Skip to main content

Drug therapy for hereditary cancers

Abstract

Tumors arising in patients with hereditary cancer syndromes may have distinct drug sensitivity as compared to their sporadic counterparts. Breast and ovarian neoplasms from BRCA1 or BRCA2 mutation carriers are characterized by deficient homologous recombination (HR) of DNA, that makes them particularly sensitive to platinum compounds or inhibitors of poly (ADP-ribose) polymerase (PARP). Outstandingly durable complete responses to high dose chemotherapy have been observed in several cases of BRCA-related metastatic breast cancer (BC). Multiple lines of evidence indicate that women with BRCA1-related BC may derive less benefit from taxane-based treatment than other categories of BC patients. There is virtually no reports directly assessing drug response in hereditary colorectal cancer (CRC) patients; studies involving non-selected (i.e., both sporadic and hereditary) CRC with high-level microsatellite instability (MSI-H) suggest therapeutic advantage of irinotecan. Celecoxib has been approved for the treatment of familial adenomatous polyposis (FAP). Hereditary medullary thyroid cancers (MTC) have been shown to be highly responsive to a multitargeted tyrosine kinase inhibitor vandetanib, which exerts specific activity towards mutated RET receptor. Given the rapidly improving accessibility of DNA analysis, it is foreseen that the potential predictive value of cancer-associated germ-line mutations will be increasingly considered in the future studies.

Peer Review reports

Introduction

Search for hereditary cancer genes was always regarded as a high priority translational research with immediate health impact. It was foreseen, that the discovery of tumor predisposing mutations and the development of appropriate genetic tests will allow identifying yet healthy subjects, who are at nearly fatal risk of specific type of cancer and thus may benefit from a timely medical intervention. Since the discovery of major cancer genes in the mid 1990s, thousands of mutation carriers have been subjected to intensive surveillance programs in order to secure early diagnosis of the disease [1]. The preventive surgery has been applied in some instances, that led to a proven reduction of cancer-specific mortality [2]. While the initial practical focus of cancer genetic research was limited to various aspects of cancer detection and prevention, it is now getting increasingly recognized that hereditary tumors may have distinct bioclinical characteristics and thus require tailored treatment strategies.

Breast-ovarian cancer syndrome

The best-known hereditary cancer genes, BRCA1 and BRCA2, contribute to substantial share of breast and ovarian tumor incidence around the globe, and have been studied with significant level of comprehension [3]. Mutations in other relevant genes, such as CHEK2, PALB2 (FANCN), ATM, NBN (NBS1), BRIP1 (FANCJ, BACH1), BLM, are less frequent and have not been subjected yet to systematic clinical studies.

BRCA1 and BRCA2

BRCA1 and BRCA2 genes play a central role in the repair of double-strand DNA breaks by homologous recombination (HR). Cancers in BRCA heterozygous individuals arise due to somatic inactivation of the remaining wild-type allele of the gene. This provides a critical biological difference between cancerous and normal cells: while BRCA-driven tumors are characterized by HR deficiency, normal tissues from the same individuals retain non-affected BRCA allele and the ability to cope with DNA damage [4, 5]. Although BRCA1- and BRCA2-related cancers demonstrate somewhat distinct picture of genetic abnormalities [6, 7], they both have increased number of gross chromosomal aberrations and therefore higher tumor grade [8–11]. Cells carrying multiple genetic lesions due to HR defect are normally eliminated by p53-guided defense mechanisms; there are convincing evidence from both human studies and murine models that p53 inactivation is an absolute prerequisite for the propagation of BRCA-dysfunctional tumors cells [12–15].

BRCA1 has a wider spectrum of functions than BRCA2[16, 17]. In addition to DNA repair, BRCA1 is involved in breast cell differentiation and transcriptional regulation of the estrogen receptor (ER) [18]. It has been repeatedly shown, that the majority of BRCA1-mutated breast carcinomas (BC) do not express ER, while the hormonal receptor pattern in BRCA2-associated BC is similar to sporadic cases [19]. BRCA1 is also essential for the mitotic spindle checkpoint as it triggers cellular suicide in response to microtubule damage [20].

Preclinical studies

A large number of preclinical studies aimed to assess specific sensitivity of BRCA1- and BRCA2-defective cells to various anticancer agents. Surprisingly, while the effect of individual compounds has been repeatedly evaluated in diverse model systems (Table 1), there was virtually no attempt to compare clinically relevant combinations of the drugs (Table 2). This may constitute a critical gap between preclinical and clinical research, as single-agent therapy is almost never used as initial treatment of breast or ovarian cancers. It is highly likely, that the standard combinations of cytotoxic compounds produce distinct spectrum of DNA lesions and therefore mediate distinct responses of BRCA-deficient cells when compared to the same drugs acting alone.

Table 1 Drug sensitivity of breast-ovarian cancer syndrome related tumors: preclinical evidence
Table 2 Drug sensitivity of breast-ovarian cancer syndrome related tumors: clinical evidence

There is an excellent consistency in the literature regarding high sensitivity of both BRCA1- and BRCA2-deficient cells to cisplatin and other platinum derivatives [21–40]. It is believed that the DNA crosslinks caused by platinating agents ultimately require homologous recombination to correctly repair DNA damage, so the BRCA-inactive cells cannot cope with this class of drugs. Similar consistency has been observed for another DNA-crosslinking agent, mitomycin C [28, 33, 36, 41–44].

Controversial data have been obtained for doxorubicin, a widely used anthracycline antibiotic with multiple mechanisms of action. Doxorubicin causes double-strand breaks in the target DNA, so it may be particularly effective for the cells lacking error-free repair of this type of lesion. Some investigations demonstrated high sensitivity of BRCA-deficient cells to doxorubicin [24, 45, 46], while other reports described entirely opposite findings [26, 33, 34. 36]. Another topoisomerase II inhibitor, etoposide, showed selective efficacy against BRCA-defective cell in all but one studies [23–25, 36, 47, 48].

Analysis of microtubule poisons produced even more complicated picture. It has been repeatedly demonstrated that BRCA1-deficient cells are significantly less sensitive to taxanes or vinca alkaloids than cells with preserved BRCA1 function [23, 25, 26, 49–51]. Although these observations are in good agreement with the established role of BRCA1 in cellular response to microtubule damage [20], one cannot ignore the existence of sound contradictory data. Zhou et al. [52] reported increased sensitivity of BRCA1-mutated ovarian cancer (OC) cell line to paclitaxel as compared to isogenic cells with reconstituted BRCA1 function. Tassone et al. [53] showed high sensitivity of BRCA1-deficient breast cancer cells to vinorelbine and argued that the differences in the mechanism of action between various microtubule interfering drugs have to be considered while interpreting the results of BRCA1 studies. DeLigio and Zorio [20] commented that the tissue origin of the BRCA1-mutated cells may be essential in determining the response to taxanes and vinca alkaloids. BRCA2 preclinical studies suggested little impact of the status this gene in determining the response to microtubule interfering agents [27, 33, 46, 47].

Alkylating agents are almost always included in the standard schemes for the treatment of breast and ovarian cancers. Surprisingly, this class of drugs has not been subjected to systematic studies in BRCA-deficient model systems. Single-agent cyclophosphamide showed only slight antitumor activity against BRCA1-mutated human breast cancer xenografts growing in nude mice [31]. At the same time, high-throughput pharmaceutical screen involving BRCA2-deficent vs. BRCA2-proficient mouse mammary tumor cell lines identified alkylating agents (chlorambucil, melphalane, nimustine) as the most potent and specific inhibitors of cell growth; furthermore, high efficacy of these drugs was confirmed in animal experiments [38].

Topoisomerase I inhibitors are rarely used for the treatment of breast cancer, but included in some therapeutic schemes for ovarian cancer. High sensitivity to these drugs was suggested for both BRCA1- [24, 54] and BRCA2-defective cells [38, 46, 55], although controversial results have been reported as well [27].

There is a good agreement in the literature that single-agent antimetabolites, 5-fluorouracil and gemcitabine, do not exert specific action against BRCA-deficient tumors [24, 33, 46]. In contrast, 6-thioguanine was identified by a chemical library screen as the most potent antagonist of BRCA2-mutated cells [46].

There is a growing number of studies demonstrating pronounced efficacy of specific inhibitors of poly (ADP-ribose) polymerase (PARP) against BRCA-deficient cancers [31, 33, 38–40, 56–60]. It is suggested that inactivation of PARP interferes with the repair of spontaneous DNA single-strand breaks. In the normal cells these lesions are converted to double-strand breaks during DNA replication and then eliminated by homologous recombination. Since hereditary cancers are deficient for HR due to loss of both alleles of either BRCA1 or BRCA2, they cannot eliminate double-strand breaks by error-free mechanism. As result, cancers arising in BRCA carriers are selectively sensitive to PARP inhibitors, while the normal tissues from the same individuals retain a non-affected BRCA allele and are therefore capable to compensate the consequences of decreased PARP activity [61, 62]. PARP inhibitors appear to be the only class of drugs which was assessed in preclinical models in combination with other anticancer agents; synergism of PARP with platinum compounds and alkylating agents has been reported [31, 33, 38, 39, 59].

Breast cancer

The majority of BRCA2- and a certain fraction of BRCA1-related BC express estrogen and/or progesterone receptors and are therefore expected to benefit from endocrine therapy. While a couple of studies examined the chemopreventive impact of tamoxifen or oophorectomy in BRCA carriers [63] and some investigators analyzed benefits from the adjuvant use of estrogen antagonists [64], there is no reports assessing the role of BRCA status in determining the effect of endocrine intervention in neoadjuvant or metastatic setting.

Data on the efficacy of conventional chemotherapeutic schemes in BRCA-related vs. sporadic breast cancers are summarized in the Table 2. Several research groups reported outcomes of anthracycline-based therapy. Delagoge et al. [65], Chappuis et al. [66], Warner et al. [67], Hubert et al. [68], Fourquet et al. [69] and Byrski et al. [70] provided evidence for remarkable sensitivity of BRCA1/2-related cancers to the neoadjuvant anthracycline-containing regimens. Interestingly, Hubert et al. [68] and Wong Wong Keet et al. [71] observed worse outcomes in BRCA2 vs. BRCA1 carriers. The data of Petit et al. [72] are in strong disagreement with the above observations: in their study only 2/12 (17%) of BRCA1 carriers receiving 5-fluorouracil, epirubicin and cyclophosphamide achieved pathologic complete response (pCR), while pCR was detected in 21/43 (49%) sporadic triple-negative BC.

The only available study of metastatic BC included patients treated by either anthracycline-based or CMF (CMF-like) therapy [73]. In contrast to neoadjuvant series of Hubert et al. [68] and Wong Wong Keet et al. [71], significantly improved outcomes were detected in BRCA2 but not BRCA1 carriers. Low efficacy of CMF therapy in BRCA1-related BC was also described by Byrski et al. [70].

Several investigators analyzed the use of taxane-containing schemes. Byrski et al. [70] observed low rate of pCR in patients with BRCA1-mutated BC receiving neoadjuvant combination of doxorubicin and docetaxel. Moiseyenko et al. [74] reported lack of response of chemonaive triple negative BRCA1-linked BC to the epirubicin-docetaxel doublet. Wysocki et al. [75] genotyped 19 non-responders to docetaxel and revealed as many as 5 (26%) BRCA1 carriers. Kriege et al. [76] confirmed poor efficacy of docetaxel in BRCA1 carriers, while BRCA2-related BC did not fare worse as compared to sporadic controls. It appears, that the preclinical and clinical evidence warning against the early use of taxanes for the treatment of BRCA1-related subtype of BC has already achieved a critical threshold; probably, specifically designed retrospective studies assessing BRCA1 status in distinct categories of taxane users may accelerate further understanding of this issue [77].

The experience of the use of single-agent cisplatin for the treatment of hereditary BC is still limited to BRCA1 carriers. Byrski et al. [70] reported 10/12 (83%) pCR in patients undergoing neoadjuvant treatment. Silver et al. [78] used cisplatin for the preoperative treatment of triple-negative BC; both BRCA1 carriers included in the study demonstrated pCR. Moiseyenko et al. [74] observed major response to cisplatin in a patient whose metastatic BC was insensitive to the upfront anthracycline-taxane combination. The development of resistance to cisplatin may involve the appearance of secondary mutations, which restore BRCA1 reading frame and therefore function of the corresponding protein [61, 79]. One may expect that the newly acquired BRCA1 proficiency will result in sensitization of the cancer cells to estrogen antagonists and taxanes [74].

There are a few case reports on the extremely successful use of high dose chemotherapy in metastatic BRCA-related BC. Since BRCA-deficient BC are particularly sensitive to DNA damaging agents, use of intensive cytotoxic treatment may provide additional benefit to this category of patients. Furthermore, high dose therapy is likely to counteract tumor adaptation to the drugs, e.g. to induce rapid killing of cancer cells and therefore decrease the probability of developing secondary BRCA1-restoring mutations. It is also important to consider that BRCA-driven cancers are often characterized by young age at onset, i.e. the majority of these patients would retain sufficient health conditions to qualify for a risky medical intervention. Huang et al. [80] described a patient with metastatic BRCA2-related BC, who was treated by high dose chemotherapy and remains disease-free for more than 11 years. Vollebergh et al. [81] presented long term outcomes for 40 metastatic BC patients treated by high dose chemotherapy. 6 patients remained on complete remission for 56+ - 150+ months, and all these 6 patients demonstrated chromosomal imbalances characteristic for BRCA1-related cancers. Complete long term responders included 1 out of 2 BRCA1- and 1 out of 2 BRCA2-carriers.

The only prospective clinical trial specifically designed for BRCA carriers evaluated the efficacy of the PARP inhibitor olaparib (AZD2281, KU0059436) [82]. The study included metastatic breast cancer patients, who progressed on the standard chemotherapy schemes. When olaparib was given 400 mg twice daily, objective response and disease stabilization were observed in 11/27 (41%) and 12/27 (44%) patients, respectively. Median progression-free survival approached to 5.7 months.

In agreement with preclinical findings, cisplatin and olaparib clearly outperform conventional treatment schemes when administered to BRCA1-driven BC cases. However, both these drugs have limited duration of response, so their use may require the addition of other anticancer agents [31, 33, 38, 39, 59].

Ovarian cancer

BRCA-deficiency in cancer cell can be caused either by germ-line mutation followed by the "second hit", or by somatic inactivation of the BRCA1 gene. BRCA-inactive tumors constitute the minority of breast cancers (up to 10-15%), and are usually accumulated among family history positive or triple-negative cases. In contrast to BC, the majority of ovarian carcinomas have signs of BRCA inactivation, commonly defined in the literature as "BRCA ness" [9, 15]. Frequent BRCA-deficiency in OC appears to be a plausible explanation of the clinical success of platinum-based schemes in the treatment of this disease.

Three studies compared response to the standard chemotherapeutic regimens in BRCA1/2- mutated vs. sporadic OC cases [83–85]. These reports provide consistent evidence for higher sensitivity of BRCA-driven OC to platinum-containing treatments as compared to the mutation-negative tumors. Interestingly, prolonged tumor responses were documented both for taxane-free schemes and for the combination of platinating drugs with paclitaxel [83–86].

Two independent large trials evaluated the efficacy of olaparib in BRCA-mutated OC patients, who experienced prior chemotherapy [87, 88]. Audeh et al. [87] observed objective response in 33% and stable disease in 36% of women receiving olaparib at dose 400 mg twice daily. Fong et al. [88] reported tumor response in 40% and disease stabilization in 6% patients, respectively; as expected, higher efficacy of olaparib was documented in those cases, which retained sensitivity to platinum-based therapy.

Other genes and other tumors

Hereditary BC research led to identification of several genes other than BRCA1 and BRCA2. CHEK2 appears to be the most studied gene of this class. It confers elevated risk of breast cancer, while its heterozygous occurrence among ovarian cancer patients is not elevated [89, 90]. CHEK2-mutated BC frequently express estrogen receptor [91, 92]. Inactivation of CHEK2 by RNA-interference increased cell sensitivity to PARP inhibition [93]. The only available clinical observation describes BC progression in 2 out of 3 CHEK2 carriers, who were treated by neoadjuvant single-agent epirubicin, while this outcome was rare (8/104, 8%) in the non-carriers [94].

PALB2 (Partner And Localizers of BRCA2) has been proven to cause breast and pancreatic hereditary cancer [95, 96]. In agreement with BRCA2-related function of the PALB2, pancreatic cancer xenografts obtained from a PALB2 carrier demonstrated pronounced sensitivity to cisplatin and mitomycin C [97]. Importantly, excellent concordance between in vitro and clinical data was observed for this patient: his poorly differentiated ductal adenocarcinoma of the pancreas failed standard gemcitabine therapy, but demonstrated durable tumor response after mitomycin C or cisplatin administration [97]. Increased drug sensitivity of pancreatic tumors obtained from BRCA2 carriers was described in several other case reports [98–100]. Therefore, hereditary pancreatic cancers have clearly more favorable pattern of drug response as compared to sporadic cases. Similarly, excellent treatment effect lasting for more than 10 years was documented for BRCA2-related advanced lung cancer, which was treated by mitomycin C, cisplatin, and vincristine [101]. Another BRCA2 carrier, who suffered from castration-resistant prostate cancer, showed durable marker response and resolution of bone metastases after the administration of olaparib [102]. Vesprini et al. [103] have described a case of metastatic BRCA2-related prostate cancer, which was treated by cisplatin after becoming insensitive to androgen ablation. This therapy resulted in normalization of prostate-specific antigen level and symptomatic relief for period of 8 months; docetaxel was administered after the disease progression, and also led to an evident tumor response.

Sokolenko et al. [104] have recently revealed a role of BLM gene mutations in hereditary predisposition to breast cancer. This study included 5 patients treated by conventional neoadjuvant therapy; nearly complete pathological response was observed in 3 cases, while the remaining 2 women showed partial reduction of the tumor mass.

Preclinical data suggest specific drug sensitivity pattern for the cells with inactivated NBN (NBS1) and BRIP1 (FANCJ, BACH1) genes [29, 93]. It may turn to be difficult to validate these findings in the clinical setting, due to rarity and population-specific distribution of mutations in the mentioned genes.

Hereditary non-polyposis colorectal cancer

Hereditary non-polyposis colorectal cancer (HNPCC) is caused by germ-line mutations in MLH1, MSH2, PMS2 and MSH6 genes. Virtually all tumors from HNPCC mutation carriers are characterized by the defect of mismatch repair (MMR), which is manifested by so-called high-level microsatellite instability (MSI-H). MSI-H occurs in up to 15% of colorectal cancers (CRC), however the majority of the microsatellite-unstable carcinomas are sporadic; hereditary CRC constitute approximately one fifth of MSI-H cases and account for only 2-3% of the total CRC incidence. Mutations in HNPCC-related genes may also predispose to a number of non-colonic tumors, including endometrial, gastric, urothelial, ovarian and some other neoplasms [105, 106].

Given the rarity of hereditary CRC and the requirement of expensive multigene test for its definite diagnosis, the collection of clinical series for this disease represents a challenge. Instead, there is an intensive research focusing on MSI-H tumors as a distinct CRC entity; although sporadic and hereditary MSI-H CRC tumors share essential bioclinical features, many experts warn against combined analysis of these two tumor subsets. It is emphasized, that while hereditary CRC affect relatively young subjects, sporadic MSI-H cases are accumulated among elderly patients. Hereditary CRC arise due to mutational inactivation of the MLH1, MSH2, PMS2 or MSH6; sporadic MSI-H tumors are usually driven by methylation of the MLH1 gene promoter that may be a consequence of wide-spread abnormalities of epigenetic regulation ("methylator phenotype"). For unknown reason, BRAF mutations occur only in sporadic but not in hereditary MSI-H tumors [106, 107].

Although MSI-H tumors tend to be poorly differentiated, they are usually characterized by favorable disease course. In particular, MSI-H tumors show relatively low relapse rates after potentially curative surgery [108, 109]; in accordance with this, only 4% of advanced CRC have MSI-H phenotype [110]. As result, MSI-H cases are exceptionally rare in trials involving metastatic CRC, therefore the direct clinical assessment of their chemosensitivity is highly complicated. The majority of treatment response data for MSI-H cases is derived from the adjuvant trials, where the reliable discrimination between prognostic and predictive significance of a given parameter is not always possible. Another critical issue concerns technical aspects of determination of microsatellite instability. The existing approaches for detection of MSI-H phenotype are not fully standardized and may be a subject of significant interlaboratory variations [111]. In particular, there is a debate concerning the inclusion of dinucleotide microsatellite markers in the "Bethesda panel", which is frequently used for MSI-H diagnosis [112]. Many opinion leaders insist, that only mononucleotide markers (e.g., BAT26) allow to differentiate between true MSI-H and irrelevant mutational noise; hence, consideration of dinucleotide loci may increase the frequency of false-positive MSI-H detection and further compromise the conclusions of clinical trials [111].

Sensitivity of MMR-deficient cells to various anticancer drugs has been a subject of multiple laboratory studies. It is important to acknowledge, that naturally occurring MSI-H cancer cells have highly increased mutation rate and therefore accumulate significant number of "secondary" genetic lesions; depending of the spectrum of the target genes, these secondary lesions may substantially modify the response to treatment modalities [110, 113]. Furthermore, inactivation of distinct MMR genes, e.g. MSH2 and MLH1, may result in distinct patterns of chemosensitivity [114].

Most of preclinical studies point at resistance of MSI-H cells to 5-fluorouracil (5-FU) [110, 115]. MSI-H status is also associated with low sensitivity to cisplatin, carboplatin, 6-thioguanine, however these compounds are anyway not engaged in routine CRC treatment [110, 115–117]. While MMR-deficiency is associated with non-response to cisplatin and carboplatin, the third platinating drug, oxaliplatin, does not require functional MMR for its action [118]. Several studies have demonstrated specific sensitivity of MSI-H cells to irinotecan; it has been shown, that the response to topoisomerase I poisons may be mediated by the presence of secondary mutations in the double strand break response genes MRE11 and RAD50 [113]. Screen of the library of cytotoxic drugs has identified methotrexate as selective inhibitor of MSH2-deficient cells; MLH1-defective cells did not show specific sensitivity to this compound [114]. Martin et al. [119, 120] have recently identified PINK1 kinase and several DNA polymerases as potential targets in cells with mutated MMR genes.

There is a good consensus in the literature that MSI-H CRC patients do not benefit from 5-fluorouracil based adjuvant therapy [121]. Some reports have suggested even worse outcome in treated vs. non-treated patients; it is speculated that ineffective adjuvant therapy may compromise natural immune response to MSI-H cells [110, 122, 123]. One of the adjuvant patient series specifically included hereditary CRC cases, and also demonstrated lack of benefit from 5-fluorouracil [124]. Given an improved prognosis of MSI-H tumors, it is generally agreed that adjuvant therapy should be omitted for the stage II microsatellite unstable CRC [110, 125, 126]. Combination of 5-fluorouracil with oxaliplatin has been recently incorporated in the guidelines for adjuvant treatment of stage III CRC; as only a few MSI-H patients with follow-up are currently available, it is impossible to draw conclusions from the existing data sets [127, 128]. Trials with irinotecan did not qualify this drug for the use in adjuvant setting; however, the analysis of subset of patients with MSI-H has demonstrated, that this specific category of CRC patients may benefit from addition of irinotecan to fluorouracil and leucovorin [129].

Data on the use of chemotherapy for advanced MSI-H CRC are limited by a few small studies. Liang et al. [130] and Brueckl et al. [131] reported improved response of microsatellite unstable CRC to the 5-fluorouracil-based therapy. There is conflicting information regarding the role of MSI status in determining response to the combination of 5-fluorouracil and oxaliplatin combination [132–134]. Several reports suggested increased response rate of MSI-H CRC to irinotecan [135–137], although this statement was disputed by the recent study of Kim et al. [138].

Burn et al. [139] analyzed the effect of aspirin and resistant starch, given either alone or in combination, on the occurrence of colorectal neoplasia in the MLH1, MSH2 or MSH6 mutation carriers. Despite encouraging preclinical and epidemiological evidence, neither of these compounds influenced the risk of adenoma formation during the four years of the study.

Familial adenomatous polyposis

Familial adenomatous polyposis (FAP) is manifested by multiple polyps, which cause severe gastrointestinal symptoms and frequently progress into cancer lesions. Classical FAP is caused by a dominant germ-line mutation of the APC gene. Some patients bear an attenuated form of this disease; mild manifestation of FAP may indicate the involvement of another genetic lesion, i.e. homozygous inactivation of MUTYH gene [140]. Development of colonic adenomas usually involves activation of cyclooxygenase 2. Clinical trial involving the specific inhibitor of this enzyme, celecoxib (Onsenal), demonstrated 28% reduction of the number of polyps and 30.7% reduction of the sum of polyp diameters in patients receiving this drug at 400 mg twice daily for 6 months [141]. Based on the results of this trial, celecoxib has been approved for the treatment of FAP. However the safety of its long term-use is questioned by reports revealing elevated rate of cardiovascular events in patients receiving therapeutically effective dose of the drug [142]. Earlier studies demonstrated beneficial effect of sulindac, a non-steroidal anti-inflammatory drug; the results of these trials may be revisited, given that the main adverse effect of this drug, i.e. gastrointestinal toxicity, is medically manageable [143].

Hereditary medullary thyroid cancer

Hereditary medullary thyroid cancer (MTC) is caused by germ-line mutation in RET tyrosine kinase. It can be a part of multiple endocrine neoplasia (MEN) type 2A (MEN2A) or type 2B (MEN2B) syndromes, or manifest as a single-organ lesion (familial medullary thyroid cancer, FMTC) [144]. A novel multitargeted tyrosine kinase inhibitor vandetanib (ZD6474, Zactima) demonstrates specific activity against mutated RET and inhibits growth of RET-transformed cancer cells [145]. A clinical trial involving 30 patients with hereditary MTC, who received 300 mg vandetanib daily, demonstrated objective tumor response in 6/30 (20%) and disease stabilization for more than 24 weeks in 16/30 (53%) cases, respectively [146]. Precise measurement of the change of tumor size revealed the reduction of the lesions in 25/30 (83%) patients; the estimated median progression-free survival approached to 27.9 months [146]. Comparable results were obtained in another trial, which utilized 100 mg daily dosage of this drug [147].

Sporadic phenocopies of hereditary cancers

Hereditary neoplasms make relatively little contribution in the total cancer incidence. Nevertheless, advances in the treatment of this category of tumors may have broader practical implications, as many sporadic tumors develop phenotype similar to hereditary cancers. This issue was particularly intensively discussed in breast cancer research, owing to substantial overlap between BRCA1-related and triple-negative BC [14]. Given that BRCA1 may be inactivated not only by germ-line but also by somatic alterations, several investigations suggested to use BRCA1 expression as predictive marker of response to platinum-based and taxane-based therapy [78, 148–152]. Other approaches are based on the detection of consequences of either BRCA deficiency or other critical defects of homologous recombination; in particular, it has been observed that tumors with presumably impaired repair of DNA double-strand breaks show characteristic pattern of acquired mutations [153, 154]. Similarly to BRCA1, the mutations of RET oncogene are observed not only in hereditary, but also in sporadic medullary thyroid carcinomas; it is expected, that at least a subset of RET-driven non-hereditary MTC should respond to vandetanib therapy [144]. While for some tumor types clinical experience is translated from familial cancers to their phenocopies, the reverse flow is observed in colorectal cancer research; as already mentioned above, virtually all data on drug response have been obtained not on a genuine hereditary CRC, but on its phenocopy, i.e. MSI-H tumors; this limitation has to be considered by medical oncologists [106, 107].

Conclusions and perspectives

Patients with hereditary tumors often benefit from distinct drugs as compared to sporadic cases. The detection of cancer-predisposing germ-line mutations among the participants of clinical trials has rarely been considered, due to significant cost of genetic testing. Given the rapidly increasing accessibility of DNA analysis, it is foreseen that a large number of germ-line mutation carriers will be included in forthcoming trials and/or identified within retrospective collections of biological material. The analysis of correlations between genotype and drug response may substantially improve treatment outcomes, both for hereditary cancer patients and for subjects bearing phenocopies of familial tumors.

Abbreviations

AC:

doxorubicin and cyclophosphamide

BC:

breast cancer

CMF:

cyclophosphamide, methotrexate and fluorouracil

cCR:

clinical complete response

CR:

complete response

CRC:

colorectal cancer

FAC:

5-fluorouracil, doxorubicin and cyclophosphamide

FAP:

familial adenomatous polyposis

FEC:

5-fluorouracil, epirubicin and cyclophosphamide

HNPCC:

hereditary non-polyposis colorectal cancer

HR:

homologous recombination

MMR:

mismatch repair

MTC:

medullary thyroid cancer

MSI-H:

high-level microsatellite instability

OC:

ovarian cancer

OR:

objective response

OS:

overall survival

PARP:

poly (ADP-ribose) polymerase

pCR:

pathological complete response

PD:

progressive disease

PFS:

progression-free survival

PR:

partial response

SD:

stable disease.

References

  1. Bermejo-Pérez MJ, Márquez-Calderón S, Llanos-Méndez A: Effectiveness of preventive interventions in BRCA1/2 gene mutation carriers: a systematic review. Int J Cancer 2007, 121: 225–231. 10.1002/ijc.22817

    PubMed  Google Scholar 

  2. Domchek SM, Friebel TM, Singer CF, Evans DG, Lynch HT, Isaacs C, Garber JE, Neuhausen SL, Matloff E, Eeles R, Pichert G, Van t'veer L, Tung N, Weitzel JN, Couch FJ, Rubinstein WS, Ganz PA, Daly MB, Olopade OI, Tomlinson G, Schildkraut J, Blum JL, Rebbeck TR: Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA 2010, 304: 967–975. 10.1001/jama.2010.1237

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kurian AW: BRCA1 and BRCA2 mutations across race and ethnicity: distribution and clinical implications. Curr Opin Obstet Gynecol 2010, 22: 72–78. 10.1097/GCO.0b013e328332dca3

    PubMed  Google Scholar 

  4. Fasano J, Muggia F: Breast cancer arising in a BRCA-mutated background: therapeutic implications from an animal model and drug development. Ann Oncol 2009, 20: 609–614. 10.1093/annonc/mdn669

    CAS  PubMed  Google Scholar 

  5. Trainer AH, Lewis CR, Tucker K, Meiser B, Friedlander M, Ward RL: The role of BRCA mutation testing in determining breast cancer therapy. Nat Rev Clin Oncol 2010, 7: 708–717. 10.1038/nrclinonc.2010.175

    CAS  PubMed  Google Scholar 

  6. van Beers EH, van Welsem T, Wessels LF, Li Y, Oldenburg RA, Devilee P, Cornelisse CJ, Verhoef S, Hogervorst FB, van't Veer LJ, Nederlof PM: Comparative genomic hybridization profiles in human BRCA1 and BRCA2 breast tumors highlight differential sets of genomic aberrations. Cancer Res 2005, 65: 822–827.

    CAS  PubMed  Google Scholar 

  7. Joosse SA, Brandwijk KI, Mulder L, Wesseling J, Hannemann J, Nederlof PM: Genomic signature of BRCA1 deficiency in sporadic basal-like breast tumors. Genes Chromosomes Cancer 2011, 50: 71–81. 10.1002/gcc.20833

    CAS  PubMed  Google Scholar 

  8. Lakhani SR, Manek S, Penault-Llorca F, Flanagan A, Arnout L, Merrett S, McGuffog L, Steele D, Devilee P, Klijn JG, Meijers-Heijboer H, Radice P, Pilotti S, Nevanlinna H, Butzow R, Sobol H, Jacquemier J, Lyonet DS, Neuhausen SL, Weber B, Wagner T, Winqvist R, Bignon YJ, Monti F, Schmitt F, Lenoir G, Seitz S, Hamman U, Pharoah P, Lane G, Ponder B, Bishop DT, Easton DF: Pathology of ovarian cancers in BRCA1 and BRCA2 carriers. Clin Cancer Res 2004, 10: 2473–2481. 10.1158/1078-0432.CCR-1029-3

    CAS  PubMed  Google Scholar 

  9. Turner N, Tutt A, Ashworth A: Hallmarks of 'BRCAness' in sporadic cancers. Nat Rev Cancer 2004, 4: 814–819. 10.1038/nrc1457

    CAS  PubMed  Google Scholar 

  10. Bane AL, Beck JC, Bleiweiss I, Buys SS, Catalano E, Daly MB, Giles G, Godwin AK, Hibshoosh H, Hopper JL, John EM, Layfield L, Longacre T, Miron A, Senie R, Southey MC, West DW, Whittemore AS, Wu H, Andrulis IL, O'Malley FP: BRCA2 mutation-associated breast cancers exhibit a distinguishing phenotype based on morphology and molecular profiles from tissue microarrays. Am J Surg Pathol 2007, 31: 121–128. 10.1097/01.pas.0000213351.49767.0f

    PubMed  Google Scholar 

  11. Stefansson OA, Jonasson JG, Johannsson OT, Olafsdottir K, Steinarsdottir M, Valgeirsdottir S, Eyfjord JE: Genomic profiling of breast tumours in relation to BRCA abnormalities and phenotypes. Breast Cancer Res 2009, 11: R47. 10.1186/bcr2334

    PubMed  PubMed Central  Google Scholar 

  12. Yarden RI, Papa MZ: BRCA1 at the crossroad of multiple cellular pathways: approaches for therapeutic interventions. Mol Cancer Ther 2006, 5: 1396–1404. 10.1158/1535-7163.MCT-05-0471

    CAS  PubMed  Google Scholar 

  13. Holstege H, Joosse SA, van Oostrom CT, Nederlof PM, de Vries A, Jonkers J: High incidence of protein-truncating TP53 mutations in BRCA1-related breast cancer. Cancer Res 2009, 69: 3625–3633. 10.1158/0008-5472.CAN-08-3426

    CAS  PubMed  Google Scholar 

  14. Jaspers JE, Rottenberg S, Jonkers J: Therapeutic options for triple-negative breast cancers with defective homologous recombination. Biochim Biophys Acta 2009, 1796: 266–280.

    CAS  PubMed  Google Scholar 

  15. Hennessy BT, Timms KM, Carey MS, Gutin A, Meyer LA, Flake DD, Abkevich V, Potter J, Pruss D, Glenn P, Li Y, Li J, Gonzalez-Angulo AM, McCune KS, Markman M, Broaddus RR, Lanchbury JS, Lu KH, Mills GB: Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol 2010, 28: 3570–3576. 10.1200/JCO.2009.27.2997

    PubMed  PubMed Central  Google Scholar 

  16. Gudmundsdottir K, Ashworth A: The roles of BRCA1 and BRCA2 and associated proteins in the maintenance of genomic stability. Oncogene 2006, 25: 5864–5874. 10.1038/sj.onc.1209874

    CAS  PubMed  Google Scholar 

  17. Tutt A, Ashworth A: Can genetic testing guide treatment in breast cancer? Eur J Cancer 2008, 44: 2774–2780. 10.1016/j.ejca.2008.10.009

    CAS  PubMed  Google Scholar 

  18. Hosey AM, Gorski JJ, Murray MM, Quinn JE, Chung WY, Stewart GE, James CR, Farragher SM, Mulligan JM, Scott AN, Dervan PA, Johnston PG, Couch FJ, Daly PA, Kay E, McCann A, Mullan PB, Harkin DP: Molecular basis for estrogen receptor alpha deficiency in BRCA1-linked breast cancer. J Natl Cancer Inst 2007, 99: 1683–1694. 10.1093/jnci/djm207

    CAS  PubMed  Google Scholar 

  19. Atchley DP, Albarracin CT, Lopez A, Valero V, Amos CI, Gonzalez-Angulo AM, Hortobagyi GN, Arun BK: Clinical and pathologic characteristics of patients with BRCA-positive and BRCA-negative breast cancer. J Clin Oncol 2008, 26: 4282–4288. 10.1200/JCO.2008.16.6231

    PubMed  Google Scholar 

  20. DeLigio JT, Zorio DA: Early growth response 1 (EGR1): a gene with as many names as biological functions. Cancer Biol Ther 2009, 8: 1889–1892. 10.4161/cbt.8.20.9804

    CAS  PubMed  Google Scholar 

  21. Husain A, He G, Venkatraman ES, Spriggs DR: BRCA1 up-regulation is associated with repair-mediated resistance to cis-diamminedichloroplatinum(II). Cancer Res 1998,58(6):1120–1123.

    CAS  PubMed  Google Scholar 

  22. Bhattacharyya A, Ear US, Koller BH, Weichselbaum RR, Bishop DK: The breast cancer susceptibility gene BRCA1 is required for subnuclear assembly of Rad51 and survival following treatment with the DNA cross-linking agent cisplatin. J Biol Chem 2000, 275: 23899–23903. 10.1074/jbc.C000276200

    CAS  PubMed  Google Scholar 

  23. Lafarge S, Sylvain V, Ferrara M, Bignon YJ: Inhibition of BRCA1 leads to increased chemoresistance to microtubule-interfering agents, an effect that involves the JNK pathway. Oncogene 2001, 20: 6597–6606. 10.1038/sj.onc.1204812

    CAS  PubMed  Google Scholar 

  24. Fedier A, Steiner RA, Schwarz VA, Lenherr L, Haller U, Fink D: The effect of loss of Brca1 on the sensitivity to anticancer agents in p53-deficient cells. Int J Oncol 2003, 22: 1169–1173.

    CAS  PubMed  Google Scholar 

  25. Quinn JE, Kennedy RD, Mullan PB, Gilmore PM, Carty M, Johnston PG, Harkin DP: BRCA1 functions as a differential modulator of chemotherapy-induced apoptosis. Cancer Res 2003, 63: 6221–6228.

    CAS  PubMed  Google Scholar 

  26. Tassone P, Tagliaferri P, Perricelli A, Blotta S, Quaresima B, Martelli ML, Goel A, Barbieri V, Costanzo F, Boland CR, Venuta S: BRCA1 expression modulates chemosensitivity of BRCA1-defective HCC1937 human breast cancer cells. Br J Cancer 2003, 88: 1285–1291. 10.1038/sj.bjc.6600859

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Samouëlian V, Maugard CM, Jolicoeur M, Bertrand R, Arcand SL, Tonin PN, Provencher DM, Mes-Masson AM: Chemosensitivity and radiosensitivity profiles of four new human epithelial ovarian cancer cell lines exhibiting genetic alterations in BRCA2, TGFbeta-RII, KRAS2, TP53 and/or CDNK2A. Cancer Chemother Pharmacol 2004, 54: 497–504. 10.1007/s00280-004-0843-9

    PubMed  Google Scholar 

  28. van der Heijden MS, Brody JR, Gallmeier E, Cunningham SC, Dezentje DA, Shen D, Hruban RH, Kern SE: Functional defects in the fanconi anemia pathway in pancreatic cancer cells. Am J Pathol 2004, 165: 651–657. 10.1016/S0002-9440(10)63329-9

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Bartz SR, Zhang Z, Burchard J, Imakura M, Martin M, Palmieri A, Needham R, Guo J, Gordon M, Chung N, Warrener P, Jackson AL, Carleton M, Oatley M, Locco L, Santini F, Smith T, Kunapuli P, Ferrer M, Strulovici B, Friend SH, Linsley PS: Small interfering RNA screens reveal enhanced cisplatin cytotoxicity in tumor cells having both BRCA network and TP53 disruptions. Mol Cell Biol 2006, 26: 9377–9386. 10.1128/MCB.01229-06

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Xing D, Orsulic S: A mouse model for the molecular characterization of brca1-associated ovarian carcinoma. Cancer Res 2006, 66: 8949–8953. 10.1158/0008-5472.CAN-06-1495

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ, Bontcheva-Diaz VD, Cox BF, DeWeese TL, Dillehay LE, Ferguson DC, Ghoreishi-Haack NS, Grimm DR, Guan R, Han EK, Holley-Shanks RR, Hristov B, Idler KB, Jarvis K, Johnson EF, Kleinberg LR, Klinghofer V, Lasko LM, Liu X, Marsh KC, McGonigal TP, Meulbroek JA, Olson AM, Palma JP, et al.: ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res 2007, 13: 2728–2737. 10.1158/1078-0432.CCR-06-3039

    CAS  PubMed  Google Scholar 

  32. Rottenberg S, Nygren AO, Pajic M, van Leeuwen FW, van der Heijden I, van de Wetering K, Liu X, de Visser KE, Gilhuijs KG, van Tellingen O, Schouten JP, Jonkers J, Borst P: Selective induction of chemotherapy resistance of mammary tumors in a conditional mouse model for hereditary breast cancer. Proc Natl Acad Sci USA 2007, 104: 12117–12122. 10.1073/pnas.0702955104

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Evers B, Drost R, Schut E, de Bruin M, van der Burg E, Derksen PW, Holstege H, Liu X, van Drunen E, Beverloo HB, Smith GC, Martin NM, Lau A, O'Connor MJ, Jonkers J: Selective inhibition of BRCA2-deficient mammary tumor cell growth by AZD2281 and cisplatin. Clin Cancer Res 2008, 14: 3916–3925. 10.1158/1078-0432.CCR-07-4953

    CAS  PubMed  Google Scholar 

  34. Shafee N, Smith CR, Wei S, Kim Y, Mills GB, Hortobagyi GN, Stanbridge EJ, Lee EY: Cancer stem cells contribute to cisplatin resistance in Brca1/p53-mediated mouse mammary tumors. Cancer Res 2008, 68: 3243–3250. 10.1158/0008-5472.CAN-07-5480

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Hay T, Matthews JR, Pietzka L, Lau A, Cranston A, Nygren AO, Douglas-Jones A, Smith GC, Martin NM, O'Connor M, Clarke AR: Poly(ADP-ribose) polymerase-1 inhibitor treatment regresses autochthonous Brca2/p53-mutant mammary tumors in vivo and delays tumor relapse in combination with carboplatin. Cancer Res 2009, 69: 3850–3855. 10.1158/0008-5472.CAN-08-2388

    CAS  PubMed  Google Scholar 

  36. Santarosa M, Del Col L, Tonin E, Caragnano A, Viel A, Maestro R: Premature senescence is a major response to DNA cross-linking agents in BRCA1-defective cells: implication for tailored treatments of BRCA1 mutation carriers. Mol Cancer Ther 2009, 8: 844–854.

    CAS  PubMed  Google Scholar 

  37. Tassone P, Di Martino MT, Ventura M, Pietragalla A, Cucinotto I, Calimeri T, Bulotta A, Neri P, Caraglia M, Tagliaferri P: Loss of BRCA1 function increases the antitumor activity of cisplatin against human breast cancer xenografts in vivo. Cancer Biol Ther 2009, 8: 648–653.

    CAS  PubMed  Google Scholar 

  38. Evers B, Schut E, van der Burg E, Braumuller TM, Egan DA, Holstege H, Edser P, Adams DJ, Wade-Martins R, Bouwman P, Jonkers J: A high-throughput pharmaceutical screen identifies compounds with specific toxicity against BRCA2-deficient tumors. Clin Cancer Res 2010, 16: 99–108. 10.1158/1078-0432.CCR-09-2434

    CAS  PubMed  Google Scholar 

  39. Drew Y, Mulligan EA, Vong WT, Thomas HD, Kahn S, Kyle S, Mukhopadhyay A, Los G, Hostomsky Z, Plummer ER, Edmondson RJ, Curtin NJ: Therapeutic potential of poly(ADP-ribose) polymerase inhibitor AG014699 in human cancers with mutated or methylated BRCA1 or BRCA2. J Natl Cancer Inst 2011, 103: 334–346. 10.1093/jnci/djq509

    CAS  PubMed  Google Scholar 

  40. Kortmann U, McAlpine JN, Xue H, Guan J, Ha G, Tully S, Shafait S, Lau A, Cranston AN, O'Connor MJ, Huntsman DG, Wang Y, Gilks CB: Tumor growth inhibition by olaparib in BRCA2 germline-mutated patient-derived ovarian cancer tissue xenografts. Clin Cancer Res 2011, 17: 783–791. 10.1158/1078-0432.CCR-10-1382

    CAS  PubMed  Google Scholar 

  41. Yu VP, Koehler M, Steinlein C, Schmid M, Hanakahi LA, van Gool AJ, West SC, Venkitaraman AR: Gross chromosomal rearrangements and genetic exchange between nonhomologous chromosomes following BRCA2 inactivation. Genes Dev 2000, 14: 1400–1406.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Moynahan ME, Cui TY, Jasin M: Homology-directed dna repair, mitomycin-c resistance, and chromosome stability is restored with correction of a Brca1 mutation. Cancer Res 2001, 61: 4842–4850.

    CAS  PubMed  Google Scholar 

  43. van der Heijden MS, Brody JR, Dezentje DA, Gallmeier E, Cunningham SC, Swartz MJ, DeMarzo AM, Offerhaus GJ, Isacoff WH, Hruban RH, Kern SE: In vivo therapeutic responses contingent on Fanconi anemia/BRCA2 status of the tumor. Clin Cancer Res 2005, 11: 7508–7515. 10.1158/1078-0432.CCR-05-1048

    CAS  PubMed  Google Scholar 

  44. Yun J, Zhong Q, Kwak JY, Lee WH: Hypersensitivity of Brca1-deficient MEF to the DNA interstrand crosslinking agent mitomycin C is associated with defect in homologous recombination repair and aberrant S-phase arrest. Oncogene 2005, 24: 4009–4016.

    CAS  PubMed  Google Scholar 

  45. Brodie SG, Xu X, Qiao W, Li WM, Cao L, Deng CX: Multiple genetic changes are associated with mammary tumorigenesis in Brca1 conditional knockout mice. Oncogene 2001, 20: 7514–7523. 10.1038/sj.onc.1204929

    CAS  PubMed  Google Scholar 

  46. Issaeva N, Thomas HD, Djureinovic T, Jaspers JE, Stoimenov I, Kyle S, Pedley N, Gottipati P, Zur R, Sleeth K, Chatzakos V, Mulligan EA, Lundin C, Gubanova E, Kersbergen A, Harris AL, Sharma RA, Rottenberg S, Curtin NJ, Helleday T: 6-thioguanine selectively kills BRCA2-defective tumors and overcomes PARP inhibitor resistance. Cancer Res 2010, 70: 6268–6276. 10.1158/0008-5472.CAN-09-3416

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Abbott DW, Freeman ML, Holt JT: Double-strand break repair deficiency and radiation sensitivity in BRCA2 mutant cancer cells. J Natl Cancer Inst 1998, 90: 978–985. 10.1093/jnci/90.13.978

    CAS  PubMed  Google Scholar 

  48. Treszezamsky AD, Kachnic LA, Feng Z, Zhang J, Tokadjian C, Powell SN: BRCA1- and BRCA2-deficient cells are sensitive to etoposide-induced DNA double-strand breaks via topoisomerase II. Cancer Res 2007, 67: 7078–7081. 10.1158/0008-5472.CAN-07-0601

    CAS  PubMed  Google Scholar 

  49. Mullan PB, Quinn JE, Gilmore PM, McWilliams S, Andrews H, Gervin C, McCabe N, McKenna S, White P, Song YH, Maheswaran S, Liu E, Haber DA, Johnston PG, Harkin DP: BRCA1 and GADD45 mediated G2/M cell cycle arrest in response to antimicrotubule agents. Oncogene 2001, 20: 6123–6131. 10.1038/sj.onc.1204712

    CAS  PubMed  Google Scholar 

  50. Chabalier C, Lamare C, Racca C, Privat M, Valette A, Larminat F: BRCA1 downregulation leads to premature inactivation of spindle checkpoint and confers paclitaxel resistance. Cell Cycle 2006, 5: 1001–1007. 10.4161/cc.5.9.2726

    CAS  PubMed  Google Scholar 

  51. Promkan M, Liu G, Patmasiriwat P, Chakrabarty S: BRCA1 modulates malignant cell behavior, the expression of survivin and chemosensitivity in human breast cancer cells. Int J Cancer 2009, 125: 2820–2828. 10.1002/ijc.24684

    CAS  PubMed  Google Scholar 

  52. Zhou C, Smith JL, Liu J: Role of BRCA1 in cellular resistance to paclitaxel and ionizing radiation in an ovarian cancer cell line carrying a defective BRCA1. Oncogene 2003, 22: 2396–2404. 10.1038/sj.onc.1206319

    CAS  PubMed  Google Scholar 

  53. Tassone P, Blotta S, Palmieri C, Masciari S, Quaresima B, Montagna M, D'Andrea E, Eramo OP, Migale L, Costanzo F, Tagliaferri P, Venuta S: Differential sensitivity of BRCA1-mutated HCC1937 human breast cancer cells to microtubule-interfering agents. Int J Oncol 2005, 26: 1257–1263.

    CAS  PubMed  Google Scholar 

  54. Zander SA, Kersbergen A, van der Burg E, de Water N, van Tellingen O, Gunnarsdottir S, Jaspers JE, Pajic M, Nygren AO, Jonkers J, Borst P, Rottenberg S: Sensitivity and acquired resistance of BRCA1;p53-deficient mouse mammary tumors to the topoisomerase I inhibitor topotecan. Cancer Res 2010, 70: 1700–1710. 10.1158/0008-5472.CAN-09-3367

    CAS  PubMed  Google Scholar 

  55. Rahden-Staroń I, Szumił M, Grosicka E, Kraakman van der Zwet M, Zdzienicka MZ: Defective Brca2 influences topoisomerase I activity in mammalian cells. Acta Biochim Pol 2003, 50: 139–144.

    PubMed  Google Scholar 

  56. Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ, Helleday T: Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005, 434: 913–917. 10.1038/nature03443

    CAS  PubMed  Google Scholar 

  57. Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, Martin NM, Jackson SP, Smith GC, Ashworth A: Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434: 917–921. 10.1038/nature03445

    CAS  PubMed  Google Scholar 

  58. McCabe N, Lord CJ, Tutt AN, Martin NM, Smith GC, Ashworth A: BRCA2-deficient CAPAN-1 cells are extremely sensitive to the inhibition of Poly (ADP-Ribose) polymerase: an issue of potency. Cancer Biol Ther 2005, 4: 934–936. 10.4161/cbt.4.9.2141

    CAS  PubMed  Google Scholar 

  59. Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AO, Zander SA, Derksen PW, de Bruin M, Zevenhoven J, Lau A, Boulter R, Cranston A, O'Connor MJ, Martin NM, Borst P, Jonkers J: High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci USA 2008, 105: 17079–17084. 10.1073/pnas.0806092105

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Goldberg MS, Xing D, Ren Y, Orsulic S, Bhatia SN, Sharp PA: Nanoparticle-mediated delivery of siRNA targeting Parp1 extends survival of mice bearing tumors derived from Brca1-deficient ovarian cancer cells. Proc Natl Acad Sci USA 2011, 108: 745–750. 10.1073/pnas.1016538108

    CAS  PubMed  Google Scholar 

  61. Ashworth A: Drug resistance caused by reversion mutation. Cancer Res 2008, 68: 10021–10023. 10.1158/0008-5472.CAN-08-2287

    CAS  PubMed  Google Scholar 

  62. Underhill C, Toulmonde M, Bonnefoi H: A review of PARP inhibitors: from bench to bedside. Ann Oncol 2011, 22: 268–279. 10.1093/annonc/mdq322

    CAS  PubMed  Google Scholar 

  63. Narod SA: BRCA mutations in the management of breast cancer: the state of the art. Nat Rev Clin Oncol 2010, 7: 702–707. 10.1038/nrclinonc.2010.166

    CAS  PubMed  Google Scholar 

  64. Foulkes WD, Goffin J, Brunet JS, Bégin LR, Wong N, Chappuis PO: Tamoxifen may be an effective adjuvant treatment for BRCA1-related breast cancer irrespective of estrogen receptor status. J Natl Cancer Inst 2002, 94: 1504–1506.

    PubMed  Google Scholar 

  65. Delaloge S, Pélissier P, Kloos I, Bressac de Paillerets B, Mathieu MC, Chompret A, Noguès C, Lortholary A, Piketty AC, Spielmann M: BRCA1-linked breast cancer (BC) is highly more chemosensitive than its BRCA2-linked or sporadic counterparts. Ann Oncol 2002,13(Suppl 5):34.

    Google Scholar 

  66. Chappuis PO, Goffin J, Wong N, Perret C, Ghadirian P, Tonin PN, Foulkes WD: A significant response to neoadjuvant chemotherapy in BRCA1/2 related breast cancer. J Med Genet 2002, 39: 608–610. 10.1136/jmg.39.8.608

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Warner E, Trudeau M, Holloway C: Sensitivity of BRCA-1-related breast cancer to neoadjuvant chemotherapy: practical implications. Breast J 2003, 9: 507–508. 10.1046/j.1524-4741.2003.09624.x

    PubMed  Google Scholar 

  68. Hubert A, Mali B, Hamburger T, Rottenberg Y, Uziely B, Peretz T, Kadouri L: Response to neo-adjuvant chemotherapy in BRCA1 and BRCA2 related stage III breast cancer. Fam Cancer 2009, 8: 173–177. 10.1007/s10689-008-9223-7

    CAS  PubMed  Google Scholar 

  69. Fourquet A, Stoppa-Lyonnet D, Kirova YM, Sigal-Zafrani B, Asselain B, Institut Curie Breast Cancer Study Group; Institut Curie Breast Ovary Cancer Risk Study Group: Familial breast cancer: clinical response to induction chemotherapy or radiotherapy related to BRCA1/2 mutations status. Am J Clin Oncol 2009, 32: 127–131. 10.1097/COC.0b013e31817f9e1c

    CAS  PubMed  Google Scholar 

  70. Byrski T, Gronwald J, Huzarski T, Grzybowska E, Budryk M, Stawicka M, Mierzwa T, Szwiec M, Wisniowski R, Siolek M, Dent R, Lubinski J, Narod S: Pathologic complete response rates in young women with BRCA1-positive breast cancers after neoadjuvant chemotherapy. J Clin Oncol 2010, 28: 375–379. 10.1200/JCO.2008.20.7019

    CAS  PubMed  Google Scholar 

  71. Wong Wong Keet A, Al-Rafae M, Chappuis PO, Brunet JS, Ghadirian P, Foulkes WD: Long-term outcome after neo-adjuvant chemotherapy for breast cancer in BRCA1/2 carriers. Int J Cancer 2009, 125: 2236–2238. 10.1002/ijc.24596

    PubMed  Google Scholar 

  72. Petit T, Wilt M, Rodier J, Muller D, Ghnassia J, Dufour P, Fricker J: Are BRCA1 mutations a predictive factor for anthracycline-based neoadjuvant chemotherapy response in triple-negative breast cancers? J Clin Oncol 2007,25(Suppl):abstract 580.

    Google Scholar 

  73. Kriege M, Seynaeve C, Meijers-Heijboer H, Collee JM, Menke-Pluymers MB, Bartels CC, Tilanus-Linthorst MM, Blom J, Huijskens E, Jager A, van den Ouweland A, van Geel B, Hooning MJ, Brekelmans CT, Klijn JG: Sensitivity to first-line chemotherapy for metastatic breast cancer in BRCA1 and BRCA2 mutation carriers. J Clin Oncol 2009, 27: 3764–3771. 10.1200/JCO.2008.19.9067

    PubMed  Google Scholar 

  74. Moiseyenko VM, Protsenko SA, Brezhnev NV, Maximov SY, Gershveld ED, Hudyakova MA, Lobeiko OS, Gergova MM, Krzhivitskiy PI, Semionov II, Matsko DE, Iyevleva AG, Sokolenko AP, Sherina NY, Kuligina ESh, Suspitsin EN, Togo AV, Imyanitov EN: High sensitivity of BRCA1-associated tumors to cisplatin monotherapy: report of two cases. Cancer Genet Cytogenet 2010, 197: 91–94. 10.1016/j.cancergencyto.2009.10.003

    CAS  PubMed  Google Scholar 

  75. Wysocki PJ, Korski K, Lamperska K, Zaluski J, Mackiewicz A: Primary resistance to docetaxel-based chemotherapy in metastatic breast cancer patients correlates with a high frequency of BRCA1 mutations. Med Sci Monit 2008, 14: SC7-SC10.

    CAS  PubMed  Google Scholar 

  76. Kriege M, Jager A, Hooning MJ, Huijskens E, Blom J, van Deurzen CH, Bontenbal M, Collee JM, Menke-Pluijmers MB, Martens JW, Seynaeve C: The efficacy of taxane chemotherapy for metastatic breast cancer in BRCA1 and BRCA2 mutation carriers. Cancer 2011, in press.

    Google Scholar 

  77. Imyanitov EN: Breast cancer therapy for BRCA1 carriers: moving towards platinum standard? Hered Cancer Clin Pract 2009, 7: 8. 10.1186/1897-4287-7-8

    PubMed  PubMed Central  Google Scholar 

  78. Silver DP, Richardson AL, Eklund AC, Wang ZC, Szallasi Z, Li Q, Juul N, Leong CO, Calogrias D, Buraimoh A, Fatima A, Gelman RS, Ryan PD, Tung NM, De Nicolo A, Ganesan S, Miron A, Colin C, Sgroi DC, Ellisen LW, Winer EP, Garber JE: Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol 2010, 28: 1145–1153. 10.1200/JCO.2009.22.4725

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Swisher EM, Sakai W, Karlan BY, Wurz K, Urban N, Taniguchi T: Secondary BRCA1 mutations in BRCA1-mutated ovarian carcinomas with platinum resistance. Cancer Res 2008, 68: 2581–2586. 10.1158/0008-5472.CAN-08-0088

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Huang F, Kushner YB, Langleben A, Foulkes WD, Medscape: Eleven years disease-free: role of chemotherapy in metastatic BRCA2-related breast cancer. Nat Rev Clin Oncol 2009, 6: 488–492. 10.1038/nrclinonc.2009.90

    CAS  PubMed  Google Scholar 

  81. Vollebergh MA, Nederlof PM, Wessels LF, Schmidt MK, Joosse SA, van Beers E, Froklage F, Holtkamp M, Schrama JG, Wesseling J, Hauptmann M, de Bruin M, Rodenhuis S, Linn SC: Predicting response to alkylating chemotherapy in breast cancer patients using array comparative genomic hybridization. Cancer Res 2009,69(Suppl 1):abstract 6050.

    Google Scholar 

  82. Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, Friedlander M, Arun B, Loman N, Schmutzler RK, Wardley A, Mitchell G, Earl H, Wickens M, Carmichael J: Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet 2010, 376: 235–244. 10.1016/S0140-6736(10)60892-6

    CAS  PubMed  Google Scholar 

  83. Cass I, Baldwin RL, Varkey T, Moslehi R, Narod SA, Karlan BY: Improved survival in women with BRCA-associated ovarian carcinoma. Cancer 2003, 97: 2187–2195. 10.1002/cncr.11310

    CAS  PubMed  Google Scholar 

  84. Tan DS, Rothermundt C, Thomas K, Bancroft E, Eeles R, Shanley S, Ardern-Jones A, Norman A, Kaye SB, Gore ME: "BRCAness" syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol 2008, 26: 5530–5536. 10.1200/JCO.2008.16.1703

    PubMed  Google Scholar 

  85. Vencken PM, Kriege M, Hoogwerf D, Beugelink S, van der Burg ME, Hooning MJ, Berns EM, Jager A, Collée M, Burger CW, Seynaeve C: Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol 2011, 22: 1346–1352. 10.1093/annonc/mdq628

    CAS  PubMed  Google Scholar 

  86. Melichar B, Fridrichová P, Tomsová M, Malírová E: Repeat chemosensitivity of epithelial ovarian carcinoma in a BRCA1 mutation carrier to paclitaxel/platinum combination chemotherapy. Eur J Gynaecol Oncol 2009, 30: 323–325.

    CAS  PubMed  Google Scholar 

  87. Audeh MW, Carmichael J, Penson RT, Friedlander M, Powell B, Bell-McGuinn KM, Scott C, Weitzel JN, Oaknin A, Loman N, Lu K, Schmutzler RK, Matulonis U, Wickens M, Tutt A: Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet 2010, 376: 245–251. 10.1016/S0140-6736(10)60893-8

    CAS  PubMed  Google Scholar 

  88. Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, De Greve J, Lubinski J, Shanley S, Messiou C, A'Hern R, Tutt A, Ashworth A, Stone J, Carmichael J, Schellens JH, de Bono JS, Kaye SB: Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol 2010, 28: 2512–2519. 10.1200/JCO.2009.26.9589

    CAS  PubMed  Google Scholar 

  89. Weischer M, Bojesen SE, Ellervik C, Tybjaerg-Hansen A, Nordestgaard BG: CHEK2*1100delC genotyping for clinical assessment of breast cancer risk: meta-analyses of 26,000 patient cases and 27,000 controls. J Clin Oncol 2008, 26: 542–548. 10.1200/JCO.2007.12.5922

    PubMed  Google Scholar 

  90. Suspitsin EN, Sherina NY, Ponomariova DN, Sokolenko AP, Iyevleva AG, Gorodnova TV, Zaitseva OA, Yatsuk OS, Togo AV, Tkachenko NN, Shiyanov GA, Lobeiko OS, Krylova NY, Matsko DE, Maximov SY, Urmancheyeva AF, Porhanova NV, Imyanitov EN: High frequency of BRCA1, but not CHEK2 or NBS1 (NBN), founder mutations in Russian ovarian cancer patients. Hered Cancer Clin Pract 2009, 7: 5. 10.1186/1897-4287-7-5

    PubMed  PubMed Central  Google Scholar 

  91. de Bock GH, Schutte M, Krol-Warmerdam EM, Seynaeve C, Blom J, Brekelmans CT, Meijers-Heijboer H, van Asperen CJ, Cornelisse CJ, Devilee P, Tollenaar RA, Klijn JG: Tumour characteristics and prognosis of breast cancer patients carrying the germline CHEK2*1100delC variant. J Med Genet 2004, 41: 731–735. 10.1136/jmg.2004.019737

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Cybulski C, Huzarski T, Byrski T, Gronwald J, Debniak T, Jakubowska A, Górski B, Wokołrczyk D, Masojć B, Narod SA, Lubiński J: Estrogen receptor status in CHEK2-positive breast cancers: implications for chemoprevention. Clin Genet 2009, 75: 72–78. 10.1111/j.1399-0004.2008.01111.x

    CAS  PubMed  Google Scholar 

  93. McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, Giavara S, O'Connor MJ, Tutt AN, Zdzienicka MZ, Smith GC, Ashworth A: Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res 2006, 66: 8109–8115. 10.1158/0008-5472.CAN-06-0140

    CAS  PubMed  Google Scholar 

  94. Chrisanthar R, Knappskog S, Løkkevik E, Anker G, Østenstad B, Lundgren S, Berge EO, Risberg T, Mjaaland I, Maehle L, Engebretsen LF, Lillehaug JR, Lønning PE: CHEK2 mutations affecting kinase activity together with mutations in TP53 indicate a functional pathway associated with resistance to epirubicin in primary breast cancer. PLoS One 2008, 3: e3062. 10.1371/journal.pone.0003062

    PubMed  PubMed Central  Google Scholar 

  95. Jones S, Hruban RH, Kamiyama M, Borges M, Zhang X, Parsons DW, Lin JC, Palmisano E, Brune K, Jaffee EM, Iacobuzio-Donahue CA, Maitra A, Parmigiani G, Kern SE, Velculescu VE, Kinzler KW, Vogelstein B, Eshleman JR, Goggins M, Klein AP: Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 2009, 324: 217. 10.1126/science.1171202

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Bogdanova N, Sokolenko AP, Iyevleva AG, Abysheva SN, Blaut M, Bremer M, Christiansen H, Rave-Fränk M, Dörk T, Imyanitov EN: PALB2 mutations in German and Russian patients with bilateral breast cancer. Breast Cancer Res Treat 2011, 126: 545–550. 10.1007/s10549-010-1290-4

    PubMed  Google Scholar 

  97. Villarroel MC, Rajeshkumar NV, Garrido-Laguna I, De Jesus-Acosta A, Jones S, Maitra A, Hruban RH, Eshleman JR, Klein A, Laheru D, Donehower R, Hidalgo M: Personalizing cancer treatment in the age of global genomic analyses: PALB2 gene mutations and the response to DNA damaging agents in pancreatic cancer. Mol Cancer Ther 2011, 10: 3–8. 10.1158/1535-7163.MCT-10-0893

    CAS  PubMed  Google Scholar 

  98. Chalasani P, Kurtin S, Dragovich T: Response to a third-line mitomycin C (MMC)-based chemotherapy in a patient with metastatic pancreatic adenocarcinoma carrying germline BRCA2 mutation. JOP 2008, 9: 305–308.

    PubMed  Google Scholar 

  99. James E, Waldron-Lynch MG, Saif MW: Prolonged survival in a patient with BRCA2 associated metastatic pancreatic cancer after exposure to camptothecin: a case report and review of literature. Anticancer Drugs 2009, 20: 634–8. 10.1097/CAD.0b013e32832b511e

    CAS  PubMed  Google Scholar 

  100. Fogelman DR, Wolff RA, Kopetz S, Javle M, Bradley C, Mok I, Cabanillas F, Abbruzzese JL: Evidence for the efficacy of Iniparib, a PARP-1 inhibitor, in BRCA2-associated pancreatic cancer. Anticancer Res 2011, 31: 1417–1420.

    PubMed  Google Scholar 

  101. Moule R, Sohaib A, Eeles R: Dramatic response to platinum in a patient with cancer with a germline BRCA2 mutation. Clin Oncol (R Coll Radiol) 2009, 21: 444–447.

    CAS  Google Scholar 

  102. Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ, Ashworth A, Carmichael J, Kaye SB, Schellens JH, de Bono JS: Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009, 361: 123–134. 10.1056/NEJMoa0900212

    CAS  PubMed  Google Scholar 

  103. Vesprini D, Narod SA, Trachtenberg J, Crook J, Jalali F, Preiner J, Sridhar S, Bristow RG: The therapeutic ratio is preserved for radiotherapy or cisplatin treatment in BRCA2-mutated prostate cancers. Can Urol Assoc J 2011, 5: E31-E35.

    PubMed  PubMed Central  Google Scholar 

  104. Sokolenko AP, Iyevleva AG, Preobrazhenskaya EV, Mitiushkina NV, Abysheva SN, Suspitsin EN, Kuligina ESh, Gorodnova TV, Pfeifer W, Togo AV, Turkevich EA, Ivantsov OA, Voskresenskiy DV, Dolmatov GD, Bit-Sava EM, Matsko DE, Semiglazov VF, Fichtner I, Larionov AA, Kuznetsov SG, Antoniou AC, Imyanitov EN: High prevalence and breast cancer predisposing role of the BLM c.1642 C>T (Q548X) mutation in Russia. Int J Cancer, in press.

  105. Boland CR, Goel A: Microsatellite instability in colorectal cancer. Gastroenterology 2010, 138: 2073–2087. 10.1053/j.gastro.2009.12.064

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Vilar E, Gruber SB: Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol 2010, 7: 153–162. 10.1038/nrclinonc.2009.237

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Clark AJ, Barnetson R, Farrington SM, Dunlop MG: Prognosis in DNA mismatch repair deficient colorectal cancer: are all MSI tumours equivalent? Fam Cancer 2004, 3: 85–91.

    CAS  PubMed  Google Scholar 

  108. Popat S, Hubner R, Houlston RS: Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol 2005, 23: 609–618.

    CAS  PubMed  Google Scholar 

  109. Guastadisegni C, Colafranceschi M, Ottini L, Dogliotti E: Microsatellite instability as a marker of prognosis and response to therapy: a meta-analysis of colorectal cancer survival data. Eur J Cancer 2010, 46: 2788–2798. 10.1016/j.ejca.2010.05.009

    CAS  PubMed  Google Scholar 

  110. Hewish M, Lord CJ, Martin SA, Cunningham D, Ashworth A: Mismatch repair deficient colorectal cancer in the era of personalized treatment. Nat Rev Clin Oncol 2010, 7: 197–208. 10.1038/nrclinonc.2010.18

    PubMed  Google Scholar 

  111. Laghi L, Bianchi P, Malesci A: Differences and evolution of the methods for the assessment of microsatellite instability. Oncogene 2008, 27: 6313–6321. 10.1038/onc.2008.217

    CAS  PubMed  Google Scholar 

  112. Perucho M: Correspondence re: C.R. Boland et al., A National Cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res., 58: 5248–5257, 1998. Cancer Res 1999, 59: 249–256.

    CAS  PubMed  Google Scholar 

  113. Vilar E, Scaltriti M, Balmaña J, Saura C, Guzman M, Arribas J, Baselga J, Tabernero J: Microsatellite instability due to hMLH1 deficiency is associated with increased cytotoxicity to irinotecan in human colorectal cancer cell lines. Br J Cancer 2008, 99: 1607–1612. 10.1038/sj.bjc.6604691

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Martin SA, McCarthy A, Barber LJ, Burgess DJ, Parry S, Lord CJ, Ashworth A: Methotrexate induces oxidative DNA damage and is selectively lethal to tumour cells with defects in the DNA mismatch repair gene MSH2. EMBO Mol Med 2009, 1: 323–337. 10.1002/emmm.200900040

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Valentini AM, Armentano R, Pirrelli M, Caruso ML: Chemotherapeutic agents for colorectal cancer with a defective mismatch repair system: the state of the art. Cancer Treat Rev 2006, 32: 607–618. 10.1016/j.ctrv.2006.08.001

    CAS  PubMed  Google Scholar 

  116. Papouli E, Cejka P, Jiricny J: Dependence of the cytotoxicity of DNA-damaging agents on the mismatch repair status of human cells. Cancer Res 2004, 64: 3391–3394. 10.1158/0008-5472.CAN-04-0513

    CAS  PubMed  Google Scholar 

  117. Yamane K, Schupp JE, Kinsella TJ: BRCA1 activates a G2-M cell cycle checkpoint following 6-thioguanine-induced DNA mismatch damage. Cancer Res 2007, 67: 6286–6292. 10.1158/0008-5472.CAN-06-2205

    CAS  PubMed  Google Scholar 

  118. Fink D, Zheng H, Nebel S, Norris PS, Aebi S, Lin TP, Nehmé A, Christen RD, Haas M, MacLeod CL, Howell SB: In vitro and in vivo resistance to cisplatin in cells that have lost DNA mismatch repair. Cancer Res 1997, 57: 1841–1845.

    CAS  PubMed  Google Scholar 

  119. Martin SA, Hewish M, Sims D, Lord CJ, Ashworth A: Parallel high throughput RNA interference screens identify PINK1 as a potential therapeutic target for the treatment of DNA mismatch repair deficient cancers. Cancer Res 2011, 71: 1836–1848. 10.1158/0008-5472.CAN-10-2836

    CAS  PubMed  Google Scholar 

  120. Martin SA, McCabe N, Mullarkey M, Cummins R, Burgess DJ, Nakabeppu Y, Oka S, Kay E, Lord CJ, Ashworth A: DNA polymerases as potential therapeutic targets for cancers deficient in the DNA mismatch repair proteins MSH2 or MLH1. Cancer Cell 2010, 17: 235–248. 10.1016/j.ccr.2009.12.046

    CAS  PubMed  PubMed Central  Google Scholar 

  121. des Guetz G, Schischmanoff O, Nicolas P, Perret GY, Morere JF, Uzzan B: Does microsatellite instability predict the efficacy of adjuvant chemotherapy in colorectal cancer? A systematic review with meta-analysis. Eur J Cancer 2009, 45: 1890–1896. 10.1016/j.ejca.2009.04.018

    CAS  PubMed  Google Scholar 

  122. Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, Tu D, Redston M, Gallinger S: Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med 2003, 349: 247–257. 10.1056/NEJMoa022289

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Sargent DJ, Marsoni S, Monges G, Thibodeau SN, Labianca R, Hamilton SR, French AJ, Kabat B, Foster NR, Torri V, Ribic C, Grothey A, Moore M, Zaniboni A, Seitz JF, Sinicrope F, Gallinger S: Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol 2010, 28: 3219–3226. 10.1200/JCO.2009.27.1825

    CAS  PubMed  PubMed Central  Google Scholar 

  124. de Vos tot Nederveen Cappel WH, Meulenbeld HJ, Kleibeuker JH, Nagengast FM, Menko FH, Griffioen G, Cats A, Morreau H, Gelderblom H, Vasen HF: Survival after adjuvant 5-FU treatment for stage III colon cancer in hereditary nonpolyposis colorectal cancer. Int J Cancer 2004, 109: 468–471. 10.1002/ijc.11712

    CAS  PubMed  Google Scholar 

  125. Sinicrope FA, Sargent DJ: Clinical implications of microsatellite instability in sporadic colon cancers. Curr Opin Oncol 2009, 21: 369–373. 10.1097/CCO.0b013e32832c94bd

    CAS  PubMed  PubMed Central  Google Scholar 

  126. de la Chapelle A, Hampel H: Clinical relevance of microsatellite instability in colorectal cancer. J Clin Oncol 2010, 28: 3380–3387. 10.1200/JCO.2009.27.0652

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Kim ST, Lee J, Park SH, Park JO, Lim HY, Kang WK, Kim JY, Kim YH, Chang DK, Rhee PL, Kim DS, Yun H, Cho YB, Kim HC, Yun SH, Lee WY, Chun HK, Park YS: Clinical impact of microsatellite instability in colon cancer following adjuvant FOLFOX therapy. Cancer Chemother Pharmacol 2010, 66: 659–667. 10.1007/s00280-009-1206-3

    CAS  PubMed  Google Scholar 

  128. Zaanan A, Cuilliere-Dartigues P, Guilloux A, Parc Y, Louvet C, de Gramont A, Tiret E, Dumont S, Gayet B, Validire P, Fléjou JF, Duval A, Praz F: Impact of p53 expression and microsatellite instability on stage III colon cancer disease-free survival in patients treated by 5-fluorouracil and leucovorin with or without oxaliplatin. Ann Oncol 2010, 21: 772–780. 10.1093/annonc/mdp383

    CAS  PubMed  Google Scholar 

  129. Bertagnolli MM, Niedzwiecki D, Compton CC, Hahn HP, Hall M, Damas B, Jewell SD, Mayer RJ, Goldberg RM, Saltz LB, Warren RS, Redston M: Microsatellite instability predicts improved response to adjuvant therapy with irinotecan, fluorouracil, and leucovorin in stage III colon cancer: Cancer and Leukemia Group B Protocol 89803. J Clin Oncol 2009, 27: 1814–1821. 10.1200/JCO.2008.18.2071

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Liang JT, Huang KC, Lai HS, Lee PH, Cheng YM, Hsu HC, Cheng AL, Hsu CH, Yeh KH, Wang SM, Tang C, Chang KJ: High-frequency microsatellite instability predicts better chemosensitivity to high-dose 5-fluorouracil plus leucovorin chemotherapy for stage IV sporadic colorectal cancer after palliative bowel resection. Int J Cancer 2002, 101: 519–525. 10.1002/ijc.10643

    CAS  PubMed  Google Scholar 

  131. Brueckl WM, Moesch C, Brabletz T, Koebnick C, Riedel C, Jung A, Merkel S, Schaber S, Boxberger F, Kirchner T, Hohenberger W, Hahn EG, Wein A: Relationship between microsatellite instability, response and survival in palliative patients with colorectal cancer undergoing first-line chemotherapy. Anticancer Res 2003, 23: 1773–1777.

    CAS  PubMed  Google Scholar 

  132. des Guetz G, Mariani P, Cucherousset J, Benamoun M, Lagorce C, Sastre X, Le Toumelin P, Uzzan B, Perret GY, Morere JF, Breau JL, Fagard R, Schischmanoff PO: Microsatellite instability and sensitivitiy to FOLFOX treatment in metastatic colorectal cancer. Anticancer Res 2007, 27: 2715–2719.

    CAS  PubMed  Google Scholar 

  133. Müller CI, Schulmann K, Reinacher-Schick A, Andre N, Arnold D, Tannapfel A, Arkenau H, Hahn SA, Schmoll SH, Porschen R, Schmiegel W, Graeven U, AIO Colorectal Study Group: Predictive and prognostic value of microsatellite instability in patients with advanced colorectal cancer treated with a fluoropyrimidine and oxaliplatin containing first-line chemotherapy. A report of the AIO Colorectal Study Group. Int J Colorectal Dis 2008, 23: 1033–1039. 10.1007/s00384-008-0504-2

    PubMed  Google Scholar 

  134. Kim ST, Lee J, Park SH, Park JO, Lim HY, Kang WK, Kim JY, Kim YH, Chang DK, Rhee PL, Kim DS, Yun H, Cho YB, Kim HC, Yun SH, Chun HK, Lee WY, Park YS: The effect of DNA mismatch repair (MMR) status on oxaliplatin-based first-line chemotherapy as in recurrent or metastatic colon cancer. Med Oncol 2010, 27: 1277–1285. 10.1007/s12032-009-9374-x

    CAS  PubMed  Google Scholar 

  135. Fallik D, Borrini F, Boige V, Viguier J, Jacob S, Miquel C, Sabourin JC, Ducreux M, Praz F: Microsatellite instability is a predictive factor of the tumor response to irinotecan in patients with advanced colorectal cancer. Cancer Res 2003, 63: 5738–5744.

    CAS  PubMed  Google Scholar 

  136. Charara M, Edmonston TB, Burkholder S, Walters R, Anne P, Mitchell E, Fry R, Boman B, Rose D, Fishel R, Curran W, Palazzo J: Microsatellite status and cell cycle associated markers in rectal cancer patients undergoing a combined regimen of 5-FU and CPT-11 chemotherapy and radiotherapy. Anticancer Res 2004, 24: 3161–3167.

    CAS  PubMed  Google Scholar 

  137. Bendardaf R, Lamlum H, Ristamäki R, Korkeila E, Syrjänen K, Pyrhönen S: Mismatch repair status is a predictive factor of tumour response to 5-fluorouracil and irinotecan chemotherapy in patients with advanced colorectal cancer. Tumour Biol 2007, 28: 212–220. 10.1159/000107417

    CAS  PubMed  Google Scholar 

  138. Kim JE, Hong YS, Ryu MH, Lee JL, Chang HM, Lim SB, Kim JH, Jang SJ, Kim MJ, Yu CS, Kang YK, Kim JC, Kim TW: Association between deficient mismatch repair system and efficacy to irinotecan-containing chemotherapy in metastatic colon cancer. Cancer Sci 2011, in press.

    Google Scholar 

  139. Burn J, Bishop DT, Mecklin JP, Macrae F, Möslein G, Olschwang S, Bisgaard ML, Ramesar R, Eccles D, Maher ER, Bertario L, Jarvinen HJ, Lindblom A, Evans DG, Lubinski J, Morrison PJ, Ho JW, Vasen HF, Side L, Thomas HJ, Scott RJ, Dunlop M, Barker G, Elliott F, Jass JR, Fodde R, Lynch HT, Mathers JC, CAPP2 Investigators: Effect of aspirin or resistant starch on colorectal neoplasia in the Lynch syndrome. N Engl J Med 2008, 359: 2567–2578. 10.1056/NEJMoa0801297

    CAS  PubMed  Google Scholar 

  140. Half E, Bercovich D, Rozen P: Familial adenomatous polyposis. Orphanet J Rare Dis 2009, 4: 22. 10.1186/1750-1172-4-22

    PubMed  PubMed Central  Google Scholar 

  141. Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Gordon GB, Wakabayashi N, Saunders B, Shen Y, Fujimura T, Su LK, Levin B: The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med 2000, 342: 1946–1952. 10.1056/NEJM200006293422603

    CAS  PubMed  Google Scholar 

  142. Bertagnolli MM, Eagle CJ, Zauber AG, Redston M, Breazna A, Kim K, Tang J, Rosenstein RB, Umar A, Bagheri D, Collins NT, Burn J, Chung DC, Dewar T, Foley TR, Hoffman N, Macrae F, Pruitt RE, Saltzman JR, Salzberg B, Sylwestrowicz T, Hawk ET, Adenoma Prevention with Celecoxib Study Investigators: Five-year efficacy and safety analysis of the Adenoma Prevention with Celecoxib Trial. Cancer Prev Res (Phila) 2009, 2: 310–321. 10.1158/1940-6207.CAPR-08-0206

    CAS  Google Scholar 

  143. Vasen HF, Möslein G, Alonso A, Aretz S, Bernstein I, Bertario L, Blanco I, Bülow S, Burn J, Capella G, Colas C, Engel C, Frayling I, Friedl W, Hes FJ, Hodgson S, Järvinen H, Mecklin JP, Møller P, Myrhøi T, Nagengast FM, Parc Y, Phillips R, Clark SK, de Leon MP, Renkonen-Sinisalo L, Sampson JR, Stormorken A, Tejpar S, Thomas HJ, et al.: Guidelines for the clinical management of familial adenomatous polyposis (FAP). Gut 2008, 57: 704–713. 10.1136/gut.2007.136127

    CAS  PubMed  Google Scholar 

  144. Frank-Raue K, Rondot S, Raue F: Molecular genetics and phenomics of RET mutations: Impact on prognosis of MTC. Mol Cell Endocrinol 2010, 322: 2–7. 10.1016/j.mce.2010.01.012

    CAS  PubMed  Google Scholar 

  145. Carlomagno F, Vitagliano D, Guida T, Ciardiello F, Tortora G, Vecchio G, Ryan AJ, Fontanini G, Fusco A, Santoro M: ZD6474, an orally available inhibitor of KDR tyrosine kinase activity, efficiently blocks oncogenic RET kinases. Cancer Res 2002, 62: 7284–7290.

    CAS  PubMed  Google Scholar 

  146. Wells SA Jr, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, Skinner M, Krebs A, Vasselli J, Schlumberger M: Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol 2010, 10: 767–772.

    Google Scholar 

  147. Robinson BG, Paz-Ares L, Krebs A, Vasselli J, Haddad R: Vandetanib (100 mg) in patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Endocrinol Metab 2010, 95: 2664–2671. 10.1210/jc.2009-2461

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Quinn JE, James CR, Stewart GE, Mulligan JM, White P, Chang GK, Mullan PB, Johnston PG, Wilson RH, Harkin DP: BRCA1 mRNA expression levels predict for overall survival in ovarian cancer after chemotherapy. Clin Cancer Res 2007, 13: 7413–7420. 10.1158/1078-0432.CCR-07-1083

    CAS  PubMed  Google Scholar 

  149. Rosell R, Perez-Roca L, Sanchez JJ, Cobo M, Moran T, Chaib I, Provencio M, Domine M, Sala MA, Jimenez U, Diz P, Barneto I, Macias JA, de Las Peñas R, Catot S, Isla D, Sanchez JM, Ibeas R, Lopez-Vivanco G, Oramas J, Mendez P, Reguart N, Blanco R, Taron M: Customized treatment in non-small-cell lung cancer based on EGFR mutations and BRCA1 mRNA expression. PLoS One 2009, 4: e5133. 10.1371/journal.pone.0005133

    PubMed  PubMed Central  Google Scholar 

  150. Font A, Taron M, Gago JL, Costa C, Sánchez JJ, Carrato C, Mora M, Celiz P, Perez L, Rodríguez D, Gimenez-Capitan A, Quiroga V, Benlloch S, Ibarz L, Rosell R: BRCA1 mRNA expression and outcome to neoadjuvant cisplatin-based chemotherapy in bladder cancer. Ann Oncol 2011, 22: 139–144.

    CAS  PubMed  Google Scholar 

  151. Su C, Zhou S, Zhang L, Ren S, Xu J, Zhang J, Lv M, Zhang J, Zhou C: ERCC1, RRM1 and BRCA1 mRNA expression levels and clinical outcome of advanced non-small cell lung cancer. Med Oncol, in press.

  152. Papadaki C, Tsaroucha E, Kaklamanis L, Lagoudaki E, Trypaki M, Tryfonidis K, Mavroudis D, Stathopoulos E, Georgoulias V, Souglakos J: Correlation of BRCA1, TXR1 and TSP1 mRNA expression with treatment outcome to docetaxel-based first-line chemotherapy in patients with advanced/metastatic non-small-cell lung cancer. Br J Cancer 2011, 104: 316–323. 10.1038/sj.bjc.6606027

    CAS  PubMed  Google Scholar 

  153. Holstege H, Horlings HM, Velds A, Langerød A, Børresen-Dale AL, van de Vijver MJ, Nederlof PM, Jonkers J: BRCA1-mutated and basal-like breast cancers have similar aCGH profiles and a high incidence of protein truncating TP53 mutations. BMC Cancer 2010, 10: 654. 10.1186/1471-2407-10-654

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Vollebergh MA, Lips EH, Nederlof PM, Wessels LF, Schmidt MK, van Beers EH, Cornelissen S, Holtkamp M, Froklage FE, de Vries EG, Schrama JG, Wesseling J, van de Vijver MJ, van Tinteren H, de Bruin M, Hauptmann M, Rodenhuis S, Linn SC: An aCGH classifier derived from BRCA1-mutated breast cancer and benefit of high-dose platinum-based chemotherapy in HER2-negative breast cancer patients. Ann Oncol 2011, 22: 1561–1570. 10.1093/annonc/mdq624

    CAS  PubMed  Google Scholar 

  155. Gallmeier E, Kern SE: Absence of specific cell killing of the BRCA2-deficient human cancer cell line CAPAN1 by poly(ADP-ribose) polymerase inhibition. Cancer Biol Ther 2005, 4: 703–706. 10.4161/cbt.4.7.1909

    CAS  PubMed  Google Scholar 

  156. Kloos I, Delaloge S, Chompret A, Noguès C, Bressac de Paillerets B, Spielmann M: BRCA1/2-linked breast carcinoma is a highly chemosensitive disease. Proc Am Soc Clin Oncol 2001, 20: abstract 1862.

    Google Scholar 

  157. Melichar B, Fridrichová P, Lukesová S, Mergancová J, Urminská H, Ryska A, Foretová L: Pathological complete response after primary chemotherapy in a mother and daughter with hereditary breast carcinoma: two case reports. Eur J Gynaecol Oncol 2008, 29: 188–190.

    CAS  PubMed  Google Scholar 

  158. Rhiem K, Wappenschmidt B, Bosse K, Köppler H, Tutt AN, Schmutzler RK: Platinum sensitivity in a BRCA1 mutation carrier with advanced breast cancer. Clin Oncol (R Coll Radiol) 2009, 21: 448–450.

    CAS  Google Scholar 

  159. Byrski T, Huzarski T, Dent R, Gronwald J, Zuziak D, Cybulski C, Kladny J, Gorski B, Lubinski J, Narod SA: Response to neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat 2009, 115: 359–363. 10.1007/s10549-008-0128-9

    CAS  PubMed  Google Scholar 

  160. Byrski T, Gronwald J, Huzarski T, Grzybowska E, Budryk M, Stawicka M, Mierzwa T, Szwiec M, Wi÷niowski R, Siolek M, Narod SA, Lubinski J, Polish Hereditary Breast Cancer Consortium: Response to neo-adjuvant chemotherapy in women with BRCA1-positive breast cancers. Breast Cancer Res Treat 2008, 108: 289–296. 10.1007/s10549-007-9600-1

    CAS  PubMed  Google Scholar 

  161. Leunen K, Cadron I, Van Gorp T, Amant F, Berteloot P, Neven P, Legius E, Vergote I: Does paclitaxel-carboplatin chemotherapy in a dose-dense regimen enhance survival of BRCA-related ovarian cancer patients? Int J Gynecol Cancer 2009, 19: 1501–154. 10.1111/IGC.0b013e3181bb703f

    PubMed  Google Scholar 

Download references

Acknowledgements

This work has been supported by the Russian Federation for Basic Research (grants 10-04-92110, 10-04-92601, 11-04-00227), the Federal Agency for Science and Innovations (contract 02.740.11.0780), the Commission of the European Communities (grant PITN-GA-2009-238132) and the Government of Moscow (grant 15/11).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Evgeny N Imyanitov.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

Both authors contributed to the literature search, data analysis and manuscript preparation. Both authors read and approved the final manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Imyanitov, E.N., Moiseyenko, V.M. Drug therapy for hereditary cancers. Hered Cancer Clin Pract 9, 5 (2011). https://doi.org/10.1186/1897-4287-9-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1897-4287-9-5

Keywords