8th International Inter University Scientific Meeting
Academy of Studenica
NEW TRENDS IN DIAGNOSTICS AND THERAPY OF MALIGNANT TUMORS
Organizer: Institute of Oncology Sremska Kamenica, Yugoslavia
Co-organizers:
Institute for Oncology and Radiology, Belgrade, Yugoslavia;
"Aristotel School", Thessaloniki, Greece
President: Prof.Dr. Vladimir Vit. Baltiæ
ISSN 1450-708

Content
5 /2001
 
THE ROLE OF EPIDERMAL GROWTH FACTOR RECEPTOR FAMILY IN THE MOLECULAR PATHOGENESIS AND TREATMENT OF BREAST CANCER
K. T. Papazisis
Breast Cancer Biology Group, Cancer Research UK, London SE1 9RT
 
  Keywords: Breast cancer; HER; HER2; Herceptin; Protein tyrosine kinases  
  Receptor Protein Tyrosine Kinases (rPTKs)
PTKs are a group of enzymes that catalyze the transfer of the g-phosphate unit from ATP to a tyrosine residue. They are involved in the regulation of cellular proliferation, differentiation and apoptosis. Human genome encodes for more than 150 PTKs and at least 18 are well-characterized oncogenes.
There are two distinct groups of protein tyrosine kinases: receptor PTKs and non-receptor (cytoplasmic) PTKs. Receptor PTKs are glycoproteins with an extracellular (aminoterminal) domain, a short transmembrane domain and an intracellular (carboxytelic) domain. Intracellular domain consists of specific subdomains for phosphorylation, interaction with other proteins of the downstream signal transduction pathways and the kinase subunit. All rPTKs (except Insulin-like Growth Factor Receptor - IGFR) exist in the cellular membrane in the form of monomers. Ligand association leads to receptor dimerization (or oligomerization), eterophosphoryliosis of the cytoplasmic tails of the receptors and activation of the downstream cascades by means of kinase activation and "opening" of the adaptor modules that are capable to recruit the components of signal transduction.
Epidermal Growth Factor Receptor Famamily
There are four members of human epidermal growth factor receptor (HER) family [HER-1 (EGFR or c-erbB), HER-2 (c-erbB-2), HER-3 and HER-4] and several ligands. EGF is the ligand for HER-1, whilst TGFa, neuregulins (NRGs), epiregulin (EPI), betacellulin (BTC), and other related molecules interact with the other three receptors. The interaction of ligand with the receptor induces conformational changes of the later, which result to receptor dimerization, either with a same receptor (homodimerization) or with another receptor of the family (heretodimerization). Downstream messages include PI3-kinase, the Grb2-ras pathway, PLC-g and MAPK pathway activation. The outcome of these complex networks is the induction of cellular proliferation and the inhibition of apoptosis.
The intracellular domain of HER family receptors has been described in detail. There is a catalytic subunit and several SH2 (src homology regions 2) and PTB (phosphotyrosine-binding) motifs. SH2 domains are 100 amino acid-long motifs that interact with phosphotyrosine residues depending on the 3-6 carboxy-terminal adjacent amino acids. On the contrary, PTB domains interact with phosphotyrosines depending on the amino-terminal adjacent sequence. Several adaptor proteins are recruited to the receptors, like Grb2, Grb7, Shc, Crk and Gab1, kinases like PI3K and Src, phospholipases (PLC-g1) and phosphatases (SHP1 and SHP2). The net outcome of this complex network depends on several factors, as the type of ligand (or ligands) that activated the receptors, ligand concentration and the kind of dimer that was formed (determined both by the ligand and by the degree of expression of the receptor molecules).
It is interesting that HER2 (which has the most active PTK subunit) has not any known ligand (and this way HER2/HER2 dimers cannot be normally formed) but heterodimerizes with other receptors of the family, when they bind to ligand. The dimerization with HER2 leads to signal stabilization (i.e. prolongation) due to decreased intracellular traffic of the dimers, increased affinity to the ligand and signal amplification. On the other hand HER3 has not any kinase activity (HER3/HER3 dimers can be formed but are inactive) but is the only receptor of the HER family that can recruit PI3K effectively because it has many p85 (the adaptor domain of PI3K) binding sites. This way, the heterodimer HER2/HER3 is the most potent dimer in inducing cellular proliferation and survival.
HER family in breast cancer
HER2 is overexpressed in almost a third of breast cancer cases. The overexpression of HER2 in other cancer types has been also documented but only in breast cancer there is firm evidence for its role in pathogenesis and therapy. On a cellular level, HER2 overexpression can lead to increased cellular proliferation (by inducing cyclin D expression and inhibiting p27KIP1 levels), decreased apoptosis and resistance to cytotoxic treatment and reduced expression of several adhesion molecules (a2-integrins, cadherins) that increases the metastatic potential.
In breast cancer patients, increased HER2 expression is usually the result of gene amplification (usually an increased copy number up to ´10) that results to protein overexpression (´10- ´100) in the cytoplasmic membrane. However there are single-copy overexpressors as well as patients with gene amplification without protein overexpression. The significance of the two later observations is not clear, but some studies have shown that gene amplification without protein overexpression is also a bad prognostic indicator, possibly due to amplification of adjacent genes (as topoisomerase-IIa). Finally, some patients express an alternatively spliced form that lacks a small part just carboxytelic to the transmembrane domain that leads to constant HER2 dimerization and activation. The proteolytical cleavage of the extracellular domain can also lead to constant dimerization and activation whilst the extracellular domain can be detected (by immunoabsorbent assays) in the serum and used as a tumor marker.
In the clinical setting, HER2 overexpression is a bad prognostic indicator leading to
1) Decreased disease-free survival and
2) Decreased overall survival in node-positive patients.
It is also a predictive factor because it results in
1) Resistance to CMF chemotherapy
2) Resistance to antiestrogen hormonal treatment
3) A better response to anthracyclin-containing regimens (possibly because of the co-expression of the target protein for anthracyclins topoisomerase-IIa)
4) Response to a new anti-HER2 monoclonal antibody treatment (trastuzumab, Herceptin®)
However, because HER2 is not usually signaling in the absence of the other HER family members, the oncogenic potential of HER2 depends largely on the kind of heterodimers that forms with HER1,3 and 4. Among HER2-positive breast cancer patients, those with overexpression of HER1 or/and HER3 are the ones that belong to worse prognosis group. The overexpression of HER4 on the other hand seems to improve the prognosis of HER2-positive patients. Much work is yet to be done in analyzing by detail the effect of the HER-family differential expression in breast cancer, using large-scale studies with standardized and approved techniques.
HER2-targeted therapy for breast cancer
Trastuzumab (Herceptin, Roche) was developed as a humanized anti-HER2 monoclonal antibody that targets the extracellular (juxta-membrane) domain of HER2. Trastuzumab treatment results in
• Inhibition of HER2 expression (inducing endocytosis of the HER2/HERx/trastuzumab complex and subsequently proteolytic cleavage of HER2)
• Inhibition of HER2 phosphoryliosis
• Induction of HER3 cleavage when it is in dimers with HER2
• Increased expression of p27KIP1 and decreased expression of cyclin D1 (resulting in inhibition of proliferation in G0/G1). The decreasing on cyclin D1 expression leaves unbound p27KIP1 to bind and inactivate cyclin E also, resulting in G1 arrest.
• Inhibition of Rb phosphorylation
• Inhibition of extracellular cleavage of HER2
• Reduction in VEGF expression
• Antigen-depended cell cyctotoxicity (ADCC) against HER2+ve cells
In clinical terms, these effects can be translated to a higher response rate, longer disease-free survival and possibly to a longer overall survival as well. The combination of trastuzumab with chemotherapeutic drugs (taxanes as paclitaxel and docetaxel, cis-platinum, etc) is proven synergistic in vitro and in vivo.
However, only one third of HER2+ve patients will eventually respond and benefit from herceptin treatment in the clinical practice. An explanation for this is the overexpression and activation of other "parallel" signal transduction pathways (HER1, FGFR1) or the independence of the proliferation and survival signal from the receptor regulation, due to mutations in downstream signals that render them unresponsive to HER2 downregulation. On the other hand, HER2+ve tumors do not represent a single group, since expression of the other three members of the HER family is largely influencing the final outcome. In this way, HER3+ve patients or patients with active PI3K (as can be demonstrated with anti p-akt immunohistochemistry) are more likely to respond to herceptin treatment. There is still a long way to identify the patients with a higher probability to respond to anti-HER2 treatment, to standardize the most effective ways of anti-HER treatment and combinations with other therapeutic modalities and to improve anti-HER treatment possibly by using small molecule PTK inhibitors with high anti-HER potency.

SELECTED REFERENCES
1. Annals of Oncology 12 (Suppl. 1): S3-S107, 2001
2. Seminars in Oncology 29 (Suppl 11): 4-70, 2002
3. Bange J, Zwick E, Ullrich A. Molecular targets for breast cancer therapy and prevention. Nature Med. 7: 548-552, 2001
4. Gullick WJ, Srinivasan R. The type 1 growth factor receptor family: new ligands and receptors and their role in breast cancer. Breast Cancer Res. Treat. 52: 43-53, 1998
5. Hung M-C, Lau Y-K. Basic science of HER-2/neu: A review. Semin. Oncol. 26 (suppl 12): 51-59, 1999
6. Miles, DW. Update on HER-2 as a target for cancer therapy. Herceptin in the clinical setting. Breast Cancer Res. 3: 380-384, 2001 7. Piccart MJ, Di Leo A, Hamilton A. HER2: a "predictive factor" ready to use in the daily management of breast cancer patients? Eur. J. Cancer 36: 1755-1761, 2000
8. Robinson DR, Wu Y-M, Lin S-F. The protein tyrosine kinase family of the human genome. Oncogene 19: 5548-5557, 2000
9. Sliwkowski MX, Lofgren JA, Lewis GD, Hotaling TE, Fendly BM, Fox JA. Nonclinical studies addressing the mechanism of action of trastuzumab (Herceptin). Semin. Oncol. 26 (suppl 12): 60-70, 1999
10. Stern, DF. Tyrosine kinase signaling in breast cancer. Breast Cancer Res. 2: 176-183, 2000
11. Strawn LM, Shawver LK. Tyrosine kinases in disease: overview of kinase inhibitors as therapeutic agents and current drugs in clinical trials. Exp. Opin. Invest. Drugs 7: 553-573, 1998
 
© Academy of Studenica, 2002