Next Article in Journal
Genome-Wide Identification and Analysis Uncovers the Potential Role of JAZ and MYC Families in Potato under Abiotic Stress
Next Article in Special Issue
Mutation Hotspots Found in Bladder Cancer Aid Prediction of Carcinogenic Risk in Normal Urothelium
Previous Article in Journal
Bridging Gaps in HDR Improvement: The Role of MAD2L2, SCAI, and SCR7
Previous Article in Special Issue
Germline Variants in 32 Cancer-Related Genes among 700 Chinese Breast Cancer Patients by Next-Generation Sequencing: A Clinic-Based, Observational Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Analysis of Multiple Primary Malignant Tumors in Women with Breast and Ovarian Cancer

by
Alina Savkova
1,2,3,*,
Lyudmila Gulyaeva
1,2,
Aleksey Gerasimov
1,4 and
Sergey Krasil’nikov
2,3
1
Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
2
V. Zelman Institute for the Medicine and Psychology, Novosibirsk State University, Novosibirsk 630090, Russia
3
E. Meshalkin National Medical Research Center of Ministry of Health of Russian Federation, Novosibirsk 630055, Russia
4
Novosibirsk Region Clinical Oncology Center, Novosibirsk 630108, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6705; https://doi.org/10.3390/ijms24076705
Submission received: 25 February 2023 / Revised: 28 March 2023 / Accepted: 29 March 2023 / Published: 4 April 2023
(This article belongs to the Special Issue Linking Genomic Changes with Cancer in the NGS Era)

Abstract

:
Familial cancer syndromes, which are commonly caused by germline mutations in oncogenes and tumor suppressor genes, are generally considered to be the cause of primary multiple malignant neoplasias (PMMNs). Using targeted genomic sequencing, we screened for eight germline mutations: BRCA1 185delAG, BRCA1 T300G, BRCA1 2080delA, BRCA1 4153delA, BRCA1 5382insC, BRCA2 6174delT, CHEK2 1100delC, and BLM C1642T, which provoke the majority of cases of hereditary breast and ovary cancer syndrome (HBOC), in genomic (blood) DNA from 60 women with PMMNs, including breast (BC) and/or ovarian cancer(s) (OC). Pathogenic allelic forms were discovered in nine samples: in seven instances, it was BRCA1 5382insC, and in the following two, BRCA1 4153delA and BRCA1 T300G. The age of onset in these patients (46.8 years) was younger than in the general Russian population (61.0) for BC but was not for OC: 58.3 and 59.4, correspondingly. There were invasive breast carcinomas of no special type and invasive serous ovarian carcinomas in all cases. Two or more tumors of HBOC-spectrum were only in five out of nine families of mutation carriers. Nevertheless, every mutation carrier has relatives who have developed malignant tumors.

1. Introduction

PMMNs are defined as two or more histologically distinct malignancies that are not induced by metastasis, recurrence, or local spread within one individual.
If the second cancer was detected within 6 months after the first one, this means that PMMNs developed synchronously. Otherwise, the interval between them exceeds 6 months; it can be concluded that tumors occurred metachronously [1,2].
In the case of the development of three tumors, diagnoses are metachronous-metachronous, metachronous-synchronous, synchronous-metachronous, or synchronous-synchronous malignant neoplasms.
Nowadays, the absolute number of PMMNs and their proportion among newly diagnosed malignant neoplasias are on the rise due to improved diagnostics, the aging of the population, and the increased lifespan of cancer patients thanks to successful treatment methods [1,3]. Unfortunately, radiotherapy and certain types of pharmacotherapy also produce immunosuppressive and carcinogenic effects [1,4,5].
The pathogenesis of multiple and single tumors has similar mechanisms. Each person has a greater or lesser hereditary predisposition to the development of cancer, due to the presence in the genome of some germline oncogenic mutations with high, medium, and low degrees of penetrance. During life, under the influence of external (radiation or some carcinogenic substances) and internal (replication mistakes) factors, random somatic mutations occur in DNA to be added to germline ones. The combination of hereditary and acquired mutations in oncogenes and tumor suppressor genes initiates the process of carcinogenesis in the respective cells at an earlier or later age [2].
A decline in immune surveillance associated with aging, long-term stress, chronic inflammation, hormonal changes, certain infections, or immunosuppressive effects of treatment allows the tumor to continue its development. The whole sequence of events may repeat more than once in the same organism [4,5,6,7,8].
The development of metachronous tumors can be considered as an independent process. The simultaneous occurrence of malignant neoplasms may be associated with the action of the same triggering factor, which leads to the development of two or more tumors without any time gap [2,3].
In the Novosibirsk region, the incidence of PMNMs (34.6 per 100,000) exceeds the Russian average (24.8 per 100,000). This may stem from environmental conditions, in particular, high natural background radiation. Environmental influences, reproductive history, infections, behavioral and cultural factors (nutrition, smoking, alcohol consumption), working conditions, and potentially carcinogenic treatment—these risk factors are highly significant, as they lead to the occurrence of somatic mutations [1,4]. Nonetheless, a hereditary predisposition remains one of the main reasons for PMMNs’ development.
Individuals carrying some germline mutations in oncogenes and tumor suppressor genes typically have a family history of cancer, develop a neoplasm at a young age, and often have PMMNs. Furthermore, each mutation leads to the occurrence of tumors of the corresponding spectrum; therefore, some hereditary oncological syndromes have been characterized (for example, HBOC syndrome, Li-Fraumeni syndrome, syndrome of multiple endocrine neoplasia, etc.).
There are hotspots in several genes, mutations in which lead to developing breast, ovarian, pancreatic, gastric, and prostate cancers, and hematological neoplasms [9,10].
If there is a history of BC and OC in the family, as a rule, its members are carriers of one of the following allelic variants: BRCA1 5382insC, BRCA1 4153delA, BRCA1 185delAG, BRCA1 T300G, BRCA1 2080delA, BRCA2 6174delT, CHEK2 1100delC, CHEK2 I157T, and BLM C1642T in Eastern Europe and in Russia [11,12,13,14,15].
All of these genes (and their corresponding proteins) are involved in the regulation of the cell cycle and may also influence the effects of sex hormones in hormone-dependent tissues [16,17,18,19,20].
In addition, a multiplicative interaction between several pathogenic mutations can also play a key role [21]. As regards hereditary BRCA1/2 mutations, tumors develop in accordance with the classical two-hit model, whereas other mechanisms act in the case of CHEK2 and BLM [22].
Thus, PMMNs are highly heterogeneous in their causes, triggering factors, and clinical manifestations (as well as malignancies in general). In order to identify the main patterns of pathology and prognosis, we analyzed all the necessary information about 60 female patients with PMMNs from the Novosibirsk region with BC, OC, or both: age of manifestation, interval between previous and subsequent malignancies, their localizations, treatments, and family cancer histories. Using targeted sequencing, we screened their genomic DNA for eight germline mutations most commonly associated with the development of the HBOC syndrome in this geographical area.
With the development and practical application of personalized therapies, such as poly(ADP-ribose)polymerase inhibitors (PARP inhibitors) for the treatment of BRCA1/2-mutation carriers, and other modern technologies, such as gene theranostics, it is possible to improve the survival and quality of life for patients predisposed to PMMNs [19,23].

2. Results

2.1. Patients’ Characteristics

In our group, there were 49 double primaries and 11 triple primaries.
Forty-eight women had BC, fifteen of them had non-metastatic ones in both mammary glands (in each individual instance, malignant tumors were histologically different or had a discrepancy in hormone-receptor status, or a time gap was at least 5 years), eight had BC and OC, and two had cancers in both mammary glands and ovarian one. Beyond that, 12 patients had OC with another malignant neoplasia(s).
Contralateral BC, OC, and other reproductive tract malignancies (five endometrial carcinomas, five cervical cancers, four uterine sarcomas, one granulosa cell tumor, one borderline ovarian tumor, and one vaginal sarcoma), kidney and digestive tract malignancies (four stomach and two colorectal cancers) more commonly occurred in patients with BC (Figure 1a).
BC, other reproductive tract malignancies (four endometrial carcinomas and one cervical cancer), and thyroid cancers prevailed in patients with OC (Figure 1b).
The age of onset of the first tumor ranged from 27 to 76 years, with an average of 52.7 years. With regard to the second one, it was detected between 30 and 77 years, with a mean age of 58.7 years. The third tumor appeared between 39 and 84 years, with an average of 64.8 years (Figure 2).
Synchronous tumors were developed in 12 patients, metachronous in 37 patients, metachronous-metachronous ones in 9 patients, and metachronous-synchronous in 2 patients. Thus, 23.3% of tumors progressed simultaneously (Figure 3).
Potentially carcinogenic treatments for the previous tumor were performed on 41 (85.4%) of 48 patients with metachronous tumors. In 20 cases, it was chemotherapy using substances that are carcinogenic to humans (Group 1 according to the classification of the International Agency for Research on Cancer (IARC)); in 1 case, treatment included probably carcinogenic to humans agents (Group 2A); in 8 cases, radiotherapy was applied; and in 13, complex treatment: carcinogenic substances (Group 1) along with radiotherapy.
Some of the therapeutic methods used lead to DNA damage and immunosuppression (alkylating agents and radiation); others interfere with the action of sex hormones (tamoxifen) [5,24].
We classified 28 family histories as not aggravated and 15 as aggravated by HBOC-spectrum malignancies. In 11 cases, there were at least two malignant tumors in relatives, but less than two of them belonged to the HBOC-spectrum (Figure 4).

2.2. Targeted Sequencing

Pathogenic allelic variants were discovered in nine samples (Table 1). In seven cases, there was BRCA1 5382insC (rs80357906), in one case, BRCA1 4153delA (rs80357711), and in another one, BRCA1 T300G (rs28897672).
We consider it appropriate to give examples of mutation carriers’ family histories, paying attention to the fact that not all of them are aggravated by HBOC-spectrum malignancies (see Figure 5 and Figure 6 below).

3. Discussion

A combination of many factors leads to the development of PMMNs. However, some patients’ characteristics such as the age of tumor onset, family history of cancer, time gap between previous and subsequent malignancies, localizations, and potentially carcinogenic treatments, are most important for understanding the general patterns of pathology and prognosis.
Of the causes, the most significant is the carriage of oncogenic mutations with high and medium degrees of penetrance. Due to the contribution of such cases to the statistics, tumors in patients with PMMNs develop at a younger age than the average age of the cancer manifestation in the same geographical area.
So, the mean age of first (52.7 years) and even second (58.7 years) neoplasms in the group was lower than the age of cancer manifestation in Russia (63.9 years for women in 2019). The same trend is noted by other authors [25,26]. The mean age of BC for the first and subsequent tumors (64 malignancies, 56.1 years) is lower than the average in Russia (61.0 years). The mean age of OC (22 malignancies, 54.6 years) is also lower than the average in Russia (59.4 years). The difference is about five years.
The mean age of BC manifestation in BRCA mutation carriers was 46.8 years (nine cases). This result is consistent with data from a large study that included 3797 BRCA1 mutation carriers. The average age was 40 years in that study [27]. Thus, BC in BRCA1 mutation carriers develops 15–20 years earlier than in the general population. The mean age of OC onset (58.3 years, five cases) appears not to differ from that of the general population (59.4 in Russia in 2019). Other studies do not contradict our data [28].
In all examined patients, 23.3% of tumors had simultaneous progression. The rate of synchronous primary malignancies during the past ten years in Russia is 26.1–30.1. In the Novosibirsk region, its share is about 25%. In other countries, the proportion is nearly the same [3,4]. It seems probable that a single triggering factor can lead to the development of several tumors at the same time in a patient with a cancer predisposition. Sometimes it is a big problem to find an anticancer therapy that covers both cancer types without increased toxicity and pharmacological interactions [1]. In addition, many widely used methods have potentially carcinogenic effects.
An association between certain types of treatment (chemotherapy and radiotherapy) and the development of a subsequent metachronous tumor is proven. Such therapy increases the lifetime risk of leukemia, kidney cancer, and some other malignancies [4,29]. Radiotherapy increases the risk of thyroid cancer and subsequent malignancies of breast, bone, connective tissue, and lung at the area of exposure [1]. Two patients from the group developed leukemia several years after the complex treatment, and two developed kidney tumors. It is interesting to note that all of them have an aggravated family history, and potentially carcinogenic treatment is an additional risk factor in these cases.
The most common PMMNs are gender-specific [3]. The same proportion is observed for single malignancies. Sex steroid hormones act as growth factors. The peculiarities of the metabolism of these hormones and their action on the receptors in the tissues are associated with the presence of hereditary and acquired mutations in several genes (BRCA1/2, CHEK2, CYP1A1, CYP19, SULT1, and others), reproductive history, and hormone therapy use.
Sex hormones and the expression of their respective receptors affect the growth of tumors of the ovaries, breast, reproductive tract, and colon [3,30]. Many studies indicate a relationship between the patient’s reproductive history and the development of tumors in these organs [1]. Of course, the role of sex hormones in carcinogenesis in the digestive tract is still debatable. However, the risk of colorectal cancer is higher in those patients who had tumors of the breast or reproductive tract [31], and the risk of BC is higher in patients with a history of gastric, colon, endometrial, or ovarian cancer [32]. In addition, the hormonal treatment of a primary BC increases the risk of these types of cancer [1].
The cumulative proportion of combinations with such tumors for the BC is 83.9%, and for the OC, 70.4%.
C. Frank et al. described that shared environmental risk factors (spousal risk) have less of an impact on familial risk than shared genes. The strongest correlation was observed in relatives for prostate, breast, and colorectal cancer. The authors note that many of the underlying genes are still unknown. Well-known mutations in highly penetrant genes explain a small proportion of the genetic basis of cancers, whereas familial aggregation has been suspected for almost all cancers [33,34].
An aggravated family history is considered as an additional basis for mutation screening in the cases of prostate, colon, endometrium, and ovary cancer [34]. However, the current guidelines for HBOC syndrome (the Mainstreaming Cancer Genetics (MCG) criteria) do not mention a family history of cancer as an important basis for testing, but only as an additional one [35].
Only five (1, 3, 6, 7, 8) out of nine family histories were aggravated by tumors of the HBOC spectrum.
Nevertheless, every mutation carrier has relatives who have developed malignant tumor(s). Therefore, a non-aggravated family history does not guarantee the absence of pathogenic variants, and cancer-based MCG criteria appear to be more effective than previous ones [36].
All BRCA1 mutation carriers, except patient 8, identify themselves with Russian nationality. Patient 8 is Ashkenazi Jewish but has a mutation BRCA1 T300G classified as not typical in representatives of this nationality [36].
The carriage of each specific mutation leads to the development of the corresponding histological types of tumors. In all well-described cases, BRCA mutation carriers have had invasive breast carcinomas of no special type, also called invasive ductal carcinomas (grade II or III), and invasive serous ovarian carcinomas. These histological types of BC and OC develop in 80% and 67% of cases, respectively, in BRCA1 mutation carriers [27]. Patients 4 and 7 also had non-HBOC tumors. Patient 4 developed serous endometrial carcinoma 12 years after treatment of BC with tamoxifen. However, not only tamoxifen but also germline BRCA1 5382insC may increase the risk of this histopathological subtype of uterine cancer [37]. Patient 7 developed uterine leiomyosarcoma at the age of 62, i.e., 30 and 12 years after the first and the second primary BCs, respectively. This patient had never been treated with tamoxifen, which could also exacerbate the risk of uterine sarcoma [24]. Therefore, the development of sarcoma in the patient may be associated with BRCA1 5382insC. This relationship was described by Laitman Y. et al. [37].
All of the newly identified carriers of mutations and their relatives received personal recommendations for surveillance. The possibilities for the prevention of malignant tumors are extremely limited and consist of the preventive removal of an organ (while it is still healthy) in which a tumor is expected to appear. After the operation, its function can be replaced. For carriers of highly penetrant BRCA1/2 mutations, bilateral salpingo-oophorectomy or more frequent screening is recommended. This translates to a reduction in OC and BC-specific mortality [38]. PARP inhibitors, furthermore, were added to the treatment regimens of these patients.

4. Materials and Methods

4.1. Patients and Their Histories

In our study, we collected all essential information about 60 female patients with PMMNs who had taken a part in the research: age of manifestation, interval between first and subsequent malignancies, their localizations, treatments, and family cancer histories.
Each of them received treatment for malignant neoplasias in the Novosibirsk Region Clinical Oncology Center or in E. Meshalkin National Medical Research Center of the Ministry of Health of the Russian Federation, Novosibirsk. Each patient had a history of two or three malignant neoplasms and had developed BC, OC, or both. Only patients who lived all their lives in the Novosibirsk region were included in the study. Therefore, the environmental impact (in particular, the influence of background radiation and insolation) was nearly the same for all patients.
The family cancer history was drawn up for concrete patients; information about their relatives’ cancer histories was assembled. The criterion for classifying a family history as aggravated was the presence of two or more tumors from the HBOC spectrum in first-degree, second-degree, and third-degree relatives (parents, grandparents, siblings, uncles, and aunts).

4.2. Morphological Examination

The material for the morphological study was obtained during the cytoreductive surgery. The morphological examination was carried out using light-optical microscopy (Zeiss Axio-Imager.M2 microscope (Carl Zeiss Microscopy GmbH, 07745, Jena, Germany)) with staining of sections by hematoxylin and eosin.

4.3. Targeted Sequencing

Then, we estimated the frequency of pathogenic variants in eight locations: BRCA1 185delAG (p.Glu23fs; rs80357914), BRCA1 T300G (p.Cys61Gly; rs28897672), BRCA1 2080delA (p.Tyr655fs; rs80357522), BRCA1 4153delA (p.Glu1346fs; rs80357711), BRCA1 5382insC (p.Gln1756fs; rs80357906), BRCA2 6174delT (p.Ser1982fs; rs80359550), CHEK2 1100delC (p.Thr367fs; rs555607708), and BLM C1642T (p.Gln548Ter, rs200389141) for women who had developed PMMNs.
From each patient, 2 mL of venous blood was obtained. Samples were collected between November 2017 and April 2018. Ethylenediaminetetraacetic acid was used for safety. Blood samples were stored at −20 °C until further use. The isolation of genomic DNA was performed using the classical phenol-chloroform method.
Isolated DNA was quantified with the QuantumDNA 211 Kit (Evrogen, Moskow, Russia). DNA concentration ranged from 1.1 to 67.4 ng/mL.
A TruSight Cancer Illumina panel (Illumina Inc, San Diego, CA, USA) was used for the targeted genomic sequencing. Raw sequencing read quality was assessed using FastQC v. 0.11.8 (Babraham Bioinformatics, Babraham Institute, Babraham, Cambridgeshire, UK). The reads were trimmed for quality (less than Q20), and adapters were removed using Trimmomatic. Alignment of reads to the reference human genome GRCh37.75/hg19 (Ensembl) (EMBL’s European Bioinformatics Institute, Hinxton, Cambridgeshire, UK) was performed with a Burrows-Wheeler Aligner (BWA), v. 0.7.17 (by Li H. and Durbin R, Wellcome Sanger Institute, Hinxton, Cambridgeshire, UK). To report alignment statistics and determine read duplicates, we applied Sequence Alignment Map (SAM) tools and Picard tools. Base quality score recalibration was carried out with a Genome Analysis Toolkit 4 (GATK4) (v. 4.1.2) (Broad Institute, Cambridge, MA, USA), and dbSNP (build 144) (National Center for Biotechnology Information, Bethesda, MD, USA). Variant calling was performed with GATK (v. 4.1.2) HaplotypeCaller. We excluded false positives using StrandBiasBySample, StrandOddsRatio, and BaseQualityRankSumTest annotations (Broad Institute, Cambridge, MA, USA), as well as mis-sequenced single-nucleotide variants in polyN motifs, such as GGGTG > GGGGG, CCCCG > CCCCC, and others. For functional annotation of variants, ANNOVAR was used.

5. Conclusions

One of the most relevant tasks of clinical oncology is the prediction of new tumors in patients predisposed to cancer.
Obviously, risk-reducing surgery cannot be applied widely. For most patients, only the early detection of cancer, while it is still curable, is acceptable. To predict the localization of a tumor, the clinician needs to know about the presence of oncogenic mutations in the patient’s genome, his family cancer history, and the features of already developed neoplasm(s). The integrated use of clinical and laboratory methods for examining patients, including targeted sequencing, can bring success in the early diagnosis and personalized treatment of tumors in patients with PMMNs and their relatives. This approach can increase the length and quality of their lives.

Author Contributions

Conceptualization, L.G. and S.K.; methodology, L.G.; software, L.G.; validation, L.G. and S.K.; formal analysis, A.S. and A.G.; investigation, A.S. and A.G.; resources, S.K. and A.G.; data curation, L.G.; writing—original draft preparation, A.S.; writing—review and editing, L.G.; visualization, A.S.; supervision, L.G. and S.K.; project administration, L.G.; funding acquisition, L.G. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by budgetary financing project FGMU-2022-0004, N1021050601085-9-1.6.4; 3.1.6.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, Russia, 630117 (protocol code 18, date of approval 15 July 2022).

Informed Consent Statement

Written informed consent was obtained from all patients involved in the study. Published data do not allow the identification of patients, including by the patients themselves.

Data Availability Statement

Epidemiological data for Russia and the Novosibirsk region are taken from open annual reports of the Ministry of Health: The state of oncological care for the population of Russia (from 2011 to 2021).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vogt, A.; Schmid, S.; Heinimann, K.; Frick, H.; Herrmann, C.; Cerny, T.; Omlin, A. Multiple primary tumours: Challenges and approaches, a review. ESMO Open 2017, 2, e000172. [Google Scholar] [CrossRef] [Green Version]
  2. Testori, A.; Cioffi, U.; De Simone, M.; Bini, F.; Vaghi, A.; Lemos, A.A.; Ciulla, M.M.; Alloisio, M. Multiple primary synchronous malignant tumors. BMC Res. Notes 2015, 8, 730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Tanjak, P.; Suktitipat, B.; Vorasan, N.; Juengwiwattanakitti, P.; Thiengtrong, B.; Songjang, C.; Therasakvichya, S.; Laiteerapong, S.; Chinswangwatanakul, V. Risks and cancer associations of metachronous and synchronous multiple primary cancers: A 25-year retrospective study. BMC Cancer 2021, 21, 1045. [Google Scholar] [CrossRef] [PubMed]
  4. Pan, S.-Y.; Huang, C.-P.; Chen, W.-C. Synchronous/Metachronous Multiple Primary Malignancies: Review of Associated Risk Factors. Diagnostics 2022, 12, 1940. [Google Scholar] [CrossRef]
  5. Rice, J.M. Immunosuppression. In Tumour Site Concordance and Mechanisms of Carcinogenesis; Baan, R.A., Stewart, B.W., Straif, K., Eds.; International Agency for Research on Cancer: Lyon, France, 2019; pp. 159–162. [Google Scholar]
  6. Hong, H.; Wang, Q.; Li, J.; Liu, H.; Meng, X.; Zhang, H. Aging, Cancer and Immunity. J. Cancer 2019, 10, 3021–3027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Falcinelli, M.; Thaker, P.H.; Lutgendorf, S.K.; Conzen, S.D.; Flaherty, R.L.; Flint, M.S. The Role of Psychologic Stress in Cancer Initiation: Clinical Relevance and Potential Molecular Mechanisms. Cancer Res. 2021, 81, 5131–5140. [Google Scholar] [CrossRef] [PubMed]
  8. Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 1492. [Google Scholar] [CrossRef]
  9. Daly, M.B.; Pal, T.; Berry, M.P.; Buys, S.S.; Dickson, P.; Domchek, S.M.; Elkhanany, A.; Friedman, S.; Goggins, M.; Hutton, M.L.; et al. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 77–102. [Google Scholar] [CrossRef] [PubMed]
  10. Mohamad, H.B.; Apffelstaedt, J.P. Counseling for male BRCA mutation carriers—A review. Breast 2008, 17, 441–450. [Google Scholar] [CrossRef]
  11. Gorodetska, I.; Serga, S.; Lahuta, T.; Ostapchenko, L.; Demydov, S.; Khranovska, N.; Skachkova, O.; Inomistova, M.; Kolesnik, O.; Svintsitsky, V.; et al. Prevalence of two BRCA1 mutations, 5382insC and 300T > G, in ovarian cancer patients from Ukraine. Fam. Cancer 2017, 16, 471–476. [Google Scholar] [CrossRef]
  12. Foretová, L.; Navrátilová, M.; Svoboda, M.; Vašíčková, P.; Hrabincová, E.S.; Házová, J.; Kleiblová, P.; Kleibl, Z.; Macháčková, E.; Palácová, M.; et al. Recommendations for Preventive Care for Women with Rare Genetic Cause of Breast and Ovarian Cancer. Klin. Onkol. 2019, 32, 6–13. [Google Scholar] [CrossRef]
  13. Cybulski, C.; Kluźniak, W.; Huzarski, T.; Wokołorczyk, D.; Kashyap, A.; Rusak, B.; Stempa, K.; Gronwald, J.; Szymiczek, A.; Bagherzadeh, M.; et al. The spectrum of mutations predisposing to familial breast cancer in Poland. Int. J. Cancer 2019, 145, 3311–3320. [Google Scholar] [CrossRef]
  14. Bogdanova, N.; Togo, A.V.; Ratajska, M.; Kluźniak, W.; Takhirova, Z.; Tarp, T.; Prokofyeva, D.; Bermisheva, M.; Yanus, G.A.; Gorodnova, T.V.; et al. Prevalence of the BLM nonsense mutation, p.Q548X, in ovarian cancer patients from Central and Eastern Europe. Fam. Cancer 2014, 14, 145–149. [Google Scholar] [CrossRef] [PubMed]
  15. Sokolenko, A.P.; Iyevleva, A.G.; Preobrazhenskaya, E.V.; Mitiushkina, N.V.; Abysheva, S.N.; Suspitsin, E.N.; Kuligina, E.S.; Gorodnova, T.V.; Pfeifer, W.; Togo, A.V.; et al. High prevalence and breast cancer predisposing role of the BLM c.1642 C > T (Q548X) mutation in Russia. Int. J. Cancer 2011, 130, 2867–2873. [Google Scholar] [CrossRef]
  16. Wang, L.; Di, L.-J. BRCA1 and Estrogen/Estrogen Receptor in Breast Cancer: Where They Interact? Int. J. Biol. Sci. 2014, 10, 566–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Malone, J.L.; Nelson, A.C.; Lieberman, R.; Anderson, S.; Holt, J.T. Oestrogen-mediated phosphorylation and stabilization of BRCA2 protein in breast. J. Pathol. 2008, 217, 380–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Cybulski, C.; Huzarski, T.; Byrski, T.; Gronwald, J.; Dębniak, T.; Jakubowska, A.; Górski, B.; Wokołorczyk, D.; Masojć, B.; Narod, S.; et al. Estrogen receptor status in CHEK2-positive breast cancers: Implications for chemoprevention. Clin. Genet. 2009, 75, 72–78. [Google Scholar] [CrossRef]
  19. Arora, A.; Abdel-Fatah, T.M.; Agarwal, D.; Doherty, R.; Moseley, P.M.; Aleskandarany, M.A.; Green, A.R.; Ball, G.; Alshareeda, A.T.; Rakha, E.A.; et al. Transcriptomic and Protein Expression Analysis Reveals Clinicopathological Significance of Bloom Syndrome Helicase (BLM) in Breast Cancer. Mol. Cancer Ther. 2015, 14, 1057–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Turk, A.A.; Wisinski, K.B. PARP inhibitors in breast cancer: Bringing synthetic lethality to the bedside. Cancer 2018, 124, 2498–2506. [Google Scholar] [CrossRef]
  21. Sokolenko, A.P.; Bogdanova, N.; Kluzniak, W.; Preobrazhenskaya, E.V.; Kuligina, E.S.; Iyevleva, A.G.; Aleksakhina, S.N.; Mitiushkina, N.V.; Gorodnova, T.V.; Bessonov, A.A.; et al. Double heterozygotes among breast cancer patients analyzed for BRCA1, CHEK2, ATM, NBN/NBS1, and BLM germ-line mutations. Breast Cancer Res. Treat. 2014, 145, 553–562. [Google Scholar] [CrossRef]
  22. Suspitsin, E.N.; Yanus, G.A.; Sokolenko, A.P.; Yatsuk, O.S.; Zaitseva, O.A.; Bessonov, A.A.; Ivantsov, A.O.; Heinstein, V.A.; Klimashevskiy, V.F.; Togo, A.V.; et al. Development of breast tumors in CHEK2, NBN/NBS1 and BLM mutation carriers does not commonly involve somatic inactivation of the wild-type allele. Med. Oncol. 2014, 31, 828. [Google Scholar] [CrossRef] [PubMed]
  23. Panda, S.; Hajra, S.; Kaushik, A.; Rubahn, H.; Mishra, Y.; Kim, H. Smart nanomaterials as the foundation of a combination approach for efficient cancer theranostics. Mater. Today Chem. 2022, 26, 101182. [Google Scholar] [CrossRef]
  24. Homer, L.; Muller, M.; Dupré, P.-F.; Lucas, B.; Pradier, O. Uterine sarcoma associated with tamoxifen use after breast cancer: Review of the pathogenesis. J. Gynécologie Obs. Biol. La Reprod. 2009, 38, 629–633. [Google Scholar] [CrossRef] [PubMed]
  25. Bagri, P.K.; Singh, D.; Singhal, M.K.; Singh, G.; Mathur, G.; Jakhar, S.L.; Beniwal, S.; Sharma, N.; Kumar, H.S.; Sharma, A.; et al. Double Primary Malignancies: A Clinical & Pathological Analysis Report from a Regional Cancer Institute in India. Iran. J. Cancer Prev. 2014, 7, 66–72. [Google Scholar]
  26. Xiao, L.; Cao, T.; Ou, J.; Liang, W. Clinical characteristics and prognostic analysis of multiple primary malignant neoplasms in female patients with breast cancer or genitalia malignancies. PeerJ 2022, 10, e13528. [Google Scholar] [CrossRef] [PubMed]
  27. Mavaddat, N.; Barrowdale, D.; Andrulis, I.L.; Domchek, S.M.; Eccles, D.; Nevanlinna, H.; Ramus, S.J.; Spurdle, A.; Robson, M.; Sherman, M.; et al. Pathology of Breast and Ovarian Cancers among BRCA1 and BRCA2 Mutation Carriers: Results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiol. Biomark. Prev. 2012, 21, 134–147. [Google Scholar] [CrossRef] [Green Version]
  28. Biglia, N.; Sgandurra, P.; Bounous, V.E.; Maggiorotto, F.; Piva, E.; Pivetta, E.; Ponzone, R.; Pasini, B. Ovarian cancer in BRCA1 and BRCA2 gene mutation carriers: Analysis of prognostic factors and survival. Ecancermedicalscience 2016, 10, 639. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, C.; Shen, Y.; Zhang, Y.; Guo, F.; Li, Q.; Zhang, H.; Han, X.; Zhao, H.; Yang, Z. Metachronous Multiple Primary Carcinoma With Acute Promyelocytic Leukemia: 2 Cases Report and Literature Review. Front. Oncol. 2022, 12, 893319. [Google Scholar] [CrossRef]
  30. Zervoudakis, A.; Strickler, H.D.; Park, Y.; Xue, X.; Hollenbeck, A.; Schatzkin, A.; Gunter, M.J. Reproductive History and Risk of Colorectal Cancer in Postmenopausal Women. J. Natl. Cancer Inst. 2011, 103, 826–834. [Google Scholar] [CrossRef] [Green Version]
  31. Jang, J.-H.; Cotterchio, M.; Gallinger, S.; Knight, J.A.; Daftary, D. Family history of hormonal cancers and colorectal cancer risk: A case-control study conducted in Ontario. Int. J. Cancer 2009, 125, 918–925. [Google Scholar] [CrossRef] [Green Version]
  32. Zheng, G.; Sundquist, J.; Sundquist, K.; Ji, J. Family history of breast cancer as a second primary malignancy in relatives: A nationwide cohort study. BMC Cancer 2021, 21, 1210. [Google Scholar] [CrossRef] [PubMed]
  33. Frank, C.; Fallah, M.; Ji, J.; Sundquist, J.; Hemminki, K. The population impact of familial cancer, a major cause of cancer. Int. J. Cancer 2013, 134, 1899–1906. [Google Scholar] [CrossRef]
  34. Frank, C.; Fallah, M.; Sundquist, J.; Hemminki, A.; Hemminki, K. Population Landscape of Familial Cancer. Sci. Rep. 2015, 5, 12891. [Google Scholar] [CrossRef] [Green Version]
  35. Kemp, Z.; Turnbull, A.; Yost, S.; Seal, S.; Mahamdallie, S.; Poyastro-Pearson, E.; Warren-Perry, M.; Eccleston, A.; Tan, M.-M.; Teo, S.H.; et al. Evaluation of Cancer-Based Criteria for Use in Mainstream BRCA1 and BRCA2 Genetic Testing in Patients with Breast Cancer. JAMA Netw. Open 2019, 2, e194428. [Google Scholar] [CrossRef] [Green Version]
  36. Tennen, R.I.; Laskey, S.B.; Koelsch, B.L.; McIntyre, M.H.; Tung, J.Y. Identifying Ashkenazi Jewish BRCA1/2 founder variants in individuals who do not self-report Jewish ancestry. Sci. Rep. 2020, 10, 7669. [Google Scholar] [CrossRef] [PubMed]
  37. Laitman, Y.; Michaelson-Cohen, R.; Levi, E.; Chen-Shtoyerman, R.; Reish, O.; Josefsberg Ben-Yehoshua, S.; Bernstein-Molho, R.; Keinan-Boker, L.; Rosengarten, O.; Silverman, B.G.; et al. Uterine cancer in Jewish Israeli BRCA1/2 mutation carriers. Cancer 2018, 125, 698–703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Gasparri, M.L.; Taghavi, K.; Fiacco, E.; Zuber, V.; Di Micco, R.; Gazzetta, G.; Valentini, A.; Mueller, M.D.; Papadia, A.; Gentilini, O.D. Risk-Reducing Bilateral Salpingo-Oophorectomy for BRCA Mutation Carriers and Hormonal Replacement Therapy: If It Should Rain, Better a Drizzle than a Storm. Medicina 2019, 55, 415. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The other invasion sites: (a) in patients with BC; (b) in patients with OC. Three combinations per case are considered for triple primary malignancies.
Figure 1. The other invasion sites: (a) in patients with BC; (b) in patients with OC. Three combinations per case are considered for triple primary malignancies.
Ijms 24 06705 g001
Figure 2. Ages of patients at the time malignant tumors were first detected.
Figure 2. Ages of patients at the time malignant tumors were first detected.
Ijms 24 06705 g002
Figure 3. Intervals between the previous and the next malignancy. Two intervals per case are considered for triple primary malignancies.
Figure 3. Intervals between the previous and the next malignancy. Two intervals per case are considered for triple primary malignancies.
Ijms 24 06705 g003
Figure 4. Aggravation of family histories by HBOC-spectrum cancers and other malignancies.
Figure 4. Aggravation of family histories by HBOC-spectrum cancers and other malignancies.
Ijms 24 06705 g004
Figure 5. Family history of patient 3, aggravated by tumors of HBOC-spectrum. Persons who developed a tumor of HBOC-spectrum are marked in dark gray; relatives with other malignancies are marked in light gray.
Figure 5. Family history of patient 3, aggravated by tumors of HBOC-spectrum. Persons who developed a tumor of HBOC-spectrum are marked in dark gray; relatives with other malignancies are marked in light gray.
Ijms 24 06705 g005
Figure 6. Family history of patient 9, not aggravated by tumors of HBOC-spectrum.
Figure 6. Family history of patient 9, not aggravated by tumors of HBOC-spectrum.
Ijms 24 06705 g006
Table 1. Mutation carriers and their family histories.
Table 1. Mutation carriers and their family histories.
The First TumorThe Second TumorThe Third Tumor
CaseHistological Type TNM StageAgeHistological TypeTNM StageAgeHistological TypeTNM StageAgeFamily Cancer History: Localization and Age
Patients with BRCA1 5382insC
1.HGSOC 1 T1a
N0
M0
55IBC-NST 2 (grade II)T1a
N0
M0
56 PMMNs: BC + OC (up to 50), E(M)C (52)
2.IBC-NST (grade III)T1a
N0
M0
32HGSOCT3
N0
M0
58 CRC 3 (70),
BC (44), esophageal cancer (37)
3.IBC-NST (grade II; sin.)T2
N0
M0
38IBC-NST (grade III; dex.)T2
N0
M0
40 BC (34),
BC (50),
bone sarcoma (48), lung cancer (31)
4.IBC-NST, (grade II)T2
N0
M0
48LG 4 serous E(M)C 5T1b
N0
M0
60 BC (32),
lung cancer (77)
5.IBC-NST (grade II; sin.)T2
N0
M0
48IBC-NST (grade II; dex.)T2
N0
M0
59HGSOCT1b
N0
M0
60stomach cancer (50)
6.IBC-NST (grade II; sin.)T1
N0
M0
54IBC-NST (grade III; dex.)T1
N1
M0
59HGSOCT3c
N0
M0
64BC (48),
BC (60)
7.BCT2
N1
M0
32IBC-NST (grade II)T1a
N0
M0
50Uterine leiomyosarcomaT2
N0
M0
62Bilateral BC (41);
thyroid cancer (52)
Patient with BRCA1 T300G
8.IBC-NST (grade II; sin.)T2
N1
M0
59IBC-NST (grade II; dex.)T2
N1
M0
63 BC (60), stomach cancer (74), prostate cancer (52)
Patient with BRCA1 4153delA
9.IBC-NST (grade II)T2
N1
M0
49HGSOCT3
N0
M0
52 CRC (78), cancer of unknown primary site (19), malignant brain tumor (51), nasopharyngeal carcinoma (48)
1 HGSOC—high-grade serous ovarian carcinoma; 2 IBC-NST—invasive breast carcinoma of no special type; 3 CRC—colorectal cancer; 4 LG—low-grade; 5 E(M)C—endometrial cancer (carcinoma).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Savkova, A.; Gulyaeva, L.; Gerasimov, A.; Krasil’nikov, S. Genetic Analysis of Multiple Primary Malignant Tumors in Women with Breast and Ovarian Cancer. Int. J. Mol. Sci. 2023, 24, 6705. https://doi.org/10.3390/ijms24076705

AMA Style

Savkova A, Gulyaeva L, Gerasimov A, Krasil’nikov S. Genetic Analysis of Multiple Primary Malignant Tumors in Women with Breast and Ovarian Cancer. International Journal of Molecular Sciences. 2023; 24(7):6705. https://doi.org/10.3390/ijms24076705

Chicago/Turabian Style

Savkova, Alina, Lyudmila Gulyaeva, Aleksey Gerasimov, and Sergey Krasil’nikov. 2023. "Genetic Analysis of Multiple Primary Malignant Tumors in Women with Breast and Ovarian Cancer" International Journal of Molecular Sciences 24, no. 7: 6705. https://doi.org/10.3390/ijms24076705

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop