A Case Report of RRBP1-ALK Fusion Gene-Positive Epithelioid Inflammatory Myofibroblastic Sarcoma With Collagenous Stroma

DOI: https://doi.org/10.21203/rs.3.rs-801086/v1

Abstract

Background: Epithelioid inflammatory myofibroblastic sarcoma (EIMS) is a rare variant of inflammatory myofibroblastic tumors with distinct morphological features and malignant clinical behavior. ALK is positive in all EIMS cases, the RANBP2-ALK fusion gene has been detected in most EIMSs, but several RRBP1-ALK and EML4-ALK fusion genes have also recently been reported.

Case presentation: Here, we report a case of RRBP1-ALK fusion-positive EIMS in the colon. The patient was a 26-year-old man. A pelvic mass was found in a routine physical examination and was completely resected. Microscopically, the tumor cells were rounded and epithelioid in shape. Diffuse collagenous stroma and focal inflammatory infiltration were present. ALK was positive, with a pattern of cytoplasmic staining with perinuclear accentuation, and the RRBP1-ALK fusion gene was detected by targeted next generation sequencing. The patient began to take crizotinib from October 2019 to July 2020, and the patient is presently in good condition. No synchronous or metachronous metastases was found.

Conclusions: EIMSs with different ALK fusion partner genes have unique staining patterns, which may provide clues for diagnosing EIMS. EIMS cases with RRBP1-ALK fusion gene and with diffuse collagenous stroma may have a good prognosis.

Background

Based on evidence to date, IMT is listed in the WHO classification as an intermediate-grade tumor that is prone to local recurrence but of low metastatic risk[1]. However, several studies have shown that some IMTs are morphologically atypical and clinically malignant. Thus, epithelioid inflammatory myofibroblastic sarcoma (EIMS) has recently emerged as a histologically distinctive and clinically aggressive variant, as reported by Marino-Enriquez et al. in 2011. EIMS has a poor prognosis and is associated with the rapid development of local recurrence and metastasis. EIMS exhibits predominantly large round or epithelioid cells with vesicular nuclei and prominent nucleoli, with a minor spindle cell component. The cells are distributed in sheets or clusters within a stroma that is often myxoid[2]. The RANBP2-ALK fusion gene has been detected in most EIMSs, but several RRBP1-ALK and EML4-ALK fusion genes have also recently been reported[36].

Case Presentation

The patient was a 26-year-old man. A pelvic mass was found in a routine physical examination at a local hospital in January 2019, and he had no obvious symptoms. The patient came to our hospital in February 2019, and CT showed an irregular solid nodule on the left side of the pelvis, with a maximum cross-section of 7 × 3.6 cm (Fig. 1a). Although the internal reinforcement was uneven, the boundaries of the tumor were clear. The tumor was located close to the intestinal wall. The radiologist concluded that it might be a stromal tumor but that a source of mesenchymal tissues could not be excluded. Pelvic mass resection was performed, and the pathological result was epithelioid inflammatory myofibrosarcoma with RRBP1-ALK gene fusion (indicating poor prognosis; close follow-up was recommended). The patient began taking crizotinib after the surgery and is currently in good condition.

The tumor was located on the serosal side of the intestinal wall and invaded muscular layer (Fig. 1b) which was composed of scattered well-circumscribed polygonal and epithelioid tumor cells with distinct nucleoli and an eosinophilic cytoplasm; some of the large cells were fat and fusiform shape (Fig. 3b). The stroma of the tumor was predominantly collagenous and locally myxoid, with inflammatory cell infiltration that mainly consisted of lymphocytes and plasma cells (Fig. 2). These characteristics were different from those of typical myxoid stroma with neutrophil infiltration in most EIMSs described thus far. Mitotic activity did not exceed 1 figure per 10 high-power fields. Necrosis was not identified.

Immunohistochemical analysis revealed the tumor cells to be positive for vimentin, desmin, SMA (focal), and AE 1/AE 3 (focal) and negative for EMA, S-100, CD117, DOG-1, F8, CD31, CD21 and CD35(Fig. 5). In addition, the tumor cells were positive for ALK, exhibiting cytoplasmic stanning with perinuclear accentuation (Fig. 4), and RRBP1-ALK fusion was detected by targeted next generation sequencing (Fig. 6).

Discussion

In 2013, the 4th edition of the World Health Organization (WHO) classification of soft tissue tumors described EIMS as an IMT variant with RANBP2-ALK gene rearrangement and seemed to portend more aggressive clinical behavior, nonetheless, most conventional IMTs have a relatively indolent clinical course with a low rate of recurrence and metastasis[7]. And the latest edition of WHO in 2020 has clearly proposed that EIMS is classified as an aggressive IMT subtype of IMT, and it also mentioned that ALK immunostaining pattern varied depending on the ALK fusion partners.

To date, 40 cases of EIMS arising at any site have been documented in the English literature (Table 1); 30 affected patients were female, and 10 were male, ranging from 7 mo to 76 y in age (mean age of 34.6 y). All tumors arose within the abdomen, with the exceptions of 3 cases in the thorax (2 in the lung and 1 in the pleura) and 2 in the pelvis (1 in the rectum and 1 in the ovary). The EIMSs range in size from 4.3 to 26 cm (mean size of 12.91 cm).

Table 1

Clinicopathological features of EIMSs described in previous reports

Case

Fusion type

Age/Sex

Site

Size (cm)

Treatment

Recurrence

Metastasis

Follow up

Source

1

RANBP2-ALK

7y/M

Abdominal cavity

NA

SE, CT

Yes (5w, 5 m)

No

NED (5 m)

Ma et al. [12]

2

RANBP2-ALK

7 m/M

Mesentery and omentum

11

SE

Yes (5w, 5 m)

No

AWD (8 m)

 

3

RANBP2-ALK

2y/M

Abdominal cavity

10

SE

No

No

NED (36 m)

Patel et al. [13]

4

RANBP2-ALK

34y/M

Liver

8

SE

Yes (5 m)

No

DOD (5.5 m)

Chen et al. [14]

5

RANBP2-ALK

44y/M

Omentum

NA

SE, CT, ALKi

Yes (5 m)

Liver (12 m)

AWD (39 m)

Butrynski et al. [9]

6

RANBP2-ALK

41y/M

Omentum

26

SE, CT, ALKi

Yes (12 m)

Liver (12 m)

NED (40 m)

Marino-Enriquez et al. [4]

7

RANBP2-ALK

59y/M

Mesentery of small bowel

15

SE, CT

Yes

No

DOD (12 m)

 

8

RANBP2-ALK

6y/M

Omentum

10.5

SE, CT

Yes

No

AWD (3 m)

 

9

RANBP2-ALK

28y/M

Mesentery of small bowel

NA

NA

NA

NA

NA

 

10

RANBP2-ALK

63y/M

Mesentery of small bowel

25

SE, CT

Yes

No

DOD (3 m)

 

11

RANBP2-ALK

42y/M

Intra-abdominal

NA

SE, CT

Yes

No

AWD (13 m)

 

12

RANBP2-ALK

7 m/M

Peritoneum

10

SE, CT, RT

Yes

No

DOD (36 m)

 

13

RANBP2-ALK

40y/M

Peritoneum

8

SE, CT, RT

Yes

Lung, liver, lymph node

NA

 

14

RANBP2-ALK

31y/M

Mesentery of small bowel

17.5

SE, CT

Yes

No

DOD (11 m)

 

15

RANBP2-ALK

6y/M

Omentum and mesentery

14

SE

NA

NA

NA

 

16

RANBP2-ALK

39y/M

Mesentery of small bowel

15

SE

NA

NA

NA

 

17

RANBP2-ALK

57y/M

Pleura or chest wall

NA

ALKi

NA

NA

NA

Kozu et al.[15]

18

RANBP2-ALK

19y/F

Mesentery of small bowel

19

SE

Yes(9w)

No

DOD (12w)

Li et al. [16]

19

RANBP2-ALK

39y/M

Mesentery of colon

15

SE, CT

Yes (4 m)

No

AWD (12 m)

 

20

RANBP2-ALK

22y/M

Mesentery of small bowel

6

SE, CT, ALKi

Yes (3 m, 4 m)

No

AWD (14 m)

Kimbara et al. [17]

21

RANBP2-ALK

37/F

Rectum

5

SE

No (8 m)

No

NED (8 m)

Lin Yu et al.[4]

22

RANBP2-ALK

55/M

Mesentery of ileum

11

SE, CT

Yes (2 m)

No

NED (10 m)

 

23

RANBP2-ALK

22/M

Mesentery of colon

20

SE, ALKi

Yes (2 m)

No

AWD (14 m)

 

24

RANBP2-ALK

58/F

Omentum

5.5

SE, CT

Yes (2 m)

No

DOD (8 m)

 

25

RANBP2-ALK

15/F

Transverse colon

12

SE

No (7 m)

No

NED (7 m)

 

26

RANBP2-ALK

42/M

Abdominal cavity

NA

ALKi

Yes (8 m)

NA

AWD (40 m)

Jen-Chieh Lee et al.[18]

27

RANBP2-ALK

34/M

Liver

NA

NA

Yes

NA

DOD (5 m)

 

28

RANBP2-ALK

76/F

Abdominal cavity

NA

NA

Yes

NA

DOD (4 m)

 

29

RANBP2-ALK

62/M

Omentum

25

SE, CT

NA

NA

DOD (2 m)

Jen-Chieh Lee et al.[19]

30

RANBP2-ALK

30/M

Abdominal cavity

10

SE

Yes (4 m)

NA

DOD (8 m)

 

31

RANBP2-ALK

16/F

Lung

8

SE, CT, ALKi

Yes (1 m)

NA

AWD (33 m)

 

32

RANBP2-ALK

22/M

Mesentery of colon

10.42

SE, ALKi

NO

NO

AWD (16 m)

Liu, Q.et al. [20]

33

RANBP2-ALK

47/F

Mesentery of colon

7.5

SE, CT

Yes (2 m)

liver, lungs

DOD (6 m)

Wu, H.et al.[21]

34

RANBP2-ALK

15/F

Ovary

NA

SE, CT, ALKi

Yes

rectosigmoid colon, uterine serosa,

DOD(2y)

Fang, H.et al.[22]

35

RANBP2-ALK

35/F

Stomach

NA

SE

No

No

NED (11m)

Xu, P.et al. [23]

36

RRBP1-ALK

62/M

Abdominal cavity

NA

NA

NA

NA

DOD (2m)

ZLee, J. C. et al[3]

37

RRBP1-ALK

26/M

Abdominal cavity

NA

NA

Yes(7m,16m)

intra-abdominal dissemination

AWD (16m)

 

38

RRBP1-ALK

39/F

Abdominal cavity

NA

NA

Yes(36m)

intra-abdominal and pleural dissemination

DOD(36m)

 

39

EML4-ALK

45/M

Abdominal cavity

20

NA

Yes(1m)

Liver,spleen, small intestine, right pleural cavity

DOD(6w)

.Jiang, Q., et al[5]

40

EML4-ALK

67/M

Lung

4.3

NA

NA

Brain, liver

DOD (57m)

Sokai, A., et al. [6]

ALKi ALK inhibitor, AWD alive with disease, CT chemotherapy, DOD dead of disease, NA not available, NED no evidence of disease, RT radiation therapy, SE surgical excision

As cases of EIMS are rare, the diagnosis should only be made based on strict histological and clinical manifestations. EIMS has distinctive morphological features: the tumor is typically characterized by loosely arrayed, round or epithelioid neoplastic cells with vesicular nuclei, prominent large nucleoli and an amphophilic to eosinophilic cytoplasm distributed in a widespread myxoid stroma. The striking feature of EIMS is the presence of obvious inflammatory infiltrates frequently composed of neutrophils[4]; however, this was not found in our case, with the infiltrate being mostly composed of lymphocytes and plasma cells. Furthermore, the stroma in our case was predominantly collagenous and locally myxoid, and some of the epithelial tumor cells with a spindle-like morphology may be squeezed by the dense collagen fibers in the stroma, which is different from the typically round epithelioid tumor cells in loosely myxoid stroma. Marino-Enriquez et al. has reported 2 cases with predominantly collagenous stroma and 2 cases with mixed collagenous and myxoid stroma[2], however, there is a lack of detailed description of morphology and prognosis.

Immunohistochemically, ALK is positive in all EIMSs. Desmin expression is diffuse and strong in almost all cases. In addition, the tumor displays variable expression of CD30, SMA and CK[4]. RANBP2-ALK is the most common fusion gene involved, but two other fusion gene types have recently been, including RRBP1-ALK and EML4-ALK. When ALK fuses with different partner genes, unique stanning patterns arise. For example, tumors with RANBP2-ALK show a distinctive nuclear membranous localization, owing to the roles of RANBP2 in the nuclear pore complex. All 3 cases of RRBP1-ALK fusion specifically displayed cytoplasmic staining with perinuclear accentuation by ALK immunohistochemistry, as RRBP1 is a coiled-coil protein that functions in interactions between ribosomes and the endoplasmic reticulum and in microtubule binding[3]. Cases with EML4-ALK show cytoplasmic staining under the membrane, consistent with the distribution of EML4 in the cell[5]; Richards MW reported that the EML4-ALK fusion protein localizes in the cytoplasm and is possibly associated with microtubules[8]. The unique stanning patterns of different fusion partner genes can be clues for diagnosing EIMS.

Three patients (21, 25 and 35 in Table 1) with the RANBP2-ALK fusion gene experienced no metastasis or recurrence and lived with no evidence of disease as of the follow-up date, which was inconsistent with the malignant clinical features of EIMS in previous articles. Therefore, it is possible that the RANBP2-ALK fusion gene was be involved in conventional IMT, as EML4 was considered a typical partner gene in conventional IMT until it was first confirmed in EIMS in 2017.

EIMS appears to have a poor prognosis with a short survival time after surgical treatment and following chemotherapy and/or radiotherapy; it is also prone to relapse and metastasis. The effectiveness of alternative treatment modalities, such as radiotherapy, chemotherapy, and steroids, is uncertain. Butrynski reported a case of IMT in a patient carrying the RANBP2-ALK fusion gene who was treated with the ALK inhibitor crizotinib after surgical tumor resection, with no evidence of recurrence after 19 months[9]. Kurihara-Hosokawa and Fujiya also recently presented a patient with recurrent EIMS who continued to be alive with disease at 14 months after surgical treatment and administration of crizotinib[10, 11]. Therefore, some researchers have suggested that surgery and the ALK inhibitor crizotinib may become the standard treatment for EIMS positive for the RANBP2-ALK fusion gene [21]. The current case was also treated with the ALK inhibitor crizotinib after tumor resection, and he at present shows no evidence of recurrence and metastasis.

Conclusion

In conclusion, we encountered an unusual case of RRBP1-ALK fusion gene-positive colon EIMS with a collagenous stroma. To the best of our knowledge, this may be the fourth case of EIMS involving such a fusion type. EIMS with different ALK partner fusion genes exhibit unique staining patterns, which may be clues for diagnosing EIMS. ALK inhibitors are a promising treatment for this aggressive tumor.

Abbreviations

EIMS: Epithelioid inflammatory myofibroblastic sarcoma; IMT: inflammatory myofibroblastic tumors; ALK: Anaplastic lymphoma kinase; WHO: World Health Organization

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Written informed consent for the publication of this article and any accompanying images was obtained from the family members of the patient.

Availability of data and materials

As a case report, all data generated or analyzed are included in this article.

Competing interests

The authors declare that they have no competing interests.

Funding

This project was supported by the CAMS Innovation Fund for Medical Sciences (CIFMS) (No. 2016-I2M-3-001).

Authors' contributions

NC searched the literatures and wrote the majority of the paper. LYX provided the interesting case that we reported, as well as guidance and editing throughout the writing process. All authors have read and approved the final manuscript.

Acknowledgements

None.

References

  1. Coffin CM, Humphrey PA, Dehner LP. Extrapulmonary inflammatory myofibroblastic tumor: a clinical and pathological survey. Semin Diagn Pathol. 1998;15:85–101.
  2. Marino-Enriquez A, Wang WL, Roy A, Lopez-Terrada D, Lazar AJ, Fletcher CD, et al. Epithelioid inflammatory myofibroblastic sarcoma: An aggressive intra-abdominal variant of inflammatory myofibroblastic tumor with nuclear membrane or perinuclear ALK. Am J Surg Pathol. 2011;35:135–44.
  3. ZLee JC, Li CF, Huang HY, Zhu MJ, Marino-Enriquez A, Lee CT, et al. ALK oncoproteins in atypical inflammatory myofibroblastic tumours: novel RRBP1-ALK fusions in epithelioid inflammatory myofibroblastic sarcoma. J Pathol. 2017;241:316–23.
  4. 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;11:67.
  5. Jiang Q, Tong HX, Hou YY, Zhang Y, Li JL, Zhou YH, et al. Identification of EML4-ALK as an alternative fusion gene in epithelioid inflammatory myofibroblastic sarcoma. Orphanet J Rare Dis. 2017;12:97.
  6. Sokai A, Enaka M, Sokai R, Mori S, Mori S, Gunji M, et al. Pulmonary inflammatory myofibroblastic tumor harboring EML4-ALK fusion gene. Jpn J Clin Oncol. 2014;44:93–6.
  7. Fu X, Jiang J, Tian XY, Li Z. Pulmonary epithelioid inflammatory myofibroblastic sarcoma with multiple bone metastases: case report and review of literature. Diagn Pathol. 2015;10:106.
  8. Richards MW, O'Regan L, Roth D, Montgomery JM, Straube A, Fry AM, et al. Microtubule association of EML proteins and the EML4-ALK variant 3 oncoprotein require an N-terminal trimerization domain. Biochem J. 2015;467:529–36.
  9. Butrynski JE, D'Adamo DR, Hornick JL, Dal Cin P, Antonescu CR, Jhanwar SC, et al. Crizotinib in ALK-rearranged inflammatory myofibroblastic tumor. N Engl J Med. 2010;363:1727–33.
  10. Fujiya M, Kohgo Y. ALK inhibition for the treatment of refractory epithelioid inflammatory myofibroblastic sarcoma. Intern Med (Tokyo Japan). 2014;53:2177–8.
  11. Kurihara-Hosokawa K, Kawasaki I, Tamai A, Yoshida Y, Yakushiji Y, Ueno H, et al. Epithelioid inflammatory myofibroblastic sarcoma responsive to surgery and an ALK inhibitor in a patient with panhypopituitarism. Intern Med (Tokyo Japan). 2014;53:2211–4.
  12. Ma Z, Hill DA, Collins MH, Morris SW, Sumegi J, Zhou M, et al. Fusion of ALK to the Ran-binding protein 2 (RANBP2) gene in inflammatory myofibroblastic tumor. Genes Chromosom Cancer. 2003;37:98–105.
  13. Patel AS, Murphy KM, Hawkins AL, Cohen JS, Long PP, Perlman EJ, et al. RANBP2 and CLTC are involved in ALK rearrangements in inflammatory myofibroblastic tumors. Cancer Genet Cytogenet. 2007;176:107–14.
  14. Chen ST, Lee JC. An inflammatory myofibroblastic tumor in liver with ALK and RANBP2 gene rearrangement: combination of distinct morphologic, immunohistochemical, and genetic features. Hum Pathol. 2008;39:1854–8.
  15. Kozu Y, Isaka M, Ohde Y, Takeuchi K, Nakajima T. Epithelioid inflammatory myofibroblastic sarcoma arising in the pleural cavity. General thoracic cardiovascular surgery. 2014;62:191–4.
  16. Li J, Yin WH, Takeuchi K, Guan H, Huang YH, Chan JK. Inflammatory myofibroblastic tumor with RANBP2 and ALK gene rearrangement: a report of two cases and literature review. Diagn Pathol. 2013;8:147.
  17. Kimbara S, Takeda K, Fukushima H, Inoue T, Okada H, Shibata Y, et al. A case report of epithelioid inflammatory myofibroblastic sarcoma with RANBP2-ALK fusion gene treated with the ALK inhibitor, crizotinib. Jpn J Clin Oncol. 2014;44:868–71.
  18. ALK oncoproteins in. atypical inflammatory myofibroblastic tumours: novel RRBP1-ALK fusions in epithelioid inflammatory myofibroblastic sarcoma.
  19. Lee JC, Wu JM, Liau JY, Huang HY, Lo CY, Jan IS, et al. Cytopathologic features of epithelioid inflammatory myofibroblastic sarcoma with correlation of histopathology, immunohistochemistry, and molecular cytogenetic analysis. Cancer Cytopathol. 2015;123:495–504.
  20. Liu Q, Kan Y, Zhao Y, He H, Kong L. Epithelioid inflammatory myofibroblastic sarcoma treated with ALK inhibitor: a case report and review of literature. Int J Clin Exp Pathol. 2015;8:15328–32.
  21. Wu H, Meng YH, Lu P, Ning HY, Hong L, Kang XL, et al. Epithelioid inflammatory myofibroblastic sarcoma in abdominal cavity: a case report and review of literature. Int J Clin Exp Pathol. 2015;8:4213–9.
  22. Fang H, Langstraat CL, Visscher DW, Folpe AL, Schoolmeester JK. Epithelioid Inflammatory Myofibroblastic Sarcoma of the Ovary With RANB2-ALK Fusion: Report of a Case. International journal of gynecological pathology: official journal of the International Society of Gynecological Pathologists. 2018;37:468–72.
  23. Xu P, Shen P, Jin Y, Wang L, Wu W. Epithelioid inflammatory myofibroblastic sarcoma of stomach: diagnostic pitfalls and clinical characteristics. Int J Clin Exp Pathol. 2019;12:1738–44.