Next Article in Journal
Atom Search Optimization with Deep Learning Enabled Arabic Sign Language Recognition for Speaking and Hearing Disability Persons
Next Article in Special Issue
Fibroid Removal after Myomectomy: An Overview on the Problems of Power Morcellation
Previous Article in Journal
Assessing Barriers Encountered by Women in Cervical Cancer Screening and Follow-Up Care in Urban Bolivia, Cochabamba
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Advances on Prevention and Screening of Gynecologic Tumors: Are We Stepping Forward?

1
Department of Medical and Surgical Sciences and Translational Medicine, Sapienza University, 00189 Rome, Italy
2
Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Policlinico Umberto I, 00185 Rome, Italy
3
Gynecologic Oncology Unit, Department of Experimental Clinical Oncology, IRCSS-Regina Elena National Cancer Institute, 00144 Rome, Italy
4
Unit of Gynecologic Oncology, ARNAS “Civico-Di Cristina-Benfratelli”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
5
Obstetrics and Gynecological Unit, Department of Woman’s and Child’s Health, San Camillo-Forlanini Hospital, 00152 Rome, Italy
6
Gynecology Division, Department of Medical and Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Rome, Italy
*
Author to whom correspondence should be addressed.
Healthcare 2022, 10(9), 1605; https://doi.org/10.3390/healthcare10091605
Submission received: 10 August 2022 / Revised: 22 August 2022 / Accepted: 23 August 2022 / Published: 24 August 2022
(This article belongs to the Special Issue Gynecological Cancer Diagnosis and Prevention)
According to 2020 comprehensive global cancer statistics published by the International Agency for Research on Cancer, gynecologic malignancies accounted overall for 16.5% of 8.2 million estimated new cancer cases in women [1]. Gynecological cancers represent an ongoing source of concern due to their high incidence and cancer-related mortality [2,3,4].
Although significant therapeutic advances occurred in recent years, patients’ outcomes still remained poor. Indeed, the adoption of innovative treatments (including targeted agents with potential anticancer effects such as antiangiogenic agents, poly (ADP-ribose) polymerase (PARP) inhibitors, tumor-intrinsic signaling pathway inhibitors, and immune checkpoint inhibitors) are still reserved in patients with advanced or recurrent disease [2,3,4,5].
There are three approaches used to manage cancers, based on the concept of primary, secondary, and tertiary prevention. The primary prevention approach focuses on preventing disease before it develops (i.e., prophylactic treatment); secondary prevention attempts to detect the disease as early as possible (i.e., early detection); and tertiary prevention is directed at managing a present disease (i.e., treatment) [6,7,8,9].
Cervical cancer represents an ideal model in which to apply primary, secondary, and tertiary preventions. Prophylactic vaccination against HPV aims to reduce the burden of HPV infection and HPV-related lesions (including both precancerous lesions and overt cervical cancer) [6,7]. Vaccination (including bivalent, quadrivalent, and nonavalent vaccines) reduce the incident and persistent infections from HPV types included in the vaccines. Cross protection against other HPV types, not included in the vaccine, is shown only in vaccines that have the adjuvant substance 04 (AS04) [6,7]. Accumulating evidence suggests that the adoption of the vaccines resulted in a significant decrease in HPV infection, HPV lesions, and cervical cancer [6,7,8,9]. In particular, the most recent Cochrane systematic review about the topic clarified that both nonavalent and quadrivalent vaccines offer similar protection against a combined outcome of cervical, vaginal, and vulval precancer lesions or cancer [10].
Secondary prevention (i.e., screening) is widely adopted to prevent cervical cancer. Screening is defined as the identification of early signs of a specific disease in apparently ‘healthy’ people who do not have any symptoms, to provide early detection and to reduce mortality. The adoption of Pap smear and HPV testing resulted in a significant increase in the diagnosis of cervical dysplasia and a significant decrease in cervical cancer. Moreover, mobile health interventions effectively improve cancer screening rates in the general population, but they are necessary for the poorest countries. New screening methods, such as p16/Ki67 [11], HPV self-testing, and the use of artificial intelligence in colposcopic assessment, should be disseminated [7,8]. It is useful to focus on precancerous lesions in order to create tools for women regarding their risk of persistence/recurrence after primary conization and identify categories at higher risk compared with the other ones [9]. Tertiary prevention in cervical cancer includes its treatment with surgery (radical hysterectomy), radiotherapy, and systemic treatments (chemotherapy, bevacizumab, and immunotherapy) [12]. This can be considered of paramount importance, since a recent systematic review and meta-analysis found that women treated for cervical intraepithelial neoplasia (CIN) have a considerably higher risk of later being diagnosed with cervical and other HPV-related cancers compared with the general population: in particular, the higher risk of cervical cancer lasts for at least 20 years after treatment and is higher for women of more than 50 years of age [13]. In this context, a significant risk reduction of developing recurrent CIN after surgical excision and HPV vaccination was shown compared to surgical excision only [14]. Since cervical cancer is a predictable disease, we need more efforts to implement prevention, screening, and early diagnosis in cervical cancer.
Unfortunately, the implementation of primary and secondary prevention in other gynecological cancers is not optimal at present.
Endometrial cancer occurs in postmenopausal women with an average age at diagnosis of 60 years, and the risk factors associated with this tumor are widely known [15]. Although there is no standardized screening test for endometrial cancer, prevention is possible following healthy behaviors (prevention of obesity, weight gain, metabolic syndrome, and diabetes). As confirmed by a recent systematic review and meta-analysis, early detection strategies focused on women with post-menopausal bleeding have the potential to capture as many as 90% of endometrial cancers [16].
Furthermore, the identification of women with hyperplasia and atypia is of paramount importance to reduce the burden of endometrial cancer. The identification of women suffering with abnormal uterine bleeding, and with those with abnormal endometrial thickness (evaluated by trans vaginal ultrasound (TVS)) [17] is making screening and early diagnosis possible, even in endometrial cancer patients [18]. Finding a screening test for endometrial cancer is challenging, but early detection is more and more possible. This should be considered a research priority, since a recent meta-analysis of prospective studies found that adherence to cancer prevention guidelines was negatively related to endometrial cancer risk [19].
TVS screening for endometrial cancer has good sensitivity in postmenopausal women, as shown by a case–control study within the United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) cohort [20]. Recent studies have shown that urine CA125 displays potential as a diagnostic marker for symptomatic women with suspected endometrial cancer [21], but more robust molecular markers are still to be implemented in the clinical practice [22].
Ovarian cancer is the deadliest gynecologic cancer, despite the incidence of this tumor being relatively low. Currently, ovarian cancer is a target of intense research because it is often not discovered until the disease is advanced, which causes significant mortality [23]. Screening is not currently recommended in the general population, and some countries offer surveillance of high-risk women, with specific genetic patterns and positive family history, with a lifetime ovarian cancer risk of 10% or more [24].
CA125 is the typical biomarker of serous ovarian cancer used in recent decades. The application of this tumor biomarker has changed using longitudinal algorithms in recent years, trying to be more effective [25]. In addition, pooled data from a recent and well-designed meta-analysis confirmed the overall diagnostic value of Serum Human Epididymis Protein 4 (HE4) in ovarian cancer [26]. In addition to these biomarkers, accumulating evidence from other meta-analyses suggests that future strategies for early detection of ovarian cancer may be based on non-invasive evaluation of circulating tumor DNA [27], as well as circulating microRNA profiling [28,29].
The randomized controlled trial UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) has shown that the use of longitudinal CA125-based multimodal screening (MMS), or annual TVS (USS) is not useful for reducing mortality for OC. However, the number of women affected by low-volume invasive epithelial ovarian and peritoneal cancer (stage I, II, and IIIa) was higher in the MMS group than in the ultrasound counterpart [30]. The Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) confirmed that there was no difference in number of deaths for OC between the screen and no screen arms, analyzing 78,286 women at a median follow-up of 14.7 years [31]. In the same way, UK FOCSS Phase I had the same results, showing that annual screening is ineffective for the general population when it comes to detecting early stage disease [32]. Although no screening strategies have been identified to definitively decrease ovarian cancer mortality at the moment, a strategy based on the longitudinal CA125 profile and second-line TVS performed by skilled operators can guide to earlier diagnosis of OC in the general population, not only in the high-risk group. Notably, The US Food and Drugs Administration (FDA) highlighted that “Health professionals should not recommend ovarian cancer screening tests to women who do not have any symptoms because of the high possibility of unreliable results” and, more importantly, that “there are currently no screening tests for ovarian cancer that are sensitive enough to reliably screen for ovarian cancer” [33].
In this context, are screening techniques and prevention strategies for gynecologic cancers adequate? For cervical cancer, effective screening methods exist, although they are underused. For endometrial and ovarian cancers, no effective screening methods exist.
There is an urgent need to identify novel strategies to detect all gynecologic tumors as early as possible, thus reducing mortality and improving the quality of care.

Author Contributions

Conceptualization, A.G. and O.D.; writing—original draft preparation, A.S.L.; writing—review and editing, G.B., V.C.; visualization, E.V.; supervision, L.M., D.C., M.G.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Makker, V.; Colombo, N.; Casado Herráez, A.; Santin, A.D.; Colomba, E.; Miller, D.S.; Fujiwara, K.; Pignata, S.; Baron-Hay, S.; Ray-Coquard, I.; et al. Lenvatinib plus Pembrolizumab for Advanced Endometrial Cancer. N. Engl. J. Med. 2022, 386, 437–448. [Google Scholar] [CrossRef] [PubMed]
  3. Colombo, N.; Dubot, C.; Lorusso, D.; Caceres, M.V.; Hasegawa, K.; Shapira-Frommer, R.; Tewari, K.S.; Salman, P.; Hoyos Usta, E.; Yañez, E.; et al. Pembrolizumab for Persistent, Recurrent, or Metastatic Cervical Cancer. N. Engl. J. Med. 2021, 385, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  4. 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. 2019, 381, 2416–2428. [Google Scholar] [CrossRef]
  5. Di Tucci, C.; Schiavi, M.C.; Faiano, P.; D’Oria, O.; Prata, G.; Sciuga, V.; Giannini, A.; Palaia, I.; Muzii, L.; Panici, P.B. Therapeutic Vaccines and Immune Checkpoints Inhibition Options for Gynecological Cancers. Crit. Rev. Oncol./Hematol. 2018, 128, 30–42. [Google Scholar] [CrossRef]
  6. D’Oria, O.; Corrado, G.; Laganà, A.S.; Chiantera, V.; Vizza, E.; Giannini, A. New Advances in Cervical Cancer: From Bench to Bedside. Int. J. Environ. Res. Public Health 2022, 19, 7094. [Google Scholar] [CrossRef]
  7. Bogani, G.; Sopracordevole, F.; Di Donato, V.; Ciavattini, A.; Ghelardi, A.; Lopez, S.; Simoncini, T.; Plotti, F.; Casarin, J.; Serati, M.; et al. High-Risk HPV-Positive and -Negative High-Grade Cervical Dysplasia: Analysis of 5-Year Outcomes. Gynecol. Oncol. 2021, 161, 173–178. [Google Scholar] [CrossRef]
  8. Poniewierza, P.; Panek, G. Cervical Cancer Prophylaxis-State-of-the-Art and Perspectives. Healthcare 2022, 10, 1325. [Google Scholar] [CrossRef]
  9. Bogani, G.; Lalli, L.; Sopracordevole, F.; Ciavattini, A.; Ghelardi, A.; Simoncini, T.; Plotti, F.; Casarin, J.; Serati, M.; Pinelli, C.; et al. Development of a Nomogram Predicting the Risk of Persistence/Recurrence of Cervical Dysplasia. Vaccines 2022, 10, 579. [Google Scholar] [CrossRef]
  10. Bergman, H.; Buckley, B.S.; Villanueva, G.; Petkovic, J.; Garritty, C.; Lutje, V.; Riveros-Balta, A.X.; Low, N.; Henschke, N. Comparison of Different Human Papillomavirus (HPV) Vaccine Types and Dose Schedules for Prevention of HPV-Related Disease in Females and Males. Cochrane Database Syst. Rev. 2019, 2019. [Google Scholar] [CrossRef]
  11. Valenti, G.; Vitale, S.G.; Tropea, A.; Biondi, A.; Laganà, A.S. Tumor Markers of Uterine Cervical Cancer: A New Scenario to Guide Surgical Practice? Updat. Surg. 2017, 69, 441–449. [Google Scholar] [CrossRef] [PubMed]
  12. Raglan, O.; Kalliala, I.; Markozannes, G.; Cividini, S.; Gunter, M.J.; Nautiyal, J.; Gabra, H.; Paraskevaidis, E.; Martin-Hirsch, P.; Tsilidis, K.K.; et al. Risk Factors for Endometrial Cancer: An Umbrella Review of the Literature. Int. J. Cancer 2019, 145, 1719–1730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kalliala, I.; Athanasiou, A.; Veroniki, A.A.; Salanti, G.; Efthimiou, O.; Raftis, N.; Bowden, S.; Paraskevaidi, M.; Aro, K.; Arbyn, M.; et al. Incidence and Mortality from Cervical Cancer and Other Malignancies after Treatment of Cervical Intraepithelial Neoplasia: A Systematic Review and Meta-Analysis of the Literature. Ann. Oncol. 2020, 31, 213–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jentschke, M.; Kampers, J.; Becker, J.; Sibbertsen, P.; Hillemanns, P. Prophylactic HPV Vaccination after Conization: A Systematic Review and Meta-Analysis. Vaccine 2020, 38, 6402–6409. [Google Scholar] [CrossRef] [PubMed]
  15. Bogani, G.; Donato, V.D.; Scambia, G.; Landoni, F.; Ghezzi, F.; Muzii, L.; Panici, P.B.; Raspagliesi, F.; Investigator of the Italian Gynecological Cancer Study Group; Bogani, G.; et al. Practice Patterns and 90-Day Treatment-Related Morbidity in Early-Stage Cervical Cancer. Gynecol. Oncol. 2022; in press. [Google Scholar] [CrossRef]
  16. Clarke, M.A.; Long, B.J.; Del Mar Morillo, A.; Arbyn, M.; Bakkum-Gamez, J.N.; Wentzensen, N. Association of Endometrial Cancer Risk With Postmenopausal Bleeding in Women: A Systematic Review and Meta-Analysis. JAMA Intern. Med. 2018, 178, 1210–1222. [Google Scholar] [CrossRef] [Green Version]
  17. Spagnol, G.; Noventa, M.; Bonaldo, G.; Marchetti, M.; Vitagliano, A.; Laganà, A.S.; Cavallin, F.; Scioscia, M.; Saccardi, C.; Tozzi, R. 3D Transvaginal Ultrasound vs Magnetic Resonance Imaging for Preoperative Staging of Myometrial and Cervical Invasion in Patients with Endometrial Cancer: Systematic Review and Meta-Analysis. Ultrasound Obstet. Gynecol. 2022. [CrossRef]
  18. Njoku, K.; Abiola, J.; Russell, J.; Crosbie, E.J. Endometrial Cancer Prevention in High-Risk Women. Best Pract. Res. Clin. Obstet. Gynaecol. 2020, 65, 66–78. [Google Scholar] [CrossRef]
  19. Sun, H.; Chang, Q.; Liu, Y.-S.; Jiang, Y.-T.; Gong, T.-T.; Ma, X.-X.; Zhao, Y.-H.; Wu, Q.-J. Adherence to Cancer Prevention Guidelines and Endometrial Cancer Risk: Evidence from a Systematic Review and Dose-Response Meta-Analysis of Prospective Studies. Cancer Res. Treat. 2021, 53, 223–232. [Google Scholar] [CrossRef]
  20. Jacobs, I.; Gentry-Maharaj, A.; Burnell, M.; Manchanda, R.; Singh, N.; Sharma, A.; Ryan, A.; Seif, M.W.; Amso, N.N.; Turner, G.; et al. Sensitivity of Transvaginal Ultrasound Screening for Endometrial Cancer in Postmenopausal Women: A Case-Control Study within the UKCTOCS Cohort. Lancet Oncol. 2011, 12, 38–48. [Google Scholar] [CrossRef]
  21. Njoku, K.; Barr, C.E.; Sutton, C.J.J.; Crosbie, E.J. Urine CA125 and HE4 for the Triage of Symptomatic Women with Suspected Endometrial Cancer. Cancers 2022, 14, 3306. [Google Scholar] [CrossRef] [PubMed]
  22. Benati, M.; Montagnana, M.; Danese, E.; Mazzon, M.; Paviati, E.; Garzon, S.; Laganà, A.S.; Casarin, J.; Giudici, S.; Raffaelli, R.; et al. Aberrant Telomere Length in Circulating Cell-Free DNA as Possible Blood Biomarker with High Diagnostic Performance in Endometrial Cancer. Pathol. Oncol. Res. 2020, 26, 2281–2289. [Google Scholar] [CrossRef] [PubMed]
  23. Nash, Z.; Menon, U. Ovarian Cancer Screening: Current Status and Future Directions. Best Pract. Res. Clin. Obstet. Gynaecol. 2020, 65, 32–45. [Google Scholar] [CrossRef] [PubMed]
  24. Saccardi, C.; Zovato, S.; Spagnol, G.; Bonaldo, G.; Marchetti, M.; Alessandrini, L.; Tognazzo, S.; Guerriero, A.; Vitagliano, A.; Laganà, A.S.; et al. Efficacy of Risk-Reducing Salpingo-Oophorectomy in BRCA1-2 Variants and Clinical Outcomes of Follow-up in Patients with Isolated Serous Tubal Intraepithelial Carcinoma (STIC). Gynecol. Oncol. 2021, 163, 364–370. [Google Scholar] [CrossRef]
  25. Jacobs, I.J.; Menon, U.; Ryan, A.; Gentry-Maharaj, A.; Burnell, M.; Kalsi, J.K.; Amso, N.N.; Apostolidou, S.; Benjamin, E.; Cruickshank, D.; et al. Ovarian Cancer Screening and Mortality in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A Randomised Controlled Trial. Lancet 2016, 387, 945–956. [Google Scholar] [CrossRef] [Green Version]
  26. Huang, J.; Chen, J.; Huang, Q. Diagnostic Value of HE4 in Ovarian Cancer: A Meta-Analysis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2018, 231, 35–42. [Google Scholar] [CrossRef]
  27. Lu, Y.; Li, L. The Prognostic Value of Circulating Tumor DNA in Ovarian Cancer: A Meta-Analysis. Technol. Cancer Res. Treat. 2021, 20, 15330338211043784. [Google Scholar] [CrossRef]
  28. Zhang, L.; Hu, C.; Huang, Z.; Li, Z.; Zhang, Q.; He, Y. In Silico Screening of Circulating Tumor DNA, Circulating MicroRNAs, and Long Non-Coding RNAs as Diagnostic Molecular Biomarkers in Ovarian Cancer: A Comprehensive Meta-Analysis. PLoS ONE 2021, 16, e0250717. [Google Scholar] [CrossRef]
  29. Wu, L.; Shang, W.; Zhao, H.; Rong, G.; Zhang, Y.; Xu, T.; Zhang, J.; Huang, P.; Wang, F. In Silico Screening of Circulating MicroRNAs as Potential Biomarkers for the Diagnosis of Ovarian Cancer. Dis. Markers 2019, 2019, 7541857. [Google Scholar] [CrossRef] [Green Version]
  30. Menon, U.; Gentry-Maharaj, A.; Burnell, M.; Singh, N.; Ryan, A.; Karpinskyj, C.; Carlino, G.; Taylor, J.; Massingham, S.K.; Raikou, M.; et al. Ovarian Cancer Population Screening and Mortality after Long-Term Follow-up in the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS): A Randomised Controlled Trial. Lancet 2021, 397, 2182–2193. [Google Scholar] [CrossRef]
  31. Pinsky, P.F.; Yu, K.; Kramer, B.S.; Black, A.; Buys, S.S.; Partridge, E.; Gohagan, J.; Berg, C.D.; Prorok, P.C. Extended Mortality Results for Ovarian Cancer Screening in the PLCO Trial with Median 15years Follow-Up. Gynecol. Oncol. 2016, 143, 270–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Rosenthal, A.N.; Fraser, L.; Manchanda, R.; Badman, P.; Philpott, S.; Mozersky, J.; Hadwin, R.; Cafferty, F.H.; Benjamin, E.; Singh, N.; et al. Results of Annual Screening in Phase I of the United Kingdom Familial Ovarian Cancer Screening Study Highlight the Need for Strict Adherence to Screening Schedule. J. Clin. Oncol. 2013, 31, 49–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Safety Alerts for Human Medical Products > Ovarian Cancer Screening Tests: Safety Communication–FDA Recommends Against Use. Available online: https://www.fdanews.com/ext/resources/files/2016/09/09-08-16-FDAsafetynotice.pdf?1473536862 (accessed on 9 August 2022).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giannini, A.; Bogani, G.; Vizza, E.; Chiantera, V.; Laganà, A.S.; Muzii, L.; Salerno, M.G.; Caserta, D.; D’Oria, O. Advances on Prevention and Screening of Gynecologic Tumors: Are We Stepping Forward? Healthcare 2022, 10, 1605. https://doi.org/10.3390/healthcare10091605

AMA Style

Giannini A, Bogani G, Vizza E, Chiantera V, Laganà AS, Muzii L, Salerno MG, Caserta D, D’Oria O. Advances on Prevention and Screening of Gynecologic Tumors: Are We Stepping Forward? Healthcare. 2022; 10(9):1605. https://doi.org/10.3390/healthcare10091605

Chicago/Turabian Style

Giannini, Andrea, Giorgio Bogani, Enrico Vizza, Vito Chiantera, Antonio Simone Laganà, Ludovico Muzii, Maria Giovanna Salerno, Donatella Caserta, and Ottavia D’Oria. 2022. "Advances on Prevention and Screening of Gynecologic Tumors: Are We Stepping Forward?" Healthcare 10, no. 9: 1605. https://doi.org/10.3390/healthcare10091605

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