treatment. Abbreviations: PD, progressive disease; PR, partial remission; SD, stable disease; STUMP, smooth muscle tumor of uncertain malignant potential; SUV, standardized uptake value.

Figure 5

Responses to ALK inhibitors. A, Initial partial remission, followed by disease progression after crizotinib and ceritinib treatment in inflammatory epithelioid cell sarcoma (patient 1). B, Response to ceritinib in metastatic undifferentiated sarcoma (patient 4). C, A metabolic response to alectinib in metastatic STUMP (patient 6) in both the spine (upper) and iliac bone (lower). Bold lines denote the duration of ALK-targeted treatment. Abbreviations: PD, progressive disease; PR, partial remission; SD, stable disease; STUMP, smooth muscle tumor of uncertain malignant potential; SUV, standardized uptake value.

Close modal

We described the clinicopathologic and molecular properties of patients with ALK-positive mesenchymal tumors and their response to ALK-targeted therapies. Genetic rearrangement of ALK was identified in 5 of 6 available cases and was not observed in patient 3, in whom ALK-driven oncogenic pathways were suggested by transcriptomic analysis. The oncogenic action of ALK depends on the ligand-independent activation of the intracytoplasmic tyrosine kinase moiety at the 3′ end of ALK. In ALK-translocated neoplasms, such tyrosine kinase regions are dissociated and fuse with new partners at the 5′ end, as identified in patients 5 (TNS1-ALK), 6 (PRKAR1A-ALK), and 7 (KIF5B-ALK) of the present study.5,11  Expression of ALK and/or its translocation were key to changing the diagnoses of patients 1 through 3. In addition, we suggest that altered expression of ALK could be found outside of conventional IMT/EIMS. For instance, we discovered positivity for ALK in a handful of poorly defined mesenchymal and histiocytic neoplasms, including 3 inflammatory epithelioid cell sarcomas exhibiting features of EIMS, undifferentiated sarcoma, and a histiocytic neoplasm of soft tissue. Positive ALK immunolabeling could lead to further molecular studies and ALK-targeted treatments, which demonstrated promising results in patients with metastatic sarcomas included in this study. ALK expression in tumors is closely related to the underlying genetic alterations of ALK.11,21  Therefore, it is plausible to argue that immunohistochemical investigation of ALK is an efficient method that can aid in the correct diagnosis of unusual mesenchymal tumors and in the selection of personalized treatment for aggressive sarcomas.

Constitutively activated ALK enhances the proliferation, survival, and migration of tumor cells through multiple pathways, including Ras-MAPK, PI3K-Akt, phospholipase Cγ, and Janus kinase (JAK)–signal transducer and activator of transcription (STAT) signals, or less commonly, through epigenetic deregulation.5,11  This pathogenicity centered around ALK has become a basis for targeted therapies in ALK-driven diseases, including mesenchymal neoplasms. For example, crizotinib, a small-molecule inhibitor of ALK and other tyrosine kinases, is a treatment of choice for ALK-rearranged non–small cell lung cancer and has also become the standard treatment for locally advanced or metastatic ALK-positive IMT/EIMS.19,21,3133  Consistent with this, the ALK-translocated inflammatory epithelioid cell sarcoma in patient 1 showed robust shrinkage after crizotinib treatment, which was followed by similar responsiveness to ceritinib.33  In addition, as seen in patients 4 and 6, who had response to ALK inhibitors, off-label treatment with ALK inhibitors has resulted in treatment effects in ALK-driven mesenchymal tumors, such as STUMP, leiomyosarcoma, rhabdomyosarcoma, and myofibroblastic sarcoma, in vitro and in vivo.16,20,34,35  To our knowledge, this is the first report to share the real-world experience of responses of patients with ALK-positive undifferentiated sarcoma to ALK inhibitors.

Case 3 is an unusual inflammatory sarcoma with features of EIMS that did not reveal any signs of differentiation. Although there was no structural variation in ALK, the ALK gene was significantly overexpressed, consistent with the immunohistochemical staining for ALK. Moreover, we demonstrated enrichment of the PI3K-Akt and MAPK pathways, upregulation of molecular hallmarks related to proliferation, including E2F targets and G2/M checkpoint proteins, and downregulation of Akt and ALK pathways in this tumor, suggesting that ALK, which was overexpressed, functioned as a driver oncogene in this patient. Similar immunohistochemical and genetic discrepancies were previously reported in ALK-positive IMT.18  In addition to translocation, activating events in the ALK gene, including mutation, amplification, or epigenetic dysregulation, have been reported in different sarcomas.3638  The exact genetic alteration of ALK in this patient has yet to be confirmed by further investigation.

Like in EIMS, patients 1 through 3 had epithelioid or polygonal tumor cells and patients 1 and 3 also had infiltrating inflammatory cells. However, there were some discrepancies between inflammatory epithelioid cell sarcomas diagnosed in patients 1 through 3 and EIMS. For example, EIMS has been typified by recurrent RANBP2-ALK or RRBP1-ALK translocation, ALK immunoreactivity in the nuclear membrane or perinuclear cytoplasm, and frequent expression of desmin and CD30.17,39  In patients 1 through 3, however, ALK translocation, which was studied in 1 patient, and such immunohistochemical characteristics were not observed, as ALK was expressed predominantly in the cytoplasm and/or cytoplasmic membrane in all patients, desmin was expressed in 1 patient (33%), and CD30 was nonreactive in 2 included patients (0%). In addition, patients 2 and 3 had tumors in the lung and buttock, respectively, which are unusual sites for EIMS.17  Therefore, we classified these tumors as ALK-positive inflammatory epithelioid cell sarcomas with features of EIMS, although the data on ALK-positive epithelioid cell sarcoma are very limited.40  In addition, ALK immunostaining associated with RANBP2/RRBP1-ALK–rearranged EIMS was shown in ALK-positive undifferentiated sarcoma (nuclear membrane; patient 4) and ALK-positive atypical fibrohistiocytic tumor (nucleus; patient 7) in the present study. This result suggests that other fusion partners may also produce immunohistochemical patterns similar to those related to the molecular topology of RANBP2/RRBP1-ALK fusions.11,17,39 

The ALK-positive histiocytic neoplasm of patient 5 is a rare, ill-defined entity. ALK-positive histiocytosis, which has been reported in fewer than 20 patients, is possibly an appropriate classification for this neoplasm.12,15  In agreement with the findings for patient 5, ALK-positive histiocytosis may also diffusely express ALK, show hemophagocytosis, and involve distant organs.10,12,15,41 KIF5B, and less frequently, COL1A2, EML4, or TPM3, have been identified as partners of ALK fusion in this rare entity.10,12  We found a novel transcript, PRKAR1A-ALK, in ALK-positive histiocytic neoplasm, which was reported in ALK-positive IMT.18  ALK inhibitors, as previously reported in ALK-positive histiocytosis, might be a promising treatment option for this patient in the future.12  It is worth mentioning that unlike what has been described in ALK-positive histiocytosis, patient 5 had a large and infiltrative soft tissue mass that exhibited cytologic atypia and presented with lymph node metastasis; this patient also had to undergo a second excision for persistent disease, even after systemic treatment following wide excision, and did not show any hematologic abnormality. On the contrary, ALK-positive histiocytosis is usually reported to have an indolent clinical course, even in disseminated conditions.12  Therefore, we reasoned that the ALK-positive histiocytic neoplasm in patient 5 might represent a distinct entity that may benefit from more intensive treatments than what has been described for ALK-positive histiocytosis.12  In addition, histiocytic sarcoma could be another differential diagnosis for case 5, which may be distinguished by the absence of “monster” cells and the presence of ALK expression in our patient.42 

Patient 7, who was diagnosed with ALK-positive atypical fibrohistiocytic tumor, had a KIF5B-ALK fusion, and intriguingly, this is the same fusion that is frequently observed in ALK-positive histiocytosis.12,43  Histiocytic differentiation by immunohistochemical staining in patient 7 might signify a connection of this fusion to histiocytic differentiation. The main differential diagnosis for this patient was ALK-positive IMT, although myofibroblastic differentiation was absent.23  For example, the same KIF5B-ALK fusion was discovered in an ALK-positive spindle cell tumor of the brain that was negative for SMA and desmin and that was classified as IMT by the authors.44  Epithelioid fibrous histiocytoma, which is characterized by consistent expression of ALK and ALK gene rearrangements, also requires diagnostic consideration.13  In addition to its classical exophytic, well-circumscribed, and epithelioid morphologies, nontypical forms containing spindle cells admixed with inflammatory cells have been recognized.45  ALK-positive skin tumors encompass a wide spectrum of diseases, from benign to malignant tumors, which may share pathologic features.13  The results of the present study would aid in overcoming this diagnostic challenge in the future.

We investigated ALK expression by using a standardized method across all patients. However, the small number of patients included in this study limited the detailed determination of the immunohistochemical-genetic associations of ALK.5  In addition, comprehensive assessment of the types of ALK fusions implicated in the response to ALK-targeted therapy in ALK-positive mesenchymal tumors is urgently needed. Finally, the unambiguous classification of ALK-positive undifferentiated (patient 4), histiocytic (patient 5), and fibrohistiocytic (patient 7) neoplasms has not yet been determined owing to their rarity, which warrants further study.

1.
Shiota
M,
Fujimoto
J,
Semba
T,
Satoh
H,
Yamamoto
T,
Mori
S.
Hyperphosphorylation of a novel 80 kDa protein-tyrosine kinase similar to Ltk in a human Ki-1 lymphoma cell line, AMS3
.
Oncogene
.
1994
;
9
(6)
:
1567
1574
.
2.
Amatu
A,
Somaschini
A,
Cerea
G,
et al
Novel CAD-ALK gene rearrangement is drugable by entrectinib in colorectal cancer
.
Br J Cancer
.
2015
;
113
(12)
:
1730
1734
.
3.
Soda
M,
Choi
YL,
Enomoto
M,
et al
Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer
.
Nature
.
2007
;
448
(7153)
:
561
566
.
4.
Kusano
H,
Togashi
Y,
Akiba
J,
et al
Two cases of renal cell carcinoma harboring a novel STRN-ALK fusion gene
.
Am J Surg Pathol
.
2016
;
40
(6)
:
761
769
.
5.
Ducray
SP,
Natarajan
K,
Garland
GD,
Turner
SD,
Egger
G.
The transcriptional roles of ALK fusion proteins in tumorigenesis
.
Cancers (Basel)
.
2019
;
11
(8)
:
1074
.
6.
Beltran
B,
Castillo
J,
Salas
R,
et al
ALK-positive diffuse large B-cell lymphoma: report of four cases and review of the literature
.
J Hematol Oncol
.
2009
;
2
:
11
.
7.
Busam
KJ,
Kutzner
H,
Cerroni
L,
Wiesner
T.
Clinical and pathologic findings of Spitz nevi and atypical Spitz tumors with ALK fusions
.
Am J Surg Pathol
.
2014
;
38
(7)
:
925
933
.
8.
Perron
E,
Pissaloux
D,
Charon Barra
C,
et al
Melanocytic myxoid spindle cell tumor with ALK rearrangement (MMySTAR): report of 4 cases of a nevus variant with potential diagnostic challenge
.
Am J Surg Pathol
.
2018
;
42
(5)
:
595
603
.
9.
Griffin
CA,
Hawkins
AL,
Dvorak
C,
Henkle
C,
Ellingham
T,
Perlman
EJ.
Recurrent involvement of 2p23 in inflammatory myofibroblastic tumors
.
Cancer Res
.
1999
;
59
(12)
:
2776
2780
.
10.
Ross
JS,
Ali
SM,
Fasan
O,
et al
ALK fusions in a wide variety of tumor types respond to anti-ALK targeted therapy
.
Oncologist
.
2017
;
22
(12)
:
1444
1450
.
11.
Hallberg
B,
Palmer
RH.
Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology
.
Nat Rev Cancer
.
2013
;
13
(10)
:
685
700
.
12.
Chang
KTE,
Tay
AZE,
Kuick
CH,
et al
ALK-positive histiocytosis: an expanded clinicopathologic spectrum and frequent presence of KIF5B-ALK fusion
.
Mod Pathol
.
2019
;
32
(5)
:
598
608
.
13.
Hornick
JL.
Cutaneous soft tissue tumors: how do we make sense of fibrous and “fibrohistiocytic” tumors with confusing names and similar appearances?
Mod Pathol
.
2020
;
33
(suppl 1)
:
56
65
.
14.
Marušić
Z,
Ko
JS,
Billings
SD.
Cutaneous fibrohistiocytic/fibroblastic tumours: an update
.
Diagn Histopathol
.
2018
;
24
(8)
:
288
300
.
15.
Chan
JK,
Lamant
L,
Algar
E,
et al
ALK+ histiocytosis: a novel type of systemic histiocytic proliferative disorder of early infancy
.
Blood
.
2008
;
112
(7)
:
2965
2968
.
16.
Devereaux
KA,
Kunder
CA,
Longacre
TA.
ALK-rearranged tumors are highly enriched in the STUMP subcategory of uterine tumors
.
Am J Surg Pathol
.
2019
;
43
(1)
:
64
74
.
17.
Yu
L,
Liu
J,
Lao
IW,
Luo
Z,
Wang
J.
Epithelioid inflammatory myofibroblastic sarcoma: a clinicopathological, immunohistochemical and molecular cytogenetic analysis of five additional cases and review of the literature
.
Diagn Pathol
.
2016
;
11
(1)
:
67
.
18.
Lovly
CM,
Gupta
A,
Lipson
D,
et al
Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions
.
Cancer Discov
.
2014
;
4
(8)
:
889
895
.
19.
Casanova
M,
Brennan
B,
Alaggio
R,
et al
Inflammatory myofibroblastic tumor: the experience of the European pediatric Soft Tissue Sarcoma Study Group (EpSSG)
.
Eur J Cancer
.
2020
;
127
:
123
129
.
20.
Davis
LE,
Nusser
KD,
Przybyl
J,
et al
Discovery and characterization of recurrent, targetable ALK fusions in leiomyosarcoma
.
Mol Cancer Res
.
2019
;
17
(3)
:
676
685
.
21.
Theilen
TM,
Soerensen
J,
Bochennek
K,
et al
Crizotinib in ALK(+) inflammatory myofibroblastic tumors—current experience and future perspectives
.
Pediatr Blood Cancer
.
2018
;
65
(4)
:
e26920
.
22.
Vargas-Madueno
F,
Gould
E,
Valor
R,
Ngo
N,
Zhang
L,
Villalona-Calero
MA.
EML4-ALK rearrangement and its therapeutic implications in inflammatory myofibroblastic tumors
.
Oncologist
.
2018
;
23
(10)
:
1127
1132
.
23.
Yamamoto
H.
Inflammatory myofibroblastic tumour
.
In:
WHO Classification of Tumours Editorial Board, ed. Soft Tissue and Bone Tumours. 5th ed
.
Lyon, France
:
International Agency for Research on Cancer (IARC);
2020
:
109
111
.
World Health Organization Classification of Tumours; vol 3.
24.
Schwartz
LH,
Litiere
S,
de Vries
E,
et al
RECIST 1.1-update and clarification: from the RECIST committee
.
Eur J Cancer
.
2016
;
62
:
132
137
.
25.
Bankhead
P,
Loughrey
MB,
Fernandez
JA,
et al
QuPath: open source software for digital pathology image analysis
.
Sci Rep
.
2017
;
7
(1)
:
16878
.
26.
Jung
M,
Kim
S,
Lee
JK,
et al
Clinicopathological and preclinical findings of NUT carcinoma: a multicenter study
.
Oncologist
.
2019
;
24
(8)
:
e740
e748
.
27.
Kim
H,
Shim
HS,
Kim
L,
et al
Guideline recommendations for testing of ALK gene rearrangement in lung cancer: a proposal of the Korean cardiopulmonary pathology study group
.
Korean J Pathol
.
2014
;
48
(1)
:
1
9
.
28.
Kanehisa
M,
Goto
S.
KEGG: Kyoto Encyclopedia of Genes and Genomes
.
Nucleic Acids Res
.
2000
;
28
(1)
:
27
30
.
29.
Subramanian
A,
Tamayo
P,
Mootha
VK,
et al
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
.
2005
;
102
(43)
:
15545
15550
.
30.
Martin
JA,
Wang
Z.
Next-generation transcriptome assembly
.
Nat Rev Genet
.
2011
;
12
(10)
:
671
682
.
31.
Schoffski
P,
Sufliarsky
J,
Gelderblom
H,
et al
Crizotinib in patients with advanced, inoperable inflammatory myofibroblastic tumours with and without anaplastic lymphoma kinase gene alterations (European Organisation for Research and Treatment of Cancer 90101 CREATE): a multicentre, single-drug, prospective, non-randomised phase 2 trial
.
Lancet Respir Med
.
2018
;
6
(6)
:
431
441
.
32.
Mosse
YP,
Voss
SD,
Lim
MS,
et al
Targeting ALK with crizotinib in pediatric anaplastic large cell lymphoma and inflammatory myofibroblastic tumor: a Children's Oncology Group Study
.
J Clin Oncol
.
2017
;
35
(28)
:
3215
3221
.
33.
Trahair
T,
Gifford
AJ,
Fordham
A,
et al
Crizotinib and surgery for long-term disease control in children and adolescents with ALK-positive inflammatory myofibroblastic tumors
.
JCO Precis Oncol
.
2019
;
3
:
1
11
.
34.
Jiao
XD,
Liu
K,
Xu
M,
et al
Metastatic low-grade sarcoma with CARS-ALK fusion dramatically responded to multiple ALK tyrosine kinase inhibitors: a case report with comprehensive genomic analysis
.
Oncologist
.
2020
;
25
:
1
6
.
35.
Felkai
L,
Banusz
R,
Kovalszky
I,
et al
The presence of ALK alterations and clinical relevance of crizotinib treatment in pediatric solid tumors
.
Pathol Oncol Res
.
2019
;
25
(1)
:
217
224
.
36.
George
RE,
Sanda
T,
Hanna
M,
et al
Activating mutations in ALK provide a therapeutic target in neuroblastoma
.
Nature
.
2008
;
455
(7215)
:
975
978
.
37.
Corao
DA,
Biegel
JA,
Coffin
CM,
et al
ALK expression in rhabdomyosarcomas: correlation with histologic subtype and fusion status
.
Pediatr Dev Pathol
.
2009
;
12
(4)
:
275
283
.
38.
Huang
TT,
Gonzales
CB,
Gu
F,
et al
Epigenetic deregulation of the anaplastic lymphoma kinase gene modulates mesenchymal characteristics of oral squamous cell carcinomas
.
Carcinogenesis
.
2013
;
34
(8)
:
1717
1727
.
39.
Lee
JC,
Li
CF,
Huang
HY,
et al
ALK oncoproteins in atypical inflammatory myofibroblastic tumours: novel RRBP1-ALK fusions in epithelioid inflammatory myofibroblastic sarcoma
.
J Pathol
.
2017
;
241
(3)
:
316
323
.
40.
Ishibashi
Y,
Miyoshi
H,
Hiraoka
K,
et al
Anaplastic lymphoma kinase protein expression, genetic abnormalities, and phosphorylation in soft tissue tumors: phosphorylation is associated with recurrent metastasis
.
Oncol Rep
.
2015
;
33
(4)
:
1667
1674
.
41.
Huang
HY,
Gheorghe
G,
North
PE,
Suchi
M.
Expanding the phenotype of ALK-positive histiocytosis: a report of 2 cases
.
Pediatr Dev Pathol
.
2018
;
21
(5)
:
449
455
.
42.
Hung
YP,
Qian
X.
Histiocytic sarcoma
.
Arch Pathol Lab Med
.
2020
;
144
(5)
:
650
654
.
43.
Diamond
EL,
Durham
BH,
Haroche
J,
et al
Diverse and targetable kinase alterations drive histiocytic neoplasms
.
Cancer Discov
.
2016
;
6
(2)
:
154
165
.
44.
Maruggi
M,
Malicki
DM,
Levy
ML,
Crawford
JR.
A novel KIF5B-ALK fusion in a child with an atypical central nervous system inflammatory myofibroblastic tumour
.
BMJ Case Rep.
2018
;
2018(2018):bcr-2018-226431.
45.
Kazakov
DV,
Kyrpychova
L,
Martinek
P,
et al
ALK gene fusions in epithelioid fibrous histiocytoma: a study of 14 cases, with new histopathological findings
.
Am J Dermatopathol
.
2018
;
40
(11)
:
805
814
.

Author notes

This work was supported by the Seoul National University Cancer Research Institute Research Program (grant No. 0431-20190014 [to Jeon]) funded by the Seoul National University, Republic of Korea.

Supplemental digital content is available for this article at https://meridian.allenpress.com/aplm in the December 2022 table of contents.

The authors have no relevant financial interest in the products or companies described in this article.

Jeon and Lee contributed equally to this work.

Supplementary data