Defects in the Fanconi anemia (FA) pathway occur in subsets of diverse human cancers. The hypersensitivity of FA pathway-deficient cells to DNA interstrand cross-linking and possibly other agents renders these genes attractive targets for a genotype-based, individualized anticancer therapy. A prerequisite before clinical trials is the validation and quantification of this hypersensitivity in suitable preclinical pharmacogenomic models. In addition, the effects of combinational therapy need to be evaluated and novel agents sought. We discuss here the pitfalls and limitations in the interpretation of common FA models when applied to the validation of FA gene defects as therapeutic targets. In general, all preclinical models are prone to certain artifacts and, thus, promising results in a single or few models rarely translate into clinical success. Nevertheless, the extraordinary robustness of FA pathway-deficient cells to interstrand cross-linking agents, which are observable in virtually any model independent of species, cell type, or technique used to engineer the gene defect, in various in vitro and in vivo settings, renders these gene defects particularly attractive for targeted therapy. Clinical trials are now under way.

The Fanconi anemia (FA) gene family consists of at least 12 genes (FANCA, FANCB, FANCC, BRCA2/FANCD1, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ, FANCL, FANCM; refs. 112). Biallelic mutations of these genes cause FA, a rare recessive disorder comprising congenital skeletal abnormalities, progressive bone marrow failure, and increased cancer risk (13). FA gene defects also occur in solid tumors among the general population; BRCA2 is mutated in pancreatic, breast, ovarian, and other cancers (8); FANCC and FANCG are mutated in some pancreatic cancers (1416); and epigenetic gene silencing of FANCF occurs in a variety of tumors (1719).

The FA proteins act in a common pathway, distal parts of which interact with regulators of cell cycle control and DNA repair, especially the repair of DNA interstrand cross-links and double-strand breaks (Fig. 1). The formation of the nuclear FA core complex, comprising Fanca, Fancb, Fancc, Fance, Fancf, Fancg, Fancl, Fancm, and one yet unidentified 100-kDa protein (Faap100), depends on the integrity of all proteins involved (11, 20, 21). The FA complex becomes activated on DNA damage, as coordinated by DNA damage sensor proteins such as ataxia telangiectasia mutated (ATM) or ataxia telangiectasia mutated and Rad3-related (ATR) (13, 2225). This activation causes the monoubiquitination of Fancd2 (26), which is subsequently targeted to nuclear foci. Fancd2 colocalizes and interacts with Brca2 (27, 28) and several other DNA repair proteins, including Blm, Brca1, Nbn, Pcna, Rad51, and Rpa2 (2934). A direct function of the FA pathway in DNA repair is further supported by Fancj being identical to the DNA helicase Brip1 (Brca1-interacting protein; refs. 10, 35, 36) and Fancm representing the human orthologue of the bacterial DNA repair protein Hef (11, 37).

Fig. 1.

Schematic representation of the FA pathway. For clarity, interactions of FA core complex proteins with DNA repair focus proteins (99) are not depicted.

Fig. 1.

Schematic representation of the FA pathway. For clarity, interactions of FA core complex proteins with DNA repair focus proteins (99) are not depicted.

Close modal

In contrast to tumors of FA patients, FA pathway-deficient tumors arising in the general population have a biallelic FA gene defect whereas the patients' other cells do not, thus representing a tumor-specific absolute biochemical difference (38). FA pathway-deficient cells are hypersensitive to certain therapeutics, particularly interstrand cross-linking agents (13) and some poly(ADP-ribose) polymerase inhibitors (3944). Their sensitivity to irradiation remains controversial (45, 46). An important challenge will be the identification and development of novel agents to which FA pathway-deficient cells are hypersensitive. If those agents elicited hypersensitivity via a different mechanism of action than interstrand cross-linking agents, they might permit synergistic effects when used in combination. This would allow a reduction of dosage and thus of the harmful side effects of interstrand cross-linking agents. Likewise, combinational uses of the known agents exerting hypersensitivity deserve investigation.

Potential options for genotype-specific anticancer therapies arising from FA pathway inactivation in tumors will require suitable preclinical models, for which stringent and universal validation criteria have not yet been established (47). Various models of FA gene defects have been developed, each of which has certain limitations when used for pharmacogenomic studies.

Pharmacogenomic FA models

Human versus other species. The progress in understanding the molecular functions of the FA genes is largely owed to the successful development and analysis of models of FA gene defects in a variety of species (including human, mouse, hamster, chicken, frog, and zebrafish). For pharmacogenomic studies, the use of human cells may be desirable to eliminate any artifacts attributable to interspecies variability, as most proximal FA genes (constituting the FA core complex) are structurally and functionally not highly conserved. As an example, the chicken FANCG gene sequence shares only 39% similarity to human FANCG, and its function is not complemented by the human gene (48). In contrast, the more distal FANCD2 is better conserved (9) and BRCA2 is highly conserved within the BRC-repeat regions (49).

Malignant versus nonmalignant cells. The FA genes are involved in the regulation of cell cycle controls, DNA repair, and genome maintenance, features that are expected to differ between malignant and nonmalignant cells. FA models that use cancer cells may therefore be preferable for pharmacogenomic studies because it is uncertain whether the FA phenotype of nonmalignant cells can be fully extrapolated to malignant cells (46). Table 1 lists many of the presently available human cancer FA lines.

Table 1.

Human FA cancer cell models

Cell lineTissueGene defectOrigin of null stateAvailable controlsLimitationsRef.
RKO FANCC−/− Colorectal adenocarcinoma FANCC (homozygous Δexon 10) Engineered in vitro Parental/heterozygote clones (physiologic gene expression) Defect not naturally selected, microsatellite unstable cancer cells (46) 
RKO FANCG−/− Colorectal adenocarcinoma FANCG (homozygous Δexon 8) Engineered in vitro Parental/heterozygote clones (physiologic gene expression) Defect not naturally selected, microsatellite unstable cancer cells (46) 
Hs766T Pancreatic adenocarcinoma FANCG (313G>T + LOH*) Somatically acquired in vivo Derivatives overexpressing FANCG or other lines Either gene overexpression or lack of isogenic controls (15, 86) 
PL11 Pancreatic adenocarcinoma FANCC (homozygous deletion) Somatically acquired in vivo Derivatives overexpressing FANCC or other lines Either gene overexpression or lack of isogenic controls (15, 86) 
CAPAN1 Pancreatic adenocarcinoma BRCA2 (6174ΔT + LOH*) Somatically acquired in vivo Derivatives overexpressing BRCA2 or other lines Either gene overexpression or lack of isogenic controls (15, 86, 88, 89) 
TOV-21G Ovarian adenocarcinoma FANCF (epigenetic silencing) Unknown Derivatives overexpressing FANCF or other lines Either gene overexpression or lack of isogenic controls (19) 
OHSU-974 Head and neck squamous cell carcinoma FANCA? Germ line (i.e., before tumorigenesis) Other lines Lack of isogenic controls (100) 
VU1365 Head and neck squamous cell carcinoma FANCA Germ line (i.e., before tumorigenesis) Derivatives overexpressing FANCA or other lines Either gene overexpression or lack of isogenic controls (100) 
VU1131 Head and neck squamous cell carcinoma FANCC (67ΔG) Germ line (i.e., before tumorigenesis) Derivatives overexpressing FANCC or other lines Either gene overexpression or lack of isogenic controls (100) 
FA-AML1 Acute myeloic leukemia cells BRCA2 (8415G>T, 8732C>A, reverting mutation 8731T>G) Somatically reverted germ line null state during tumorigenesis Other lines No FA phenotype due to reversion of the null state (101) 
SB1690CB Acute myeloic leukemia cells BRCA2 (3827ΔGT, IVS7 + 2T>G) Germ line (i.e., before tumorigenesis) Other lines Lack of isogenic controls (102) 
Cell lineTissueGene defectOrigin of null stateAvailable controlsLimitationsRef.
RKO FANCC−/− Colorectal adenocarcinoma FANCC (homozygous Δexon 10) Engineered in vitro Parental/heterozygote clones (physiologic gene expression) Defect not naturally selected, microsatellite unstable cancer cells (46) 
RKO FANCG−/− Colorectal adenocarcinoma FANCG (homozygous Δexon 8) Engineered in vitro Parental/heterozygote clones (physiologic gene expression) Defect not naturally selected, microsatellite unstable cancer cells (46) 
Hs766T Pancreatic adenocarcinoma FANCG (313G>T + LOH*) Somatically acquired in vivo Derivatives overexpressing FANCG or other lines Either gene overexpression or lack of isogenic controls (15, 86) 
PL11 Pancreatic adenocarcinoma FANCC (homozygous deletion) Somatically acquired in vivo Derivatives overexpressing FANCC or other lines Either gene overexpression or lack of isogenic controls (15, 86) 
CAPAN1 Pancreatic adenocarcinoma BRCA2 (6174ΔT + LOH*) Somatically acquired in vivo Derivatives overexpressing BRCA2 or other lines Either gene overexpression or lack of isogenic controls (15, 86, 88, 89) 
TOV-21G Ovarian adenocarcinoma FANCF (epigenetic silencing) Unknown Derivatives overexpressing FANCF or other lines Either gene overexpression or lack of isogenic controls (19) 
OHSU-974 Head and neck squamous cell carcinoma FANCA? Germ line (i.e., before tumorigenesis) Other lines Lack of isogenic controls (100) 
VU1365 Head and neck squamous cell carcinoma FANCA Germ line (i.e., before tumorigenesis) Derivatives overexpressing FANCA or other lines Either gene overexpression or lack of isogenic controls (100) 
VU1131 Head and neck squamous cell carcinoma FANCC (67ΔG) Germ line (i.e., before tumorigenesis) Derivatives overexpressing FANCC or other lines Either gene overexpression or lack of isogenic controls (100) 
FA-AML1 Acute myeloic leukemia cells BRCA2 (8415G>T, 8732C>A, reverting mutation 8731T>G) Somatically reverted germ line null state during tumorigenesis Other lines No FA phenotype due to reversion of the null state (101) 
SB1690CB Acute myeloic leukemia cells BRCA2 (3827ΔGT, IVS7 + 2T>G) Germ line (i.e., before tumorigenesis) Other lines Lack of isogenic controls (102) 
*

LOH, loss of heterozygosity.

Complementation analysis was not conclusive (100).

Complementation group was determined by retroviral complementation (100).

Natural selection versus artificial engineering. Gene defects conferring a selective cellular advantage can cause coincidental detrimental effects (“reduction of fitness”), which, if evolving naturally, must be outweighed by the gained advantage. The mutational profile of a cancer thus represents the balance of selective pressures for and against any given mutation; both directions of selection can operate on a given mutated gene (50, 51).1

1

Gallmeier E, Hucl T, Calhoun ES, et al. Gene-specific selection against fanconi anemia gene inactivation in human cancer cells. Submitted for publication, 2006.

Accordingly, cancer cells that have been naturally selected for a FA gene defect are thought to differ significantly from cells on which this defect was superimposed. The extent of a gene knockout–induced reduction of fitness varies strongly among the FA genes, with only a few being essential under many experimental circumstances. Mice having a gene knockout of the proximal genes FANCA, FANCC, and FANCG have normal neonatal viability and no gross growth abnormalities (5257). In contrast, mice with null mutations of the distal gene BRCA2 have embryonic lethality, whereas truncating mutations that retain the conserved BRC repeats can confer a viable phenotype (5864). Some mice having defects in the distal FANCD2 gene are perinatally lethal, depending on the background strain, which suggests the presence of a modifier locus (65). Fancd2-deficient zebrafish embryos develop a smaller body due to extensive apoptosis (66). In human cancer cells, we found the engineered disruption of the proximal FA genes FANCC and FANCG, but not of the distal FANCD2 and BRCA2, to yield viable clones (46).1 Consistently, mutations of the proximal FANCC and FANCG, but not of the distal FANCD2, are reported. Despite BRCA2 being mutated in some tumors, its complete inactivation might predominantly be an adverse event (51, 67) because BRCA2 mutations occur late during tumorigenesis (68), almost exclusively yield truncated Brca2 proteins that retain partial function, and are mainly of germ line origin, even in apparently sporadic tumors (6972). Therefore, tissue culture models in which FA gene defects are artificially created might not accurately reflect the phenotype of this defect in a natural setting, depending on the extent of reduction of fitness and compensatory evolution associated (i.e., high for the distal FANCD2 and BRCA2 genes, low for the proximal FA genes). Consequently, depending on the gene of interest, models using cells that had been naturally selected for a FA defect are preferable for pharmacogenomic studies.

On-target versus off-target effects. On-target effects need to be distinguished from off-target effects, although a specific FA gene defect defines the model. Whereas on-target effects are immediate and directly caused by the FA gene defect, off-target effects comprise artifacts resulting from the particular technique applied to manipulate FA gene function, including unintended drug interferences, oligonucleotide interactions, squelching, threshold effects, etc. Whereas on-target effects are reproducible, off-target effects can seem to be random and are not reliably reproducible. FA models using random chemical mutagenesis, small interfering RNA technology, or nonphysiologic exogenous gene overexpression seem to be particularly prone to off-target effects.

Options for manipulating FA gene function in isogenic cells. The development of isogenic cell lines, in which a gene defect is artificially created or, conversely, the function of a defective gene is artificially restored, overcomes the problem of managing uncontrolled variation inherent to nonisogenic models. Nevertheless, the confirmation of a relationship using outbred nonisogenic cells provides a useful assurance of robustness and argues against the results being dominated by idiosyncratic phenomena.

Gene overexpression. Some FA models compare conditions of constitutive gene overexpression to null expression. In neither condition is physiologic gene function necessarily assessed, for it may be exaggerated, mitigated, or not present at all. In addition, when using human lymphocytes or fibroblasts from FA patients, even the immortalization step, which involves viral tumorigenic proteins, could introduce nonphysiologic influences. Despite these concerns, robust FA phenotypes, such as deficient Fancd2 monoubiquitination, loss of Fancd2 focus formation, and hypersensitivity to interstrand cross-linking agents, are routinely reversed by complementation through gene overexpression.

Specific (targeted) gene knockout and knock-in. The preferable comparison of physiologic to null expression of FA genes can be achieved by either endogenously creating a gene defect in cells proficient (knockout) or reverting a gene defect in cells deficient for the respective gene (knock-in). As these defects are artificially created, there is little opportunity for compensatory evolution in these models and, thus, effects due to a reduction of fitness sometimes predominate, depending on the gene of interest. In addition, somatic cell gene targeting is mainly done in (diploid) cancer cells having microsatellite instability. Microsatellite unstable cells, however, do not display chromosomal instability and therefore do not reflect the majority of tumors. Furthermore, the mismatch repair defect of microsatellite unstable cells might complicate the interpretation of DNA repair-related FA phenotypes. Another largely neglected problem of knockout models, which is particularly obvious in cancer cells, is clonal variability. “Isogenic” derivatives can comprise variable genotypes among different clones. Thus, more than one gene knockout clone needs to be created and compared with parental and heterozygous control cells. Technical improvements have recently simplified this formerly highly laborious approach (7375).

Unspecific (random) gene knockout. Models using chemical random mutagenesis have lesser gene specificity than single gene knockout models. For example, N-ethyl-N-nitrosurea–mutagenized Chinese hamster cells having defects in the hamster homologues of FANCA, FANCG, and BRCA2 (7682) are expected to involve many, perhaps hundreds of thousands, simultaneous genetic mutations. Thus, gene specificity of a phenotypic alteration can only be inferred when multiple independent clones can be analyzed. Even then, the magnitude of these phenotypic alterations can potentially be misleadingly high (82).

RNA interference. RNA interference models, comparing physiologic gene expression to acutely reduced, but not absent, gene expression (75, 83), have become increasingly popular, but might not be the best choice for pharmacogenomic studies. A major disadvantage is that any effects dependent on the complete absence of a gene will be missed. Another concern is that methods using transient transfections will examine unselected cells. Therefore, compensatory evolution cannot occur and a reduction of fitness may predominate, depending on the gene of interest. Furthermore, it is difficult to apply suitable controls for RNA interference, especially because off-target effects are common (84) and cannot be predictably recreated or monitored by matched RNA reagents. Finally, off-target effects frequently induce a toxic phenotype (29% probability according to ref. 85) and therefore interfere with cell survival read-outs of pharmacogenomic studies. RNA interference techniques have only been rarely used to mimic FA gene defects, partly due to the fact that nonmalignant cells having “true” FA gene defects are readily available from FA patients.

The robust hypersensitivity of FA pathway-deficient cells to interstrand cross-linking agents

Pharmacogenomic FA models differ with regard not only to the above features but also to where the FA defect evolved (in vitro or in vivo) and whether the cellular drug sensitivity was tested in vitro or in vivo through the xenografting of cell lines in mice (86, 87). Each combination offers a different constellation of effects and artifacts. Notably, despite the huge variety of unique FA models, which are all prone to different artifacts, the hypersensitivity of FA pathway-deficient cells to interstrand cross-linking agents holds true in virtually all of these models. The extent of hypersensitivity is best described by the IC50 ratios between FA pathway-proficient and FA pathway-deficient cells, here termed the pharmacogenomic window. The width of this pharmacogenomic window depends on the specific interstrand cross-linking agent used and might reflect the proportional contribution of interstrand cross-links to the overall toxicity of the drug (46). IC50 ratios of ≥10-fold, generally regarded as promising for further clinical evaluation, can be achieved in preclinical FA cancer models on treatment with several interstrand cross-linking agents (e.g., melphalan, mitomycin C, and carboplatin; ref. 46).

The particular problem of modeling BRCA2 defects in tumors

CAPAN1, the only BRCA2-deficient human cancer cell line. Only one cancer cell line identified to date harbors a naturally selected, inactivating BRCA2 mutation (6174ΔT and loss of the second allele). The pancreatic cancer line CAPAN1 is cumbersome for pharmacogenomic studies because it has low transfectability, poor clonogenicity, and slow growth in culture, the latter presumably representing an on-target, reduction-of-fitness phenotype. Perhaps for similar reasons, CAPAN1 cells are more sensitive towards a variety of agents than other pancreatic cancer lines,2

2

Unpublished observations.

constituting in most instances a false positive result. Therefore, nonisogenic cancer lines do not represent suitable controls for CAPAN1. Isogenic controls were provided through the exogenous overexpression of wild-type BRCA2 (88). Paradoxically, the growth rate in these derivatives is even lower, which may be an off-target effect due to unphysiologically high levels of BRCA2 expression. Very recently, additional BRCA2-overexpressing CAPAN1 derivatives were reported and follow-up studies are anticipated (89). Due to the above, the CAPAN1 cell line is thus best suited as a positive control in pharmacogenomic studies, whereas tests of specificity are not afforded.

BRCA2 knockout models in nonhuman species. No engineered human gene knockout model exists for BRCA2, probably because the BRCA2-null state usually is lethal (46, 51).1BRCA2 knockout models in mouse, hamster, and chicken cells do exist, but are of unclear relevance for pharmacogenomic studies of human cancers. In mice, null mutations of BRCA2 are embryonic lethal, a phenotype seeming to be less severe in a p53-null background (61). In contrast, truncating mutations that retain certain BRC repeats can confer a viable phenotype (5864). In hamster cells, the compound heterozygote BRCA2 line V-C8, derived from (random) chemical mutagenesis of V79 cells, harbors two different nonsense BRCA2 mutations (82). In chicken DT40 cells, the complete disruption of BRCA2 did not yield viable clones (90), whereas truncating mutations, which spared the first two BRC repeats, only decreased the cell growth rate in these cells (91). Engineered BRCA2 loss thus variably affects different species, presumably due to the different balance of cellular selection, reduction of fitness, and compensatory evolution, which operates after gene targeting.

A model mimicking a non-FA patient having a BRCA2-deficient cancer. A model that might closely reflect non-FA patients having BRCA2-deficient cancers was established by Ludwig et al. (92). BRCA2−/− mammary tumors in BRCA2-proficient mice were obtained through conditional tissue-specific gene targeting. This model has not yet been applied to pharmacogenomic studies.

Requirement of rapid diagnostic assays for FA defects in cancer

Defects in the distal FA pathway are diagnosed by direct sequencing for germ line mutations in blood samples, which are commercially available for BRCA2 (such as Myriad Genetics, Salt Lake City, UT). Screening for proximal FA pathway inactivation in tumors remains problematic. As a clinical test, direct sequencing would be laborious and expensive due to the number and size of the many proximal FA genes. Furthermore, one would miss large deletions (especially relevant for FANCA; ref. 93) or epigenetic inactivation (especially relevant for FANCF; refs. 1719, 94). Chromosome breakage on treatment with diepoxybutane, which is the clinical test applied for the diagnosis of FA, is only feasible for pure and actively growing cancer cells (i.e., cell lines). Similarly, Fancd2 immunoblots using primary cancer tissue are hampered by contaminating normal tissue and therefore also require cell lines. Cell lines, however, cannot yet be quickly and reliably grown from primary tumors. Thus, other techniques to enable screening for proximal FA gene defects in tumors need to be developed.

Despite increasing interest in targeted therapies in recent years, there are few examples of successful genotype-based anticancer therapy, including imatinib (Gleevec) for the treatment of chronic myeloid leukemia (95, 96) and gefitinib (Iressa) for the treatment of non–small-cell lung cancers harboring EGFR mutations (97, 98). As discussed in this review, any preclinical pharmacogenomic model has limitations and is prone to certain artifacts. Most pharmacogenomic findings are therefore strongly model-dependent and are rarely generalizable over several models. Thus, they often translate poorly into clinical success. In stark contrast, the hypersensitivity of FA pathway-deficient cells to interstrand cross-linking agents holds true in virtually any FA model, using different species, cell types, and techniques, in various in vivo and in vitro settings. This extremely robust pharmacogenomic phenomenon might therefore represent one of the most promising avenues for individualized anticancer therapy to date. Clinical trials are now under way.

We thank Jonathan R. Brody, Eric S. Calhoun, Tomas Hucl, and Jordan M. Winter for critically reading the manuscript.

1
de Winter JP, Waisfisz Q, Rooimans MA, et al. The Fanconi anaemia group G gene FANCG is identical with XRCC9.
Nat Genet
1998
;
20
:
281
–3.
2
de Winter JP, Rooimans MA, van Der Weel L, et al. The Fanconi anaemia gene FANCF encodes a novel protein with homology to ROM.
Nat Genet
2000
;
24
:
15
–6.
3
de Winter JP, Leveille F, van Berkel CG, et al. Isolation of a cDNA representing the Fanconi anemia complementation group E gene.
Am J Hum Genet
2000
;
67
:
1306
–8.
4
Lo Ten Foe JR, Rooimans MA, Bosnoyan-Collins L, et al. Expression cloning of a cDNA for the major Fanconi anaemia gene, FAA.
Nat Genet
1996
;
14
:
320
–3.
5
Strathdee CA, Gavish H, Shannon WR, Buchwald M. Cloning of cDNAs for Fanconi's anaemia by functional complementation.
Nature
1992
;
356
:
763
–7.
6
Meetei AR, de Winter JP, Medhurst AL, et al. A novel ubiquitin ligase is deficient in Fanconi anemia.
Nat Genet
2003
;
35
:
165
–70. Epub 2003 Sep 14.
7
Meetei AR, Levitus M, Xue Y, et al. X-linked inheritance of Fanconi anemia complementation group B.
Nat Genet
2004
;
36
:
1219
–24.
8
Howlett NG, Taniguchi T, Olson S, et al. Biallelic inactivation of BRCA2 in Fanconi anemia.
Science
2002
;
297
:
606
–9.
9
Timmers C, Taniguchi T, Hejna J, et al. Positional cloning of a novel Fanconi anemia gene, FANCD2.
Mol Cell
2001
;
7
:
241
–8.
10
Levitus M, Waisfisz Q, Godthelp BC, et al. The DNA helicase BRIP1 is defective in Fanconi anemia complementation group J.
Nat Genet
2005
;
37
:
934
–5.
11
Meetei AR, Medhurst AL, Ling C, et al. A human ortholog of archaeal DNA repair protein Hef is defective in Fanconi anemia complementation group M.
Nat Genet
2005
;
37
:
958
–63.
12
Levitus M, Rooimans MA, Steltenpool J, et al. Heterogeneity in Fanconi anemia: evidence for 2 new genetic subtypes.
Blood
2004
;
103
:
2498
–503.
13
D'Andrea AD, Grompe M. The Fanconi anaemia/BRCA pathway.
Nat Rev Cancer
2003
;
3
:
23
–34.
14
van der Heijden MS, Yeo CJ, Hruban RH, Kern SE. Fanconi anemia gene mutations in young-onset pancreatic cancer.
Cancer Res
2003
;
63
:
2585
–8.
15
van der Heijden MS, Brody JR, Gallmeier E, et al. Functional defects in the Fanconi anemia pathway in pancreatic cancer cells.
Am J Pathol
2004
;
165
:
651
–7.
16
Couch FJ, Johnson MR, Rabe K, et al. Germ line Fanconi anemia complementation group C mutations and pancreatic cancer.
Cancer Res
2005
;
65
:
383
–6.
17
Marsit CJ, Liu M, Nelson HH, Posner M, Suzuki M, Kelsey KT. Inactivation of the Fanconi anemia/BRCA pathway in lung and oral cancers: implications for treatment and survival.
Oncogene
2004
;
23
:
1000
–4.
18
Narayan G, Arias-Pulido H, Nandula SV, et al. Promoter hypermethylation of FANCF: disruption of Fanconi anemia-BRCA pathway in cervical cancer.
Cancer Res
2004
;
64
:
2994
–7.
19
Taniguchi T, Tischkowitz M, Ameziane N, et al. Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors.
Nat Med
2003
;
9
:
568
–74.
20
Fei P, Yin J, Wang W. New advances in the DNA damage response network of Fanconi anemia and BRCA proteins. FAAP95 replaces BRCA2 as the true FANCB protein.
Cell Cycle
2005
;
4
:
80
–6.
21
Thomashevski A, High AA, Drozd M, et al. The Fanconi anemia core complex forms four complexes of different sizes in different subcellular compartments.
J Biol Chem
2004
;
279
:
26201
–9.
22
Andreassen PR, D'Andrea AD, Taniguchi T. ATR couples FANCD2 monoubiquitination to the DNA-damage response.
Genes Dev
2004
;
18
:
1958
–63.
23
Taniguchi T, Garcia-Higuera I, Xu B, et al. Convergence of the Fanconi anemia and ataxia telangiectasia signaling pathways.
Cell
2002
;
109
:
459
–72.
24
Wang X, D'Andrea AD. The interplay of Fanconi anemia proteins in the DNA damage response.
DNA Repair (Amst)
2004
;
3
:
1063
–9.
25
Pichierri P, Rosselli F. The DNA crosslink-induced S-phase checkpoint depends on ATR-CHK1 and ATR-NBS1-2 pathways.
EMBO J
2004
;
23
:
1178
–87.
26
Garcia-Higuera I, Taniguchi T, et al. Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway.
Mol Cell
2001
;
7
:
249
–62.
27
Wang X, Andreassen PR, D'Andrea AD. Functional interaction of monoubiquitinated FANCD2 and BRCA2/FANCD1 in chromatin.
Mol Cell Biol
2004
;
24
:
5850
–62.
28
Montes de Oca R, Andreassen PR, Margossian SP, et al. Regulated interaction of the Fanconi anemia protein, FANCD2, with chromatin.
Blood
2005
;
105
:
1003
–9.
29
Pichierri P, Franchitto A, Rosselli F. BLM and the FANC proteins collaborate in a common pathway in response to stalled replication forks.
EMBO J
2004
;
23
:
3154
–63.
30
Nakanishi K, Taniguchi T, Ranganathan V, et al. Interaction of FANCD2 and NBS1 in the DNA damage response.
Nat Cell Biol
2002
;
4
:
913
–20.
31
Pichierri P, Averbeck D, Rosselli F. DNA cross-link-dependent RAD50/MRE11/NBS1 subnuclear assembly requires the Fanconi anemia C protein.
Hum Mol Genet
2002
;
11
:
2531
–46.
32
Taniguchi T, Garcia-Higuera I, Andreassen PR, Gregory RC, Grompe M, D'Andrea AD. S-phase-specific interaction of the Fanconi anemia protein, FANCD2, with BRCA1 and RAD51.
Blood
2002
;
100
:
2414
–20.
33
Hussain S, Wilson JB, Medhurst AL, et al. Direct interaction of FANCD2 with BRCA2 in DNA damage response pathways.
Hum Mol Genet
2004
;
13
:
1241
–8.
34
Howlett NG, Taniguchi T, Durkin SG, D'Andrea AD, Glover TW. The Fanconi anemia pathway is required for the DNA replication stress response and for the regulation of common fragile site stability.
Hum Mol Genet
2005
;
14
:
693
–701.
35
Levran O, Attwooll C, Henry RT, et al. The BRCA1-interacting helicase BRIP1 is deficient in Fanconi anemia.
Nat Genet
2005
;
37
:
931
–3.
36
Bridge WL, Vandenberg CJ, Franklin RJ, Hiom K. The BRIP1 helicase functions independently of BRCA1 in the Fanconi anemia pathway for DNA crosslink repair.
Nat Genet
2005
;
37
:
953
–7.
37
Mosedale G, Niedzwiedz W, Alpi A, et al. The vertebrate Hef ortholog is a component of the Fanconi anemia tumor-suppressor pathway.
Nat Struct Mol Biol
2005
;
12
:
763
–71.
38
Hahn SA, Hoque AT, Moskaluk CA, et al. Homozygous deletion map at 18q21.1 in pancreatic cancer.
Cancer Res
1996
;
56
:
490
–4.
39
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
–6.
40
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
–6.
41
Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase.
Nature
2005
;
434
:
913
–7.
42
Farmer H, McCabe N, Lord CJ, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy.
Nature
2005
;
434
:
917
–21.
43
De Soto JA, Wang X, Tominaga Y, et al. The inhibition and treatment of breast cancer with poly(ADP-ribose) polymerase (PARP-1) inhibitors.
Int J Biol Sci
2006
;
2
:
179
–85.
44
De Soto JA, Deng CX. PARP-1 inhibitors: are they the long-sought genetically specific drugs for BRCA1/2-associated breast cancers?
Int J Med Sci
2006
;
3
:
117
–23.
45
Kalb R, Duerr M, Wagner M, et al. Lack of sensitivity of primary Fanconi's anemia fibroblasts to UV and ionizing radiation.
Radiat Res
2004
;
161
:
318
–25.
46
Gallmeier E, Calhoun ES, Rago C, et al. Targeted disruption of FANCC and FANCG in human cancer provides a preclinical model for specific therapeutic options.
Gastroenterology
2006
;
130
:
2145
–54.
47
Benson JD, Chen YN, Cornell-Kennon SA, et al. Validating cancer drug targets.
Nature
2006
;
441
:
451
–6.
48
Yamamoto K, Ishiai M, Matsushita N, et al. Fanconi anemia FANCG protein in mitigating radiation- and enzyme-induced DNA double-strand breaks by homologous recombination in vertebrate cells.
Mol Cell Biol
2003
;
23
:
5421
–30.
49
Sharan SK, Bradley A. Murine Brca2: sequence, map position, and expression pattern.
Genomics
1997
;
40
:
234
–41.
50
Cunningham SC, Gallmeier E, Hucl T, et al. A detrimental phenotype as a counterweight to the evolution of tumor-suppressor loss in tumorigenesis: explorations in MKK4-Null cancer cells. Cancer Res 2006;66:5560–4.
51
Van der Heijden MS, Brody JR, Elghalbzouri-Maghrani E, Zdzienicka MZ, Kern SE. Does tumorigenesis select for or against mutations of the DNA repair-associated genes BRCA2 and MRE11? Considerations from somatic mutations in microsatellite unstable (MSI) gastrointestinal cancers.
BMC Genet
2006
;
7
:
3
.
52
Chen M, Tomkins DJ, Auerbach W, et al. Inactivation of Fac in mice produces inducible chromosomal instability and reduced fertility reminiscent of Fanconi anaemia.
Nat Genet
1996
;
12
:
448
–51.
53
Cheng NC, van de Vrugt HJ, van der Valk MA, et al. Mice with a targeted disruption of the Fanconi anemia homolog Fanca.
Hum Mol Genet
2000
;
9
:
1805
–11.
54
Koomen M, Cheng NC, van de Vrugt HJ, et al. Reduced fertility and hypersensitivity to mitomycin C characterize Fancg/Xrcc9 null mice.
Hum Mol Genet
2002
;
11
:
273
–81.
55
Whitney MA, Royle G, Low MJ, et al. Germ cell defects and hematopoietic hypersensitivity to γ-interferon in mice with a targeted disruption of the Fanconi anemia C gene.
Blood
1996
;
88
:
49
–58.
56
Wong JC, Alon N, McKerlie C, Huang JR, Meyn MS, Buchwald M. Targeted disruption of exons 1 to 6 of the Fanconi Anemia group A gene leads to growth retardation, strain-specific microphthalmia, meiotic defects and primordial germ cell hypoplasia.
Hum Mol Genet
2003
;
12
:
2063
–76.
57
Yang Y, Kuang Y, De Oca RM, et al. Targeted disruption of the murine Fanconi anemia gene, Fancg/Xrcc9.
Blood
2001
;
98
:
3435
–40.
58
Bennett LM, McAllister KA, Blackshear PE, et al. BRCA2-null embryonic survival is prolonged on the BALB/c genetic background.
Mol Carcinog
2000
;
28
:
174
–83.
59
Connor F, Bertwistle D, Mee PJ, et al. Tumorigenesis and a DNA repair defect in mice with a truncating Brca2 mutation.
Nat Genet
1997
;
17
:
423
–30.
60
Friedman LS, Thistlethwaite FC, Patel KJ, et al. Thymic lymphomas in mice with a truncating mutation in Brca2.
Cancer Res
1998
;
58
:
1338
–43.
61
Ludwig T, Chapman DL, Papaioannou VE, Efstratiadis A. Targeted mutations of breast cancer susceptibility gene homologs in mice: lethal phenotypes of Brca1, Brca2, Brca1/Brca2, Brca1/p53, and Brca2/p53 nullizygous embryos.
Genes Dev
1997
;
11
:
1226
–41.
62
McAllister KA, Bennett LM, Houle CD, et al. Cancer susceptibility of mice with a homozygous deletion in the COOH-terminal domain of the Brca2 gene.
Cancer Res
2002
;
62
:
990
–4.
63
Sharan SK, Morimatsu M, Albrecht U, et al. Embryonic lethality and radiation hypersensitivity mediated by Rad51 in mice lacking Brca2.
Nature
1997
;
386
:
804
–10.
64
Suzuki A, de la Pompa JL, Hakem R, et al. Brca2 is required for embryonic cellular proliferation in the mouse.
Genes Dev
1997
;
11
:
1242
–52.
65
Houghtaling S, Timmers C, Noll M, et al. Epithelial cancer in Fanconi anemia complementation group D2 (Fancd2) knockout mice.
Genes Dev
2003
;
17
:
2021
–35. Epub 2003 Jul 2031.
66
Liu TX, Howlett NG, Deng M, et al. Knockdown of zebrafish Fancd2 causes developmental abnormalities via p53-dependent apoptosis.
Dev Cell
2003
;
5
:
903
–14.
67
Daniels MJ, Wang Y, Lee M, Venkitaraman AR. Abnormal cytokinesis in cells deficient in the breast cancer susceptibility protein BRCA2.
Science
2004
;
306
:
876
–9.
68
Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications.
Am J Pathol
2000
;
156
:
1767
–71.
69
Goggins M, Schutte M, Lu J, et al. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas.
Cancer Res
1996
;
56
:
5360
–4.
70
Hahn SA, Greenhalf B, Ellis I, et al. BRCA2 germline mutations in familial pancreatic carcinoma.
J Natl Cancer Inst
2003
;
95
:
214
–21.
71
Murphy KM, Brune KA, Griffin C, et al. Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, BRCA2 in familial pancreatic cancer: deleterious BRCA2 mutations in 17%.
Cancer Res
2002
;
62
:
3789
–93.
72
Ozcelik H, Schmocker B, Di Nicola N, et al. Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients.
Nat Genet
1997
;
16
:
17
–8.
73
Topaloglu O, Hurley PJ, Yildirim O, Civin CI, Bunz F. Improved methods for the generation of human gene knockout and knockin cell lines.
Nucleic Acids Res
2005
;
33
:
e158
.
74
Kohli M, Rago C, Lengauer C, Kinzler KW, Vogelstein B. Facile methods for generating human somatic cell gene knockouts using recombinant adeno-associated viruses.
Nucleic Acids Res
2004
;
32
:
e3
.
75
Hudson DF, Morrison C, Ruchaud S, Earnshaw WC. Reverse genetics of essential genes in tissue-culture cells: “dead cells talking.”
Trends Cell Biol
2002
;
12
:
281
–7.
76
Arwert F, Rooimans MA, Westerveld A, Simons JW, Zdzienicka MZ. The Chinese hamster cell mutant V-H4 is homologous to Fanconi anemia (complementation group A).
Cytogenet Cell Genet
1991
;
56
:
23
–6.
77
Zdzienicka MZ, Simons JW. Mutagen-sensitive cell lines are obtained with a high frequency in V79 Chinese hamster cells.
Mutat Res
1987
;
178
:
235
–44.
78
Overkamp WJ, Rooimans MA, Neuteboom I, Telleman P, Arwert F, Zdzienicka MZ. Genetic diversity of mitomycin C-hypersensitive Chinese hamster cell mutants: a new complementation group with chromosomal instability.
Somat Cell Mol Genet
1993
;
19
:
431
–7.
79
Kraakman-van der Zwet M, Overkamp WJ, van Lange RE, et al. Brca2 (XRCC11) deficiency results in radioresistant DNA synthesis and a higher frequency of spontaneous deletions.
Mol Cell Biol
2002
;
22
:
669
–79.
80
Liu N, Lamerdin JE, Tucker JD, et al. The human XRCC9 gene corrects chromosomal instability and mutagen sensitivities in CHO UV40 cells.
Proc Natl Acad Sci U S A
1997
;
94
:
9232
–7.
81
Wilson JB, Johnson MA, Stuckert AP, et al. The Chinese hamster FANCG/XRCC9 mutant NM3 fails to express the monoubiquitinated form of the FANCD2 protein, is hypersensitive to a range of DNA damaging agents and exhibits a normal level of spontaneous sister chromatid exchange.
Carcinogenesis
2001
;
22
:
1939
–46.
82
Wiegant WW, Overmeer RM, Godthelp BC, van Buul PP, Zdzienicka MZ. Chinese hamster cell mutant, V-C8, a model for analysis of Brca2 function.
Mutat Res
2006
;
600
:
79
–88.
83
Harborth J, Elbashir SM, Bechert K, Tuschl T, Weber K. Identification of essential genes in cultured mammalian cells using small interfering RNAs.
J Cell Sci
2001
;
114
:
4557
–65.
84
Birmingham A, Anderson EM, Reynolds A, et al. 3′ UTR seed matches, but not overall identity, are associated with RNAi off-targets.
Nat Methods
2006
;
3
:
199
–204.
85
Fedorov Y, Anderson EM, Birmingham A, et al. Off-target effects by siRNA can induce toxic phenotype.
RNA
2006
;
12
:
1188
–96.
86
van der Heijden MS, Brody JR, Dezentje DA, et al. In vivo therapeutic responses contingent upon Fanconi/BRCA2 tumor status.
Clin Cancer Res
2005
;
11
:
7508
–15.
87
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
–85.
88
Wang SC, Shao R, Pao AY, Zhang S, Hung MC, Su LK. Inhibition of cancer cell growth by BRCA2.
Cancer Res
2002
;
62
:
1311
–4.
89
Wang H, Han H, Von Hoff DD. Targeting a loss-of-function mutation in the BRCA2 gene in pancreatic cancer. In: 97th AACR Annual Meeting, Washington DC, April 1-5, 2006.
90
Warren M, Lord CJ, Masabanda J, Griffin D, Ashworth A. Phenotypic effects of heterozygosity for a BRCA2 mutation.
Hum Mol Genet
2003
;
12
:
2645
–56.
91
Hatanaka A, Yamazoe M, Sale JE, et al. Similar effects of Brca2 truncation and Rad51 paralog deficiency on immunoglobulin V gene diversification in DT40 cells support an early role for Rad51 paralogs in homologous recombination.
Mol Cell Biol
2005
;
25
:
1124
–34.
92
Ludwig T, Fisher P, Murty V, Efstratiadis A. Development of mammary adenocarcinomas by tissue-specific knockout of Brca2 in mice.
Oncogene
2001
;
20
:
3937
–48.
93
Morgan NV, Tipping AJ, Joenje H, Mathew CG. High frequency of large intragenic deletions in the Fanconi anemia group A gene.
Am J Hum Genet
1999
;
65
:
1330
–41.
94
Tischkowitz M, Ameziane N, Waisfisz Q, et al. Bi-allelic silencing of the Fanconi anaemia gene FANCF in acute myeloid leukaemia.
Br J Haematol
2003
;
123
:
469
–71.
95
Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells.
Nat Med
1996
;
2
:
561
–6.
96
Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med
2001
;
344
:
1031
–7.
97
Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.
N Engl J Med
2004
;
350
:
2129
–39.
98
Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.
Science
2004
;
304
:
1497
–500.
99
Gurtan AM, D'Andrea AD. Dedicated to the core: understanding the Fanconi anemia complex.
DNA Repair (Amst)
2006
;
5
:
1119
–25.
100
van Zeeburg HJ, Snijders PJ, Pals G, et al. Generation and molecular characterization of head and neck squamous cell lines of Fanconi anemia patients.
Cancer Res
2005
;
65
:
1271
–6.
101
Ikeda H, Matsushita M, Waisfisz Q, et al. Genetic reversion in an acute myelogenous leukemia cell line from a Fanconi anemia patient with biallelic mutations in BRCA2.
Cancer Res
2003
;
63
:
2688
–94.
102
Meyer S, Fergusson WD, Oostra AB, et al. A cross-linker-sensitive myeloid leukemia cell line from a 2-year-old boy with severe Fanconi anemia and biallelic FANCD1/BRCA2 mutations.
Genes Chromosomes Cancer
2005
;
42
:
404
–15.