somatic mutations and human breast cancer. a status report

7
1582 Somatic Mutations and Human Breast Cancer A Status Report Robert Callahan, PhD,* Craig S. Cropp, MD,* Giorgio R. Merlo, PhD,* Daniel S. Liscia, MD,t Albert0 P. M. Cappa, MD,t and Rosette Lidereau, PhD$ A systematic study of primay human breast tumor DNA demonstrated that three proto-oncogenes or regions of the genome (c-myc, int-2, and c-erbB2) are frequently am- plified and that there is loss of heterozygosity (LOH) on chromosomes lp(37%), lq(20%), 3p(30%), 7(41%), llp(ZO%), 13q(30%), 17p(49%), 17q(29%),and 18q(34%). Specific subsets of tumors can be defined based on the particular collection of mutations they contain. For in- stance, LOH on chromosomes Ilp, 17p, and 18q fre- quently occurs in the same tumor. A search for putative tumor suppressor genes within the regions of the genome affected by LOH has been started. In a comprehensive molecular analysis of the p53 gene on chromosome 17p, 46% of the tumors contained a point mutation in the p53 gene. Cancer 1992; 691582-1588. An important characteristic of human breast cancer is its etiologic heterogeneity due to factors in the patient history and physiologic status.'-3 It seems likely that some of these factors, such as menstrual status, repro- ductive history, family history, long-term treatment with estrogens, diet, and previous atypical benign breast disease, provide a selective environment for the clonal outgrowth of cells which contain somatic muta- tions. One biological consequence of these mutations may be the development of neoplasia through the un- coupling of normal mammary gland development. Other consequences could be the provision of atypical nonmalignant as well as tumor cells with a selective Presented at the American Cancer Society and American Joint Committee on Cancer Workshop on Molecular Markers in the Classi- fication and Staging of Cancer, Atlanta, Georgia, December 13-14, 1990. From the *National Cancer Institute, Bethesda, MD; tAnatomic Pathology Section, S. Giovanni Hospital, Torino, Italy; and the $Centre Rene' Huguenin, St. Cloud, France. Address for reprints: Robert Callahan, PhD, National Cancer Institute, Bethesda, MD 20892. Accepted for publication September 10, 1991. growth advantage, the means to metastasize to distant organ sites, or evasion of host immunosurveillance. Be- cause of this heterogeneity and the multiple events nec- essary for most epithelial tumors, it seems likely that multiple genetic mutations act in concert to produce an invasive breast carcinoma with the ability to metasta- size to distant organ sites. Consistent with this view, cytogenetic analysis of primary human breast tumor cells in culture has demon- strated frequent somatic alterations. These include an- euploidy (either gain or loss of entire chromosomes) and rearrangements affecting chromosomes lq (trans- locations), 6q (deletions and translocations), 7p (trans- locations, pericentric inversions, and isochromosomes), and 1 l q trans location^).^ More recently, we and other laboratories have begun to survey the human genome in primary breast tumors at a molecular level to identify frequent somatic mutations of specific genes or alter- ations of specific regions of the cellular g e n ~ m e . ~ , ~ One goal of these studies has been to determine whether specific mutations are linked to clinical parameters in the patients history, including disease outcome, or whether the gene products can be used as targets for possible therapeutic intervention. In this report we sum- marize the results of our analysis, and discuss their po- tential implication in understanding the biology of the disease and its clinical management. Our studies have focused on comparing the DNA of 280 tumors with normal lymphocyte DNA from the same patients. The specimens were collected from two different hospitals and were composed primarily of in- vasive ductal carcinomas. Currently we have exten- sively surveyed 14 of the 41 nonacrocentric chromo- some arms in these tumor DNA samples with recombi- nant DNA probes which detect either proto-oncogenes or restriction fragment length polymorphisms (RFLP) of anonymous cellular loci. Eleven frequently occurring somatic mutations have been identified as discussed below (Table 1).

Upload: robert-callahan

Post on 06-Jun-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Somatic mutations and human breast cancer. A Status Report

1582

Somatic Mutations and Human Breast Cancer A Status Report

Robert Callahan, PhD,* Craig S. Cropp, MD,* Giorgio R. Merlo, PhD,* Daniel S. Liscia, M D , t Albert0 P. M . Cappa, M D , t and Rosette Lidereau, PhD$

A systematic study of primay human breast tumor DNA demonstrated that three proto-oncogenes or regions of the genome (c-myc, int-2, and c-erbB2) are frequently am- plified and that there is loss of heterozygosity (LOH) on chromosomes lp(37%), lq(20%), 3p(30%), 7(41%), llp(ZO%), 13q(30%), 17p(49%), 17q(29%), and 18q(34%). Specific subsets of tumors can be defined based on the particular collection of mutations they contain. For in- stance, LOH on chromosomes Ilp, 17p, and 18q fre- quently occurs in the same tumor. A search for putative tumor suppressor genes within the regions of the genome affected by LOH has been started. In a comprehensive molecular analysis of the p53 gene on chromosome 17p, 46% of the tumors contained a point mutation in the p53 gene. Cancer 1992; 691582-1588.

An important characteristic of human breast cancer is its etiologic heterogeneity due to factors in the patient history and physiologic status.'-3 It seems likely that some of these factors, such as menstrual status, repro- ductive history, family history, long-term treatment with estrogens, diet, and previous atypical benign breast disease, provide a selective environment for the clonal outgrowth of cells which contain somatic muta- tions. One biological consequence of these mutations may be the development of neoplasia through the un- coupling of normal mammary gland development. Other consequences could be the provision of atypical nonmalignant as well as tumor cells with a selective

Presented at the American Cancer Society and American Joint Committee on Cancer Workshop on Molecular Markers in the Classi- fication and Staging of Cancer, Atlanta, Georgia, December 13-14, 1990.

From the *National Cancer Institute, Bethesda, MD; tAnatomic Pathology Section, S. Giovanni Hospital, Torino, Italy; and the $Centre Rene' Huguenin, St. Cloud, France.

Address for reprints: Robert Callahan, PhD, National Cancer Institute, Bethesda, MD 20892.

Accepted for publication September 10, 1991.

growth advantage, the means to metastasize to distant organ sites, or evasion of host immunosurveillance. Be- cause of this heterogeneity and the multiple events nec- essary for most epithelial tumors, it seems likely that multiple genetic mutations act in concert to produce an invasive breast carcinoma with the ability to metasta- size to distant organ sites.

Consistent with this view, cytogenetic analysis of primary human breast tumor cells in culture has demon- strated frequent somatic alterations. These include an- euploidy (either gain or loss of entire chromosomes) and rearrangements affecting chromosomes l q (trans- locations), 6q (deletions and translocations), 7p (trans- locations, pericentric inversions, and isochromosomes), and 1 l q trans location^).^ More recently, we and other laboratories have begun to survey the human genome in primary breast tumors at a molecular level to identify frequent somatic mutations of specific genes or alter- ations of specific regions of the cellular g e n ~ m e . ~ , ~ One goal of these studies has been to determine whether specific mutations are linked to clinical parameters in the patients history, including disease outcome, or whether the gene products can be used as targets for possible therapeutic intervention. In this report we sum- marize the results of our analysis, and discuss their po- tential implication in understanding the biology of the disease and its clinical management.

Our studies have focused on comparing the DNA of 280 tumors with normal lymphocyte DNA from the same patients. The specimens were collected from two different hospitals and were composed primarily of in- vasive ductal carcinomas. Currently we have exten- sively surveyed 14 of the 41 nonacrocentric chromo- some arms in these tumor DNA samples with recombi- nant DNA probes which detect either proto-oncogenes or restriction fragment length polymorphisms (RFLP) of anonymous cellular loci. Eleven frequently occurring somatic mutations have been identified as discussed below (Table 1).

Page 2: Somatic mutations and human breast cancer. A Status Report

Somatic Mutations and Breast Cancer/Callahan et al. 1583

Table 1. Associations Between Genetic Alterations and Clinical Parameters in Patients With Breast Cancer

Frequency Proto-oncogene (YO) Clinical parameters P value Reference no.

c-myc 32 Patient age > 50 yr < 0.02 Escot et a!.' c-erbB2 10 None Ali et al.I7 int-2 16 Local recurrence or distal metastasis < 0.002 Lidereau et al." lp32-pter 26 Decreased survival after relapse < 0.011 Bieche ef a/.34 lq22-q32 20 None Merlo et a/.35 3p21-p25 30 ER-negative tumors Hitopath Grade I11 < 0.019 Ali et ~ 1 . ~ ~

11~11 .2 -~15 .5 20 ER/PR-negative tumors histopath. Grade I11 < 0.002 Ali et a1.,37 Theillet et a1.38

13q14.1(RB-1) 30 Histopath Grade I11 < 0.031 Merlo and Callahad9 1 7 ~ 1 3 47 High proliferative index < 0.022 Merlo et 17q 32 ER-negative tumors < 0.020 Leone et d.'* 18q21-qter 34 Histopath Grade I11 < 0.040 Cropp et al."

ER: estrogen receptor; PR: progesterone receptor; Histopath: histopathologic.

< 0.0001

< 0.006

Amplification of Regions of the Cellular Genome

Amplification of specific regions of the cellular genome is a dominant mutation which is associated with the activation or over-expression of the target gene located within the amplification unit.7 In our study amplifica- tion of three regions of the human genome were de- tected. One region on chromosome 8q, containing the c-myc proto-oncogene, was amplified in 32% of the breast tumors.' This gene is related to the retroviral v- myc on~ogene.~ Frozen sections of primary human breast tumors containing amplification of c-myc were shown by in situ RNA:RNA hybridization to contain high levels of c-myc RNA." In these tumors only the tumor cells contained c-myc RNA. The second affected chromosomal region, located on chromosome 1 lq13, was found to be amplified in 16% of the tumors."*" This region contains the int-2 and hst/K-FGF proto-on- cogenes which are members of the fibroblast growth factor (FGF) gene In mouse mammary tu- mors the mouse mammary tumor virus (MMTV) fre- quently activates these genes by insertion mutagene- sis.15 Again using in situ RNA:RNA hybridization, we were able to detect int-2 RNA, but not hst/K-FGF RNA, in frozen sections from tumors containing an amplifica- tion of these genes.16 This result suggests that int-2 am- plification has been selected in these samples during tumor development. A third region on chromosome 17q, containing the c-erbB2 proto-oncogene, was am- plified in 10% of the turn or^.'^ The c-erbB2 proto-onco- gene is a member of the epidermal growth factor recep- tor gene family.'8-20 In the rat homologue (neu onco- gene), point mutations altering the transmembrane domain of the protein confer on it the capability to ma- lignantly transform tissue culture cells. Although, we have not determined whether amplification of c-erbB2

is associated with elevated levels of the gene product, several other laboratories have found such an associa- tion."

The association between the amplification of spe- cific genes and RNA or protein expression, however, is not always p e r f e ~ t . ~ For instance, over-expression of c-myc and c-erbB2 in primary breast tumors has been observed in the absence of gene amplification. In the case of c-myc over-expression, this may in part reflect heavy lymphoplasma-cellular infiltration of the tumor in which the lymphocytes, but not the tumor cells, ex- press c-myc RNA." Among other explanations is the possibility that contaminating normal tissue in the biopsy material has masked the presence of gene am- plification in the tumor cells. The association between amplification of sequences on chromosome l l q and int-2 RNA expression is also controversial. For instance, one study found that both genes were expressed when they were amplified in breast tumors." In another study, the expression of neither gene was detected in tumors in which they were am~lified.'~ Instead, a closely linked anonymous gene was coamplified and e~pressed.'~

In vivo models to study how activation of these genes contributes to breast cancer have been devel- oped. Transgenic mouse strains containing as trans- genes the c-myc proto-oncogene, the rat neu oncogene, or the in t-2 proto-oncogene showed either altered mammary gland development or a high incidence of spontaneous mammary tumors. For instance, trans- genic mouse strains containing the int-2 transgene de- velop multifocal preneoplastic hyperplasia of the mam- mary gland which can give rise to focal mammary tu- m o r ~ . ~ ~ Other transgenic mouse strains containing c-myc or the neu transgenes have a high incidence of focal mammary tumor^.^^-^^ However, with the excep-

Page 3: Somatic mutations and human breast cancer. A Status Report

1584 CANCER Supplement March 15,2992, Volume 69, NO. 6

tion of one unique strain of neu transgenic mice,27 which expresses high levels of the oncoprotein in the mammary gland and develops multifocal mammary tu- mors, none of these activated transgenes appears to be alone sufficient to induce mammary neoplasia. In this setting, it seems that additional mutations are required for mammary carcinoma development in mice.

Loss of Heterozygosity

In primary human breast tumors loss of heterozygosity (LOH) represents the most frequent type of mutation. LOH appears to occur as a consequence of either inter- stitial deletions, chromosome loss, or aberrant mitotic recombination events and is a common feature of many kinds of malignan~ies.~',~~ It is thought to mask the pres- ence of a "tumor suppressor" gene(s) located within the corresponding affected region on the homologous c h r o m o s ~ m e . ~ ~ , ~ ~ The retinoblastoma gene (RB-1) is re- garded as the paradigm for this type of cellular gene. Inactivation of the RB-1 gene may occur as a conse- quence of two independent mutation events. Com- monly, one normal allele is lost as a result of LOH, whereas the other allele contains either a small deletion or a point mutation which inactivates the gene product. In our panel of breast tumor DNA, LOH was frequently detected on chromosomes lp,34 lq,35 3 ~ , 3 ~ 1 1p,37*38 13q,39 17p, 17q, and 18q (Table l).40,41

Sat0 et aL4' have "allelotyped" a panel of primary breast carcinoma DNA by examining one RFLP per au- tosomal chromosome arm for LOH. They found that the most frequently affected chromosomes were 13q (21%), 16q (38%), and 17p (52%). The frequency of LOH on the other chromosome arms ranged from 0% to 19%. The low frequency of LOH on some chromo- some arms could be interpreted as evidence for nonspe- cific or "background" genetic alterations sustained dur- ing tumor progression. Their findings were similar to other studies on colorectal and renal cell carcinomas, in that most chromosome arms were affected to a varying extent by LOH.43,44 However, the reported frequencies of LOH on these chromosome arms could have been significantly underestimated in these studies since only a single RFLP was tested per chromosome arm. For in- stance, the apparent frequency of LOH for a particular chromosome arm could be reduced if the target tumor- suppressor gene is located at some distance from the site of the RFLP. For this reason our strategy has been to examine multiple RFLP which are located at regular intervals along each chromosome arm.

Although our study is incomplete, we have consis- tently found that the frequency of LOH varies signifi- cantly and in an ordered manner between individual RFLP on the same chromosome arm. This type of pat-

tern of LOH is consistent with the presence of a tumor- suppressor gene near the locus having the highest fre- quency of LOH. Cellular heterogeneity of the tumor is another factor which could affect the apparent fre- quency of mutations. The heterogeneity could be a con- sequence of contaminating normal tissue in the biopsy material or heterogeneity of tumor cells containing the particular mutation. Currently, therefore, it is probably premature to make any definitive conclusions on whether the large number of regions of the genome which are affected by LOH are a consequence of the nonspecific accumulation of mutations during tumor progre~sion,~~ or reflect a progressive selection of muta- tions which contribute to tumor progression.

Although more extensive and systematic studies will be required to distinguish between these possibili- ties, several observations suggest to us that the com- plexity of mutations contributing to the evolution of human breast carcinoma will be high. For instance, if the high frequency of LOH at different regions of the cellular genome was a consequence rather than a con- tributing factor in breast tumor development, the muta- tions might be expected to occur independent of one another. In fact we have found 11 pairs of mutations which occur together at a statistically significant fre- q~ency .~ ' On this basis two subsets of tumors within our tumor panel can be distinguished: one of these fre- quently contains LOH on chromosomes l l p , 17p and 18q; whereas in the other subset chromosomes lp, 13q, and 18q are affected by LOH. Similar findings have been reported in other studies of primary breast, colon, and lung carcinoma^.^^*^^-^^ In addition, the activation or inactivation of eight cellular genes by somatic muta- tions has been found in mouse mammary t ~ m o r s . ' ~ , ~ ' - ~ ~ Taken together these findings suggest that, depending on the history and physiologic status of the patient, groups of collaborating mutations may be selected dur- ing tumor development.

Currently, only two of the probable target tumor- suppressor genes affected by LOH in primary breast tumors have been identified. They both are believed to be involved in the normal suppression of cellular prolif- eration during development. The target for LOH on chromosome 13q appears to be the RB- 1 gene.53-55 This gene encodes a nuclear phosphoprotein which has the capability of binding to DNA.56 Although the level of the protein is constant throughout the cell cycle, the extent of its phosphorylation fluctuate^.^^-^' It is thought that the dephosphorylated form is normally associated with the suppression of cellular prolifera- tion. On chromosome 17p, the p53 gene appears to rep- resent another target tumor-suppressor gene for LOH in primary breast tumors.

Our group41 and others62-66 have found that 10% to

Page 4: Somatic mutations and human breast cancer. A Status Report

Somatic Mutations and Breast Cancer/Callahan et al. 1585

46% of primary breast tumors contain point mutations in the p53 gene. In our study three of 28 tumors con- tained LOH spanning the p53 gene as well as a point mutation in the remaining p53 allele, as determined by sequencing tumor-derived p53 cDNA clones. More- over, in other studies a germline mutation of the p53 gene has been shown to be linked to the Li-Fraumeni hereditary cancer syndrome,67 in which breast cancer is a major component. The p53 gene also encodes a DNA- binding nuclear pho~phoprotein,~',~~ which is capable of activating the transcription of a reporter gene.70*7' Unlike the RB protein, the levels of the p53 protein do fluctuate during the cell c y ~ l e . ~ * , ~ ~ Some mutant p53 genes have been shown to have transforming activity in ~ i t r o . ~ ~ Two consequences of these mutations are an increase in the half-life of the protein and a loss their ability to stimulate transcription of a reporter gene.71*75 Currently, however, whether the RB and p53 proteins have a direct or indirect involvement in the regulation of the cell cycle or differentiation remains to be deter- mined.

Other cellular genes which may represent targets for LOH in primary breast tumors include the nm23- H176 and deleted in colon carcinoma (DCC)77 genes on chromosomes 17q and 18q, respectively. The nm23-H1 gene is a candidate metastasis suppressor gene, whose expression is down-regulated in highly metastatic tu- mor ~ e l l s . ~ ~ , ~ ' , ' ~ This gene encodes a 17-kd protein that shares homology with nucleoside diphosphate (NDP) kinases.80,s1 It has been suggested that the nm23-H1 protein may regulate certain G-protein mediated events. LOH at nm23-Hl was detected 64% of the tu- mor DNA in our tumor panels2 Studies are underway to determine whether loss of expression of the nm23-H1 gene is linked to LOH at this locus. The DCC gene en- codes a protein that is structurally related to several neural cell adhesion molecules and cell surface glyco- proteins.77 This gene is expressed in most if not all nor- mal tissues, yet in human colon carcinomas where the gene is frequently affected by LOH, its expression is decreased or absent. A loss of this gene product could contribute to malignancy by altering normal cell-cell or cell-basement membrane interactions. Currently, how- ever, a correlation between LOH at the DCC gene and a decrease or absence of its expression has not been dem- onstrated in primary human breast tumors. It may also be pertinent that the hereditary cancer syndromes which are associated with a high incidence of either breast and ovarian or only breast carcinomas are linked to chromosomes lps3 and 1 7q,84 respectively. Whether the genes affected by these hereditary mutations are also the targets genes for LOH in sporadic breast cancer remains to be determined.

Potential Usefulness of Mutations or Their Gene Products as Indicators of Disease Outcome

Serious efforts have been made by several laboratories to determine whether specific mutations are associated with particular aspects of the patients history, charac- teristics of the tumor, or disease o ~ t c o m e . ~ , ~ * ' ~ Particular emphasis has been placed on disease outcome in axil- lary node-negative patients, since there are few individ- ual breast tumor markers that adequately discriminate patients at high risk for relapse.86 The associations found in our own studies are summarized in Table 1. The only mutation which was associated ( P < 0.002) with local recurrence or distal metastasis was amplifica- tion of the int-2 gene. Borg et dS7 have recently re- ported a similar association between int-2 amplification and poor prognosis in breast cancer patients. In addi- tion, we have found that LOH on chromosome l p is associated ( P < 0.011) with shortened survival for pa- tients after relapse.34 Six other mutations (LOH on chromosomes 3p, l lp , 13q, 17p, 17q, and 18q) were associated with the more aggressive tumors that were histopathologic Grade 3, estrogen receptor negative, or had a high proliferative index.

In general, however, the potential clinical useful- ness of the mutations or their gene products as indica- tors of probable disease outcome is in q u e s t i ~ n . ~ , ~ * ~ ~ There are several underlying factors in the methodol- ogy and analysis of the results used by different labora- tories which have contributed to this problem. Currently there is no standardization of reagents or methodologies for detecting mutations or expression of the gene products between laboratories. In addition the care taken to remove normal tissue from the biopsy material before the extraction of DNA, RNA, or protein varies between laboratories. Due to this factor the fre- quency of particular mutations may be significantly under estimated in some studies. The composition of the various tumor panels with respect to associated clinical parameters and individual characteristics of the tumors can also vary significantly between laboratories. A related variable between different retrospective stud- ies is the treatment the patients received after surgery. In some studies they all received the same treatment, whereas in others it was varied and uncontrolled.

Another problem is the preoccupation with P val- ues in the statistical analysis of the results. In some stud- ies this has led to an over-interpretation of the signifi- cance of the data. The P values depend in part on sam- ple size and do not measure the power of an association. Thus, tumor panels which are small in size are restricted in the ability to detect anything but a large effect. Alternatively, a relatively small association with little clinical importance could have an impressive P

Page 5: Somatic mutations and human breast cancer. A Status Report

1586 CANCER Supplement March 25, 2992, Volume 69, No. 6

value in a sufficiently large panel of tumors. We, there- fore, view our studies as well as those of others as pilot studies directed at hypothesis development rather than their confirmation.

Future Possibilities

Clearly this field holds promise for the development of new prognostic indicators that could be useful in the management of the disease. However, before this goal can be reached, a standardization of the reagents and methodologies as well as the development of compara- ble tumor panels must occur. A second area which holds promise for the future is the development of ther- apeutic approaches based on the over-expressed gene products of mutated cellular genes or the gene products of mutated cellular genes or the gene products of dis- tant cellular genes expressed as a consequence of a par- ticular mutation. For instance, it seems likely that sev- eral laboratories will begin therapeutic clinical trials in the near future using monoclonal antibodies prepared against the c-erbB2 protein as a vehicle for radionu- clides or toxins. In the near future it may be possible to develop recombinant vaccinia vectors containing one of the activated genes (i.e., int-2) whose gene product is displayed on the tumor cell surface but not on the sur- face of normal cells.88 An alternative approach is sug- gested by the achievement of antiviral protection in vivo by peptide-induced cytotoxic T - ~ e l l s . ~ ~ , ~ ~ A similar approach using a peptide corresponding to a mutated H-rus p21 protein was able to stimulate CD4-positive T-cells to specifically recognize cells expressing the mu- tated protein sequence.” Thus, it may be possible to develop a therapeutic approach in which class I or class I1 major histocompatibility molecule restricted cytotoxic T-lymphocytes are stimulated with peptides corre- sponding to the mutated portion of the p53 protein or other potential point mutated tumor-associated and tu- mor-suppresor gene products.

References

1.

2.

3.

4.

5 .

Hams JR, Hellman S, Canellos GP, Fisher B. Cancer of the breast. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer Principles and Practice of Oncology. Philadelphia: JB Lippin-

Gompel G, van Kerkem C. The breast. In: Silverberg S, ed. Prin- ciples of Surgical Pathology, vol. 1. New York: Wiley Medical,

Lynch HT, Albano WA, Danes S et al. Genetic predisposition to breast cancer. Cancer 1984; 53:612-622. Trent JM. Cytogenetic and molecular biologic alterations in hu- man breast cancer: A review. Breast Cancer Res Treat 1985;

Callahan R. Genetic alterations in primary breast cancer. Breast Cancer Res Treat 1989; 13:191-203.

cott, 1982; 1119-1178.

1983; 245-255.

5~221-229.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

la .

19.

20.

21.

22.

23.

24.

25.

26.

Callahan R, Campbell G. Mutations in human breast cancer: An overview, Natl Cancer lnst 1989; 81:1780-1786. Alitalo K, Schwab M. Oncogene amplification in tumor cells. Adv Cancer Res 1986; 47235-277. Escot C, Theillet C, Lidereau R et al. Genetic alterations of the c-myc proto-oncogene in human primary breast carcinomas. Proc Nut1 Acad Sci U S A 1986; 83:4834-4838. Teich N, Wyke J, Mak T et al. Pathogenesis of retrovirus induced disease. In: Weiss R, Teich N, Varmus HE, Coffin J, eds. Molecu- lar Biology of Tumor Viruses: RNA Tumor Viruses. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 1982; 845-856. Mariani-Costantini R, Escot C, Theillet C et al. I n situ myc ex- pression and genomic status of the c-myc locus in infiltrating ductal carcinomas of the breast. Cancer Res 1988; 48:199-205. Lidereau R, Callahan R, Dickson C et al. Amplification of the int-2 gene in primary human breast tumors. Oncogene Res 1988;

Ali IU, Merlo G, Lidereau R, Callahan R. The amplification unit on chromosome llq13 in aggressive primary human breast tu- mors contains the bcl-1, int-2, and hst loci. Oncogene 1989; 4:89- 92. Dickson C, Peters G. Potential oncogene product related to growth factor. Nature 1987; 326:833. Yoshida T, Miyagawa K, Odagiri H et al. Genomic sequence of hst/KS transforming gene encoding a protein homologous to fibroblast growth factors and the int-2 encoded protein. Proc Nut1 Acad Sci U S A 1987; 84:7305-7309. Peters G. Oncogenes at viral integration sites. Cell Growth D i f

Liscia DS, Merlo G, Garrett C et al. Expression of inf-2 mRNA in human tumors amplified at the inf-2 locus. Oncogene 1989;

Ali IU, Campbell G, Lidereau R, Callahan R. Lack of evidence for the prognostic significance of c-erbB-2 amplification in hu- man breast carcinoma. Oncogene Res 1988; 3:139-146. Shih C, Padney L, Murray M, Weinberg RA. Transforming genes of carcinomas and neuroblastomas introduced into mouse fibroblasts. Nature 1981; 290:261-264. Schechter AL, Stern DF, Valdyanathan L et al. The neu onco- gene: An erbB related gene encoding a 185,000-M tumor anti- gen. Nature 1984; 312:513-516. Bargmann CI, Hung MC, Weinberg RA. Multiple independent activations of the neu oncogene by a point mutation altering the transmembrane domain of p185. Cell 1986; 45:649-657. Slamon D, Godolphin W, Jones LA et al. Studies of the HER2/ neu proto-oncogene in human breast and ovarian cancer. Science

Theillet C, Le Roy X, De Laperriere 0 et a/ . Amplification of FGF related genes in human tumors: Possible involvement of hst in breast carcinomas. Oncogene 1989; 4:915-920. Fantl V, Brookes S, Smith R et a/. Characterization of the proto- oncogene int-2 and its potential for diagnosis of human breast cancer. In: Greaves M, Furth M, eds. Cancer Cell 7: Molecular Diagnosis of Human Cancer. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, 1989; 283-287. Lammie GA, Fantl V, Smith R et al. DllS287, a putative onco- gene on chromosome 1 lq13, is amplified and expressed in squa- mous cell and mammary carcinomas and linked to BCL-1. Onco- gene 1991; 6:439-444. Muller WJ, Lee FS, Dickson C, Peters G. The int-2 gene product acts as an epithelial growth factor in transgenic mice. EMBO J

Bouchard L, Lamarre L, Tremblay PJ, Jolicoeur P. Stochastic ap- pearance of mammary tumors in transgenic mice carrying the MMTVlc-neu oncogene. Cell 1989; 57:931-936.

2:285-291.

1990; 1~503-510.

4:1219-1224.

1989; 244:707-744.

1990; 9:907-913.

Page 6: Somatic mutations and human breast cancer. A Status Report

Somatic Mutations and Breast Cancer/Callahan et al. 1587

27.

28.

29.

30. 31.

32.

33.

34.

35.

36.

37.

38.

39. 40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

Muller WJ, Sinn E, Pattengale PK et al. Single step induction of mammary adenocarcinoma in transgenic mice bearing activated c-neu oncogene. Cell 1988; 54:105-115. Steward T, Pattengale P, Leder P. Spontaneous mammary ade- nocarcinomas in transgenic mice that carry and express MMTV/ myc fusion genes. Cell 1984; 38:627-637. Sinn E, Muller W, Pattengale P et al. Coexpression of MMTV/v- H-ras and MMTVlc-myc genes in transgenic mice: Synergistic actions of oncogenes in vivo. Cell 1987; 49:465-475. Levine AJ. Tumor suppressor genes. Bioessays 1990; 1260-66. Seemayer TA, Cavenee WE. Biology of disease: Molecular mech- anisms of oncogenesis. Lab Znvest 1989; 60:585-599. Hollingsworth RE, Lee W-H. Tumor suppressor genes: New prospect for cancer research. J Natl Cancer Znst 1991; 83:91-96. Knudson AG. Hereditary cancers: Clue to mechanisms of carci- nogenesis. Br J Cancer 1989; 59:661-666. Bieche I, Champeme MH, Merlo G et al. Loss of heterozygosity of the L-myc oncogene in human breast tumors. Hum Genet

Merlo GR, Siddiqui J, Cropp CS et nl. Frequent alteration of the DF3 tumor-associated antigen gene in primary human breast carcinomas. Cancer Res 1989; 49:6966-6971. Ali IU, Meissner S, Lidereau R, Callahan R. Allelic deletion of c-erbA-2 proto-oncogene in human breast carcinoma signifies possible recessive mutation in member@) of steroid/thyroid hormone receptor family. ] Natl Cancer Znst 1989; 81:1815- 1820. Ali IU, Lidereau R, Theillet C, Callahan R. Reduction to homozy- gosity of genes on chromosome 11 in human breast neoplasia. Science 1987; 238:185-188. Theillet C, Lidereau R, Escot C et al. Frequent loss of a H-ras-1 allele correlates with aggressive human primary breast carci- nomas. Cancer Res 1986; 46:4776-4781. Merlo G, Callahan R. Unpublished data, 1991. Cropp C, Lidereau R, Campbell G et al. Loss of heterozygosity on chromosome 17 and 18 in breast carcinoma: Two new re- gions identified. Proc Natl Acad Sci U S A 1990; 87:7737-7741. Osborne RJ, Merlo GR, Mitsudomi T et al. Mutations in the p53 gene in primary human breast cancer. Cancer Res 1991;

Sato T, Tanigami H, Yamakawa K et al. Allelotype of breast cancer: Cumulative allele losses promote tumor progression in primary breast cancer. Cancer Res 1990; 50:7184-7189. Morita R, Ishikawa J, Tsutsumi M et al. Allelotype of renal cell carcinoma. Cancer Res 1991; 51:820-823. Vogelstein B, Fearon E, Kern BE et al. Allelotype of colorectal carcinomas. Science 1989; 244:207-211. McGuire WL, Naylor SL. Loss of heterozygosity in breast cancer: Cause or effect. ] Natl Cancer Znst 1989; 81:1764-1765. Mackay J, Elder PA, Porteous DJ e f al. Partial deletion of chro- mosome l l p in breast cancer correlates with size of primary tumor and estrogen receptor level. Br J Cancer 1988; 58:710-714. Devilee P, van den Broek M, Kuipers-Dijkshoorn N et al. At least four different chromosomal regions are involved in loss of he- terozygosity in human breast carcinoma. Genomics 1989; 5:554- 560. Yokota J, Wada M, Shimosato Y et al. Loss of heterozygosity on chromosome 3, 13, and 17 in small-cell carcinoma and on chro- mosome 3 in adenocarcinoma of the lung. Proc Natl Acad Sci U S

Vogelstein B, Fearon ER, Hamilton SR et al. Genetic alterations during colorectal tumor development. N Engl ] Med 1988;

Dandekar S, Sukumar S, Zarbl H et al. Specific activation of the cellular Harvey ras oncogene in dimethylbenzanthracene in-

1990; 85:lOl-105.

51~6194-6198.

A 1987; 84~9252-9256.

319~525-532.

51.

52. 53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

duced mouse mammary tumors. 1986; Mol Cell Biol 6:4104- 4108. Sukumar S, Notario V, Martin-Zanca D, Barbacid M. Induction of mammary carcinomas in rats by nitroso-methyl-urea involves the malignant activation of the H-ras-1 locus by single point mutations. Nature 1983; 306:658-661. Marchetti A, Callahan R. Unpublished data, 1991. Lee E, To H, Shew J et al. Inactivation of the retinoblastoma susceptibility gene in human breast cancers. Science 1988;

TAng A, Varley JM, Chakraborty S et al. Structural rearrange- ment of the retinoblastoma gene in human breast carcinoma. Science 1988; 242:263-266. Varley J, Armour J, Swallow JE et al. The retinoblastoma gene is frequently altered leading to loss of expression in primary breast tumors. Oncogene 1989; 4725-729. Lee W-H, Shew J-Y, Hong F et al. Theretinoblastoma susceptibil- ity gene encodes a nuclear phosphoprotein associated with DNA binding activity. Nature 1987; 329:642-645. Mihara K, Cao X-R, Yen A et al. Cell cycle-dependent regulation of phosphorylation of the human retinoblastoma gene product. Science 1989; 246:1300-1303. Chen PL, Scully P, Shew JY et al. Phosphorylation of the retino- blastoma gene product is modulated during the cell cycle and cellular differentiation. Cell 1989; 58:1193-1198. Buchkovich K, Duffy LA, Harlow E. The retinoblastoma protein is phosphorylated during specific stages of the cell cycle. Cell

DeCaprio ]A, Ludlow JW, Lynch D et al. The product of the retinoblastoma susceptibility gene has properties of a cell cycle regulatory element. Cell 1989; 58:1085-1095. Cross F, Roberts J, Weintraub H. Simple and complex cell cycles. Annu Rev Cell Biol 1989; 5:341-396. Bartek J, Iggo R, Gannon J, Lane DP. Genetic andimmunochemi- cal analysis of mutant p53 in human breast cancer cell lines. Oncogene 1990; 5:893-899. Bartek J, Bartkova J, Vojtesek B et al . Patterns of expression of the p53 tumour suppressor gene in human breast tissues and tumours in situ and in nitro. Znt J Cancer 1990; 46:839-844. Cattoretti G, Rilke F, Andreola S et nl. p53 expression in breast cancer. Int ] Cancer 1988; 41:178-183. Thompson AM, Steel CM, Chetty U et al. p53 gene mRNA ex- pression and chromosome 17p allele loss in breast cancer. Br ] Cancer 1990; 61:14-18. Prosser J, Thompson AM, Cranston G, Evans HJ. Evidence that p53 behaves as a tumour suppressor gene in sporadic breast tumours. Oncogene 1990; 5:1573-1579. Malkin D, Li FP, Strong LC et al. Germ line mutations in a famil- ial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990; 250:1233-1238. Levine AJ, Momand J. Tumor suppressor genes: The p53 and retinoblastoma sensitivity genes and gene products. Biochim Biophys Acta 1990; 1032119-136. Sager R. Tumor suppressor genes: The puzzle and the promise. Science 1989; 246:1406-1412. Fields S, Jang SK. Presence of a potent transcription activating sequence in the p53 protein. Science 1990; 249:1046-1049. Raycroft L, Wu HY, Lozano G. Transcriptional activation by wild-type but not transforming mutants of the p53 anti-onco- gene. Science 1990; 249:1049-1051. Bischoff JR, Friedman PN, Marshak DR et al. Human p53 is phosphorylated by p60-cdc2 and cyclin B-cdc2. Proc Natl Acad Sci U S A 1990; 87:4766-4770. Sturzbecher HW, Maimets T, Chumakov P et al. p53 interacts

241 :2 18-221.

1989; 58:1097-11105.

Page 7: Somatic mutations and human breast cancer. A Status Report

1588 CANCER Supplement March 15,1992, Volume 69, No. 6

74.

75.

76.

77.

78.

79.

80.

81.

with ~34 '~ ' ' in mammalian cells: Implication for cell cycle con- trol and oncogenesis. Oncogene 1990; 5:795-801. Eliyahu D, Michalovitz D, Eliyahu S et a/ . Wild-type p53 can inhibit oncogene-mediated focus formation. Proc Natl Acad Sci

Hinds P, Finlay C, Levine AJ. Mutation is required to activate the p53 gene for cooperation with the ras oncogene and trans- formation. ] Virol 1989; 63:739-745. Steeg PS, Bevilacqua G, Kapper L et al. Evidence for a novel gene associated with low tumor metastatic potential. ] Natl Cancer lnst 1988; 8O:ZOO-204. Fearon E, Cho KR, Nigro JM et a/. Identification of a chromo- some 18q gene that is altered in colorectal cancers. Science 1990;

Steeg PS, Bevilacqua G, Rosengard AM et al. Altered gene ex- pression in tumor metastasis: The nm23 gene. In: Schirrmacher V, Schwartz-Albiez R, eds. Cancer Metastasis: Molecular and Cellular Biology, Host Immune Responses and Perspectives for Treatment. Berlin: Springer-Verlag, 1989; 48-52. Bevilacqua G, Sobel ME, Liotta LA, Steeg PS. Association of low nrn23 RNA levels in human primary infiltrating ductal breast carcinomas with lymph node involvement and other histopatho- logical indicators of high metastatic potential. Cancer Res 1989;

Liotta LA, Steeg PS. Clues to the function of Nm23 and Awd proteins in development, signal transduction, and tumor metas- tasis provided by studies of Dictyosteliurn discoideurn. J Natl Cancer lnst 1991; 82:1170-1172. Rosengard AM, Krutzsch HC, Shearn A et al. Reduced, nm23/ Awd protein in tumor metastasis and aberrant Drosophila devel- opment. Nature 1989; 342177-179.

U S A 1989; 86~87634767.

24749-56.

49:5185-5190.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

Leone A, McBride OW, Westin A et al. Allelic deletion of nm23 in cancer. Cancer Res 1991; 512490-2493. Ferrel RE, Anderson DE, Chidambaram A et al . A genetic link- age study of familial breast-ovarian cancer. Cancer Genet Cyto- genet 1989; 38241-248. Hall JM, Lee MK, Newman B et al. Linkage of early-onset famil- ial breast cancer to chromosome 17q21. Science 1990; 250:1684- 1689. McGuire WL. Breast cancer prognostic factors: Evaluation guide- lines. ] Natl Cancer lnst 1991; 83:154-155. McGuire WL, Tandon AK, Allred DC et al. How to use prognos- tic factors in axillary node-negative breast cancer patients. ] Natl Cancer Inst 1990; 821006-1015. Borg A, Sigurdsson H, Clark GM ef a/ . Association of INTZ/HST coamplification in primary breast cancer with hormone-depen- dent phenotype and poor prognosis. Br ] Cancer 1991; 63:136- 142. Smith GL, Mackett M, Moss B. Recombinant vaccinia viruses as new live vaccines. Biotechnol Genet Engin Rev 1984; 2:383-407. Germain RW. The ins and outs of antigen processing and presen- tation. Nature 1986; 322687-689. Takahashi H, Takeshita T, Morein B et a / . Induction of CD8+ cytotoxic T cell by immunization with purified HIV-1 envelope protein in ISCOMS. Nature 1990; 344:874-875. Schultz M, Zinkernagel RM, Hengartner H. Peptide-induced nat- ural protection by cytotoxic T cells. Proc Natl Acad Sci U S A

Merlo G, Venesio T, Bernardi A et al. Loss of heterozygosity on chromosome 1 7 ~ 1 3 in breast carcinomas identifies tumors with a high proliferation index. A m ] Pathol (in press).

1991; 88~991-993.