Content » Vol 91, Issue 4

Letter to the Editor

No Evidence for ErbB4 Gene Amplification in Malignant Melanoma

Gero Brockhoff1, Marietta Bock1, Susanne Gantner2 and Christian Hafner2

Departments of 1Gynecology and Obstetrics and 2Dermatology, University of Regensburg, Franz-Josef-Strauss-Allee 11, DE-93053 Regensburg, Germany. E-mail: christian.hafner@klinik.uni-regensburg.de

Accepted January 10, 2011.

Malignant melanoma (MM) represents the most severe skin cancer (1). Somatic ErbB4 gene mutations have been identified in 19% of individuals with MM (2). These mutations result in hyperactivation of the ErbB4 receptor (2, 3). This finding indicates a pivotal role of the ErbB4 receptor tyrosine kinase (RTK) in the tumourigenesis of MM. However, oncogenic receptor activation of ErbB RTKs in human cancer is not limited to activating point mutations. Amplifications of ErbB receptor genes (ErbB1–4), resulting in receptor overexpression and ligand-independent activation have been observed in a variety of human malignancies (4). Based on this rationale we analysed potential ErbB4 gene amplification in 28 melanoma samples using fluorescence-in-situ-hybridization (FISH).

Patients and Methods

In total, 28 MM samples were analysed (17 superficial spreading melanomas, 9 nodular MMs, 2 secondary nodular MMs on the basis of a superficial spreading melanoma). The melanomas derived from 11 male and 17 female patients (mean age 60 years, age range 27–87 years). The study was performed according to the guidelines of the local ethics committee and the Declaration of Helsinki.

Sections 4 µm thick were prepared from formalin-fixed paraffin-embedded melanoma samples, as described in detail elsewhere (5, 6). In brief, for each tumour a representative tumour section was selected from a haematoxylin and eosin-stained section of the donor block. Core cylinders with a diameter of 1.5 mm each were punched from this area and deposited into a recipient paraffin block. Tissue microarray (TMA) sections were mounted on charged slides (SuperFrost™Plus; Menzel GmbH, Braunschweig, Germany). Haematoxylin and eosin stained TMA sections were used for reference histology. FISH was performed with the use of directly labelled ZytoLight SPEC HER4/2q11 dual-colour probe (ZytoVision Ltd, Bremerhaven, Germany) (Fig. 1). After probe hybridization nuclei were counterstained with anti-fading 4’,6-diamidino-2-phenylindole (DAPI) Vectashield (Vector Laboratories, Burlingame, CA, USA) and analysed by epifluorescence microscopy using the AxioImager-Z1 (Zeiss, Göttingen, Germany). Hybridization signals of 25 nuclei were manually counted on single cell basis by two independent observers.

3445fig1.tif

Fig. 1. Example images showing human ErbB4 (HER4) centromere (CEN2) fluorescence-in-situ-hybridization signals, (a) without, and (b) with low degree of chromosome 2 polysomy, respectively; (red spots = centromere signals; green spots = gene signals).

RESULTS and DISCUSSION

Dual-colour FISH, revealed no evidence for ErbB4 gene amplification (Table I). The highest ErbB4 gene/centromer-2 ratio found was 1.17. Three cases showed a low degree of chromosome-2 polysomy (nos. 5, 6 and 20). Only one specimen appeared suspicious, showing a low degree of chromosome-2 loss (no. 19). Overall we did not observe any significant alterations in ErbB4 gene copy number. Although the quantity of tissues investigated in this study is limited to 28 samples, the results indicate that, in contrast to somatic mutations, ErbB4 gene amplifications do not play a major role in MM.

Table I. ErbB4 FISH analysis of melanoma

Patient data

HER4 FISH data

No.

Sex

Age (years)

Subtype

Thickness (mm)

Localization

ErbB4 gene signals

CEN2 signals

ErbB4/CEN2 ratio

CEN2 signals ≥ 3

CEN2 signals = 2

CEN2 signals = 1

1

F

54

SSM

1.4

Trunk left

47

48

0.98

0

23

2

2

F

75

SSM

0.7

Left arm

51

49

1.04

1

22

2

3

F

68

SSM

0.7

Head right

55

55

1.00

6

16

3

4

F

76

SSM

0.8

Left lower arm

52

49

1.06

1

21

3

5a

M

70

NMM

3.0

Right lower leg

73

71

1.03

16

7

2

6a

F

84

NMM

2.4

Head left

59

66

0.89

13

11

1

7

M

71

NMM

2.0

Left lower arm

50

54

0.93

9

10

6

8

F

73

NMM

1.4

Nose

48

49

0.98

0

24

1

9

F

46

SSM

0.7

Left thigh

60

59

1.02

8

16

1

10

M

56

SSM

0.7

Right thigh

50

52

0.93

0

24

1

11

F

63

SSM

0.5

Back

50

54

0.93

5

18

2

12

F

87

NMM

6.0

Back left

49

48

1.02

0

24

1

13

M

45

SSM

0.7

Abdomen

52

51

1.02

1

22

2

14

M

27

SSM

1.1

Left lower leg

45

41

1.10

0

16

9

15

M

48

NMM

3.0

Back

51

53

0.96

4

20

1

16

F

65

SSM

0.9

Right elbow

45

50

0.90

4

16

5

17

F

45

NMM

12.0

Right elbow

51

50

1.02

3

19

3

18

M

54

sNMM

7.0

Back left

39

44

0.89

5

12

3

19b

F

34

SSM

0.6

Right thigh

41

35

1.17

0

10

15

20a

F

81

sNMM

4.0

Left arm

52

58

0.90

11

11

3

21

M

48

SSM

0.5

Back left

51

52

0.98

6

15

4

22

F

62

SSM

0.5

Right thigh

53

56

0.95

6

17

2

23

F

48

SSM

0.5

Abdomen

52

51

1.02

2

22

1

24

M

51

SSM

0.8

Left ear

47

48

0.98

0

23

2

25

F

83

SSM

1.9

Right back

48

46

1.04

1

19

5

26

M

74

NMM

2.2

Left arm

54

55

0.98

6

18

1

27

F

30

SSM

1.5

Left neck

50

48

1.04

3

17

5

28

M

60

NMM

2.7

Right knee

49

49

1.00

1

22

2

aLow polysomic tissue samples; blow degree of chromosome-2 loss.

SSM: superficial spreading melanoma; NMM: nodular malignant melanoma; sNMM: secondary NMM on the basis of a SSM; CEN: centromere.

A number of somatic mutations scattered throughout the total coding region of the ErbB4 receptor gene have been identified in 19% of patients with MM (2). All functionally analysed mutations, even those not located in the kinase encoding region, cause constitutive ligand-independent receptor activation; a finding that emphasizes the role of ErbB4 as a pivotal cancer gene in MM (7). In a screen of 19 phosphotyrosine kinases, the ErbB4 gene was found to be the most highly mutated gene, and the frequency of non-synonymous mutations has been found to be significantly higher than predicted for unselected passenger mutations. Therefore, one might conclude that the ErbB4 gene represents a hot-spot for a variety of mutations in melanomas, which in turn might act synergistically (2).

The contribution of wild-type and mutated ErbB4 to tumourigenesis and progression of MMs, however, has not been elucidated in detail. Tvorogov et al. (8) demonstrated that ErbB4 kinase malfunction does not necessarily result in loss of function of the receptor protein. A kinase-defective ErbB4 receptor might still be able to heterodimerize (for example with ErbB2) and to trigger intracellular signalling in an ErbB2 kinase-dependent manner.

Hyperactivated ErbB4 receptor does, however, not necessarily lead to enhanced cell proliferation or increased anchorage-independent growth, as shown by Pricket et al., (2) who transfected NIH-3T3 cells with mutated ErbB4. In fact, both oncogenic and tumour-suppressing signalling capacity has been attributed to differentially expressed ErbB4 isoforms, which result from alternative ErbB4 mRNA splicing (9–11). Four ErbB4 isoforms, which have been shown to be differentially expressed in malignancies of, for example, the breast (12) and the bladder (13), have not yet been analysed in MM. Even though ErbB4 receptor activity might not directly be affected by ErbB4 gene mutations, some intronic mutations might impinge on splicing of the ErbB transcript and thereby result in either pronounced survival or cell death promoting ErbB4 signalling (11).

In summary, ErbB4 gene amplification does not play a major role in MM. Nevertheless, ErbB4 represents an interesting drug target in MM, as this receptor has been shown to confer oncogenic properties by alternative genetic alterations (3, 14, 15). The role of both mutated and wild-type ErbB4, as well as the importance of differentially expressed ErbB isoforms in tumourigenesis and progression of MM still needs to be addressed.

ACKNOWLEDGEMENTS

The authors thank Dr Sven Hauke (ZytoVision GmbH, Bremerhaven, Germany) for kindly providing the dual colour Zytolight HER4/CEN2 FISH probe. We thank Eva Herschberger for excellent technical support.

The authors declare no conflicts of interest.

REFERENCES

  • Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med 2004; 351: 998–1012.
  • Prickett TD, Agrawal NS, Wei X, Yates KE, Lin JC, Wunderlich JR, et al. Analysis of the tyrosine kinome in melanoma reveals recurrent mutations in ERBB4. Nat Genet 2009; 41: 1127–1132.
  • Kurppa K, Elenius K. Mutated ERBB4: a novel drug target in metastatic melanoma? Pigment Cell Melanoma Res 2009; 22: 708–710.
  • Kamath S, Buolamwini JK. Targeting EGFR and HER-2 receptor tyrosine kinases for cancer drug discovery and development. Med Res Rev 2006; 26: 569–594.
  • Sassen A, Diermeier-Daucher S, Sieben M, Ortmann O, Hofstaedter F, Schwarz S, et al. Presence of HER4 associates with increased sensitivity to Herceptin in patients with metastatic breast cancer. Breast Cancer Res 2009; 11: R50.
  • Sassen A, Rochon J, Wild P, Hartmann A, Hofstaedter F, Schwarz S, et al. Cytogenetic analysis of HER1/EGFR, HER2, HER3 and HER4 in 278 breast cancer patients. Breast Cancer Res 2008; 10: R2.
  • Futreal PA, Coin L, Marshall M, Down T, Hubbard T, Wooster R, et al. A census of human cancer genes. Nat Rev Cancer 2004; 4: 177–183.
  • Tvorogov D, Sundvall M, Kurppa K, Hollmén M, Repo S, Johnson MS, et al. Somatic mutations of ErbB4: selective loss-of-function phenotype affecting signal transduction pathways in cancer. J Biol Chem 2009; 284: 5582–5591.
  • Määttä JA, Sundvall M, Junttila TT, Peri L, Laine VJ, Isola J, et al. Proteolytic cleavage and phosphorylation of a tumor-associated ErbB4 isoform promote ligand-independent survival and cancer cell growth. Mol Biol Cell 2006; 17: 67–79.
  • Sundvall M, Peri L, Määttä JA, Tvorogov D, Paatero I, Savisalo M, et al. Differential nuclear localization and kinase activity of alternative ErbB4 intracellular domains. Oncogene 2007; 26: 6905–6914.
  • Sundvall M, Veikkolainen V, Kurppa K, Salah Z, Tvorogov D, van Zoelen EJ, et al. Cell death or survival promoted by alternative isoforms of ErbB4. Mol Biol Cell 2010; 21: 4275–4286.
  • Junttila TT, Sundvall M, Lundin M, Lundin J, Tanner M, Härkönen P, et al. Cleavable ErbB4 isoform in estrogen receptor-regulated growth of breast cancer cells. Cancer Res 2005; 65: 1384–1393.
  • Junttila TT, Laato M, Vahlberg T, Söderström KO, Visakorpi T, Isola J, et al. Identification of patients with transitional cell carcinoma of the bladder overexpressing ErbB2, ErbB3, or specific ErbB4 isoforms: real-time reverse transcription-PCR analysis in estimation of ErbB receptor status from cancer patients. Clin Cancer Res 2003; 9: 5346–5357.
  • Hollmén M, Määttä JA, Bald L, Sliwkowski MX, Elenius K. Suppression of breast cancer cell growth by a monoclonal antibody targeting cleavable ErbB4 isoforms. Oncogene 2009; 28: 1309–1319.
  • Rudloff U, Samuels Y. A growing family: adding mutated Erbb4 as a novel cancer target. Cell Cycle 2010; 9: 1487–1503.