Next Article in Journal
Small-Molecule Fluorescent Probe for Detection of Sulfite
Next Article in Special Issue
Ion Channels and Personalized Medicine in Gynecological Cancers
Previous Article in Journal
The Role of the Adenosine System on Emotional and Cognitive Disturbances Induced by Ethanol Binge Drinking in the Immature Brain and the Beneficial Effects of Caffeine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Potential Targeted Therapies in Ovarian Cancer

by
Yagmur Sisman
1,2,
Lau Kræsing Vestergaard
1,
Douglas Nogueira Perez de Oliveira
1,
Tim Svenstrup Poulsen
1,
Tine Henrichsen Schnack
3,
Claus Høgdall
2 and
Estrid Høgdall
1,*
1
Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
2
Department of Gynecology, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
3
Department of Gynecology, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(11), 1324; https://doi.org/10.3390/ph15111324
Submission received: 1 September 2022 / Revised: 18 October 2022 / Accepted: 21 October 2022 / Published: 26 October 2022
(This article belongs to the Special Issue Personalized Medicine in Gynecological Cancer)

Abstract

:
Background: We aimed to identify somatic pathogenic and likely pathogenic mutations using next-generation sequencing (NGS). The mutational findings were held against clinically well-described data to identify potential targeted therapies in Danish patients diagnosed with high-grade serous ovarian cancer (HGSC). Methods: We characterized the mutational profile of 128 HGSC patients. Clinical data were obtained from the Danish Gynecological Database and tissue samples were collected through the Danish CancerBiobank. DNA was analyzed using NGS. Results: 47 (37%) patients were platinum-sensitive, 32 (25%) partially platinum-sensitive, 35 (27%) platinum-resistant, and three (2%) platinum-refractory, while 11 (9%) patients did not receive chemotherapy. Overall, 27 (21%) had known druggable targets. Twelve (26%) platinum-sensitive patients had druggable targets for PARP inhibitors: one for tyrosine kinase inhibitors and one for immunotherapy treatment. Eight (25%) partially platinum-sensitive patients had druggable targets: seven were eligible for PARP inhibitors and one was potentially eligible for alpesilib and hormone therapy. Seven (20%) platinum-resistant patients had druggable targets: six (86%) were potentially eligible for PARP inhibitors, one for immunotherapy, and one for erdafitinib. Conclusions: PARP inhibitors are the most frequent potential targeted therapy in HGSC. However, other targeted therapies remain relevant for investigation according to our mutational findings.

1. Introduction

Epithelial ovarian cancer (EOC) ranks fifth in cancer deaths among women, accounting for more deaths than any other cancer of the female reproductive system (cancer.org (accessed on 15th April 2022). Denmark has the second-highest incidence rate globally, and more than 500 women are diagnosed annually [1,2]. Over 70% of cases are diagnosed in an advanced stage (International Federation of Gynecology and Obstetrics (FIGO) stage III-IV) [3]. Standard initial treatment consists of cytoreductive surgery and chemotherapy (carboplatin and paclitaxel) with the addition of bevacizumab in cases with stage IV disease or residual tumors and polyadenosine diphosphate (ADP)-ribose polymerase (PARP) inhibitors as maintenance therapy in platinum-sensitive patients. Of all patients, 20–30% are resistant to chemotherapy, and 80% showing an initial response experience relapse and ultimately die of the disease. High-grade serous ovarian cancer (HGSC) is the most common subtype. accounting for approximately 70% of EOC cases and 80% of deaths [3,4].
Increasing knowledge of the molecular, genetic, and clinical aspects underlying EOC has resulted in the development of PARP inhibitors. The PARP enzymes play a critical role in repairing DNA strand breaks. PARP inhibitors prevent the repair of these single-stranded breaks. This results in double-stranded DNA breaks, as double-strand breaks cannot be repaired accurately in tumors with homologous recombination deficiency (HRD) [5]. Ultimately, the accumulation of unrepaired DNA breaks leads to cellular death. Approximately 50% of HGSC exhibit genetic or epigenetic alterations in the homologous recombination (HR) pathway 6. Most often, alterations in the homologous recombination repair (HRR) pathway are caused by BRCA mutations. However, several other HRR genes may cause HRD, such as ATM, ATR, CHEK1/2, PALB2, and RAD51 [6]. HRD status can be measured using different tests; as yet the Myriad MyChoice® CDx test is the only FDA-approved test to determine HRD status in patients with ovarian cancer 8. The Myriad MyChoice® CDx assay uses next-generation sequencing (NGS) to assess genomic instability, including detecting BRCA1/2 mutations. The test measures a genomic instability score based on an algorithmic measurement composed of loss of heterozygosity, telomeric allelic imbalance, and large-scale state transitions. HRD positivity is defined as BRCA1/2 mutations and total genomic instability score ≥42. Although the patient may not have a BRCA1/2 alteration, they may have genomic scares resulting in HRD positivity8. The evidence of PARP inhibitor treatment comes from randomized clinical trials which included platinum-sensitive patients with somatic pathogenic BRCA1/2 mutations and HRD positivity in first-line and relapsed settings [7,8]. Therefore, platinum-resistant patients do not receive PARP inhibitors in Denmark. This treatment presupposes platinum sensitivity today, which means that the treatment options are limited and a clinical challenge for these patients.
The present study aimed to identify somatic pathogenic and likely pathogenic mutations using NGS in a cohort of patients diagnosed with HGSC in order to provide an overview of possible targeted treatments based on molecular findings. The mutational findings were held against clinical data to identify potential targeted therapies in subgroups of Danish patients diagnosed with HGSC.

2. Results

2.1. Clinical Data

Clinical and pathological information for the 128 included patients is summarized in Table 1. The median age was 65 years, and 109 (85%) patients had advanced disease (FIGO III-IV). Twenty-five (19%) patients were alive at follow-up (July 2020), while 104 (81%) patients were deceased. The median overall survival (OS) was 43 months, and the median progression-free survival (PFS) was 15 months. BRCA1/2 mutated patients had a significantly higher OS (HR: 0.50, CI 95%: 0.28–0.89) (Figure S1). Forty-seven (37%) patients were platinum-sensitive, 32 (25%) patients were partially platinum-sensitive, 35 (27%) patients were platinum-resistant, and three (2%) were platinum-refractory. Eleven patients did not receive any chemotherapy due to: dying shortly after surgery (n = 4), refraining from chemotherapy (n = 2), poor general condition (n = 2), and unknown (n = 2) (Figure 1). The excluded patients did not differ clinically from the included ones.

2.2. Datamining

Initially, 560,964 variants were identified. Variants not located within an exonic region were filtered out. A filter containing validity parameters such as Phred-score, p-value, allele frequency, and coverage was used. The complete list of parameters and specific criteria are outlined in Vestergaard et al. [9]. After filtering, 1349 variants were identified for further analysis. From the variants identified, we encountered the Ion ReporterTM Software annotated variants from the National Center of Biotechnology Information (NCBI) ClinVar databases from 2019 (latest version 20190909). Therefore, all variants were manually inspected and cross-referenced with updated mutational verdicts from VarSome and ClinVar. We ended up with 226 likely pathogenic or pathogenic variants classified by ClinVar (Table S1).

2.3. Pathogenic and Likely Pathogenic Mutations

Of the patients, 118 (92%) of 128 had one or more pathogenic or likely pathogenic mutations (Table S2). Pathogenic and likely pathogenic mutations were distributed over 51 cancer-related genes, including TP53 mutations in 86% of the cases (Table 2); 87%, 91%, 97%, and 67% had pathogenic or likely pathogenic mutations among platinum-sensitive, partially platinum-sensitive, platinum-resistant, and platinum-refractory patients, respectively. All the patients (n = 11) who did not receive chemotherapy had pathogenic or likely pathogenic mutations (Table 2).

2.4. Druggable Targets

Of the patients, 27 (21%) had potentially druggable targets distributed in ten genes, including BRCA1/2 (16%) (Table 3 and Table 4). Thus, we found potential druggable targets in 26% of the platinum-sensitive patients (n = 12), 25% of the partially platinum-sensitive patients (n = 8), and 20% of the platinum-resistant patients (n = 7). No druggable targets were found in the platinum-refractory group (Figure 2).

2.5. Potential Targeted Therapies

The considered targeted therapies based exclusively on the molecular findings were PARP inhibitors (20%), tyrosine kinase inhibitors (1%), alpesilib in combination with hormone therapy (1%), erdafitinib (1%), and immunotherapy (2%) (Table 5 and Figure 2).
MSI analysis was performed in patients with POLE mutations or patients with five or more mutations. None of them was MSI high, and therefore they were not potential candidates for immunotherapy.

2.6. Mutations in the Different Platinum Response Groups

The mutational findings in the platinum response groups are listed in Table S3 and Figure S2. The platinum-sensitive, partially platinum-sensitive, and platinum-resistant patients had nine genes, including BRCA1/2 and TP53, in common (Figure S2 and Table S3).

3. Discussion

This study characterized the mutational profile of a unique population of 128 Danish HGSC patients with corresponding tumor tissue samples and complete clinical data. The mutational profiles were characterized using OncomineTM Comprehensive Assay (OCAv3). We found potential druggable targets in 26% of the platinum-sensitive patients, 25% of the partially platinum-sensitive patients, 20% of the platinum-resistant patients, and none of the refractory cases. All platinum-sensitive patients and 88% of the partially platinum-sensitive patients with druggable targets were eligible for PARP inhibitors. Of particular interest, 86% of the platinum-resistant patients with druggable targets had targets for PARP inhibitors.

3.1. Mutation Profile in Danish Ovarian Cancer Patients Compared to Other Studies

The most extensive study based on data from the TCGA database showed TP53 mutations in 88% and BRCA1/2 mutations in 23% of 316 HGSC tumors [10]. Patch et al. characterized the mutational profile of 80 HGSC patients [11]. They found TP53 mutations in 99% and BRCA1/2 mutations in 26%. The mutational differences regarding TP53 and BRCA1/2 in our study compared with TCGA and Patch et al. may have several explanations. First, our research had a therapeutic focus, opting to include only pathogenic or likely pathogenic mutations according to the American College of Medical Genetics and Genomics (ACMG) classification of variants [12]. Ten (8%) patients did not have pathogenic or likely pathogenic mutations. However, except for one, all these patients had Variants of Unknown Significance (VUS), which are dynamic variants that may change classification in the future. Neither TGCA nor Patch et al. has used the ACMG variant classification, as these studies had a broader focus on characterizing the overall mutational profile. Therefore, it is not surprising that they report a higher frequency of TP53 and BRCA1/2 mutations.
Differences in patient selection may be another explanation. In the study by Patch et al., 39% were platinum-sensitive and 61% were platinum-resistant or -refractory, while TCGA included 69% platinum-sensitive and 31% platinum-resistant patients [10,11]. In TCGA and Patch et al., platinum status was defined as resistant if the patient had progressed or recurred within 6 months after the end of the last treatment, which is consistent with our definition. Moreover, we defined relapse and PD from the best clinical evaluation based on CT/MRI/PET-CT scans, serum CA125, and patients’ symptoms. The follow-up in the two published studies is not fully described.
Finally, we used targeted sequencing of all cancer-related genes, while TCGA used whole-exome sequencing (WES) and Patch et al. used whole-genome sequencing (WGS). WES identifies variants within all coding regions, and WGS identifies variants in the whole genome. Both WES and WGS generate massive data without therapeutic relevance. In this study, we had a clinical focus on targeted therapies. Therefore, we used OCAv3, which sequences all known druggable targets today. Panel sequencing is the most cost-effective method. It can easily be implemented in a routine diagnostic setting with a fast throughput, which meets the demand for a cancer fast-track package. Furthermore, it has a higher depth, increases the statistical power of detecting variants, and lowers variant detection limits. Nonetheless, panels are dynamic and change regularly with increased knowledge about diseases to remain clinically relevant [13,14].
In 2008 Soegaard et al. established the prevalence of BRCA1/2 mutations in a population-based study of 445 EOC cases from Denmark [15]. They identified pathogenic mutations in 26 patients (5.8%), 22 mutations in BRCA1 (4.9%) and four in BRCA2 (0.9%)), compared with 16.4% in our study. In contrast to our study, only 62% of the included patients were diagnosed with serous adenocarcinomas. Furthermore, only tumors in the BRCA mutated patients were graded [15]. The study by Soegaard et al. used Sanger sequencing, while we used NGS. The sensitivity for these two methods is 20% versus 5% (Sanger/NGS), which may explain the lower frequency of reported BRCA1/2 mutations in the study by Soegaard et al. [13]. Furthermore, the identified BRCA1/2 mutations differed in the two studies, possibly because more pathogenic BRCA1/2 mutations have been identified since 2008.

3.2. Platinum Resistance and PARP Inhibitors

Based on the molecular findings, we found that six platinum-resistant patients (17%) may potentially be eligible for PARP inhibitors. Three patients had a somatic pathogenic or likely pathogenic BRCA1/2 mutation, and three patients had mutations in the ATM, CHEK1, or CDK12 genes. However, due to the European and American approvals, none of them would be offered treatment with PARP inhibitors as standard treatment [16,17]. In Denmark, PARP inhibitors are only approved as maintenance treatment for newly diagnosed advanced EOC with a BRCA1/2 mutation and partial or complete response to platinum-based chemotherapy. In the recurrence setting, only platinum-sensitive BRCA mutated or HRD patients are candidates for PARP inhibitor treatment [18].
To date, limited data is available on the activity of PARP inhibitors in platinum-resistant HGSC. No studies have examined the effect in platinum-resistant patients with mutations in genes other than BRCA1/2 (Table S4) [19,20,21,22,23,24]. The BAROCCO trial investigated the combination of olaparib and cediranib in 123 HGSC platinum-resistant patients [23]. They found a trend toward improved PFS (5.7 months vs. 3.1 months). In the ARIEL4 trial (NCT02855944), rucaparib was tested in 345 BRCA mutated patients, of whom 176 were platinum resistant. PFS was significantly higher in the rucaparib group [24]. However, the PFS has not yet been calculated specifically for platinum-resistant patients. Results are pending from the NRG-GY005 (NCT02502266) and OCTAVIA (NCT03117933) trials, which are ongoing studies comparing PARP inhibitors (alone or in combination with cedirabin) with platinum-free chemotherapy in platinum-resistant patients (Table S4). Few studies have examined whether platinum-resistant patients benefit from PARP inhibitors. Nevertheless, 17% of our platinum-resistant patients might be potentially eligible for PARP inhibitors based on their mutational profiles. PARP inhibitors represent a promising trend in response rates and PFS in platinum-resistant patients. However, results from more extensive high-quality randomized studies are needed. Unfortunately, the present cohort was not treated with PARP inhibitors; thus, response and survival estimates cannot be assessed.
Our study has several strengths. All patients were operated on and treated at the same tertiary center by gynecologic oncologists with high expertise. All clinical data were continuously updated in the Danish Gynecologic Database, and none of the patients were lost to follow-up. Furthermore, our cohort size is comparable with TCGA and Patch et al. [10,11]. However, there are limitations as well. First, PARP inhibitors may be effective in more patients than those identified here, as HRD assessment might find additional patients who would benefit from PARP inhibitors. Moreover, although the cohort is relatively large, the subgroups that can be offered potential targeted therapies based solely on molecular findings are small, resulting in statistical limitations (Figure 2). Lastly, the mutational findings in the platinum response subgroups confirm that HGSC is a highly heterogenous molecular disease (Figure S2). The number of patients with specific mutations in the subgroups was too small to perform statistical analyses (Table S3). Therefore, it was not possible to define molecular profiles in the platinum response subgroups based on our findings.
PARP inhibitors are the most frequent potential targeted therapy for HGSC patients, according to our molecular findings. This confirms the results of previous studies and the clinical practice today [10,25]. However, it is highly relevant to investigate other targeted therapies, especially in platinum-resistant and platinum-sensitive patients who no longer respond to PARP inhibitors. Therefore, extensive studies which can clarify the efficacy of PARP inhibitors in platinum-resistant patients are needed to identify molecular druggable targets. Furthermore, large basket trials such as NCI-MATCH and ProTarget with the inclusion of HGSC patients with druggable targets other than those known for PARP inhibitors are warranted. Studies investigating the efficacy of combination therapies must be prioritized to overcome resistance. Lastly, most HGSC patients have a TP53 mutation. Efficient treatments targeting TP53 can potentially improve the treatment of ovarian cancer tremendously. APR-246 is a drug that reactivates mutant p53 encoded by TP53 in cancer cells by promoting its correct wild-type folding [26,27]. However, the effect of APR-246 has not yet been examined in a large clinical trial. A Phase 1/2 Open-label, Multicenter Study is ongoing to Assess the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Efficacy of PC14586 in Patients with Advanced Solid Tumors Harboring a p53 Y220C Mutation (PYNNACLE). Although only a few patients have this specific mutation, it points toward new perspectives and directions in the future treatment of HGSC.

4. Materials and Methods

4.1. Patients and Tissue Samples

One hundred and twenty-eight out of 157 patients with a primary diagnosis of HGSC were selected from a retrospective cohort (Figure 1) [28,29]. Sequencing was conducted with DNA from the patients. Patients were registered in the Pelvic Mass study/GOVEC study, a cohort initiated in September 2004. The clinical information of the patients was registered in the Danish Gynecological Cancer Database (DGCD) [30]. Platinum resistance was defined as relapse or progressive disease (PD) within six months after chemotherapy. Patients who developed PD during treatment or within four weeks after the last cycle were considered platinum-refractory. Patients were considered platinum-sensitive if they had experienced no relapse or PD or if relapse occurred more than six months after the end of first-line chemotherapy. Relapse and PD were defined from the best clinical evaluation based on CT/MRI/PET-CT scans, serum CA125, and patients’ symptoms. Patients were followed from October 2004 until July 2020 with a minimum follow-up of 10 years. No patients were lost to follow-up. All tissue samples were registered and stored in the Danish Cancer Biobank (DCB, Bio- and GenomeBank, Denmark) and handled according to national guidelines (www.regioner.dk/rbgb (accessed on 1 September 2020). The cohort was examined and classified as HGSC by a pathologist specialized in gynecologic oncology.

4.2. DNA Extraction

A representative area of cancerous tissue in FFPE blocks was identified and extracted to ensure the high content of tumor cells by an experienced pathologist within oncogynecology. Genomic DNA was extracted using Maxwell® RSC DNA FFPE Kit (Promega). DNA concentration was quantified using the QubitTM ds DNA High-Sensitive Assay kit (Thermo Fisher Scientific, Waltham, MA, US) on the Qubit fluorometer (Thermo Fisher Scientific, Waltham, MA, US).

4.3. Library Preparation and Sequencing

Library preparation was performed manually for the OCAv3. The assay consisted of 161 cancer-related genes with the sequencing of coding areas of all known druggable targets (Thermo Fisher Scientific) [12]. PCR amplification was conducted using a DNA concentration of 20 ng as input. Sequencing was performed using the Ion S5TM XL Sequencer (Thermo Fisher Scientific). The Ion ReporterTM Software (v. 5.14) was used for initial automated analysis, and Oncomine Comprehensive v3–w4.0–DNA–Single Sample was used for analysis workflow.

4.4. Microsatellite Instability (MSI)

According to the manufacturer’s instructions, microsatellite instability (MSI) testing was performed using the PlentiPlexTM MSI PentaBase panel (v.1.9.2); 20 ng/μL extracted DNA per patient was used for analyses. MSI was evaluated individually by comparing the length of amplicons obtained in the patients’ tumor-derived DNA with the reference DNA by molecular biologists.

4.5. Data Analysis

All data analyses were conducted using Python programming language (v.3.7). Data were initially pre-filtered considering variants within exonic regions or splice site regions and classified as either single-nucleotide variants, multi-nucleotide variants, or indels. Variants passing the pre-analysis data cleaning were subsequently analyzed and annotated based on the variant filtering properties outlined in Vestergaard et al. [9]. Variants were clinically annotated using the database provided at VarSome “https://varsome.com/ (access on 13–14 October 2021). using the implemented ClinVar database. Only variants classified as pathogenic and likely pathogenic by expert panels on ClinVar were included. Files for analysis were provided from the Ion ReporterTM Software (v. 5.14). Files were downloaded without any filter chain, providing all identified variants. Potential druggable targets were known oncogenes. The observed genetic alterations were class 4 or 5 (according to ACMG variant classification [12]) with the well-known effect of an FDA-approved targeted therapy. The Ion TorrentTM OncomineTM Reporter (Thermo Fisher Scientific v.5.3.0) was used for the search. The reports from Ion TorrentTM Reporter with the clinical evidence grading are included in Section S1 in supplementary materials. All pathogenic and likely pathogenic variants were reviewed on 25th November 2021 with the Demo US and EU and Global Clinical Trials filters.
As immunotherapy recommendations are not reported in the Ion TorrentTM OncomineTM Reporter, these recommendations were found in the ESMO guidelines (25NOV2021) [25].

4.6. Statistical Analysis

All statistical analyses were performed with Python programming language (v. 3.7). Fisher’s Exact Test was used to calculate and determine potential strand bias.

5. Conclusions

PARP inhibitors are the most frequent potential targeted therapy in HGSC. However, according to our mutational findings, most patients were not candidates for PARP inhibitors. Interestingly, we found several pathogenic mutations in most of the examined tumors. Although these mutations cannot be targeted today, they may become so in the future with the development of new treatment strategies. This is important to all those not eligible for PARP inhibitors as well as in the recurrent setting, which is often characterized by multi-chemotherapy resistance. Therefore, extensive studies investigating the efficacy of PARP inhibitors in platinum-resistant patients along with basket trials with the inclusion of HGSC patients with druggable targets other than those known for PARP inhibitors are needed.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15111324/s1, Figure S1: Kaplan-Meier curve for overall survival in patient groups with BRCA1/2 mutation compared with the BRCAwt patients; Figure S2: Mutations at the gene level in the groups with druggable targets; Table S1: Proportion of classified variant for 128 HGSC patients; Table S2: Pathogenic or likely pathogenic mutations per patient; Table S3: All pathogenic and likely pathogenic mutations; Table S4: Clinical trials evaluating PARP inhibitors in platinum-resistant patients; Section S1: IonTorrentTM OncomineTM Reports used to identify druggable targets.

Author Contributions

Y.S. contributed to the conceptualization, data analysis, and writing (original draft, molecular and clinical data analysis, figure preparation, and editing), L.K.V. contributed to the conceptualization, experiments, data analysis, and writing (molecular data analysis, review and editing), D.N.P.d.O. contributed to conceptualization, experiment, data analysis, and writing (molecular data analysis, review and editing), T.S.P. contributed to conceptualization, data analysis, and writing (molecular data analysis, review and editing), T.H.S. contributed to conceptualization, data analysis, and writing (clinical data analysis, review and editing), C.H. contributed with clinical data obtained from DGCD (Danish Gynecological Database) and to the conceptualization, data analysis, and writing (design of study, clinical data analysis, review and editing), E.H. contributed with material from DCB (http://rbgb.dk/cancer/ (accessed on 1 September 2020) and to the conceptualization, data analysis, and writing (design of study, molecular data analysis, conceptualization, review and editing). All authors have read and agreed to the published version of the manuscript.

Funding

This research received a grant from The Research Council at Herlev Copenhagen University Hospital.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, including written informed consent from all subjects. The Danish National Committee has approved the study for research ethics, Capital Region (approval codes; KF01-277/03 and KF01-143/04, H15020061). All performed molecular analyzes are somatic and not germline.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent has been obtained from the patient(s) to publish this paper.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sørensen, S.M.; Bjørn, S.F.; Jochumsen, K.M.; Jensen, P.T.; Thranov, I.R.; Hare-Bruun, H.; Seibæk, L.; Høgdall, C. Danish Gynecological Cancer Database. Clin. Epidemiol. 2016, 8, 485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Torre, L.A.; Trabert, B.; DeSantis, C.E.; Miller, K.D.; Samimi, G.; Runowicz, C.D.; Gaudet, M.M.; Jemal, A.; Siegel, R.L. Ovarian cancer statistics, 2018. CA. Cancer J. Clin. 2018, 68, 284–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lheureux, S.; Gourley, C.; Vergote, I.; Oza, A.M. Epithelial ovarian cancer. Lancet 2019, 393, 1240–1253. [Google Scholar] [CrossRef] [Green Version]
  4. Kossaï, M.; Leary, A.; Scoazec, J.Y.; Genestie, C. Ovarian Cancer: A Heterogeneous Disease. Pathobiology 2018, 85, 41–49. [Google Scholar] [CrossRef]
  5. Helleday, T.; Petermann, E.; Lundin, C.; Hodgson, B.; Sharma, R.A. DNA repair pathways as targets for cancer therapy. Nat. Rev. Cancer 2008, 8, 193–204. [Google Scholar] [CrossRef] [PubMed]
  6. Mittica, G.; Ghisoni, E.; Giannone, G.; Genta, S.; Aglietta, M.; Sapino, A.; Valabrega, G. PARP Inhibitors in Ovarian Cancer. Recent Pat. Anticancer. Drug Discov. 2018, 13, 392–410. [Google Scholar] [CrossRef] [PubMed]
  7. Konstantinopoulos, P.A.; Ceccaldi, R.; Shapiro, G.I.; D’Andrea, A.D. Homologous recombination deficiency: Exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov. 2015, 5, 1137–1154. [Google Scholar] [CrossRef] [Green Version]
  8. Myriad Genetics. Available online: https://myriad.com/oncology/mychoice-cdx/ (accessed on 15 April 2022).
  9. Mirza, M.R.; Pignata, S.; Ledermann, J.A. Latest clinical evidence and further development of PARP inhibitors in ovarian cancer. Ann. Oncol. 2018, 29, 1366–1376. [Google Scholar] [CrossRef]
  10. Mirza, M.R.; Coleman, R.L.; González-Martín, A.; Moore, K.N.; Colombo, N.; Ray-Coquard, I.; Pignata, S. The forefront of ovarian cancer therapy: Update on PARP inhibitors. Ann. Oncol. 2020, 31, 1148–1159. [Google Scholar] [CrossRef]
  11. Vestergaard, L.K.; Oliveira, D.N.P.; Poulsen, T.S.; Høgdall, C.K.; Høgdall, E.V. Oncomine TM Comprehensive Assay v3 vs. Oncomine TM Com-prehensive Assay Plus. Cancers 2021, 13, 5230. [Google Scholar] [CrossRef]
  12. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef] [Green Version]
  13. Patch, A.M.; Christie, E.L.; Etemadmoghadam, D.; Garsed, D.W.; George, J.; Fereday, S.; Nones, K.; Cowin, P.; Alsop, K.; Bailey, P.J.; et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 2015, 521, 489–494. [Google Scholar] [CrossRef] [PubMed]
  14. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Crody, W.W.; Hegde, M.; Lyon, E.; Spector, E.; et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Muzzey, D.; Evans, E.A.; Lieber, C. Understanding the Basics of NGS: From Mechanism to Variant Calling. Curr. Genet. Med. Rep. 2015, 3, 158–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Altmann, A.; Weber, P.; Bader, D.; Preuß, M.; Binder, E.B.; Müller-Myhsok, B. A beginners guide to SNP calling from high-Throughput DNA-Sequencing data. Hum. Genet. 2012, 131, 1541–1554. [Google Scholar] [CrossRef]
  17. Soegaard, M.; Kjaer, S.K.; Cox, M.; Wozniak, E.; Høgdall, E.; Høgdall, C.; Blaakaer, J.; Jacobs, I.J.; Gayther, S.A.; Ramus, S.J. BRCA1 and BRCA2 mutation prevalence and clinical characteristics of a population-based series of ovarian cancer cases from Denmark. Clin. Cancer Res. 2008, 14, 3761–3767. [Google Scholar] [CrossRef] [Green Version]
  18. ESMO Guidelines Comittee. Relapsed Epithelial Ovarian Carcinoma Treatment Recommendations. 2020. Available online: https://www.esmo.org/guidelines/guidelines-by-topic/gynaecological-cancers/newly-diagnosed-and-relapsed-epithelial-ovarian-carcinoma/eupdate-ovarian-cancer-treatment-recommendations (accessed on 15 April 2022).
  19. Tew, W.P.; Lacchetti, C.; Ellis, A.; Maxian, K.; Banerjee, S.; Bookman, M.; Jones, M.B.; Lee, J.M.; Lheureux, S.; Liu, J.F.; et al. PARP Inhibitors in the Management of Ovarian Cancer: ASCO Guideline. ASCO Guidel. 2020, 38, 3468. [Google Scholar] [CrossRef]
  20. Medicinrådet. Medicinrådets Behandlingsvejledning Vedrørende Lægemidler til BRCA-Muteret Kræft I Æggestokkene, Ægge-lederne Eller Primær Kræft I Bughinden; Medicinrådet: Copenhagen, Denmark, 2020; pp. 1–10. [Google Scholar]
  21. Oronsky, B.; Ray, C.M.; Spira, A.I.; Trepel, J.B.; Carter, C.A.; Cottrill, H.M. A brief review of the management of platinum-resistant–platinum-refractory ovarian cancer. Med. Oncol. 2017, 34, 1–7. [Google Scholar] [CrossRef]
  22. Fong, P.C.; Yap, T.A.; Boss, D.S.; Carden, C.P.; Mergui-Roelvink, M.; Gourley, C.; De Greve, J.; Lubinski, J.; Shanley, S.; Messiou, C.; et al. Poly(ADP)-ribose polymerase inhibition: Frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J. Clin. Oncol. 2010, 28, 2512–2519. [Google Scholar] [CrossRef]
  23. Kaufman, B.; Shapira-Frommer, R.; Schmutzler, R.K.; Audeh, M.W.; Friedlander, M.; Balmaña, J.; Mitchell, G.; Fried, G.; Stemmer, S.M.; Hubert, A.; et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 244–250. [Google Scholar] [CrossRef]
  24. Moore, K.N.; Secord, A.A.; Geller, M.A.; Miller, D.S.; Cloven, N.G.; Fleming, G.F.; Hendrickson, A.E.W.; Azodi, M.; DiSilverstro, P.; Oza, A.M.; et al. QUADRA: A phase 2, open-label, single-arm study to evaluate niraparib in patients (pts) with relapsed ovarian cancer (ROC) in 4th or later line of therapy: Results from the tBRCAmut subset. Ann. Oncol. 2018, 29, viii332–viii358. [Google Scholar] [CrossRef]
  25. Colombo, N.; Nicoletto, M.O.; Panici, P.B.; Tognon, G.; Bologna, A.; Lissoni, A.A.; DeCensi, A.; Tomao, F.; Fossati, R.; Tettamanzi, F.; et al. BAROCCO: A randomized phase II study of weekly paclitaxel vs cediranib-olaparib combination given with continuous or intermittent schedule in patients with recurrent platinum resistant ovarian cancer (PROC). Ann. Oncol. 2019, 30, v896. [Google Scholar] [CrossRef]
  26. Kristeleit, R.; Lisyanskaya, A.; Fedenko, A.; Dvorkin, M.; de Melo, A.C.; Shparyk, Y.; Rakhmatullina, I.; Bondarenko, I.; Colombo, N.; Svintsitskiy, V.; et al. Rucaparib versus chemotherapy in patients with advanced, relapsed ovarian cancer and a deleterious BRCA mutation: Efficacy and safety from ARIEL4, a randomized phase III study. Presented Soc. Gynecol. Oncol. 2021, 162, S3–S4. [Google Scholar] [CrossRef]
  27. ESMO Guidelines. Available online: https://www.esmo.org/guidelines (accessed on 15 April 2022).
  28. Lambert, J.M.R.; Gorzov, P.; Veprintsev, D.B.; Söderqvist, M.; Segerbäck, D.; Bergman, J.; Fersht, A.R.; Hainaut, P.; Wiman, K.G.; Bykow, V.J.N.; et al. PRIMA-1 Reactivates Mutant p53 by Covalent Binding to the Core Domain. Cancer Cell 2009, 15, 376–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Mohell, N.; Alfredsson, J.; Fransson, Å.; Uustalu, M.; Byström, S.; Gullbo, J.; Hallberg, A.; Bykow, V.J.N.; Björklund, U.; Wiman, K.G.; et al. APR-246 overcomes resistance to cisplatin and doxorubicin in ovarian cancer cells. Cell Death Dis. 2015, 6, e1794. [Google Scholar] [CrossRef] [Green Version]
  30. Hentze, J.L.; Høgdall, C.; Kjær, S.K.; Blaakær, J.; Høgdall, E. Searching for new biomarkers in ovarian cancer patients: Rationale and design of a retrospective study under the Mermaid III project. Contemp. Clin. Trials Commun. 2017, 8, 167–174. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of patient inclusion.
Figure 1. Flow diagram of patient inclusion.
Pharmaceuticals 15 01324 g001
Figure 2. Potential targeted therapies in HGSC.
Figure 2. Potential targeted therapies in HGSC.
Pharmaceuticals 15 01324 g002
Table 1. Baseline characteristics of 128 patients diagnosed with HGSC.
Table 1. Baseline characteristics of 128 patients diagnosed with HGSC.
Median age in year 65 (41–89)
Median CA125 792 (9–17.160)
Median RMI4632 (63–153.432)
Median BMI 24 (15–48)
Median follow up time in months 92 (61–123)
Performance score
054 (42%)
152 (41%)
218 (14%)
34 (3%)
FIGO stage
I9 (7%)
II10 (8%)
III89 (70%)
IV20 (15%)
Residual tumor after surgery
050 (39%)
<1 cm24 (19%)
>1 cm ≤ 2 cm17 (13%)
>2 cm37 (29%)
Chemotherapy (n = 117)
Carboplatin and taxol113 (97%)
Carboplatin4 (3%)
Chemotherapy response
>12 months (sensitive)47 (40%)
6 - ≤ 12 months (partially platinum-sensitive)32 (27%)
≤6 months (resistant)35 (30%)
Refractory3 (3%)
OS in months
OS (median)43 (0.7–190)
OS FIGO stage I (median)138 (45–184)
OS FIGO stage II (median)54 (3–140)
OS FIGO stage III (median)38 (0.7–190)
OS FIGO stage IV (median)33 (0.8–180)
PFS in months
PFS (median)15 (0–118)
PFS FIGO stage I (median)74 (23–118)
PFS FIGO stage II (median)19 (0–74)
PFS FIGO stage III (median)15 (0–107)
PFS FIGO stage IV (median)10 (0–95)
Table 2. Genes with pathogenic or likely pathogenic mutations in 128 HGSC patients.
Table 2. Genes with pathogenic or likely pathogenic mutations in 128 HGSC patients.
All patients (n = 128)ARID1A
ATM *
BAP1
BRCA1 **
BRCA2 **
CDK12 *
CHEK1 *
CREBBP
CTNNB1
DDR2
EGFR *
FANCA
FANCI
FBXW7
FGFR3 *
FGFR4
FLT3
FOXL2
H3F3A
KRAS
MAP2K1
MAX
MLH1 *
MRE11
MSH2 *
MTOR
MYC
NF1
NF2
NOTCH1
NOTCH2
NOTCH3
NTRK2
NTRK3
PDGFRB
PIK3CA *
PIK3CB
POLE
PTCH1
PTPN11
RAD51
RB1
SETD2
SLX4
SMARCA4
SMARCB1
SMO
TERT
TP53 (86%)
TSC1
TSC2
Platinum-sensitive
(n = 47)
ARID1A
ATM *
BRCA1 **
BRCA2 **
CDK12 *
CREBBP
EGFR *
FANCA
FGFR4
FLT3
H3F3A
KRAS
MAP2K1
MSH2 *
NF1
NOTCH1
NOTCH3
PDGFRB
PIK3CB
PTCH1
RAD51
RB1
SMARCA4
SMARCB1
TP53 (85%)
TSC1
TSC2
Partially platinum-sensitive (n = 32)ARID1A
BRCA1 **
BRCA2 **
CREBBP
FANCA
KRAS
NF1
NOTCH1
NOTCH3
PIK3CA *
POLE
SMO
TP53 (91%)
Platinum-resistant (n = 35)ATM *
BAP1
BRCA1 **
BRCA2 **
CDK12 *
CHEK1 *
CREBBP
CTNNB1
DDR2
FANCA
FANCI
FBXW7
FGFR3 *
FOXL2
KRAS
MAX
MLH1 *
MRE11
MTOR
MYC
NF1
NF2
NOTCH1
NOTCH2
NTRK2
NTRK3
PDGFRB
POLE
PTPN11
RAD51
RB1
SETD2
SMARCA4
SMARCB1
TERT
TP53 (83%)
TSC1
Platinum-refractory (n = 3)ARID1ACREBBPTP53 (33%)
No chemotherapy
(n = 11)
NF1NOTCH1SLX4TP53 (100%)
* Druggable target in other cancers. ** Druggable target in ovarian cancer.
Table 3. Genes with druggable targets (Nov 2021).
Table 3. Genes with druggable targets (Nov 2021).
GeneCancerTherapy
ATMProstata cancerOlaparib
BRCA1Prostate cancerBevacizumab + olaparib, niraparib, olaparib, rucaparib
BRCA2Prostate cancerBevacizumab + olaparib, niraparib, olaparib, rucaparib
CDK12Prostate cancerOlaparib
CHEK1Prostate cancerOlaparib
EGFRNon-small cell lung cancerAfatinib, bevacizumab + erlotinib, bevacizumab + gefitinib, dacominitib, erlotinib + ramucirumab, gefitinib, gefitinib + chemotherapy.
FGFR3Bladder cancer, bladder urothelial carcinomaErdafitinib
MLH1
MSH2
Anaplastic thyroid cancer, bladder cancer, brain metastases from solid tumors, colorectal cancer, cutaneous melanoma, hepatocellular carcinoma, hereditary gastrointestinal cancers, hypopharyngeal cancer, larynx cancer, lung and malignant pleural mesothelioma, non-small cell lung cancer, oral cavity cancer, renal cell carcinoma, small-cell lung cancerImmunotherapy
PIK3CABreast cancerAlpelisib and hormone therapy
Table 4. Druggable targets in HGSC patients.
Table 4. Druggable targets in HGSC patients.
Platinum-sensitive (N = 47)Partially platinum-sensitive (N = 32)Platinum-resistant (N = 35)
BRCA1/2n = 10
BRCA1 p.Leu1216PhefsTer2
BRCA1 p.Asp1305AlafsTer2
BRCA1 p.Arg1699Gln
BRCA1 p.Gln494Ter
BRCA1 p.Glu1134Ter
BRCA1 p.Gln1756PorfsTer74
BRCA1 p.? splice site mutation
BRCA1 p.Arg1699Gln
BRCA1 p.Gln1756ProfsTer74
BRCA2 p.Trp2626Cys
n = 7
BRCA1 p.? splice site mutation
BRCA1 p.Ala1708Glu
BRCA1 p.Gly1738Glu
BRCA1 p.Asn609IlefsTer3
BRCA2 p.Ser1741ThrfsTer35
BRCA2 p.Ser2219Ter
BRCA2 p.Lys2909GlnfsTer16
n = 2
BRCA1 p.Glu1046Ter
BRCA1 p.Glu23ValfsTer17
BRCA1/2 and other druggable targetsn = 1
BRCA1 p.? splice site mutation
ATM p.Arg2598Ter
EGFR p.Ala289Thr
MSH2 p.Gln324Ter
MSH2 Trp345Ter
MSH2 p.Gln413Ter
n = 1
BRCA2 p.Trp3106Ter
FGFR3 p.Trp685Ter
Other druggable targetsn = 1
CDK12 p.Ser301CysfsTer5
n = 1
PIK3CA p.Glu542Lys
n = 4
ATM p.Glu1751Ter
CDK12 p.Gln602Ter
CHEK1 p.Gln318Ter
MLH1 p.Arg659Ter
Table 5. Potential targeted therapies in HGSC patients.
Table 5. Potential targeted therapies in HGSC patients.
Platinum-sensitive (N = 47)Partially platinum-sensitive (N = 32)Platinum-resistant (N = 35)
PARP inhibitorn = 12
BRCA1 chr17:41243899 p.Leu1216PhefsTer2
BRCA1 chr17:41243633 p.Asp1305AlafsTer2
BRCA1 chr17:41215947 p.Arg1699Gln
BRCA1 chr17:41246068 p.Gln494Ter
BRCA1 chr17:41244148 p.Glu1134Ter
BRCA1 chr17:41209079 p.Gln175ProfsTer74
BRCA1 chr17:41215969 p.?
BRCA1 chr17:41215947 p.Arg1699Gln
BRCA1 chr17:41209079 p.Gln1756ProfsTer74
BRCA1 chr17:41209153 p.?
BRCA2 chr13:32936732 p.Trp2626Cys
ATM chr11:108203492 p.Arg2598Ter
CDK12 chr17:37619224 p.Ser301CysfsTer5
n = 7
BRCA1 chr17:41222944 p.?
BRCA1 chr17:41215920 p.Ala1708Glu
BRCA1 chr17:41209133 p.Gly1738Glu
BRCA1 chr17:41245721 p.Asn609IlefsTer3
BRCA2 chr13:32950896 p.Lys2909GlnfsTer16
BRCA2 chr13:32915148 p.Ser2219Ter
BRCA2 chr13:32913710 p.Ser1741ThrsfTer35
n = 6
BRCA1 chr17:41244412 p.Glu1046Ter
BRCA1 chr17:41276044 p.Glu23ValfsTer17
BRCA2 chr13:32968887 p.Trp3106Ter
ATM chr11:108172448 p.Glu1751Ter
CDK12 chr17:37627889 p.Gln602Ter
CHEK1 chr11:125514014 p.Gln318Ter
Tyrosine kinase inhibitorn = 1
EGFR chr7:55221821 p.Ala289Thr
Immunotherapyn = 1
MSH2 chr2:47643462 p.Gln324Ter
MSH2 chr2:47643526 p.Trp345Ter
MSH2 chr2:47657041 p.Gln413Ter
n = 1
MLH1 chr3:37090086 p.Arg659Ter
Alpesilib and hormone therapy n = 1
PIK3CA chr3:178936082 p.Glu542Lys
Erdafitinib n = 1
FGFR3 chr4:1808296 p.Trp685Ter
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sisman, Y.; Vestergaard, L.K.; de Oliveira, D.N.P.; Poulsen, T.S.; Schnack, T.H.; Høgdall, C.; Høgdall, E. Potential Targeted Therapies in Ovarian Cancer. Pharmaceuticals 2022, 15, 1324. https://doi.org/10.3390/ph15111324

AMA Style

Sisman Y, Vestergaard LK, de Oliveira DNP, Poulsen TS, Schnack TH, Høgdall C, Høgdall E. Potential Targeted Therapies in Ovarian Cancer. Pharmaceuticals. 2022; 15(11):1324. https://doi.org/10.3390/ph15111324

Chicago/Turabian Style

Sisman, Yagmur, Lau Kræsing Vestergaard, Douglas Nogueira Perez de Oliveira, Tim Svenstrup Poulsen, Tine Henrichsen Schnack, Claus Høgdall, and Estrid Høgdall. 2022. "Potential Targeted Therapies in Ovarian Cancer" Pharmaceuticals 15, no. 11: 1324. https://doi.org/10.3390/ph15111324

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