Open Access

Precision medicine in gynecological cancer (Review)

  • Authors:
    • Aikaterini Aravantinou‑Fatorou
    • Vasiliki Epameinondas Georgakopoulou
    • Meletios Athanasios Dimopoulos
    • Michalis Liontos
  • View Affiliations

  • Published online on: January 8, 2025     https://doi.org/10.3892/br.2025.1921
  • Article Number: 43
  • Copyright: © Aravantinou‑Fatorou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The advent of personalized and precision medicine has revolutionized oncology and treatment of gynecological cancer. These innovative approaches tailor treatments to individual patient profiles beyond genetic markers considering environmental and lifestyle factors, thereby optimizing therapeutic efficacy and minimizing adverse effects. Precision medicine uses advanced genomic technologies such as next‑generation sequencing to perform comprehensive tumor profiling. This allows identification of distinct genetic mutations, expression patterns and signaling pathway alterations, revealing the complex molecular landscape of gynecological cancer such as ovarian, cervical and uterine cancer. A major challenge in treating these cancers is their inherent molecular heterogeneity, which can influence tumor behavior, therapy response and prognosis. Precision medicine aims to overcome this by identifying biomarkers and molecular drivers for targeted therapy selection. For example, the identification of breast cancer (BRCA) gene mutations in ovarian cancer has guided the use of poly (ADP‑ribose) polymerase inhibitors, leading to more effective treatments with fewer side effects. Similar targeted therapies and immunotherapies have also been developed for cervical and uterine cancer, marking progress toward personalized care. Future directions in gynecological oncology emphasize the importance of molecular profiling and development of targeted therapies. By understanding the unique molecular features of each patient, clinicians can select the most effective personalized treatment strategies to improve patient outcomes and quality of life.

1. Introduction

The landscape of cancer treatment and research has undergone notable transformation, with the emergence of personalized and precision medicine as groundbreaking approaches in the management of various types of cancer, including those affecting gynecological organs. These approaches mark a departure from the traditional one-size-fits-all strategy, paving the way for more targeted, efficient and effective interventions (1). In gynecological cancer, encompassing ovarian, cervical, uterine, vaginal and vulvar cancer, precision medicine may improve outcomes, decrease side effects and enhance patient quality of life (2).

Personalized medicine, often used interchangeably with precision medicine, involves customization of medical therapy according to the unique attributes of each patient. It extends beyond focusing on genetic profiles, encompassing a wide array of patient-specific factors, including environmental influences, lifestyle and unique disease pathways. The holistic approach of personalized medicine, which views the patient as an integral part of the treatment equation, optimizes therapeutic decisions and practices for individual patients (3).

Precision medicine emphasizes the incorporation of genetic, environmental and lifestyle information into the diagnosis, treatment and prevention of disease. This approach relies on advanced technologies and the analysis of large datasets to identify which therapies will be beneficial for patients depending on their genetic and molecular profiling (4). The goal is to select optimal therapies and interventions for patients, minimizing unnecessary treatments and focusing on those most likely to benefit the patient (5).

The present review aims to provide a comprehensive overview of the advances in precision medicine within the context of gynecological cancers, focusing on the challenges posed by tumor heterogeneity, the role of genetic alterations, and the development of targeted therapies. By discussing the current state of precision oncology and its future directions, the present study seeks to highlight the potential for improving patient outcomes through personalized treatment approaches.

2. Tumor heterogeneity in gynecological malignancy

Tumor heterogeneity, a hallmark of cancer, presents challenges in the diagnosis, treatment and management of gynecological malignancies. Heterogeneity manifests at multiple levels, including histological subtype, molecular profile and clinical behavior within gynecological cancers such as ovarian, cervical and uterine cancers (6).

Histological and molecular heterogeneity

Gynecological cancers, particularly ovarian cancer, exhibit substantial histological diversity. For example, ovarian cancer includes subtypes such as high-grade serous (HGS), endometrioid, clear cell and mucinous carcinoma, each with distinct genetic alterations, clinical presentations and responses to therapy (7). Molecular heterogeneity further complicates this landscape as tumors of the same histological subtype may show diverse genetic mutations, gene expression patterns and signaling pathway alterations (8). Advances in genomic technologies, including next-generation sequencing (NGS), enable comprehensive tumor profiling, revealing the complex molecular underpinnings and identifying potential therapeutic targets and biomarkers (8).

Functional and epigenetic heterogeneity

In addition to genetic variations, epigenetic modifications such as DNA methylation and histone modification contribute to tumor heterogeneity by regulating gene expression and cell phenotypes, leading to treatment resistance and phenotypical diversity (9). Phenotypical heterogeneity is also evident in varying cell morphology, proliferation rate and drug sensitivity, complicating treatment strategies (10). Moreover, functional heterogeneity arises from differences in cell metabolism, signaling pathway activation and immune microenvironment interactions, all of which influence tumor aggressiveness and therapeutic resistance (11-15).

3. Challenges in current treatment approaches and future directions

Despite advancements in understanding gynecological cancer and developing targeted therapy, challenges persist, particularly regarding tumor heterogeneity, treatment resistance and disparities in access to innovative treatments (16).

Treatment resistance and recurrence

A major challenge is the high rate of recurrence and development of treatment resistance, especially in advanced-stage gynecological cancer. Even with initial therapeutic success, many patients experience disease progression and become resistant to standard treatments, leading to poor long-term outcomes (16). This resistance often results from the tumor molecular heterogeneity, which necessitates alternative therapeutic strategies and development of novel targeted agents to overcome these resistance mechanisms (17).

Lack of predictive biomarkers

Another significant hurdle is the scarcity of reliable predictive biomarkers for selecting optimal treatment strategies. Although biomarkers such as breast cancer (BRCA) gene mutations in ovarian cancer have been validated for targeted therapy, many patients lack actionable mutations, limiting their treatment options (17). The identification and validation of robust biomarkers that accurately predict treatment response and guide therapeutic decisions is key (18).

Disparities in access to treatment

Disparities in access to genomic testing, targeted therapy and clinical trials are particularly pronounced among populations of developing countries. Limited access to precision medicine exacerbates disparities in outcomes and survival rates between demographic groups (18). Efforts to improve access to innovative therapy and clinical trials for all patients, regardless of socioeconomic status or geographical location, are essential for reducing these disparities (18).

Data interpretation and integration

The integration of multi-omic data and the development of computational algorithms for data analysis present opportunities and challenges. While genomic technology has enabled comprehensive molecular characterization of tumors, interpreting complex genomic data and translating research findings into clinical practice is challenging. Standardization of molecular profiling assays, data sharing initiatives and collaboration across research institutions are necessary to accelerate progress and ensure effective implementation of precision medicine (19).

4. Drivers of genetic alterations in gynecological cancer

A complex interplay of genetic alterations drives gynecological cancer, contributing to tumorigenesis, disease progression and treatment resistance. Understanding the key driver mutations and signaling pathways involved in gynecological malignancy is key for developing targeted therapy and personalized treatment strategies.

In ovarian cancer, HGS carcinoma (HGSC), the most common and aggressive subtype, is characterized by frequent mutations in genes involved in DNA repair pathways, such as BRCA1 and BRCA2(20). These mutations impair homologous recombination-mediated DNA repair, leading to genomic instability and increased susceptibility to DNA-damaging agents such as platinum-based chemotherapy and poly (ADP-ribose) polymerase (PARP) inhibitors (21). Other recurrent genetic alterations in HGSC include mutations in TP53, which occur in nearly all cases, as well as alterations in genes involved in the PI3K/AKT/mTOR pathway, such as phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) and PTEN (22). Targeted therapy directed against these genetic aberrations hold promise for improving outcomes in patients with ovarian cancer.

In endometrial cancer, the most common type of uterine cancer, molecular profiling studies have identified distinct genomic subtypes with different etiologies, clinical behavior and treatment responses (22-24). For example, 20-30% of endometrial cancer cases harbor mutations in the PTEN tumor suppressor gene, leading to dysregulated PI3K/AKT/mTOR signaling and aberrant cell proliferation (23). Other recurrent genetic alterations in endometrial cancer include mutations in PIK3CA, Catenin Beta 1 and TP53 genes, as well as microsatellite instability (MSI) and DNA mismatch repair (MMR) deficiency (24). These molecular alterations serve as potential therapeutic targets and predictive biomarkers for targeted therapy and immunotherapy in endometrial cancer.

High-risk human papilloma virus (HPV) types, particularly HPV16 and HPV18, which encode viral oncoproteins that promote cell transformation and oncogenesis, primarily drive cervical cancer (25). Integration of HPV DNA into the host genome disrupts cellular tumor suppressor pathways, such as the retinoblastoma and p53 pathways, leading to uncontrolled cell proliferation and tumor formation (26). In addition to HPV infection, cervical cancer may harbor genetic alterations in genes such as PIK3CA, PTEN and TP53, which contribute to tumor progression and treatment resistance (27). Targeting HPV-associated pathways and associated genetic alterations holds potential for improving outcomes in patients with cervical cancer.

Vulvar and vaginal cancers are relatively rare gynecological malignancies that are often associated with HPV infection, similar to cervical cancer (28). However, vulvar squamous cell carcinoma may also arise in the absence of HPV infection, with mutations in genes such as TP53, PIK3CA and fibroblast growth factor receptor 3 (FGFR3) implicated in tumorigenesis (28). In vaginal cancer, HPV infection is a common risk factor, particularly in younger patients, with mutations in TP53 and PIK3CA identified in a subset of cases (29). Targeted therapy directed against HPV-related pathways and genetic alterations offers potential avenues for improving outcomes in patients with vulvar and vaginal cancer.

5. PARP inhibitors, BRCA and homologous recombination deficiency (HRD) in gynecological cancer

PARP inhibitors exploit synthetic lethality, in which the simultaneous inhibition of two DNA repair pathways leads to cell death, to selectively target cancer cells with defects in HR-mediated DNA repair (21).

BRCA1 and BRCA2 are tumor suppressor genes involved in the repair of double-stranded DNA breaks via the HR repair pathway. Mutations in BRCA1 or BRCA2 impair this DNA repair mechanism, leading to genomic instability and increased susceptibility to DNA-damaging agents, such as platinum-based chemotherapy and PARP inhibitors (30). PARP inhibitors, such as olaparib, niraparib and rucaparib, block activity of PARP enzymes, preventing the repair of single-stranded DNA breaks and leading to accumulation of double-stranded DNA breaks in BRCA-mutated cancer cells. This synthetic lethality results in cancer cell death, sparing normal cells with intact HR repair pathways (31).

In addition to BRCA mutations, HRD, a measure of genomic instability and impaired DNA repair, is a predictive biomarker for response to PARP inhibitors in gynecological cancer (32). HRD can result from genetic alterations, including mutations in other DNA repair genes (such as RAD51 Paralog C and D), genomic rearrangement and chromosomal abnormality. Tumors with HRD are more reliant on alternative DNA repair mechanisms, such as PARP-mediated base excision repair, making them susceptible to PARP inhibition (32).

Clinical trials have demonstrated the efficacy of PARP inhibitors in the treatment of gynecological cancer, including ovarian, fallopian tube and primary peritoneal cancer (33,34). The SOLO-1 trial investigated the use of olaparib as maintenance therapy in patients with newly diagnosed advanced ovarian cancer and BRCA mutation (33). The study showed a significant improvement in progression-free survival (PFS) with olaparib compared with placebo, leading to its approval as a first-line maintenance therapy in this population (33). Similarly, the NOVA trial demonstrated the efficacy of niraparib as maintenance therapy in patients with recurrent ovarian cancer, regardless of BRCA mutation status (34). These findings have transformed the treatment landscape for gynecological cancer, offering new therapeutic options and improving outcomes for patients with BRCA mutations or HRD.

However, challenges remain in the clinical implementation of PARP inhibitors, including development of resistance mechanisms, optimal patient selection criteria and the identification of predictive biomarkers beyond BRCA mutations and HRD. Resistance to PARP inhibitors can arise through various mechanisms, such as restoration of HR repair, upregulation of alternative DNA repair pathways or acquisition of secondary mutations in PARP or other DNA repair genes (35).

6. Role of PD-L1 in gynecological cancer

Tumor and immune cells express programmed death-ligand 1 (PD-L1), a cell surface protein that serves a critical role in regulating the immune response and promoting immune evasion in gynecological cancer. PD-L1 interacts with its receptor, programmed cell death protein 1 (PD-1), on T cells, leading to T cell exhaustion and inhibition of antitumor immune responses (36). Immunotherapies, such as immune checkpoint inhibitors, target the PD-1/PD-L1 axis, a major immune checkpoint pathway for the treatment of gynecological cancer (37).

In ovarian cancer, PD-L1 expression is associated with aggressive disease characteristics, immune infiltration and poor prognosis (38). PD-L1 blockade enhances antitumor immune response and inhibits tumor growth in ovarian cancer models, providing rationale for clinical evaluation of immune checkpoint inhibitors in this disease (39). Clinical trials investigating PD-1/PD-L1 inhibitors, either as monotherapy or in combination with other agents, have shown promising results in patients with recurrent or refractory ovarian cancer, leading to their approval for this indication (40,41).

Similarly, PD-L1 expression has been reported in endometrial cancer, particularly in tumors with aggressive histological subtypes, high tumor grade and advanced-stage disease (42). The presence of PD-L1-positive immune cells in the tumor microenvironment is associated with immune evasion and treatment resistance in endometrial cancer (43). Clinical trials evaluating efficacy of PD-1/PD-L1 inhibitors in endometrial cancer have shown encouraging responses in patients with high microsatellite instability (MSI-H) or MMR-deficient (dMMR) tumors (43). These findings have led to the approval of PD-1/PD-L1 inhibitors for treatment of advanced or metastatic endometrial cancer refractory to standard therapy (44).

Chronic inflammation and HPV infection in cervical cancer upregulate PD-L1 expression, which contributes to immune suppression and tumor immune escape. High PD-L1 expression in cervical cancer is associated with advanced-stage disease, lymph node metastasis and decreased survival outcomes (45). A clinical trial investigating immune checkpoint inhibitors targeting the PD-1/PD-L1 axis in cervical cancer has demonstrated durable responses and prolonged survival in patients with advanced or recurrent disease (46). These findings support use of PD-1/PD-L1 inhibitors as a treatment option for patients with advanced or metastatic cervical cancer.

Researchers have reported PD-L1 expression in a subset of vulvar and vaginal cancer, which is associated with immune evasion and treatment resistance (47). Clinical trials evaluating immune checkpoint inhibitors in vulvar and vaginal cancer are ongoing, with preliminary results showing promising antitumor activity and manageable safety profiles (48,49). To elucidate the role of the PD-1/PD-L1 axis in these rare gynecological malignancies and optimize treatment strategies, further research is necessary.

7. Clinical trials

Clinical trials serve a pivotal role in advancing the field of gynecological oncology by evaluating novel therapeutic strategies, validating predictive biomarkers and improving patient outcomes. Data from recent clinical trials has provided valuable insight into the efficacy and safety of emerging treatments and biomarker-driven therapy for gynecological cancers (50,51).

In ovarian cancer, several landmark clinical trials have evaluated the role of targeted therapy and immunotherapies in improving outcomes for patients with advanced or recurrent disease (33,52). SOLO-1 trial showcased the efficacy of olaparib as a maintenance treatment in newly diagnosed patients with advanced ovarian cancer and BRCA mutation, resulting in a notable enhancement in PFS compared with a placebo (33). Similarly, the PAOLA-1 trial investigated the combination of olaparib and bevacizumab as first-line maintenance therapy in patients with advanced ovarian cancer, showing a significant improvement in PFS in the overall population and patients with HRD (52). These findings have led to approval of olaparib and olaparib + bevacizumab as maintenance therapies in this population (33,52).

In endometrial cancer, KEYNOTE-146 trial evaluated the efficacy of pembrolizumab, a PD-1 inhibitor, in patients with advanced or recurrent disease refractory to standard therapy (53). The study demonstrated encouraging antitumor activity with pembrolizumab, particularly in patients with MSI-H or dMMR tumors, leading to its approval as a treatment option in this population (53). Similarly, the JAVELIN Solid Tumor Trial investigated the efficacy of avelumab, another PD-L1 inhibitor, in patients with advanced or recurrent endometrial cancer, showing promising responses and manageable safety profiles in these patients (54).

In cervical cancer, the KEYNOTE-158 trial evaluated the efficacy of pembrolizumab in patients with advanced or recurrent disease refractory to standard therapy (55). The study demonstrated durable responses and prolonged survival with pembrolizumab in a subset of patients with PD-L1-positive tumors, leading to its approval as a treatment option in this population (56).

Furthermore, metastatic endometrial tumors after progression in first-line platinum-based chemotherapy are treated with combination of immunotherapy and tyrosine kinase inhibitors pembrolizumab and lenvatinib based on results of phase III trial KEYNOTE-158(56). The use of this combination is universal and does not require expression of specific biomarkers. Additionally, the GOG-240 trial investigated addition of the immune checkpoint inhibitor ipilimumab to standard chemotherapy in patients with recurrent or metastatic cervical cancer, showing improved overall survival compared with chemotherapy-alone (57).

In vulvar and vaginal cancers, clinical trials evaluating targeted therapies and immunotherapy are relatively limited due to the rarity of these malignancies. However, ongoing studies are investigating the role of immune checkpoint inhibitors, such as pembrolizumab and nivolumab, in patients with advanced or recurrent disease, with preliminary results showing promising antitumor activity and manageable safety profiles (58,59).

8. Real-world studies

Zhao et al (60) investigated the efficacy and safety of niraparib as a first-line maintenance treatment for patients with advanced ovarian cancer in a real-world setting across multiple centers in China. The findings revealed that niraparib provided significant PFS benefits, with PFS rates of 87.4, 75.9 and 63.6% at 6, 12 and 18 months, respectively. Factors such as age <65 years, BRCA mutation and successful cytoreductive surgery (R0 status) are associated with prolonged PFS. Additionally, starting niraparib at a 200 mg dose was effective and had manageable safety, with the most common severe adverse events being hematological, including decreased platelet and white blood cell counts. The study concluded that niraparib is a valuable and safe option for maintenance therapy in patients with advanced ovarian cancer, particularly when using an individualized starting dose strategy.

A retrospective cohort study in the USA evaluated the tolerability of PARP inhibitors (olaparib, niraparib and rucaparib) in patients with advanced ovarian cancer (61). The study included 824 patients and analyzed clinical events of interest such as anemia, neutropenia and nausea. The findings indicated that while PARP inhibitors are effective as maintenance therapy, they have a significant side-effect profile, which varies depending on the PARP inhibitor used. The aforementioned study highlights the importance of monitoring and managing adverse effects in real-world clinical settings (61).

Another real-world study (62) focused on treatment patterns following disease progression in patients with epithelial ovarian cancer who received PARP inhibitor maintenance therapy. The aforementioned study examined subsequent therapies and outcomes, providing valuable data on how patients fare following disease progression while receiving PARP inhibitors. The results emphasized the importance of tailoring post-progression treatment based on individual patient profiles and previous responses to therapy (62).

Huepenbecker et al (63) aimed to describe the real-world usage of immune checkpoint inhibitors in patients with advanced or recurrent endometrial cancer. The aforementioned study analyzed data from a nationwide electronic health record-derived database, focusing on patients who received systemic treatment between January 2014 and November 2020, followed until May 2021. The study included 326 patients who received their first immune checkpoint inhibitors during this period, with usage increasing significantly over time. Pembrolizumab was the most common immune checkpoint inhibitor, often combined with lenvatinib. Immune checkpoint inhibitors were administered as first-, second- or later lines of therapy, with median time to next treatment being longer when immune checkpoint inhibitors were given earlier. The study found regional and stage-associated differences in immune checkpoint inhibitor usage, and ~1/3 of patients received subsequent treatments after immune checkpoint inhibitors, with no significant difference in demographic or clinical characteristics based on post-immune checkpoint inhibitor treatment type. The findings highlight the growing role of immune checkpoint inhibitors across various treatment lines in this population (63).

Passarelli et al (23) presented a case report of a 51-year-old patient with advanced endometrioid endometrial cancer harboring a PIK3CA mutation who showed an exceptional response to alpelisib, a PI3K α-selective inhibitor. Despite multiple prior treatments, including surgery, chemotherapy and hormone therapy, the disease progressed until alpelisib was introduced. Upon starting alpelisib, the patient experienced significant clinical and radiological improvements, with a notable decrease in tumor size and levels of tumor markers. This case highlights the potential of precision medicine in treating advanced gynecological cancers by targeting specific molecular alterations such as PIK3CA mutations. The aforementioned study emphasizes the importance of genomic profiling in guiding treatment decisions and suggests that alpelisib may be a promising option for patients with similar molecular profiles (23).

9. Role of multiple targeted mutations in gynecological cancer

Gynecological cancer often harbors multiple genetic mutations and molecular alterations that contribute to tumor heterogeneity, treatment resistance and disease progression (64). Understanding the interplay between these targeted mutations is key for optimizing treatment strategies and improving outcomes for patients with gynecological malignancy.

In ovarian cancer, HGSC, the most common subtype, is characterized by extensive genomic alterations, including mutations in tumor suppressor genes (such as TP53 and BRCA1/2) and oncogenes (such as PIK3CA and KRAS) (65). The presence of multiple targeted mutations in HGSC underscores the complexity of the disease and need for personalized treatment approaches. For example, patients with concurrent mutations in BRCA1/2 and HRD may benefit from PARP inhibitors, whereas those with mutations in PIK3CA or KRAS may respond to targeted therapy against the PI3K/AKT/mTOR or MAPK signaling pathways, respectively (66). Comprehensive molecular profiling of tumors is key for identifying actionable mutations and selecting tailored treatment regimens based on the genetic makeup of each patient.

Endometrial cancer is characterized by distinct molecular subtypes with different genetic alterations and clinical behavior. The Cancer Genome Atlas has identified four molecular subtypes of endometrial cancer, including DNA polymerase epsilon ultramutated, MSI and copy number-low and -high, each with unique genomic features and prognostic implications (22). Patients with MSI-high tumors, characterized by mutations in DNA MMR genes [such as DNA mismatch repair protein (MLH1) and MSH2], may benefit from immune checkpoint inhibitors due to increased tumor mutational burden and enhanced antitumor immune responses (44). Conversely, patients with copy number-high tumors, characterized by amplifications in oncogenes such as Cyclin E1 (CCNE1) and MYC, may respond to targeted therapy directed against these genetic alterations (20). Integrating molecular subtype classification with targeted mutation profiling enables identification of personalized treatment strategies tailored to the molecular characteristics of each endometrial cancer subtype.

Persistent infection with high-risk HPV types, particularly HPV16 and HPV18, drives cervical cancer by encoding viral oncoproteins that promote cell transformation and oncogenesis (67). In addition to HPV infection, cervical cancer may harbor mutations in genes such as PIK3CA, PTEN and TP53, which contribute to tumor progression and treatment resistance (22). Patients with concurrent HPV infection and targeted mutations may exhibit distinct clinical behavior and treatment responses, highlighting the importance of comprehensive molecular profiling in guiding treatment decisions for cervical cancer.

Vulvar and vaginal cancers are relatively rare gynecological malignancies that are less well-characterized than other types of gynecological cancer at the molecular level. However, mutations in genes such as TP53, PIK3CA and FGFR3 in a subset of tumors suggest potential therapeutic targets for these diseases (28). Additionally, HPV infection is associated with a subset of vulvar and vaginal cancers, particularly in younger patients, further highlighting the role of viral oncogenesis in these malignancies (68).

10. Challenges and limitations in clinical implementation of precision medicine in gynecological cancer

The clinical implementation of precision medicine in gynecological cancer, while promising, faces several challenges. These include issues associated with access to advanced genomic testing, cost and the necessity for interdisciplinary collaboration between healthcare providers.

Access to advanced genomic testing

NGS and other molecular profiling techniques are key components of precision medicine. These technologies enable the identification of specific genetic mutations and biomarkers to guide targeted therapy. However, the availability of advanced diagnostic tools is not uniform across healthcare facilities. In low-resource settings, the lack of infrastructure and expertise required to perform comprehensive genomic analysis limits the adoption of precision medicine. Moreover, even in well-resourced settings, there can be notable disparities in access based on geographical location, with urban centers typically having more advanced diagnostic facilities compared with rural areas (1).

Cost

The high cost of genomic testing and targeted therapies can be prohibitive for patients. For example, NGS, while providing comprehensive genomic information, is expensive and its costs are often not fully covered by insurance policies. This financial burden can limit patient access to precision medicine approaches. Additionally, targeted therapy, which is developed based on specific genetic mutations identified by genomic testing, tends to be more expensive than conventional treatment. The high cost of these therapies can strain healthcare budgets and make them inaccessible. Efforts to reduce the cost of genomic testing and targeted therapy and to increase insurance coverage for these services are essential to make precision medicine more accessible (3).

Interdisciplinary collaboration

The successful implementation of precision medicine requires interdisciplinary collaboration between various healthcare providers, including oncologists, geneticists, pathologists and bioinformaticians. This collaborative approach is necessary to interpret complex genomic data accurately and integrate this information into clinical decision-making effectively. However, fostering such interdisciplinary collaboration can be challenging. It requires effective communication and coordination between diverse healthcare professionals, which can be difficult to achieve in practice. Additionally, there is need for ongoing education and training for healthcare providers to keep up with rapid advancements in genomic technology and precision medicine approaches (4).

Data interpretation and standardization

Another challenge lies in the interpretation and standardization of genomic data. The vast amount of data generated by genomic testing requires sophisticated bioinformatics tools and expertise to analyze and interpret. Variability in data interpretation and lack of standardized protocols can lead to inconsistent clinical recommendations. Therefore, establishing standardized guidelines and protocols for genomic data interpretation is key to ensure reliable and actionable insights from genomic testing (19).

Ethical and privacy concerns

The collection and storage of genomic data raise issues of patient privacy and data security. Ensuring that genetic information is protected from unauthorized access and misuse is key. Additionally, ethical considerations regarding use of genetic information, such as potential discrimination based on genetic risk factors, need to be addressed through robust regulatory frameworks (5).

Fig. 1 provides a schematic illustration of precision medicine in gynecological cancer.

11. Conclusion

The transition towards personalized and precision medicine in gynecological oncology signifies a key shift from conventional treatment paradigms, offering new potential for improved patient outcomes. Through the utilization of advanced genomic technology, such as NGS, clinicians can now unravel the complex molecular landscape of cancers affecting the gynecological organs. This detailed molecular profiling facilitates identification of unique genetic mutations and signaling pathways, enabling selection of targeted therapies tailored to the individual characteristics of each tumor.

The inherent molecular heterogeneity of gynecological cancers, presenting a notable challenge in treatment, is addressed by the ability of precision medicine to identify and target specific biomarkers and molecular drivers. Successes in ovarian, cervical and uterine cancer treatments, notably through the use of PARP inhibitors in BRCA-mutated ovarian cancer, exemplify the potential of precision medicine to enhance treatment efficacy while minimizing adverse effects. Focus on molecular profiling and development of novel targeted therapies may further refine treatment strategies to optimize outcomes and quality of life for patients with gynecological cancer. This underscores the shift towards more personalized care but also highlights potential for groundbreaking treatments in future.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

AAF and ML conceptualized the study. AAF, ML, MAD, and VEG wrote the manuscript. ML and MAD analyzed the data and provided critical revisions. All have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Use of artificial intelligence tools

Chat GPT was used to improve the readability and language of the manuscript; authors revised and edited the content produced by the AI tool as necessary, taking full responsibility for the ultimate content of the present manuscript.

References

1 

Shams M, Abdallah S, Alsadoun L, Hamid YH, Gasim R and Hassan A: Oncological horizons: The synergy of medical and surgical innovations in cancer treatment. Cureus. 15(e49249)2023.PubMed/NCBI View Article : Google Scholar

2 

Wilson EM, Eskander RN and Binder PS: Recent therapeutic advances in gynecologic oncology: A review. Cancers (Basel). 16(770)2024.PubMed/NCBI View Article : Google Scholar

3 

Stefanicka-Wojtas D and Kurpas D: Personalised medicine-implementation to the healthcare system in europe (focus group discussions). J Pers Med. 13(380)2023.PubMed/NCBI View Article : Google Scholar

4 

Hassan M, Awan FM, Naz A, deAndrés-Galiana EJ, Alvarez O, Cernea A, Fernández-Brillet L, Fernández-Martínez JL and Kloczkowski A: Innovations in genomics and big data analytics for personalized medicine and health care: A review. Int J Mol Sci. 23(4645)2022.PubMed/NCBI View Article : Google Scholar

5 

Akhoon N: Precision medicine: A new paradigm in therapeutics. Int J Prev Med. 12(12)2021.PubMed/NCBI View Article : Google Scholar

6 

Zhang A, Miao K, Sun H and Deng CX: Tumor heterogeneity reshapes the tumor microenvironment to influence drug resistance. Int J Biol Sci. 18:3019–3033. 2022.PubMed/NCBI View Article : Google Scholar

7 

Murawski M, Jagodziński A, Bielawska-Pohl A and Klimczak A: Complexity of the genetic background of oncogenesis in ovarian cancer-genetic instability and clinical implications. Cells. 13(345)2024.PubMed/NCBI View Article : Google Scholar

8 

Tsimberidou AM, Fountzilas E, Bleris L and Kurzrock R: Transcriptomics and solid tumors: The next frontier in precision cancer medicine. Semin Cancer Biol. 84:50–59. 2022.PubMed/NCBI View Article : Google Scholar

9 

Sadida HQ, Abdulla A, Marzooqi SA, Hashem S, Macha MA, Akil ASA and Bhat AA: Epigenetic modifications: Key players in cancer heterogeneity and drug resistance. Transl Oncol. 39(101821)2024.PubMed/NCBI View Article : Google Scholar

10 

Thankamony AP, Ramkomuth S, Ramesh ST, Murali R, Chakraborty P, Karthikeyan N, Varghese BA, Jaikumar VS, Jolly MK, Swarbrick A and Nair R: Phenotypic heterogeneity drives differential disease outcome in a mouse model of triple negative breast cancer. Front Oncol. 13(1230647)2023.PubMed/NCBI View Article : Google Scholar

11 

Cordani M, Dando I, Ambrosini G and González-Menéndez P: Signaling, cancer cell plasticity, and intratumor heterogeneity. Cell Commun Signal. 22(255)2024.PubMed/NCBI View Article : Google Scholar

12 

Goenka A, Khan F, Verma B, Sinha P, Dmello CC, Jogalekar MP, Gangadaran P and Ahn BC: Tumor microenvironment signaling and therapeutics in cancer progression. Cancer Commun (Lond). 43:525–561. 2023.PubMed/NCBI View Article : Google Scholar

13 

Imodoye SO, Adedokun KA and Bello IO: From complexity to clarity: unravelling tumor heterogeneity through the lens of tumor microenvironment for innovative cancer therapy. Histochem Cell Biol. 161:299–323. 2024.PubMed/NCBI View Article : Google Scholar

14 

Proietto M, Crippa M, Damiani C, Pasquale V, Sacco E, Vanoni M and Gilardi M: Tumor heterogeneity: Preclinical models, emerging technologies, and future applications. Front Oncol. 13(1164535)2023.PubMed/NCBI View Article : Google Scholar

15 

Wang Q, Shao X, Zhang Y, Zhu M, Wang FXC, Mu J, Li J, Yao H and Chen K: Role of tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med. 12:11149–11165. 2023.PubMed/NCBI View Article : Google Scholar

16 

Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS and Petrik J: The complex tumor microenvironment in ovarian cancer: Therapeutic challenges and opportunities. Curr Oncol. 31:3826–3844. 2024.PubMed/NCBI View Article : Google Scholar

17 

Radu MR, Prădatu A, Duică F, Micu R, Creţoiu SM, Suciu N, Creţoiu D, Varlas VN and Rădoi VE: Ovarian cancer: Biomarkers and targeted therapy. Biomedicines. 9(693)2021.PubMed/NCBI View Article : Google Scholar

18 

Cooper KE, Abdallah KE, Angove RSM, Gallagher KD and Bonham VL: Navigating access to cancer care: Identifying barriers to precision cancer medicine. Ethn Dis. 32:39–48. 2022.PubMed/NCBI View Article : Google Scholar

19 

Ivanisevic T and Sewduth RN: Multi-omics integration for the design of novel therapies and the identification of novel biomarkers. Proteomes. 11(34)2023.PubMed/NCBI View Article : Google Scholar

20 

Jaliffa C, Rogel U, Sen I and Singer G: Comprehensive genomic characterization in ovarian low-grade and chemosensitive and chemoresistant high-grade serous carcinomas. Oncology. 1–9. 2024.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print).

21 

Caruso G, Tomao F, Parma G, Lapresa M, Multinu F, Palaia I, Aletti G and Colombo N: Poly (ADP-ribose) polymerase inhibitors (PARPi) in ovarian cancer: Lessons learned and future directions. Int J Gynecol Cancer. 33:431–443. 2023.PubMed/NCBI View Article : Google Scholar

22 

Rios-Doria E, Momeni-Boroujeni A, Friedman CF, Selenica P, Zhou Q, Wu M, Marra A, Leitao MM Jr, Iasonos A, Alektiar KM, et al: Integration of clinical sequencing and immunohistochemistry for the molecular classification of endometrial carcinoma. Gynecol Oncol. 174:262–272. 2023.PubMed/NCBI View Article : Google Scholar

23 

Passarelli A, Ventriglia J, Pisano C, Cecere SC, Napoli MD, Rossetti S, Tambaro R, Tarotto L, Fiore F, Farolfi A, et al: The way to precision medicine in gynecologic cancers: The first case report of an exceptional response to alpelisib in a PIK3CA-mutated endometrial cancer. Front Oncol. 12(1088962)2023.PubMed/NCBI View Article : Google Scholar

24 

Li J, Xue X, Zhang Y, Ding F, Wu W, Liu C, Xu Y, Chen H, Ou Q, Shao Y, et al: The differences in immune features and genomic profiling between squamous cell carcinoma and adenocarcinoma-A multi-center study in Chinese patients with uterine cervical cancer. Gynecol Oncol. 175:133–141. 2023.PubMed/NCBI View Article : Google Scholar

25 

Song Y, Bi Z, Liu Y, Qin F, Wei Y and Wei X: Targeting RAS-RAF-MEK-ERK signaling pathway in human cancer: Current status in clinical trials. Genes Dis. 10:76–88. 2022.PubMed/NCBI View Article : Google Scholar

26 

Tewari KS, Sill MW, Long HJ III, Penson RT, Huang H, Ramondetta LM, Landrum LM, Oaknin A, Reid TJ, Leitao MM, et al: Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med. 370:734–743. 2014.PubMed/NCBI View Article : Google Scholar

27 

Friedman CF, Ravichandran V, Miller K, Vanderbilt C, Zhou Q, Iasonos A, Vivek M, Mishra P, Leitao MM Jr, Broach V, et al: Assessing the genomic landscape of cervical cancers: Clinical opportunities and therapeutic targets. Clin Cancer Res. 29:4660–4668. 2023.PubMed/NCBI View Article : Google Scholar

28 

Jenkins TM and Mills AM: Putative precancerous lesions of vulvar squamous cell carcinoma. Semin Diagn Pathol. 38:27–36. 2021.PubMed/NCBI View Article : Google Scholar

29 

Ferrandina G, Palluzzi E, Gallotta V, Gambacorta MA, Autorino R, Turco LC, Macchia G, Cosentino F, Gui B, Mattoli MV, et al: Neo-adjuvant platinum-based chemotherapy followed by chemoradiation and radical surgery in locally advanced cervical cancer (Lacc) patients: A phase II study. Eur J Surg Oncol. 44:1062–1068. 2018.PubMed/NCBI View Article : Google Scholar

30 

Pourmasoumi P, Moradi A and Bayat M: BRCA1/2 mutations and breast/ovarian cancer risk: A new insights review. Reprod Sci: Aug 6, 2024 (Epub ahead of print).

31 

Qu Y, Qin S, Yang Z, Li Z, Liang Q, Long T, Wang W, Zeng D, Zhao Q, Dai Z, et al: Targeting the DNA repair pathway for breast cancer therapy: Beyond the molecular subtypes. Biomed Pharmacother. 169(115877)2023.PubMed/NCBI View Article : Google Scholar

32 

Telli ML, Timms KM, Reid J, Hennessy B, Mills GB, Jensen KC, Szallasi Z, Barry WT, Winer EP, Tung NM, et al: Homologous recombination deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer. Clin Cancer Res. 22:3764–3773. 2016.PubMed/NCBI View Article : Google Scholar

33 

Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, Lisyanskaya A, Floquet A, Leary A, Sonke GS, et al: Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 379:2495–2505. 2018.PubMed/NCBI View Article : Google Scholar

34 

Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, Fabbro M, Ledermann JA, Lorusso D, Vergote I, et al: Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 375:2154–2164. 2016.PubMed/NCBI View Article : Google Scholar

35 

Li X and Zou L: BRCAness, DNA gaps, and gain and loss of PARP inhibitor-induced synthetic lethality. J Clin Invest. 134(e181062)2024.PubMed/NCBI View Article : Google Scholar

36 

Guo Z, Zhang R, Yang AG and Zheng G: Diversity of immune checkpoints in cancer immunotherapy. Front Immunol. 14(1121285)2023.PubMed/NCBI View Article : Google Scholar

37 

Aravantinou-Fatorou A, Andrikopoulou A, Liontos M, Fiste O, Georgakopoulou VE, Dimopoulos MA, Gavriatopoulou M and Zagouri F: Pembrolizumab in endometrial cancer: Where we stand now. Oncol Lett. 22(821)2021.PubMed/NCBI View Article : Google Scholar

38 

Hamanishi J, Mandai M, Ikeda T, Minami M, Kawaguchi A, Murayama T, Kanai M, Mori Y, Matsumoto S, Chikuma S, et al: Safety and antitumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. J Clin Oncol. 33:4015–4022. 2015.PubMed/NCBI View Article : Google Scholar

39 

Wang YM, Cai W, Xue QM, Zhang JY, Zhou L, Xiong SY and Deng H: Prognostic role of different PD-L1 expression patterns and tumor-infiltrating lymphocytes in high-grade serous ovarian cancer: A systematic review and meta-analysis. Front Immunol. 14(1234894)2023.PubMed/NCBI View Article : Google Scholar

40 

Andrikopoulou A, Liontos M, Skafida E, Koutsoukos K, Apostolidou K, Kaparelou M, Rouvalis A, Bletsa G, Dimopoulos MA and Zagouri F: Pembrolizumab in combination with bevacizumab and oral cyclophosphamide in heavily pre-treated platinum-resistant ovarian cancer. Int J Gynecol Cancer. 33:571–576. 2023.PubMed/NCBI View Article : Google Scholar

41 

Pujade-Lauraine E, Fujiwara K, Ledermann JA, Oza AM, Kristeleit R, Ray-Coquard IL, Richardson GE, Sessa C, Yonemori K, Banerjee S, et al: Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN ovarian 200): An open-label, three-arm, randomised, phase 3 study. Lancet Oncol. 22:1034–1046. 2021.PubMed/NCBI View Article : Google Scholar

42 

Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL, Watkins JC, Rodig S, Stover E, Strickland KC, D'Andrea AD, Wu CJ, et al: Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1. JAMA Oncol. 1:1319–1323. 2015.PubMed/NCBI View Article : Google Scholar

43 

Zhang C, Wang M and Wu Y: Features of the immunosuppressive tumor microenvironment in endometrial cancer based on molecular subtype. Front Oncol. 13(1278863)2023.PubMed/NCBI View Article : Google Scholar

44 

Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, et al: Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 357:409–413. 2017.PubMed/NCBI View Article : Google Scholar

45 

Heeren AM, Punt S, Bleeker MC, Gaarenstroom KN, van der Velden J, Kenter GG, de Gruijl TD and Jordanova ES: Prognostic effect of different PD-L1 expression patterns in squamous cell carcinoma and adenocarcinoma of the cervix. Mod Pathol. 29:753–763. 2016.PubMed/NCBI View Article : Google Scholar

46 

Frenel JS, Le Tourneau C, O'Neil B, Ott PA, Piha-Paul SA, Gomez-Roca C, van Brummelen EMJ, Rugo HS, Thomas S, Saraf S, et al: Safety and efficacy of pembrolizumab in advanced, programmed death ligand 1-positive cervical cancer: Results from the phase Ib KEYNOTE-028 trial. J Clin Oncol. 35:4035–4041. 2017.PubMed/NCBI View Article : Google Scholar

47 

Choschzick M, Gut A and Fink D: PD-L1 receptor expression in vulvar carcinomas is HPV-independent. Virchows Arch. 473:513–516. 2018.PubMed/NCBI View Article : Google Scholar

48 

Borella F, Preti M, Bertero L, Collemi G, Castellano I, Cassoni P, Cosma S, Carosso AR, Bevilacqua F, Gallio N, et al: Is there a place for immune checkpoint inhibitors in vulvar neoplasms? A state of the art review. Int J Mol Sci. 22(190)2020.PubMed/NCBI View Article : Google Scholar

49 

Praiss A, Navitski A, Cohen S, Tessier-Cloutier B, Broach V and O'Cearbhaill RE: Immunotherapy for recurrent or metastatic vulvar carcinoma: A case report and review of current guidelines. Gynecol Oncol Rep. 41(100982)2022.PubMed/NCBI View Article : Google Scholar

50 

Xu Y, Xiong F, Li H, Zheng H, Jiang J, Li Q, Li G, Zhao W, Li R, Li J, et al: Biomarker-driven targeted therapy in patients with recurrent platinum-resistant epithelial ovarian cancer (BRIGHT): Protocol for an open-label, multicenter, umbrella study. Int J Gynecol Cancer. 34:1461–1465. 2024.PubMed/NCBI View Article : Google Scholar

51 

Lee JY, Kim BG, Kim JW, Lee JB, Park E, Joung JG, Kim S, Choi CH and Kim HS: Korean Gynecologic Oncology Group (KGOG) investigators. Biomarker-guided targeted therapy in platinum-resistant ovarian cancer (AMBITION; KGOG 3045): A multicentre, open-label, five-arm, uncontrolled, umbrella trial. J Gynecol Oncol. 33(e45)2022.PubMed/NCBI View Article : Google Scholar

52 

Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Berger R, Fujiwara K, Vergote I, Colombo N, Mäenpää J, et al: Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 381:2416–2428. 2019.PubMed/NCBI View Article : Google Scholar

53 

Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, Gottfried M, Penel N, Hansen AR, et al: Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: Results from the phase II KEYNOTE-158 study. J Clin Oncol. 38:1–10. 2020.PubMed/NCBI View Article : Google Scholar

54 

Heery CR, O'Sullivan-Coyne G, Madan RA, Cordes L, Rajan A, Rauckhorst M, Lamping E, Oyelakin I, Marté JL, Lepone LM, et al: Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN solid tumor): A phase 1a, multicohort, dose-escalation trial. Lancet Oncol. 18:587–598. 2017.PubMed/NCBI View Article : Google Scholar

55 

Makker V, Taylor MH, Aghajanian C, Oaknin A, Mier J, Cohn AL, Romeo M, Bratos R, Brose MS, DiSimone C, et al: Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J Clin Oncol. 38:2981–2992. 2020.PubMed/NCBI View Article : Google Scholar

56 

Makker V, Colombo N, Casado Herráez A, Santin AD, Colomba E, Miller DS, Fujiwara K, Pignata S, Baron-Hay S, Ray-Coquard I, et al: Lenvatinib plus pembrolizumab for advanced endometrial cancer. N Engl J Med. 386:437–448. 2022.PubMed/NCBI View Article : Google Scholar

57 

Tewari KS, Sill MW, Penson RT, Huang H, Ramondetta LM, Landrum LM, Oaknin A, Reid TJ, Leitao MM, Michael HE, et al: Bevacizumab for advanced cervical cancer: Final overall survival and adverse event analysis of a randomised, controlled, open-label, phase 3 trial (gynecologic oncology group 240). Lancet. 390:1654–1663. 2017.PubMed/NCBI View Article : Google Scholar

58 

How JA, Jazaeri AA, Soliman PT, Fleming ND, Gong J, Piha-Paul SA, Janku F, Stephen B and Naing A: Pembrolizumab in vaginal and vulvar squamous cell carcinoma: A case series from a phase II basket trial. Sci Rep. 11(3667)2021.PubMed/NCBI View Article : Google Scholar

59 

Naumann RW, Hollebecque A, Meyer T, Devlin MJ, Oaknin A, Kerger J, López-Picazo JM, Machiels JP, Delord JP, Evans TRJ, et al: Safety and efficacy of nivolumab monotherapy in recurrent or metastatic cervical, vaginal, or vulvar carcinoma: Results from the phase I/II CheckMate 358 trial. J Clin Oncol. 37:2825–2834. 2019.PubMed/NCBI View Article : Google Scholar

60 

Zhao M, Qiu S, Wu X, Miao P, Jiang Z, Zhu T, Xu X, Zhu Y, Zhang B, Yuan D, et al: Efficacy and safety of niraparib as first-line maintenance treatment for patients with advanced ovarian cancer: Real-world data from a multicenter study in China. Target Oncol. 18:869–883. 2023.PubMed/NCBI View Article : Google Scholar

61 

Simmons D, Blank S, Nagar S, Nham T, Long G, Davis K, Wetherill G, Munley J and McLaurin K: Real-world evaluation of PARP inhibitor tolerability in advanced ovarian cancer: Retrospective US cohort study (2209). Gynecologic Oncology. 176 (Suppl 1):S262–S263. 2023.

62 

Zhang N, Zheng H, Gao Y, Shu T, Wang H and Cai Y: A real-world study of treatment patterns following disease progression in epithelial ovarian cancer patients undergoing poly-ADP-ribose polymerase inhibitor maintenance therapy. J Ovarian Res. 17(55)2024.PubMed/NCBI View Article : Google Scholar

63 

Huepenbecker S, Meyer LA, Craft M, Chan JK, Craggs C, Lambert P and Lin YG: Real-world use of immune checkpoint inhibitors in advanced or recurrent endometrial cancer. Int J Gynecol Cancer. (ijgc-2024-005541)2024.PubMed/NCBI View Article : Google Scholar : (Epub ahead of print).

64 

Testa U, Petrucci E, Pasquini L, Castelli G and Pelosi E: Ovarian cancers: Genetic abnormalities, tumor heterogeneity and progression, clonal evolution and cancer stem cells. Medicines (Basel). 5(16)2018.PubMed/NCBI View Article : Google Scholar

65 

Kotnik EN, Mullen MM, Spies NC, Li T, Inkman M, Zhang J, Martins-Rodrigues F, Hagemann IS, McCourt CK, Thaker PH, et al: Genetic characterization of primary and metastatic high-grade serous ovarian cancer tumors reveals distinct features associated with survival. Commun Biol. 6(688)2023.PubMed/NCBI View Article : Google Scholar

66 

Lliberos C, Richardson G and Papa A: Oncogenic pathways and targeted therapies in ovarian cancer. Biomolecules. 14(585)2024.PubMed/NCBI View Article : Google Scholar

67 

Pešut E, Đukić A, Lulić L, Skelin J, Šimić I, Milutin Gašperov N, Tomaić V, Sabol I and Grce M: Human papillomaviruses-associated cancers: An update of current knowledge. Viruses. 13(2234)2021.PubMed/NCBI View Article : Google Scholar

68 

Van Arsdale A, Turker L, Chang YC, Gould J, Harmon B, Maggi EC, Meshcheryakova O, Brown MP, Luong D, Van Doorslaer K, et al: Structure and transcription of integrated HPV DNA in vulvar carcinomas. NPJ Genom Med. 9(35)2024.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2025
Volume 22 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aravantinou‑Fatorou A, Georgakopoulou VE, Dimopoulos MA and Liontos M: Precision medicine in gynecological cancer (Review). Biomed Rep 22: 43, 2025.
APA
Aravantinou‑Fatorou, A., Georgakopoulou, V.E., Dimopoulos, M.A., & Liontos, M. (2025). Precision medicine in gynecological cancer (Review). Biomedical Reports, 22, 43. https://doi.org/10.3892/br.2025.1921
MLA
Aravantinou‑Fatorou, A., Georgakopoulou, V. E., Dimopoulos, M. A., Liontos, M."Precision medicine in gynecological cancer (Review)". Biomedical Reports 22.3 (2025): 43.
Chicago
Aravantinou‑Fatorou, A., Georgakopoulou, V. E., Dimopoulos, M. A., Liontos, M."Precision medicine in gynecological cancer (Review)". Biomedical Reports 22, no. 3 (2025): 43. https://doi.org/10.3892/br.2025.1921