Skip to main content
British Journal of Cancer logoLink to British Journal of Cancer
. 2003 May 13;88(10):1587–1591. doi: 10.1038/sj.bjc.6600943

Agreement between chromogenic in situ hybridisation (CISH) and FISH in the determination of HER2 status in breast cancer

L Arnould 1,*, Y Denoux 2, G MacGrogan 3, F Penault-Llorca 4, M Fiche 5, I Treilleux 6, M C Mathieu 7, A Vincent-Salomon 8, M O Vilain 9, J Couturier 8
PMCID: PMC2377115  PMID: 12771927

Abstract

Determination of the HER2/neu (HER2) status in breast carcinoma has become necessary for the selection of breast cancer patients for trastuzumab therapy. Amplification of the gene analysed by fluorescence in situ hybridisation (FISH) or overexpression of the protein determined by immunohistochemistry (IHC) are the two major methods to establish this status. A strong correlation has been previously demonstrated between these two methods. However, FISH is not always feasible in routine practice and weakly positive IHC tumours (2+) do not always correspond to a gene amplification. Our study was performed in order to evaluate the contribution of chromogenic in situ hybridisation (CISH), which enables detection of the gene copies through an immunoperoxidase reaction. CISH was performed in 79 breast carcinomas for which the HER2 status was previously determined by IHC and FISH. The results of IHC, FISH and CISH were compared for each tumour. CISH procedures were successful in 95% of our cases. Whatever the IHC results, we found a very good concordance (96%) between CISH and FISH. Our study confirms that CISH may be an alternative to FISH for the determination of the gene amplification status in 2+ tumours. Our results allow us to think that, in many laboratories, CISH may also be an excellent method to calibrate the IHC procedures or, as a quality control test, to check regularly that the IHC signal is in agreement with the gene status.

Keywords: breast cancer, HER2, gene amplification, CISH, FISH, immunohistochemistry


Determination of HER2 status has now become of major clinical importance with the advent of anti-HER2 therapy, the recombinant humanised anti-p185Her-2/neu antibody trastuzumab (Herceptin®) (Pegram et al, 1998; Cobleigh et al, 1999; Slamon et al, 2001).

Immunohistochemistry (IHC) is expected to be the best method for the determination of HER2 status, as IHC assesses the level of HER2 overexpression, which is the target of Herceptin® therapy. Moreover, the patient's selection for Herceptin® therapy is mainly based on IHC because previous studies demonstrated a good correlation between IHC results and gene status, as determined by fluorescence in situ hybridisation (FISH) (Pegram et al, 1998; Jacobs et al, 1999,2000; Couturier et al, 2000; Jimenez et al, 2000; Lebeau et al, 2001; Lehr et al, 2001). However, the HER2-IHC detection was criticised because of a lack of interlaboratory reproducibility and, furthermore, Herceptest®, a standardised IHC method, was shown to be a method with excessive sensitivity when compared to FISH (Persons et al, 1997; Bartlett et al, 2001; Tubbs et al, 2001). Even though HER2 overexpression without gene amplification was reported in 2.9–8.3% of cases (Kallioniemi et al, 1992; Persons et al, 1997; Couturier et al, 2000; Jimenez et al, 2000; Pauletti et al, 2000), discordant results between IHC and FISH were mainly observed for tumours that were scored 2+ by IHC (Persons et al, 1997; Bartlett et al, 2001; Tubbs et al, 2001). For this reason, and particularly in Europe, a confirmation of HER2 gene amplification by FISH became mandatory for a patient's inclusion in a clinical trial using Herceptin®, when the corresponding tumour is scored 2+ by IHC (Hoang et al, 2000; Ridolfi et al, 2000; Diaz, 2001; Tubbs et al, 2001; Vogel et al, 2002).

Some authors found that HER2 status determined by FISH was more reproducible (Press et al, 1994; Persons et al, 1997; Bartlett et al, 2001; Tubbs et al, 2001). Thus, these authors thought that FISH had to be proposed as the only method to select patients for Herceptin®. However, FISH is a long and expensive procedure that requires trained personnel and fluorescence microscopy.

Chromogenic in situ hybridisation (CISH) is a recently introduced technique in which the DNA probe is detected using an immunoperoxidase reaction (Tanner et al, 2000). This method is very close to FISH but does not require the use of fluorescence microscopy. Moreover, FISH signals fade within a few weeks and the FISH results have to be recorded with expensive digital systems. This is not the case for CISH staining. Owing to the similarity with IHC staining, CISH is also easier to interpret for pathologists who are not trained with fluorescence. In one study (Tanner et al, 2000), CISH was demonstrated to be well correlated with FISH.

The aims of our study were to: (a) confirm the good correlation between FISH and CISH in a nonhomogeneous series of breast tumours coming from eight different laboratories using different fixation procedures, (b) analyse this correlation according to the expression of HER2 protein analysed by IHC (c) focus on IHC 2+ cases and analyse in this situation if CISH gives the same information as FISH for the treatment of patients.

MATERIALS AND METHODS

Tumours

A total of 79 tumours were collected from eight French laboratories. Each laboratory selected cases in which IHC and FISH were previously and successfully performed. In order to analyse the discriminating power of CISH in difficult cases, tumours scored as 2+ by IHC were chosen in priority for this study. Owing to differences in the fixative procedure between the laboratories, 47 tumours were fixed in neutral-buffered formalin, 10 in Holland's bouin, and 22 in alcohol–formalin–acetic acid (AFA).

IHC

The monoclonal antibody CB11 (Novocastra, Newcastle, England) was used in 25 cases and the polyclonal antibody A485 (Dako, Glostrup, Denmark) in 44 cases. For all slides, immunostaining was scored according to the Herceptest® scoring system, which is also used in clinical trials (Cobleigh et al, 1999; Slamon et al, 2001). Negativity of normal glands was the prerequisite for interpreting the cases, according to the recommendations of the College of American Pathologists (Fitzgibbons et al, 2000).

FISH

FISH was performed in three different referent laboratories. It was performed on frozen tumour sections in 65 cases and on fixed-paraffin-embedded samples in the 14 remaining cases. FISH experiments were performed according to the protocol given by the supplier (PathVysion kit, Vysis, Downers Grove, IL or Ventana HER2 inform, Tucson, AZ). The centromeric probe of chromosome 17 was included in FISH analyses in 62 cases. In these 62 cases, HER2 amplification was determined as a ratio of HER2 and chromosome 17 centromere signal counts. As in the study previously published (Tanner et al, 2000), ratios <2 were determined as no amplification with FISH (NAF) (Figure 1B), those between 2 and 5 as low-level amplification with FISH (LAF) (Figure 1D) and those >5 as high-level amplification with FISH (HAF) (Figure 1F). In the 14 other cases, without centromeric 17 analysis, like for the CISH analysis in the study previously published (Tanner et al, 2000), HER2 gene was judged as NAF when 1–5 signals were present per nucleus. When 6–10 signals were present in more than 50% of tumour cell nuclei, the tumours were judged as LAF. Finally, tumours having more than 10 signals in more than 50% of the nuclei were judged as HAF.

Figure 1.

Figure 1

(A) CISH, only one or two signals are present in the nucleus of tumour cells (NAC). (B) Same case analysed with FISH (NAF). Pink dots correspond to HER2 probe and green dots correspond to centromere 17 probe. (C) CISH, six signals are present in the nucleus of tumour cells (LAC). (D) Same case analysed with FISH (LAF) with a ratio of HER2 dots/centromere 17 dots=3. (E) CISH, large gene copy clusters are present in the nucleus of tumour cells (HAC). (F) Same case analysed with FISH (HAF).

CISH

CISH experiments were performed according to the protocol given by the supplier (Zymed Inc., South San Francisco, CA, USA). The interpretation of the signal was that used by other authors (Tanner et al, 2000) and was only performed on invasive tumour patterns. HER2 gene was judged as nonamplified with CISH (NAC) when 1–5 signals were present per nucleus (Figure 1A).When 6–10 signals were present in more than 50% of tumour cell nuclei, the tumours were judged as having a low level of amplification with CISH (LAC) (Figure 1C). Finally, the tumours with more than 10 signals or with large gene copy clusters in more than 50% of the nuclei were judged as having a high level of HER2 gene amplification (HAC) (Figure 1E).

RESULTS

Evaluation of IHC staining, FISH and CISH signals were performed in a blinded manner.

IHC

A total of 27 (34%) tumours were defined as 0 or 1+, 29 (37%) tumours were defined as 2+, and the remaining 23 (29%) tumours were defined as 3+ (Table 1).

Table 1. HER2 gene amplification determined by FISH and CISH, according to the overexpression of HER2 protein determined by IHC.

  IHC
  0 or 1+ 2+ 3+
  (N=27) (N=29) (N=23)
FISH 27 NAF 14 NAF 11 LAF 4 HAF 1 NAF 22 HAF
             
CISH 25 NAC align="center"12 NAC 8 LAC 4 HAC 1 NAC 20 HAC
  2 WS 1 LAC 2 HAC     1 NAC
    1 HAC 1 WS     1 WS

NAF=no amplification with FISH; LAF=low level of amplification with FISH; HAF=high level of amplification with FISH; WS=without any signal; NAC=no amplification with CISH; LAC=low level of amplification with CISH; HAC=high level of amplification with CISH.

FISH

As a result of the material chosen for this study, FISH analysis was necessarily successful in all cases. In all, 41 tumours were determined as NAF, 11 tumours as LAF and 26 tumours as HAF (Table 1).

CISH

CISH was successful in 75 of the 79 tumours (94.9%). The four cases without any signal (WS) corresponded to four of the 22 tumours fixed in AFA. In these tumours, despite the use of a great variation of pretreatment procedures, no signal was present in any tumour cell. In all, 39 tumours were determined as NAC, nine as LAC and 27 as HAC (Table 1).

Comparison of CISH and FISH results

Only 75 tumours were available for this comparison (Table 1). When we compared all CISH amplifications (LAC+HAC) to all FISH amplifications (LAF+HAF), an agreement was found in 72 out of 75 (96%) tumours (Table 3). The kappa coefficient (κ) measuring agreement between the methods (0: no agreement, 1: agreement) was 0.97 (P<10−9), and if FISH was chosen as the gold standard, the sensibility of CISH would be close to 97% with a specificity of 95%. In nonproblematic IHC tumours (0, 1+ and 3+), this agreement was found in 46 out of 47 (98%) cases (κ=0.95 (P<10−9), sensibility=95% and specificity=100%). On the other hand, in problematic tumours (2+), this agreement was found in 26 out of 28 (92.8%) cases (κ=0.85 (P<5 × 10−6), sensibility=100% and specificity=85%). When we compared the level of amplification determined with the two methods, an agreement was found in 70 out of 75 (93.3%) with a κ=0.88 (P<10−9).

Table 3. Advantages and disadvantages of FISH and CISH.

FISH CISH
Pro Con
• Possibility of multicolour (HER2 and chromosome 17) • No multicolour detection
• Is the simplest, without any chromogen detection • Needs a chromogenic procedure
• Is currently the standard hybridisation procedure for HER2 • Not yet a standard hybridisation procedure for HER2
   
Con Pro
• Requires a modern and expensive fluorescence microscope • An ordinary microscope is effective
• FISH is not routinely used in pathology, and pathologists are not trained to analyse FISH signals • Pathologists are familiar with IHC signals
• Fluorescence signals can fade within several weeks • Chromogenic reaction is permanent
• Results have to be recorded with an expensive CCD camera • Regular slide storage
• Morphology is not always easy to analyse • Morphology is easier to analyse

Analysis of the cases with polysomy of chromosome 17

Centromeric probes of chromosome 17 were included in FISH analyses in 61 of the 75 cases successfully analysed with CISH. The result of the analysis of this centromere is summarised in Table 2. Using a χ2 test, we found that polysomy was statically (P<0.005) more frequently observed in IHC 2+ tumours (10 out of 16: 62.5%) than in other situations (7 out of 45: 15.55%).

Table 2. Analysis of the cases with or without polysomy of chromosome 17.

  IHC
  0 or 1+ 2+ 3+
  N=24 N=16 N=21
Normal chromosome 17 status N=18 N=6 N=20
FISH 18 HAF 4 NAF 2 LAF   1 NAF 19 HAF
CISH 18 HAC 4 NAC 2 LAC   1 NAC 18 HAC
            1 NAC
             
Chromosome 17 polysomy            
FISH N=6 N=10 N=1
  6 NAF 6 NAF 1 LAF 3 HAF   1 HAF
CISH 6 NAC 5 NAC 1 LAC 3 HAC   1 HAC
    1 LAC        

NAF=no amplification with FISH; LAF=low level of amplification with FISH; HAF=high level of amplification with FISH; WS=without any signal; NAC=no amplification with CISH; LAC=low level of amplification with CISH; HAC=high level of amplification with CISH.

DISCUSSION

Since the FDA approved Herceptin® for the treatment of metastatic breast cancer (Pegram et al, 1998; Cobleigh et al, 1999; Roche and Ingle, 1999; Slamon et al, 2001), and in order to determine whose patients might benefit from this new therapy, there has been a need to evaluate precisely the HER2 status of breast cancer specimens. The determination of this status will also be important to choose the adjuvant strategy if clinical trials including Herceptin® as adjuvant therapy give a positive result (Hortobagyi and Perez, 2001). Moreover, in the future, HER2 status may also help select patients for tyrosine kinase inhibitor therapy (Moasser et al, 2001). Two major methods (IHC and FISH) for the determination of this HER2 status have been developed all around the world but there is no consensus up to now regarding the best methods to determine this status (Thor, 2001). In two recent different clinical trials, poor concordance was found between local and central or reference IHC testing for HER2 (Paik et al, 2002; Roche et al, 2002). This poor concordance being given, the authors of these studies recommended that the HER2 status of patients included in clinical trials should be done in large-volume reference laboratories. These data also suggested an urgent need to improve the quality control programme in laboratories that use IHC testing (Fitzgibbons et al, 2000; Hoang et al, 2000; Ridolfi et al, 2000; Tubbs et al, 2001; Paik et al, 2002; Roche et al, 2002; Vogel et al, 2002). In a previous study, we showed that FISH may be used to obtain successfully a calibration of the in-house IHC technique (Vincent-Salomon et al, 2003).

CISH, a hybridisation procedure using a staining of the probe similar to IHC staining, has been previously proposed as an alternative for FISH (Tanner et al, 2000). When compared with FISH, CISH has been described as having several advantages (Tanner et al, 2000; Zhao et al, 2002). It does not require an expensive fluorescence microscope with multi-band-pass filters, CISH staining is permanent and it does not need to be recorded with an expensive CDD camera. Moreover, morphology is easier analysed on CISH slides, particularly for distinguishing invasive cancer cells and in situ components. Finally, pathologists are more familiar with the IHC signal than with the FISH signal. The advantages and disadvantages of these two hybridisation techniques are summarised in Table 3.

In order to confirm the results of this study and to precise the place of this technique in problematic IHC cases (2+), we performed a study on a group of 79 breast tumours that contained an abnormal percentage (37%) of 2+. Our study was also different from the study published earlier because the breast tissue came from different purveyor laboratories using different fixatives. CISH procedures were successful in 95% of our cases, which is identical to the results observed with FISH on paraffin sections (Lebeau et al, 2001) and very close to the results (98%) published with CISH in an unselected group of tumours (Tanner et al, 2000). It may be noticed that AFA seems to be a less effective fixative procedure for CISH, as CISH was successful in only 18 (82%) of the 22 tumours fixed in AFA and successful in all (100%) of the tumours fixed in neutral-buffered formalin or Holland's bouin. We found a very good concordance between the CISH and FISH. In terms of amplification, there was indeed an agreement between CISH and FISH in 96% of the tumours, which is almost the same as the agreement previously published (93.6%) (Tanner et al, 2000). The agreement was a little higher (98%) for nonproblematic (0, 1+, 3+) IHC cases than for problematic (2+) IHC tumours (93%). Owing to the small number (3%) of 2+ cases in their study, Tanner et al did not notice this small difference. In terms of sensitivity, when we compared the level of amplification estimated by the two methods (no amplification, low level and high level of amplification), we found that the results of CISH analyses were very close to those given by FISH, with an agreement in 93% of the tumours. According to Tanner et al (2000), the discrepancy between CISH and FISH may be because of a lower sensitivity of CISH. However, this explains only one discordant case in our study, which was found to be amplified with FISH but not with CISH. Other discrepancies may also be because of differences in the sample materials or the thickness of the slides. The most difficult situations with CISH are when 6–10 spots are present in tumour cells. Double-colour FISH analyses may give more information, particularly the ratio between HER2 signal and the number of chromosome 17, and may separate the high polysomy of chromosome 17 and the very low level of HER2 amplification. We found that polysomy of chromosome 17 is statically more frequent in IHC 2+ tumours, but only one of our discordant cases could probably be linked to this phenomenon. In routine, these situations are very infrequent and it is not proved that this distinction is relevant in terms of response to Herceptin® therapy. Clinical trials, including a large number of IHC 2+ tumours with a low level of amplification, are needed to confirm that the exact level of HER2 gene amplification is important for the patient's selection for specific therapy. Anyway, double-colour staining CISH procedures, including HER2 and chromosome 17 probes, will soon be available. The results of these new procedures would also have to be compared with the double-colour FISH analysis.

Our study confirms that CISH may be an alternative to FISH for the determination of HER2 gene status, particularly in laboratories that are not equipped or trained from fluorescence analyses. In our opinion, CISH is too expensive and too sophisticated to be an alternative to IHC screening of all the breast tumours. However, because of the good correlation between CISH and FISH, even in ambiguous IHC results, we think that it may be used for the determination of gene amplification status in IHC 2+ tumours. Owing to the poor concordance between HER2 status established in local laboratories in comparison to reference laboratories, we also think that, in many laboratories, CISH may be an excellent method to calibrate IHC procedures or, as a quality control test, to check regularly that the IHC signal is in agreement with gene status.

Acknowledgments

This work was supported in part by the ‘Ligue Bourguignonne Contre le Cancer’. We thank the other members of the GEFPICS (AJ Balaton, JP Bellocq, F Ettore, V Fridman, JP Ghnassia, JM Guinebretière, J Jacquemier, B Lannes, V Le Doussal, C Migeon, P Roger, JC Sabourin, B Sigal-Zafrani, J Simony-Lafontaine, V Veriele, MO Vilain, JJ Voigt, C Voisin-Rigaud.

Footnotes

All authors are members of the Groupe d'Etude des Facteurs de Pronostic Immunohistochimiques dans les Cancers du Sein, Fédération Nationale des Centres de Lutte Contre le Cancer, 101 rue de Tolbiac, Paris, France

References

  1. Bartlett JM, Going JJ, Mallon EA, Watters AD, Reeves JR, Stanton P, Richmond J, Donald B, Ferrier R, Cooke TG (2001) Evaluating HER2 amplification and overexpression in breast cancer. J Pathol 195: 422–428 [DOI] [PubMed] [Google Scholar]
  2. Cobleigh MA, Vogel CL, Tripathy D, Robert NJ, Scholl S, Fehrenbacher L, Wolter JM, Paton V, Shak S, Lieberman G, Slamon DJ (1999) Multinational study of the efficacy and safety of humanized anti-HER2 monoclonal antibody in women who have HER2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease. J Clin Oncol 17: 2639–2648 [DOI] [PubMed] [Google Scholar]
  3. Couturier J, Vincent-Salomon A, Nicolas A, Beuzeboc P, Mouret E, Zafrani B, Sastre-Garau X (2000) Strong correlation between results of fluorescent in situ hybridization and immunohistochemistry for the assessment of the ERBB2 (HER-2/neu) gene status in breast carcinoma. Mod Pathol 13: 1238–1243 [DOI] [PubMed] [Google Scholar]
  4. Diaz NM (2001) Laboratory testing for HER2/neu in breast carcinoma: an evolving strategy to predict response to targeted therapy. Cancer Control 8: 415–418 [DOI] [PubMed] [Google Scholar]
  5. Fitzgibbons PL, Page DL, Weaver D, Thor AD, Allred DC, Clark GM, Ruby SG, O'Malley F, Simpson JF, Connolly JL, Hayes DF, Edge SB, Lichter A, Schnitt SJ (2000) Prognostic factors in breast cancer. College of American Pathologists Consensus Statement 1999. Arch Pathol Lab Med 124: 966–978 [DOI] [PubMed] [Google Scholar]
  6. Hoang MP, Sahin AA, Ordonez NG, Sneige N (2000) HER-2/neu gene amplification compared with HER-2/neu protein overexpression and interobserver reproducibility in invasive breast carcinoma. Am J Clin Pathol 113: 852–859 [DOI] [PubMed] [Google Scholar]
  7. Hortobagyi GN, Perez EA (2001) Integration of trastuzumab into adjuvant systemic therapy of breast cancer: ongoing and planned clinical trials. Semin Oncol 28: 41–46 [DOI] [PubMed] [Google Scholar]
  8. Jacobs TW, Gown AM, Yaziji H, Barnes MJ, Schnitt SJ (1999) Comparison of fluorescence in situ hybridization and immunohistochemistry for the evaluation of HER-2/neu in breast cancer. J Clin Oncol 17: 1974–1982 [DOI] [PubMed] [Google Scholar]
  9. Jacobs TW, Gown AM, Yaziji H, Barnes MJ, Schnitt SJ (2000) HER-2/neu protein expression in breast cancer evaluated by immunohistochemistry. A study of interlaboratory agreement. Am J Clin Pathol 113: 251–258 [DOI] [PubMed] [Google Scholar]
  10. Jimenez RE, Wallis T, Tabasczka P, Visscher DW (2000) Determination of Her-2/Neu status in breast carcinoma: comparative analysis of immunohistochemistry and fluorescent in situ hybridization. Mod Pathol 13: 37–45 [DOI] [PubMed] [Google Scholar]
  11. Kallioniemi OP, Kallioniemi A, Kurisu W, Thor A, Chen LC, Smith HS, Waldman FM, Pinkel D, Gray JW (1992) ERBB2 amplification in breast cancer analyzed by fluorescence in situ hybridization. Proc Natl Acad Sci USA 89: 5321–5325 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Lebeau A, Deimling D, Kaltz C, Sendelhofert A, Iff A, Luthardt B, Untch M, Lohrs U (2001) Her-2/neu analysis in archival tissue samples of human breast cancer: comparison of immunohistochemistry and fluorescence in situ hybridization. J Clin Oncol 19: 354–363 [DOI] [PubMed] [Google Scholar]
  13. Lehr HA, Jacobs TW, Yaziji H, Schnitt SJ, Gown AM (2001) Quantitative evaluation of HER-2/neu status in breast cancer by fluorescence in situ hybridization and by immunohistochemistry with image analysis. Am J Clin Pathol 115: 814–822 [DOI] [PubMed] [Google Scholar]
  14. Moasser MM, Basso A, Averbuch SD, Rosen N (2001) The tyrosine kinase inhibitor ZD1839 (‘Iressa’) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res 61: 7184–7188 [PubMed] [Google Scholar]
  15. Paik S, Bryant J, Tan-Chiu E, Romond E, Hiller W, Park K, Brown A, Yothers G, Anderson S, Smith R, Wickerham DL, Wolmark N (2002) Real-World Performance of HER2 Testing-National Surgical Adjuvant Breast and Bowel Project Experience. J Natl Cancer Inst 94: 852–854 [DOI] [PubMed] [Google Scholar]
  16. Pauletti G, Dandekar S, Rong H, Ramos L, Peng H, Seshadri R, Slamon DJ (2000) Assessment of methods for tissue-based detection of the HER-2/neu alteration in human breast cancer: a direct comparison of fluorescence in situ hybridization and immunohistochemistry. J Clin Oncol 18: 3651–3664 [DOI] [PubMed] [Google Scholar]
  17. Pegram MD, Lipton A, Hayes DF, Weber BL, Baselga JM, Tripathy D, Baly D, Baughman SA, Twaddell T, Glaspy JA, Slamon DJ (1998) Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. J Clin Oncol 16: 2659–2671 [DOI] [PubMed] [Google Scholar]
  18. Persons DL, Borelli KA, Hsu PH (1997) Quantitation of HER-2/neu and c-myc gene amplification in breast carcinoma using fluorescence in situ hybridization. Mod Pathol 10: 720–727 [PubMed] [Google Scholar]
  19. Press MF, Hung G, Godolphin W, Slamon DJ (1994) Sensitivity of HER-2/neu antibodies in archival tissue samples: potential source of error in immunohistochemical studies of oncogene expression. Cancer Res 54: 2771–2777 [PubMed] [Google Scholar]
  20. Ridolfi RL, Jamehdor MR, Arber JM (2000) HER-2/neu testing in breast carcinoma: a combined immunohistochemical and fluorescence in situ hybridization approach. Mod Pathol 13: 866–873 [DOI] [PubMed] [Google Scholar]
  21. Roche PC, Ingle JN (1999) Increased HER2 with US Food and Drug Administration–approved antibody. J Clin Oncol 17: 434. [DOI] [PubMed] [Google Scholar]
  22. Roche PC, Suman VJ, Jenkins RB, Davidson NE, Martino S, Kaufman PA, Addo FK, Murphy B, Ingle JN, Perez EA (2002) Concordance Between Local and Central Laboratory HER2 Testing in the Breast Intergroup Trial N9831. J Natl Cancer Inst 94: 855–857 [DOI] [PubMed] [Google Scholar]
  23. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L (2001) Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med 344: 783–792 [DOI] [PubMed] [Google Scholar]
  24. Tanner M, Gancberg D, Di Leo A, Larsimont D, Rouas G, Piccart MJ, Isola J (2000) Chromogenic in situ hybridization: a practical alternative for fluorescence in situ hybridization to detect HER-2/neu oncogene amplification in archival breast cancer samples. Am J Pathol 157: 1467–1472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Thor A (2001) HER2–a discussion of testing approaches in the USA. Ann Oncol 12 (Suppl 1): S101–S107 [DOI] [PubMed] [Google Scholar]
  26. Tubbs RR, Pettay JD, Roche PC, Stoler MH, Jenkins RB, Grogan TM (2001) Discrepancies in clinical laboratory testing of eligibility for trastuzumab therapy: apparent immunohistochemical false-positives do not get the message. J Clin Oncol 19: 2714–2721 [DOI] [PubMed] [Google Scholar]
  27. Vincent-Salomon A, Mac Grogan G, Couturier J, Arnould L, Denoux Y, Fiche M, Jacquemier J, Mathieu MC, Penault-Llorca F, Rigaud C, Roger P, Treilleux I, Vilain MO, Mathoulin-Pélissier S, Le Doussal V (2003) Calibration of immunohistochemistry for assessment of HER2 in breast cancer: results of the french multicentric gefpics study. Histopathology 42: 337–347 [DOI] [PubMed] [Google Scholar]
  28. Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, Slamon DJ, Murphy M, Novotny WF, Burchmore M, Shak S, Stewart SJ, Press M (2002) Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol 20: 719–726 [DOI] [PubMed] [Google Scholar]
  29. Zhao J, Wu R, Au A, Marquez A, Yu Y, Shi Z (2002) Determination of HER2 gene amplification by chromogenic in situ hybridization (CISH) in archival breast carcinoma. Mod Pathol 15: 657–665 [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Cancer are provided here courtesy of Cancer Research UK

RESOURCES