Abstract
Precision medicine in cancer is the idea that the recognition and targeting of key genetic drivers of a patient’s tumor can permit more effective and less toxic outcomes. Point mutations that alter protein function have been primary targets. Yet in ovarian cancer, unique genetic mutations have been identified only in adult granulosa cell tumors, with a number of other point mutations present in mucinous, clear cell and endometrioid carcinoma subtypes. By contrast, the serous subtype of ovarian cancer shows many fewer point mutations but cascading defects in DNA damage repair that leads to a network of gains and losses of entire genes called somatic copy number alterations. The shuffling and selection of the thousands of genes in serous ovarian cancer has made it a complex disease to understand, but patterns are beginning to emerge based on our understanding of key cellular protein networks that may provide a better basis for future implementation of precision medicine for this most prevalent subtype of disease.
Keywords
- SCNA
- aneuploidy
- autophagy
- beclin-1
- p53
1. Introduction
When a patient asks an oncologist what tumor cells are, the frequent explanation is that the “Cancer cells are normal cells that accumulate genetic mutations, which causes them to grow out of control.” Yet the idea of what a mutation is, and what it can do, varies. It has essentially become dogma that mutations be grouped into two broad categories. One class has been described as either
This categorization has now had clinical impact. Genes that are known as
The
A second class of drivers involve SNV-mediated inactivation of
However, SOC has a further characteristic related to its poor capacity to repair DNA. SOC has the highest ratio of somatic copy number alterations (SCNAs) to SNVs for any major cancer. SCNAs are a broad group of genetic changes that encompass a myriad of short insertions, short deletions, translocations and inversions (Figure 1, left panel). SCNAs contribute to the mutational landscape of cancer, expanding the scope of changes beyond the more ‘simple’ SNVs. The impact of this on SOC malignancy will be the focus of this chapter.
2. SCNA overview and incidence
A gene normally occurs in the human nucleus twice. This normal 2N “dosage” of copy number, which originates from zygote formation, consists of one paternal gene and one maternal gene. SCNAs, which alter this occur in two types: amplifications and deletions. An amplification occurs when a chromosomal region containing a gene is copied. That gene will no longer have the normal 2N copy number, but, depending upon the number of times it is copied, could be 3N, 4N, or in cases of massive amplification, up to 200 N and more. Contrasting with this expansive range, SCNAs that result from deletions most frequently reduce the copy number to 1N. Total gene loss (0N) can occur in rare cases, and is associated with a very small fraction of the overall number of deletions. Nonetheless, these rarer SCNA-derived genotypes will obviously impact function most, since the lack of any gene copy means that the encoded protein cannot be produced. SCNAs are the most common lesions in cancer, occurring much more commonly than SNVs (Figure 1, right panel).
SCNAs occur via a variety of mechanisms in cancer [3]. Entire chromosomes may be gained/lost during cell division, generating 3N or 1N copy number status for all genes on the chromosome. This occurs due to failed cell-division checkpoints resulting in chromosome missegregation. In contrast to such gains at the total chromosome level, tiny “focal” SCNAs may alter a single gene (or even part of a gene). The most common example of this is
Within the Cancer Genome Atlas (TCGA) data sets, the presence of 3N and 1N gene copies dominate the SCNA genomic landscape. This is true across all tumors, including those tumors where SCNAs are highly prevalent, such as SOC [8]. SCNAs are prevalent in SOC. In fact, only about one third of all genes in primary tumors have a normal 2N gene dosage. Roughly a quarter of the total genes in the tumors show an extra gene copy (to 3N) and just over a third lose a gene copy (to 1N). By contrast, only 0.7% loses both gene copies (0N), while 4.2% are amplified to 4N or greater. In practice, the focus on understanding tumor biology has been only on these last two cases (total deletion and gross amplification, respectively). This has a reasonable basis; the effects of total loss or gross amplification are easiest to study.
The common gene changes (i.e., 1N and 3N) have not been the subject of focused study. Many scientists assume that the deletion, or addition, of a single gene copy has limited effect. Recessive genetic alleles are not uncommon in nature, supporting the idea that the loss of a single gene copy can be compensated for. However, the loss of a single gene may not reflect the situation in ovarian cancer, where massive genetic alteration occurs, and compensation may not be possible if the same cellular pathway is repeatedly targeted by SCNAs (Figure 2).
More than 80% of genes affected by SCNAs show concordant alteration of mRNA levels [9, 10]. For ~70% of genes, this correlates with steady-state protein levels [11]. Thus, SCNAs offer a predictable, but not absolute, indication of protein expression. This is relevant to ovarian cancer, as SCNAs modify on average 67% of the SOC genome, whereas SNVs modify only 0.12% of the average SOC genome [12]. Less than 10% of SOC patients are mutated in a targetable driver gene [12, 13].
3. Ovarian cancer and copy number alterations
It seems self-evident that an understanding of “driver SCNAs” is absolutely essential to our capacity to target the biology of the disease. Genetic disorders such as Down’s syndrome (trisomy 21) and Cri du Chat (5p monosomy) and DiGeorge Syndrome (loss of only 30–40 alleles on 22q11) clearly indicate the penetrative biology of multiple SCNA. More importantly, such lesions affect only ~2% of the genome, while SCNA in SOC affect 67% of genes. Other subtypes of ovarian cancer vary widely in their SCNA burden, but are typically much lower, and are associated with SNVs.
As most SCNA are “monoallelic” changes resulting in a 1N or 3N genotype, is there any reason to expect a phenotype, given our understanding of recessive alleles? Recurrent patterns in serous ovarian cancer suggest that frequently affected regions may be selected for as the tumor evolves. In high grade SOC, the most prevalent SNVs could have been predicted from literature preceding the genomics era. For decades, the mutation of
Aside from very infrequent gene losses paired with mutations, there are also a few SCNAs which drive cancer through amplification of oncogenes. The stem-cell transcription factor
There are plausible reasons for this. It may be that every SOC tumor is truly unique from a mutational standpoint: that those SNVs found in only one tumor nonetheless are driver genes, collaborating in ways that we understand poorly [25]. It is thus possible that drivers have already been sequenced and annotated by SCNA studies, but due to high “background” or “passenger” SCNAs it remains unclear which SCNAs are critical to the tumor’s biology [8]. The implications of this are enormous, and would necessitate an unparalleled level of personalized therapies targeting such extremely rare mutations. A second reason that SNVs have not yielded common drivers may be that further sequencing of whole genomes and epigenomes will reveal additional drivers prevalent across patients which have remained undetected by exome sequencing.
The problem investigators consistently encounter is the ubiquitous heterogeneity in SOC. Heterogeneity exists at all levels of genetics, manifesting as
Fewer genomic studies have been performed on other types of ovarian cancer. Some limited data are available on SCNAs for Clear cell and endometrioid subtypes, which share the amplification of PIK3CA and the MYC-containing 8q24 region with SOC [30, 31, 32]. Larger SCNAs encompassing whole chromosome arms rather than smaller changes dominate the clear cell ovarian cancer SCNA landscape [32]. With the exception of 17p loss (containing
Despite the limited data on these non-serous subtypes, there is good reason to expect much more data is coming soon. The copy-number arrays employed in the Cancer Genome Atlas studies sell for less than $100USD per sample, which bests the current, but constantly decreasing, cost of whole-genome sequencing. Eight oncology treatment and research centers are participating in project GENIE, which has just released 19,000 new tumor datasets to the public and will continue to grow [33]. As sequencing becomes a normal part of the treatment strategy for patients, the number of samples will likely outpace scientists’ ability to fully analyze and comprehend the complex data. Nonetheless, gathering these data is essential to progressing our understanding of the differences between cancer subtypes, which will facilitate the matching of pharmaceuticals to genotype. For now, the largest datasets exist in SOC, and will be the focus of the remainder of discussion.
4. The interplay of p53 mutation with copy number instability
Mutation in p53 has a long research history in many cancer types, and ovarian cancer is no exception. Ovarian cancer mutations within
Mechanistically this could occur via the deletion or duplication of entire chromosomes or genomes, followed by many subsequent changes enabled by the extra copies of genes, or via chromosome missegregation event, leading one or more chromosomes to acquire massive damage [41]. Either possibility can explain the high frequency of chromothripsis, a highly-disorganized form of hundreds or thousands of SCNAS, in SOC. Mutant p53 enables such mechanisms of SCNA formation by preventing the death of the cell that bears them, as missegregation directly induces p53-dependent cell-cycle arrest followed by apoptosis [42]. In one well-controlled study, ‘dominant negative’ p53 reduced the cell cycle delay associated with trisomy in mammalian cells, yet it was rare that gain of any single chromosome in those cells resulted in any proliferative advantage [43]. Thus, partial or gained p53 function may contribute. Many p53 mutations maintain partial function, while mutations such as R273H (the most common variant of TP53 found in SOC) provide a gain of function by directly impairing Mre11/ATM-dependent DNA damage responses [44].
It is likely that mutation in
5. BRCA1/2 mutations and homologous repair defects
Though few SNVs in ‘classic’ tumor genes are found in SOC relative to other cancer types, BRCA1/2 mutations are among the most frequent at ~10% [12]. BRCA genes work in coordination with dozens of other proteins to perform genome maintenance via homologous recombination [4]. The double-stranded break repair pathway begins with PARylation of the break site by PARP1, megabases of phosphorylation of H2AX and subsequent formation of Rad51 filaments. Brca1 & Brca2 bind Rad51 to stimulate strand invasion of sister chromatids during homology directed repair. While only ~10% of SOC are mutated in BRCA1 or BRCA2, it is noteworthy that 75% of patients have lost one of two alleles of BRCA1 and 57% have lost an allele of BRCA2. Very few tumors (~1.5%) have homozygous deletions in BRCA1 [12], suggesting a system of compromised (but not lost) function. In fact, mRNA expression level does not track linearly with such deletions. It remains somewhat unclear if these monoallelic deletions do have a phenotype under genotoxic stress in human cells.
Mutations in homologous repair coordinating factors are often found in serous, clear cell, endometrioid, and carcinosarcoma ovarian cancers [47, 48]. Specific mutation patterns are found within BRCA1/2 or otherwise homologous repair deficient cells. Without functional homologous repair, cells default to non-homologous end joining (NHEJ) to repair double-stranded DNA (dsDNA) lesions. NHEJ does not perfectly repair DNA, but rather often introduces small insertions or deletions along with single-nucleotide variants at the break site. These mutational marks are frequently found in non-serous ovarian cancers, yet are unlikely the main drivers of SCNA instability in serous ovarian cancer. However, NHEJ factors involved in repairing unresolved dsDNA breaks across different chromosomes, or creating translocations and other complex rearrangements, are compromised in 40%
Genetic and epigenetic changes alter
Loss in BRCA1 enables microsatellite instability in mouse models and in colorectal cancer [55], though not in ovarian cancer [56, 57]. Microsatellite instability directly leads to centrosome amplification, but SCNA instability, which may explain why it is observed in only a small minority of ovarian cancer patients, and is not linked to BRCA mutation status. Nonetheless, BRCA genes are inactivated through allelic deletions and expression modulation in ovarian cancer. Inactivation of
Coincident SCNA events enable subsequent SCNA catastrophe. BRCA1 is located within kilobases of the neighboring autophagy gene BECN1. Autophagy is a critical catabolic infrastructure that enables cellular survival, requiring only 10% or less of normal autophagy gene dose [64]. Monoallelic loss of
In summary,
6. Pathways affected by SCNAs in serous ovarian cancer
Each cancer probably evolves at least 6–10 independent oncogene or tumor suppressor alterations [69] to circumvent natural homeostatic controls known as the “Hallmarks of Cancer” [70, 71]. These hallmarks include the evasion of regulated cell death, immortalization through telomere maintenance, defects in cell cycle control, immune system suppression, and enabling of metastatic capacity through physical and metabolic means. Traditionally, it has been assumed that single gene mutations are responsible for many of these oncogenic changes. Altered p53 function promotes escape to half of these hallmarks on its own, and mutations in strong oncogenes such as Ras family members, or growth factor receptor genes such as FGFRs, Her2 and even Met supplement many of the remaining hallmarks.
Individual gene amplifications can impact serous ovarian cancer. Aside from
Recently, we analyzed single nucleotide and short ‘in frame’ deletion mutations across 120 validated oncogenes and tumor suppressors, finding that as many as 48% of serous ovarian primary tumors do not contain mutations in
To analyze this, we developed new pathway network analytics tools to identify disrupted pathways in serous ovarian cancer in this unusually unstable genetic background. Despite the high levels of heterogeneity across patients, we found that coincident gene disruptions fell along surprisingly consistent patterns tumor-to-tumor, specifically suppressing or amplifying specific cellular pathways.
6.1. Autophagy
By far the most significantly suppressed pathway which stood out as unique in serous ovarian cancer and triple negative breast cancer was macroautophagy, which is most commonly known, simply, as autophagy. The term autophagy (“
Given this critical cellular function, we considered it counter-intuitive that cancer cells would delete a wide array of autophagy genes. In fact, KRAS mutant cancers have been described as “addicted” to autophagy, particularly in hypoxic or otherwise nutrient-stressed microenvironments [78]. This interpretation has been debated [79, 80], but the fact that autophagy has been established as a tumor suppressor system [81, 82], it is not exclusive of the possibility that specific tumor genotypes can promote addiction to autophagy [78]. Mono-allelic losses in the autophagy gene
6.2. Proteosome
Interestingly, a number of other proteostasis control pathways were suppressed in serous ovarian cancer, and foremost among these is complementary to autophagy, the ubiquitin-proteasome system. The core subunits, encoded by
6.3. p53 Interactome
In addition to
6.4. Metabolism
Metabolism is fundamentally disrupted in serous ovarian cancer. This may be predicted by the observation that patients with metabolic disruptions are at risk for disease, or have a predisposition to tumors to undergo metastatic growth to adipose tissue [93, 94]. A shift to glycolysis, the Warburg effect, is a general hallmark of cancer. Glycolytic shift is considered essential to provide the many constituent molecules required for cell division: nucleotides, lipids, and amino acids, moreso than simply ATP which is produced in higher quantities by oxidative phosphorylation [95]. A metabolic pathway found to be suppressed with almost equal magnitude to autophagy was the arginine and proline metabolism pathway, particularly through deletions in
6.5. Adipocytokine
Adipocytokine signaling and
6.6. Peroxisome
An unusually altered pathway in serous ovarian cancer bridges metabolism, fatty acid oxidation and proteostasis disruption:
While each of these pathways can help to define phenotypes associated with SOC, they also have the capacity to enable development of new classes of pathway-targeted therapeutics. It may be possible in future for SCNA-modified pathways to serve as targets the same way that SNVs do, now.
7. Potential for new treatments by targeting copy number alterations
SNVs have a proven track record of targetability using small molecules. Nonetheless, in the case of SOC, new cures are unlikely to be found unless somatic copy number alterations (SCNAs) are considered. Defining this interplay will be a difficult task. It remains unclear exactly which SCNAs are most critical to SOC proliferation and metastasis. The creation of cell line models will require new methods of whole-chromosome manipulation, even as attracting pharmaceutical company support will be harder due to limited experience which such targeting strategies, as well as conservative business approaches towards eventual clinical adaptation. Nonetheless, there are reasons to be optimistic that SCNA-targeted therapeutics can be effective and that some could enter the clinic in the near future.
Consider the abundance of SCNAs in advanced SOC relative to other cancer. The successful tumors have undergone selection. The phenotypes produced include well-known hallmarks of cancer: including cell cycle defects, heightened glucose uptake [106], spontaneous proliferative immortality [107], and dysregulated autophagy [108]. The same studies identify aneuploidy-associated characteristics which present vulnerabilities particular to these unstable cells. Perhaps the most promising vulnerability is an increased reliance on protein quality control processes such as ribosome biogenesis and maintenance factors and the cellular recycling process, autophagy. Aneuploid cells require these systems to function, and may result in a general reliance on catabolic function due to the proteotoxic effect of protein-complex subunit imbalance. Early studies recognized a general, if partial, sensitivity of these cells to rapamycin [106].
Chromosome instability can incur resistance to taxanes, a common front line therapeutic for SOC [109]. Chromosomal instability endowed by docetaxel may in fact lead to subsequent additional chemoresistance [110], though it is clear that the resistant phenotype is at least initially offset by an increased sensitivity to carboplatin, the second primary chemotherapeutic co-administered with a taxane as standard of care. Although aneuploidy enables oncogenic characteristics, it offers targetable vulnerabilities as well.
The mapped SCNA patterns in SOC revealed a general fault in proteostasis control, centered on autophagy [8]. Yet these cells require autophagy to maintain viability. The delicate balance within SOC relative to normal tissue, appears to provide a therapeutic window for proteostasis-targeting agents. Since SOC cells are already severely disrupted in their proteostasis-regulatory mechanisms, further disturbance can greatly compromise survival even as normal cells readily process the insult. Given this premise, we developed the Combination of Autophagy Selective Therapeutics (COAST) method to effectively manage SOC in the lab [8]. The general approach involves directly stressing the proteostasis system, while inhibiting autophagy resolution (Figure 4).
Mice given the cocktail of five proteostasis drugs did not lose weight nor negatively alter their blood chemistry panel [111], tolerating these drugs for months of daily treatment [8]. In mouse models using recurrent human SOC cells, the proteostasis drugs out-performed platinum-taxane dual treatment. The results are consistent with previous approaches pursuing a “cyclops” hypothesis: that monoallelic deletions in cancer sensitize cancer cells to further disruption of that gene’s function [112]. As normal cells bear a full complement of all pathway genes, they are typically less sensitive to such stresses, which opens therapeutic windows for treatment.
Given that proteostasis pathways are only one type of disruption caused by SCNAs in SOC, what other SCNA-disrupted pathways might be targetable? Alluring targets may include the proteins downstream of the E3 ligases commonly deleted in SOC; inhibition of cell-cycle regulators may selectively target SOC cells even without any mutation or than copy number changes. Strong amplification of peroxisome transporters and the glycerophospholipid metabolism pathways suggest that metabolic targeting may be worthwhile.
A caveat of such designs is heterogeneity inherent in disease. Copy number instability is the result of SOC cells’ extraordinary ability to create, tolerate, and expand genomic variation. Mathematical modeling of real tumor genetic data suggests that even small tumors with low mutation rates are statistically likely to contain multiple independent clones able to resist a particular drug treatment [117, 118]. Current SOC chemotherapeutics stimulate aneuploidy. Taxanes result in chromosome missegregation and platinum agents promote translocation events due to cross-strand DNA lesions. While it is absolutely true that common chemotherapeutics have limited combination potential due to dose limiting toxicities, that does not preclude the use of highly specific drugs to be used in combination or as maintenance therapy in SOC treatment regimens. Most likely, drugs independently targeting the many SCNA-disrupted pathways may be required to completely cure a patient.
While most patients are caught late in the evolution of their disease, it may not be “too late” to treat them. A genomic analysis in highly metastatic recurrent SOC patients found that the tumors likely form a metastasis-to-metastasis spread [14]. This may explain why round after round of different chemotherapy can extend the life of SOC patients [119]. This implies that current chemotherapy is quite effective at destroying a great majority of cells, and the challenge that remains is how to complement it. The COAST strategy studied in our lab functioned equally well or better for cisplatin resistant forms of SOC [8]. Autophagy has been widely implicated in the ability of quiescent cells to survive, including in SOC [120], and has been directly shown to enable growth of Doxil resistant disease [121]. However, given that one COAST agent, chloroquine is often prescribed for the same patient for decades in high-risk malaria areas, while another, nelfinavir, is a daily long-term HIV medication with no serious side-effects, the use of COAST is warranted based on the decades of use of COAST drugs, in humans, for diseases other than cancer. The side effects are well established to be below current chemotherapeutics carboplatin, paclitaxel, and Doxil [111]. The greatest health concern may lie in kidney cells, which are also exquisitely sensitive to autophagy drugs [122].
Since the drugs target different but complementary pathways, it is feasible to design clinical trials involving either simultaneous treatment or sequential treatment, enabling a greater chance of minimized side effects. Compromised DNA repair feeds into this pathway, suggesting that the recent successes of the PARP inhibitors are not simply due to BRCA1 complementation. An expanded range of options must be aggressively explored in the near future if we are to understand how to exploit the SCNA genetics of ovarian cancer in a timely fashion.
Abbreviations
AOC | Australian Ovarian Cancer (study) |
N | the number of copies of a given gene present in a cell (e.g., 3N) |
SCNA | somatic copy number alteration |
SNV | single nucleotide variant (a point mutation) |
SOC | serous ovarian carcinoma |
TCGA | the Cancer Genome Atlas |
TP53 | tumor protein 53 kDa gene (protein is p53) |
References
- 1.
Pon JR, Marra MA. Driver and passenger mutations in cancer. Annual Review of Pathology. 2015; 10 :25-50 - 2.
Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers: Origins, consequences, and clinical use. Cold Spring Harbor Perspectives in Biology. 2010; 2 (1):a001008 - 3.
Tschaharganeh DF, Bosbach B, Lowe SW. Coordinated tumor suppression by chromosome 8p. Cancer Cell. 2016; 29 (5):617-619 - 4.
Ciccia A, Elledge SJ. The DNA damage response: Making it safe to play with knives. Molecular Cell. 2010; 40 (2):179-204 - 5.
Storlazzi CT et al. Gene amplification as double minutes or homogeneously staining regions in solid tumors: Origin and structure. Genome Research. 2010; 20 (9):1198-1206 - 6.
Vogt N et al. Molecular structure of double-minute chromosomes bearing amplified copies of the epidermal growth factor receptor gene in gliomas. Proceedings of the National Academy of Sciences of the United States of America. 2004; 101 (31):11368-11373 - 7.
Cai Y, Sablina AA. Cancer-associated chromosomal deletions: Size makes a difference. Cell Cycle. 2016; 15 (21):2850-2851 - 8.
Delaney JR et al. Haploinsufficiency networks identify targetable patterns of allelic deficiency in low mutation ovarian cancer. Nature Communications. 2017; 8 :14423 - 9.
Davoli T et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell. 2013; 155 (4):948-962 - 10.
Cai Y et al. Loss of chromosome 8p governs tumor progression and drug response by altering lipid metabolism. Cancer Cell. 2016; 29 (5):751-766 - 11.
Zhang H et al. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 2016; 166 (3):755-765 - 12.
Cancer Genome Atlas Research, N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011; 474 (7353):609-15 - 13.
Patch AM et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015; 521 (7553):489-494 - 14.
Schwarz RF et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: A phylogenetic analysis. PLoS Medicine. 2015; 12 (2):e1001789 - 15.
Kurose K et al. Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas. The American Journal of Pathology. 2001; 158 (6):2097-2106 - 16.
Baker VV et al. C-myc amplification in ovarian cancer. Gynecologic Oncology. 1990; 38 (3):340-342 - 17.
Li SB et al. Allele loss at the retinoblastoma locus in human ovarian cancer. Journal of the National Cancer Institute. 1991; 83 (9):637-640 - 18.
Sasano H et al. An analysis of abnormalities of the retinoblastoma gene in human ovarian and endometrial carcinoma. Cancer. 1990; 66 (10):2150-2154 - 19.
Fukumoto M et al. Association of Ki-ras with amplified DNA sequences, detected in human ovarian carcinomas by a modified in-gel renaturation assay. Cancer Research. 1989; 49 (7):1693-1697 - 20.
Slamon DJ et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989; 244 (4905):707-712 - 21.
Zhang X et al. Amplification and rearrangement of c-erb B proto-oncogenes in cancer of human female genital tract. Oncogene. 1989; 4 (8):985-989 - 22.
Bourhis J et al. Expression of a human multidrug resistance gene in ovarian carcinomas. Cancer Research. 1989; 49 (18):5062-5065 - 23.
Mayr D et al. Analysis of gene amplification and prognostic markers in ovarian cancer using comparative genomic hybridization for microarrays and immunohistochemical analysis for tissue microarrays. American Journal of Clinical Pathology. 2006; 126 (1):101-109 - 24.
Nakayama K et al. Amplicon profiles in ovarian serous carcinomas. International Journal of Cancer. 2007; 120 (12):2613-2617 - 25.
Krogan NJ et al. The cancer cell map initiative: Defining the hallmark networks of cancer. Molecular Cell. 2015; 58 (4):690-698 - 26.
Zack TI et al. Pan-Cancer patterns of somatic copy number alteration. Nature Genetics. 2013; 45 (10):1134-1140 - 27.
Waclaw B et al. A spatial model predicts that dispersal and cell turnover limit intratumour heterogeneity. Nature. 2015; 525 (7568):261-264 - 28.
Burrell RA et al. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013; 501 (7467):338-345 - 29.
Mutation C, Pathway Analysis C. Working group of the international cancer genome, pathway and network analysis of cancer genomes. Nature Methods. 2015; 12 (7):615-621 - 30.
Huang RY et al. Histotype-specific copy-number alterations in ovarian cancer. BMC Medical Genomics. 2012; 5 :47 - 31.
Tapper J et al. Evidence for divergence of DNA copy number changes in serous, mucinous and endometrioid ovarian carcinomas. British Journal of Cancer. 1997; 75 (12):1782-1787 - 32.
Uehara Y et al. Integrated copy number and expression analysis identifies profiles of whole-arm chromosomal alterations and subgroups with favorable outcome in ovarian clear cell carcinomas. PLoS One. 2015; 10 (6):e0128066 - 33.
Project GENIE Goes Public. Cancer Discovery. 2017; 7 (2):118 - 34.
Marks JR et al. Overexpression and mutation of p53 in epithelial ovarian cancer. Cancer Research. 1991; 51 (11):2979-2984 - 35.
Lane DP. Cancer. p53, guardian of the genome. Nature. 1992; 358 (6381):15-16 - 36.
Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997; 88 (3):323-331 - 37.
Zilfou JT, Lowe SW. Tumor suppressive functions of p53. Cold Spring Harbor Perspectives in Biology. 2009; 1 (5):a001883 - 38.
Davis MA et al. Nelfinavir is effective against human cervical cancer cells in vivo: A potential treatment modality in resource-limited settings. Drug Design, Development and Therapy. 2016; 10 :1837-1846 - 39.
Manning AL, Benes C, Dyson NJ. Whole chromosome instability resulting from the synergistic effects of pRB and p53 inactivation. Oncogene. 2014; 33 (19):2487-2494 - 40.
Delaney JR, Stupack DG. Whole genome pathway analysis identifies an association of cadmium response gene loss with copy number variation in mutant p53 bearing uterine endometrial carcinomas. PLoS One. 2016; 11 (7):e0159114 - 41.
Janssen A et al. Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science. 2011; 333 (6051):1895-1898 - 42.
Thompson SL, Compton DA. Proliferation of aneuploid human cells is limited by a p53-dependent mechanism. The Journal of Cell Biology. 2010; 188 (3):369-381 - 43.
Sheltzer JM et al. Single-chromosome gains commonly function as tumor suppressors. Cancer Cell. 2017; 31 (2):240-255 - 44.
Liu DP, Song H, Xu Y. A common gain of function of p53 cancer mutants in inducing genetic instability. Oncogene. 2010; 29 (7):949-956 - 45.
Martincorena I et al. Tumor evolution. High burden and pervasive positive selection of somatic mutations in normal human skin. Science. 2015; 348 (6237):880-886 - 46.
McCreery MQ et al. Evolution of metastasis revealed by mutational landscapes of chemically induced skin cancers. Nature Medicine. 2015; 21 (12):1514-1520 - 47.
Pennington KP et al. Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clinical Cancer Research. 2014; 20 (3):764-775 - 48.
Wang YK et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nature Genetics. 2017; 49 (6):856-865 - 49.
Lieber MR et al. Nonhomologous DNA end joining (NHEJ) and chromosomal translocations in humans. Sub-Cellular Biochemistry. 2010; 50 :279-296 - 50.
McCormick A et al. Ovarian cancers harbor defects in nonhomologous end joining resulting in resistance to Rucaparib. Clinical Cancer Research. 2016 - 51.
Sears CR, Turchi JJ. Complex cisplatin-double strand break (DSB) lesions directly impair cellular non-homologous end-joining (NHEJ) independent of downstream damage response (DDR) pathways. The Journal of Biological Chemistry. 2012; 287 (29):24263-24272 - 52.
Aly A, Ganesan S. BRCA1, PARP, and 53BP1: Conditional synthetic lethality and synthetic viability. Journal of Molecular Cell Biology. 2011; 3 (1):66-74 - 53.
Mirza MR et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. The New England Journal of Medicine. 2016; 375 (22):2154-2164 - 54.
Niraparib in recurrent ovarian cancer. New England Journal of Medicine. 2017; 376 (8):801-2 - 55.
Romanowicz-Makowska H et al. Analysis of microsatellite instability and BRCA1 mutations in patients from hereditary nonpolyposis colorectal cancer (HNPCC) family. Polish Journal of Pathology. 2005; 56 (1):21-26 - 56.
Segev Y et al. Risk factors for ovarian cancers with and without microsatellite instability. International Journal of Gynecological Cancer. 2013; 23 (6):1010-1015 - 57.
van der Looij M et al. Allelic imbalance and microsatellite instability in BRCA1 associated breast and ovarian tumors. International Journal of Oncology. 2001; 18 (4):775-780 - 58.
Arizti P et al. Tumor suppressor p53 is required to modulate BRCA1 expression. Molecular and Cellular Biology. 2000; 20 (20):7450-7459 - 59.
Wu LC et al. Identification of a RING protein that can interact in vivo with the BRCA1 gene product. Nature Genetics. 1996; 14 (4):430-440 - 60.
Deng CX. BRCA1: Cell cycle checkpoint, genetic instability, DNA damage response and cancer evolution. Nucleic Acids Research. 2006; 34 (5):1416-1426 - 61.
Xu X et al. Genetic interactions between tumor suppressors Brca1 and p53 in apoptosis, cell cycle and tumorigenesis. Nature Genetics. 2001; 28 (3):266-271 - 62.
Xu X et al. Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells. Molecular Cell. 1999; 3 (3):389-395 - 63.
Deng CX. Tumorigenesis as a consequence of genetic instability in Brca1 mutant mice. Mutation Research. 2001; 477 (1-2):183-189 - 64.
Levine B, Yuan J. Autophagy in cell death: An innocent convict? The Journal of Clinical Investigation. 2005; 115 (10):2679-2688 - 65.
Mathew R et al. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes & Development. 2007; 21 (11):1367-1381 - 66.
Zhao Z et al. UVRAG: At the crossroad of autophagy and genomic stability. Autophagy. 2012; 8 (9):1392-1393 - 67.
Godinho SA et al. Oncogene-like induction of cellular invasion from centrosome amplification. Nature. 2014; 510 (7503):167-171 - 68.
Mackeh R et al. Autophagy and microtubules—New story, old players. Journal of Cell Science. 2013; 126 (Pt 5):1071-1080 - 69.
Kandoth C et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013; 502 (7471):333-339 - 70.
Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000; 100 (1):57-70 - 71.
Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011; 144 (5):646-674 - 72.
Nilsson JA, Cleveland JL. Myc pathways provoking cell suicide and cancer. Oncogene. 2003; 22 (56):9007-9021 - 73.
Sulzmaier FJ, Jean C, Schlaepfer DD. FAK in cancer: Mechanistic findings and clinical applications. Nature Reviews. Cancer. 2014; 14 (9):598-610 - 74.
Jones SF et al. A phase I study of VS-6063, a second-generation focal adhesion kinase inhibitor, in patients with advanced solid tumors. Investigational New Drugs. 2015; 33 (5):1100-1107 - 75.
Yorimitsu T, Klionsky DJ. Autophagy: Molecular machinery for self-eating. Cell Death and Differentiation. 2005; 12 (Suppl 2):1542-1552 - 76.
Mizushima N. Autophagy: Process and function. Genes & Development. 2007; 21 (22):2861-2873 - 77.
Noda T, Suzuki K, Ohsumi Y. Yeast autophagosomes: De novo formation of a membrane structure. Trends in Cell Biology. 2002; 12 (5):231-235 - 78.
Guo JY, White E. Autophagy is required for mitochondrial function, lipid metabolism, growth, and fate of KRAS(G12D)-driven lung tumors. Autophagy. 2013; 9 (10):1636-1638 - 79.
Nyfeler B, Eng CH. Revisiting autophagy addiction of tumor cells. Autophagy. 2016; 12 (7):1206-1207 - 80.
Kroemer G, Levine B. Autophagic cell death: The story of a misnomer. Nature Reviews. Molecular Cell Biology. 2008; 9 (12):1004-1010 - 81.
Avalos Y et al. Tumor suppression and promotion by autophagy. BioMed Research International. 2014; 2014 :603980 - 82.
Jin S. p53, autophagy and tumor suppression. Autophagy. 2005; 1 (3):171-173 - 83.
Yue Z et al. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proceedings of the National Academy of Sciences of the United States of America. 2003; 100 (25):15077-15082 - 84.
Qu X et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. The Journal of Clinical Investigation. 2003; 112 (12):1809-1820 - 85.
Nezis IP et al. Autophagy as a trigger for cell death: Autophagic degradation of inhibitor of apoptosis dBruce controls DNA fragmentation during late oogenesis in Drosophila. Autophagy. 2010; 6 (8):1214-1215 - 86.
Devoy A et al. The ubiquitin-proteasome system and cancer. Essays in Biochemistry. 2005; 41 :187-203 - 87.
Devine T, Dai MS. Targeting the ubiquitin-mediated proteasome degradation of p53 for cancer therapy. Current Pharmaceutical Design. 2013; 19 (18):3248-3262 - 88.
Chen D, Dou QP. The ubiquitin-proteasome system as a prospective molecular target for cancer treatment and prevention. Current Protein & Peptide Science. 2010; 11 (6):459-470 - 89.
Torres EM et al. Identification of aneuploidy-tolerating mutations. Cell. 2010; 143 (1):71-83 - 90.
Chipuk JE et al. Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science. 2004; 303 (5660):1010-1014 - 91.
Tamura RE et al. GADD45 proteins: Central players in tumorigenesis. Current Molecular Medicine. 2012; 12 (5):634-651 - 92.
Katsura M et al. The ATR-Chk1 pathway plays a role in the generation of centrosome aberrations induced by Rad51C dysfunction. Nucleic Acids Research. 2009; 37 (12):3959-3968 - 93.
Lengyel E. Ovarian cancer development and metastasis. The American Journal of Pathology. 2010; 177 (3):1053-1064 - 94.
Clark R et al. Milky spots promote ovarian cancer metastatic colonization of peritoneal adipose in experimental models. The American Journal of Pathology. 2013; 183 (2):576-591 - 95.
Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 2009; 324 (5930):1029-1033 - 96.
Gerner EW, Meyskens FL Jr. Polyamines and cancer: Old molecules, new understanding. Nature Reviews. Cancer. 2004; 4 (10):781-792 - 97.
Altman BJ, Stine ZE, Dang CV. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nature Reviews. Cancer. 2016; 16 (10):619-634 - 98.
Wu X et al. Lipid metabolism in prostate cancer. American Journal of Clinical and Experimental Urology. 2014; 2 (2):111-120 - 99.
Jelski W, Szmitkowski M. Alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) in the cancer diseases. Clinica Chimica Acta. 2008; 395 (1-2):1-5 - 100.
Orywal K et al. The activity of class I, II, III and IV alcohol dehydrogenase isoenzymes and aldehyde dehydrogenase in ovarian cancer and ovarian cysts. Advances in Medical Sciences. 2013; 58 (2):216-220 - 101.
Lodhi IJ, Semenkovich CF. Peroxisomes: A nexus for lipid metabolism and cellular signaling. Cell Metabolism. 2014; 19 (3):380-392 - 102.
Erdmann R, Schliebs W. Peroxisomal matrix protein import: The transient pore model. Nature Reviews. Molecular Cell Biology. 2005; 6 (9):738-742 - 103.
Fang Y et al. PEX3 functions as a PEX19 docking factor in the import of class I peroxisomal membrane proteins. The Journal of Cell Biology. 2004; 164 (6):863-875 - 104.
Hoskins ER et al. Proteomic analysis of ovarian cancer proximal fluids: Validation of elevated peroxiredoxin 1 in patient peripheral circulation. PLoS One. 2011; 6 (9):e25056 - 105.
Jiang H et al. Expression of peroxiredoxin 1 and 4 promotes human lung cancer malignancy. American Journal of Cancer Research. 2014; 4 (5):445-460 - 106.
Torres EM et al. Effects of aneuploidy on cellular physiology and cell division in haploid yeast. Science. 2007; 317 (5840):916-924 - 107.
Williams BR et al. Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Science. 2008; 322 (5902):703-709 - 108.
Stingele S et al. Global analysis of genome, transcriptome and proteome reveals the response to aneuploidy in human cells. Molecular Systems Biology. 2012; 8 :608 - 109.
Swanton C et al. Chromosomal instability determines taxane response. Proceedings of the National Academy of Sciences of the United States of America. 2009; 106 (21):8671-8676 - 110.
Ogden A et al. Docetaxel-induced polyploidization may underlie chemoresistance and disease relapse. Cancer Letters. 2015; 367 (2):89-92 - 111.
Delaney JR et al. A strategy to combine pathway-targeted low toxicity drugs in ovarian cancer. Oncotarget. 2015; 6 (31):31104-31118 - 112.
Nijhawan D et al. Cancer vulnerabilities unveiled by genomic loss. Cell. 2012; 150 (4):842-854 - 113.
McCubrey JA et al. GSK-3 as potential target for therapeutic intervention in cancer. Oncotarget. 2014; 5 (10):2881-2911 - 114.
Hilliard TS et al. Glycogen synthase kinase 3beta inhibitors induce apoptosis in ovarian cancer cells and inhibit in-vivo tumor growth. Anti-Cancer Drugs. 2011; 22 (10):978-985 - 115.
Cohen P, Goedert M. GSK3 inhibitors: Development and therapeutic potential. Nature Reviews. Drug Discovery. 2004; 3 (6):479-487 - 116.
Shimizu T et al. A first-in-Asian phase 1 study to evaluate safety, pharmacokinetics and clinical activity of VS-6063, a focal adhesion kinase (FAK) inhibitor in Japanese patients with advanced solid tumors. Cancer Chemotherapy and Pharmacology. 2016; 77 (5):997-1003 - 117.
Bozic I et al. Evolutionary dynamics of cancer in response to targeted combination therapy. eLife. 2013; 2 :e00747 - 118.
Diaz LA Jr et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012; 486 (7404):537-540 - 119.
Luvero D, Milani A, Ledermann JA. Treatment options in recurrent ovarian cancer: Latest evidence and clinical potential. Therapeutic Advances in Medical Oncology. 2014; 6 (5):229-239 - 120.
Zhang Y et al. NAC1 modulates sensitivity of ovarian cancer cells to cisplatin by altering the HMGB1-mediated autophagic response. Oncogene. 2012; 31 (8):1055-1064 - 121.
Lu Z et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. The Journal of Clinical Investigation. 2008; 118 (12):3917-3929 - 122.
Kimura T et al. Chloroquine in cancer therapy: A double-edged sword of autophagy. Cancer Research. 2013; 73 (1):3-7