SciELO - Scientific Electronic Library Online

 
vol.17 número1Perfil das mulheres que recorrem a uma interrupção voluntária da gravidez num hospital terciário português: período pré e pandémico ao COVID-19Cancro durante a gravidez: visão global sobre o diagnóstico, tratamento e conduta obstétrica índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay articulos similaresSimilares en SciELO

Compartir


Acta Obstétrica e Ginecológica Portuguesa

versión impresa ISSN 1646-5830

Acta Obstet Ginecol Port vol.17 no.1 Algés mar. 2023  Epub 31-Mar-2023

 

Original Study/Estudo Original

Sentinel lymph node mapping in endometrial cancer using indocyanin green and infrared flourescence

Pesquisa de gânglio sentinela com verde de indocianina e fluorescência infravermelha no cancro do endométrio

Sara Sousa Sales1 
http://orcid.org/0000-0001-5642-0248

Ângela Melo1 

Sónia Gonçalves1 

Nuno Nogueira Martins1 

Francisco Nogueira Martins1 

1. Centro Hospitalar Tondela-Viseu, EPE. Portugal.


Abstract

Overview and Aims:

Sentinel lymph node (SLN) biopsy in endometrial cancer has emerged as an alternative to systematic lymphadenectomy, with cervical indocyanin green injection being the preferred method.

Study Design, Population and Methods:

A prospective cohort study to assess the feasibility of SLN mapping in EC using ICG and near-infrared (NIR) fluorescence is ongoing at the authors’ Gynaecological Oncology Unit, in a Portuguese level III care hospital. The authors conducted a descriptive analysis of the clinicopathologic characteristics and discuss the process of implementing a novel technique.

Results:

A minimum number of 20 patients with EC of all histologies and grades recruited until publication was established. Of all 20 patients, 12 (12/20; 60.0%) had a total laparoscopic hysterectomy plus bilateral salpingo-oophorectomy (TLH/BSO) and SLN biopsy alone, and the remaining 8 patients underwent full retroperitoneal staging (8/20; 40.0%). At least 1 SLN was detected in all patients (20/20; 100%), bilaterally in 14 (14/20; 70%) and unilaterally in 6 (6/20; 30%). In 1 case, a third SLN was identified amongst presacral nodes. No isolated para-aortic SLNs were detected. The median number of SLNs removed per patient was 2.3 (range 1-3). Out of the 20 patients, 2 (2/20; 10%) had lymph node metastases and in both cases bilateral detection of SLN did not occur. The median operative time was 240 minutes, with total SLN mapping time of approximately 40 minutes per side. Median operative time was lower among patients undergoing an SLN mapping only, compared with patients undergoing a full lymphadenectomy (219min vs 280min). No cases of ICG injection-related complications occurred. Ultrastaging of sentinel lymph nodes was performed in all cases.

Conclusions:

This is the first published series of laparoscopic sentinel lymph node biopsy using ICG and NIR fluorescence in endometrial cancer, conducted in a Portuguese hospital.

Keywords: Indocyanin green; Cervical injection; Sentinel lymph node biopsy; Lymph node mapping; Endometrial cancer

Resumo

Introdução e Objetivos:

A biópsia de gânglio sentinela (BGS) no carcinoma do endométrio surgiu como alternativa à linfadenectomia sistemática, privilegiando-se a injeção intracervical de verde de indocianina.

Métodos:

Foi realizado um estudo coorte prospetivo com o intuito de avaliar a reprodutibilidade da BGS com verde de indocianina no carcinoma do endométrio, num Hospital Português de nível III. Procedeu-se à análise descritiva das caraterísticas clinico-patológicas da população estudada e discussão do processo de implementação da nova técnica.

Resultados:

Estabeleceu-se um mínimo de 20 doentes com carcinoma do endométrio até publicação, incluindo todos os graus e tipos histológicos. Entre as 20 doentes recrutadas, 12 (12/20; 60%) foram submetidas a histerectomia total e anexectomia bilateral (HTL/AB) e BGS e as 8 restantes a estadiamento retroperitoneal completo (8/20; 40%). Foi detetado pelo menos 1 gânglio sentinela (GS) em todos os casos (20/20; 100%), bilateralmente em 14 (14/20; 70%) e unilateralmente em 6 (6/20; 30%). Um terceiro GS sagrado foi identificado num caso. Não se identificaram GS para-aórticos. O número médio de GS removidos em cada doente foi 2.3 (intervalo 1-3). Entre as 20 doentes, 2 (2/20; 10%) apresentaram metastização ganglionar e em ambos os casos não foi possível identificar bilateralmente o GS. O tempo operatório médio foi 240 minutos e o tempo dedicado ao mapeamento ganglionar 40 minutos, unilateralmente. O tempo operatório médio foi inferior nas doentes submetidas a BGS, comparativamente à linfadenectomia sistemática (219 minutos vs 280 minutos). Não houve complicações associadas à injeção do marcador. Foi realizado ultraestadiamento em todos os casos.

Conclusões:

A presente publicação é a primeira série de casos portuguesa reportada de biópsia de gânglio sentinela com verde de indocianina, por via laparoscópica no carcinoma do endométrio.

Palavras-Chave: Verde de indocianina; Injeção intracervical; Biópsia de gânglio sentinela; Estadiamento ganglionar; Carcinoma do endométrio

Introduction

Endometrial cancer (EC) is the most common gynecological malignancy in developed countries and the 4th most frequently diagnosed cancer in women, accounting for 6% of all malignancies in this population1,2. In Portugal, its incidence is 5.2/100.000 and expected to rise due to aging population and the increase in obesity and metabolic syndrome rates, which strongly correlate with an increased risk of developing endometrial cancer1,3. Upon diagnosis, most women (80%) have disease confined to the uterus (stage FIGO I) 1,3,4. Prognosis considerably worsens when regional or metastatic disease is present (5 year overall survival rates of 68% and 17%, respectively) 1. Both histology and depth of myometrium invasion are important in risk stratification into low, intermediate and high risk disease, which translates to the likelihood of lymph node metastasis and the presence of disease outside the pelvis. Disease extension is best assessed using pelvic gynecological MRI4,5. Lymph node staging defines recurrence risk and helps clinicians decide on adjuvant treatment. Surgical staging has been the standard modality for evaluation of metastatic disease, including total hysterectomy (TH) and bilateral salpingo-oophorectomy (BSO) and complete pelvic and para-aortic lymphadenectomy when indicated1,3,4. However, randomized trials did not demonstrate a survival benefit among patients with early stage EC undergoing systematic lymphadenectomy, which can be associated with significant morbidity (nerve and vascular injuries and higher incidence of postoperative lymphedema and lymphocyst formation) and longer operative time. 6,7;8-11 Sentinel lymph node (SLN) biopsy alone in EC has emerged as an alternative to systematic lymphadenectomy1,3,4;11-16. The SLN should be defined as the juxtauterine lymph node with an afferent lymph vessel in each lymphatic pathway13;17. Indocyanin green (ICG) enables the identification of both lymph vessels and lymph nodes and is associated with the highest bilateral SLN detection rate, compared to others4,11,13,15,18,19. Sentinel lymph node biopsy also allows for detection of low volume metastatic disease, in a process known as ultrastaging, thus further increasing the detection rate of metastatic disease4,17,20.

Methods

A prospective cohort study to assess the feasibility of SLN mapping in endometrial cancer using ICG and near-infrared (NIR) fluorescence is ongoing at the au-thors’ Gynaecological Oncology Unit. To our knowledge, this is the first published series of SLN mapping with ICG in endometrial cancer a Portuguese hospital. Data from all women with primary endometrial cancer considered eligible for comprehensive surgical staging by the gynecological oncology multidisciplinary team were collected and the disease managed according to the Portuguese Guidelines on Gynecological Cancer reviewed in 2020. All hystologic types of EC and cases of endometrial intraepithelial neoplasia were included. Previous studies demonstrated that the use of a SLN algorithm does not compromise the detection of stage IIIC disease in patients with deeply invasive endometrioid endometrial cancer, or in patients with serous and clear cell cancers, though the populations in which SLN biopsy are appropriate are still debatable21-25. In up to 35% of cases the final histopathological analysis reveals a higher grade lesion than the initial biopsy1. Women with synchronous cancer, gross metastatic disease, past history of allergy to intravenous or dye contrast and renal or hepatic insufficiency were excluded. Routine preoperative work-up included a pelvic MRI, a chest x-ray and computed tomography (CT) scan for low-risk and high-risk tumors, respectively, and blood tests including CA125 levels. Following a diagnostic laparoscopy and prior to exposure of the retroperitoneum, 1mL of ICG was injected into the superficial and deep (1cm) cervical stroma at 3 and 9 o’clock and clemastine was administered. One vial of 25 mg ICG solution was previously diluted in 10 ml of sterile water26,27. The fluorescent signal was then identified under NIR mode, and the SLNs excised and sent to frozen section analysis28. After SLN mapping patients underwent laparoscopic sentinel lymph node biopsy, total laparoscopic hysterectomy (TLH) and bilateral salpingo-oophorectomy. Laparoscopic pelvic lymphadenectomy (PLND) and/or pelvic and para-aortic lym-phadenectomy (PPALND) were performed based on identification of tumor risk factors and clinical judgment.

Patients with grade 1 lesions confined to the inner half of the myometrium underwent a SLN biopsy only. Ultrastaging of sentinel lymph nodes was performed in all cases, according to the protocol established with Anatomic Pathology, which included assessment of serial sections distancing 250 µ from each other and immunostaining for pankeratin AE1/AE3. In laparoscopy, the devices used were spies from Karl Storz® (Karl Storz Endoskope GmbH & Co. KG, Tuttlingen).

Ethical Approval

The study was approved by the Institution’s Ethics Committee (Centro Hospitalar Tondela-Viseu, 26th November, 2020). All patients gave their written informed consent.

Statistics

Variables derived from women’s medical records included demographic variables, body mass index, past medical and surgical history (including obstetric index and gynecological history), tumor histology, grade and stage (FIGO), imaging, surgical procedures performed, time interval from dye injection to SLN removal, total operative time, estimated blood loss, duration of hospitalization, location and number of SLNs removed, number of all nodes ressected and surgical morbidity. A minimum number of 20 patients recruited until publication was established. The authors conducted a descriptive analysis of the clinicopathologic characteristics and discuss the process of implementing a novel technique, sharing the teams’ understanding of SLN mapping with ICG and NIR fluorescence in endometrial cancer.

Results

Twenty patients (n=20) were included in the study. Clinicopathologic characteristics are summarized in Table I.

Table I Clinicopathologic characteristics. 

A TLH/BSO and SLN biopsy alone was performed on 12 patients (60.0%). Out of the remaining 8 patients, half underwent a PLND and the other half a PPALND. Of the patients undergoing a full retroperitoneal staging, 3 also underwent an omentectomy and 1 an omentectomy plus resection of a mesosigmoid implant. Cervical reinjection of indocyanin green was performed in 4 cases. The median operative time was 240 minutes, the median time interval from dye injection to SLN removal was approximately 40 minutes per side, and the estimated blood loss was 150 ml.

Median operative time was lower among patients undergoing an SLN mapping only compared with patients undergoing a full lymphadenectomy (219 min vs 280 min).

One intraoperative complication occurred in a stage IIIC1 patient undergoing full retroperitoneal staging, omentectomy plus resection of a mesosigmoid implant and laparotomic left ureteric reimplantation after iatrogenic injury. No cases of ICG injection-related complications occurred.

The median number of SLNs removed per patient was 2.3 (range 1-3) and at least one SLN was detected in all patients (20/20). Sentinel lymph nodes were detected bilaterally in 14 patients (14/20; 70%) and unilaterally in 6 patients (6/20; 30%). Data on SLN mapping are summarized in Table II.

Table II Sentinel lymph nodes mapping. 

Every patient in whom at least one SLN could be identified mapped in the pelvis. In one case, 1 additional SLN was identified in the presacral region. The distribution of the SLNs is demonstrated in Figure 1.

Figure 1 Distribution of the sentinel lymph nodes. Obturador fossa 14.7%, external iliac artery 73.5%, common iliac artery 8.8%, aortic bifurcation 3.0%. 

Figure 2 illustrates the appearance of the resected lymph node a) in conventional white light imaging and b) when fluorescence imaging is activated.

Figure 2 Appearance of resected right pelvic sentinel lymph node in a) conventional imaging, and b) fluorescence imaging. 

Out of the 20 patients, 2 (2/20; 10%) had lymph node macrometastases. No isolated tumor cells or micrometasteses were detected on ultrastaging. One patient with serous EC presented with a conglomerate nodal mass on the left hemipelvis and a positive contralateral SLN on frozen section analysis. However, migration of ICG onto the left hemipelvis was not detected. The second patient presented with invasion of the outer half of the myometrium and 1 negative SLN was detected on the left hemipelvis. However, no SLN was identified in the right hemipelvis. On the final histopathological exam, metastases were detected in 1 out of 6 pelvic right nodes.

Out of 4 patients diagnosed with endometrial intraepithelial neoplasia on biopsy, all were diagnosed with endometrial cancer on histopathological analysis of the uterus.

Discussion

In this prospective analysis, 20 women with endometrial cancer underwent laparoscopic SLN biopsy with ICG and NIR fluorescence and comprehensive surgical staging, as part of the ongoing clinical trial at the authors’ Department on SLN biopsy in EC. In the literature, there are few and limited series using ICG as a tracer for SLN biopsy in EC. Recently, a multicenter study by Moloney K. et al. on surgical competency assessment tools for SLN dissection by minimally invasive surgery on endometrial cancer was published. There is still lack of consensus about ICG concentration and volume injected.

A lower concentration of ICG was used as injection with a lower concentration and a larger volume (< 5 mg/ml, injected volume ≥ 2ml vs ≥ 5 mg/ml, injected volume < 2 ml) provide better performance of the SLN mapping27. The use of cervical injection, in 2 quadrants (3 and 9 o’clock) or 4 quadrants (3, 6, 9 and 12 o’clock) seems to have similar detection rates27;29-31.

Of all 20 patients, 12 (12/20; 60.0%) had a TLH/BSO and SLN biopsy alone and the remaining 8 patients underwent full retroperitoneal staging (8/20; 40.0%). The median number of SLNs removed per patient was 2.3, similarly to the number reported in previous studies26. At least 1 SLN was detected in all patients (20/20; 100%), bilaterally in 14 patients (14/20; 70%) and unilaterally in 6 patients (6/20; 30%). In 1 case, a third SLN was identified amongst presacral nodes. No isolated para-aortic SLNs were detected. Persson et. all’s studies on uterine lymphatic anatomy confirmed the existence of two consistent lymphatic pathways with pelvic SLNs on women with EC: an upper paracervical pathway, with draining medial external and/or obturator lymph nodes, where 95% of SLN are to be found, and a lower paracervical pathway with draining internal iliac and/or presacral lymph nodes, where the remaining 5% of SLNs will be detected. Ipsilateral ICG reinjection in case of non-display of any lymphatic pathway and keeping lymphatic pathways intact by opening avascular planes might enhance the technical success rate.

Lymphatic drainage may be impeded by cancer cells emboli, though there is still insufficient evidence of that phenomenon in gynecological cancers (endometrial or cervical). Other factors potentially influencing lymphatic drainage are history of endometriosis or prior abdominal/pelvic surgery, pelvic irradiation and obesity32.

Our preliminary data do not allow for assessment of overall and bilateral detection rates, sensitivity or negative predictive value. In a meta-analysis by Rocha, A. et all on SLN staging, sensitivity of ICG sentinel mapping ranged from 50% to 100%. In the FIRES multicentre, prospective, cohort study of 385 patients with early stage EC of all histologies and grades, 86% of patients had successful mapping of at least one SLN, and the sensitivity to detect node-positive disease reported was 97.2% and negative predictive value 99.6%29,30. The median operative time was 240 minutes, with total SLN mapping time of approximately 40 minutes per side. These results are compatible with time intervals previously reported26,27. Median operative time was lower among patients undergoing an SLN mapping only, compared with patients undergoing a full lymphadenectomy (219 min vs 280 min). No cases of ICG injection-related complications occurred.

In 12 cases (12/20, 60%), women were submitted to adjuvant treatment, mainly external pelvic radiotherapy. In 1 case, the patients’ functional status contraindicated chemotherapy and death from disease progression occurred 6 months after surgery. In all the other 19 cases the disease has not recurred after 3 years of follow-up.

Recently published ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma state that SLN biopsy is an acceptable alternative to systematic lymphadenectomy for lymph node staging in stage I/II EC, with cervical injection of ICG being the preferred method4. National guidelines recommend the use of SLN biopsy for investigation purposes only1. Owing to the paucity of data, the decision to dissect paraaortic lymph nodes likely depends on assessment of the uterine pathology, patient comorbidities and performance status and on the presence of gross pelvic nodes, as the incidence of isolated positive paraaortic lymph nodes is very low33-35. The question remains as to whether there is a therapeutic value to completion paraaortic lymphadenectomy in the setting of pelvic nodal metastasis. Additionally, the role of completion pelvic and/or paraaortic lymphadenectomy in the setting of SLN metastasis is unknown.

Conclusions

This is the first published series of laparoscopic sentinel lymph node biopsy using ICG and NIR fluorescence in endometrial cancer, conducted in a Portuguese hospital. The Society of Gynecologic Oncology’s (SGO) and SLN Working Group consensus on SLN mapping and staging in endometrial cancer recommended completing at least 20 SLN procedures with subsequent lymphadenectomy prior to adopting an SLN algorithm, due to the absence of precise learning curve in the endometrial cancer. The clinical trial is ongoing at the authors’ Department and we hope to obtain new and improved results on this novel but promising technique.

Author contribution statement

The authors confirm contribution to the paper as follows: study conception and design: Sara Sales, Ângela Melo, Sónia Gonçalves, Nuno Nogueira Martins, Francisco Nogueira Martins; data collection: Sara Sales, Nuno Nogueira Martins; analysis and interpretation of results: Sara Sales, Ângela Melo, Sónia Gonçalves, Nuno Nogueira Martins, Francisco Nogueira Martins; draft manuscript preparation: Sara Sales, Nuno Nogueira Martins. All authors reviewed the results and approved the final version of the manuscript.

Conflict of Interest

The authors have no conflicts of interest to declare.

References

1. SPG Consenso Nacional Cancro Ginecológico 2020. https://spginecologia.pt/wp-content/uploads/2021/07/spg-consenso-nacional-cancro-ginecologico-2020.pdf (accessed on November 10, 2021). [ Links ]

2. Global Cancer Observatory (Globocan). Published 2018. https://gco.iarc.fr/. [ Links ]

3. NCCN Clinical Practice Guidelines in Oncology. Uterine Neoplasms. http://www.nccn.org/professionals/physician_gls/pdf/uterine.pdf (Accessed on November, 2021). [ Links ]

4. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. https://ijgc.bmj.com/content/31/1/12.long (Accessed on November, 2021) [ Links ]

5. Beddy P, Moyle P, Kataoka M, et al. Evaluation of depth of myometrial invasion and overall staging in endometrial cancer: comparison of diffusion-weighted and dynamic contrast-enhanced MR imaging. Radiology 2012;262:530. [ Links ]

6. ASTEC study group, Kitchener H, Swart AM, et al. Efficacy of systematic pelvic lymphadenectomy in endometrial cancer (MRC ASTEC trial): a randomised study. Lancet 2009; 373:125. [ Links ]

7. Benedetti Panici P, Basile S, Maneschi F, et al. Systematic pelvic lymphadenectomy vs. no lymphadenectomy in early-stage endometrial carcinoma: randomized clinical trial. J Natl Cancer Inst 2008;100:1707. [ Links ]

8. Abu-Rustum NR, Alektiar K, Iasonos A, et al. The incidence of symptomatic lower-extremity lymphedema following treatment of uterine corpus malignancies: a 12-year experience at Memorial Sloan-Kettering Cancer Center. Gynecol Oncol 2006;103:714. [ Links ]

9. Carlson JW, Kauderer J, Hutson A, et al. GOG 244-The lymphedema and gynecologic cancer (LEG) study: Incidence and risk factors in newly diagnosed patients. Gynecol Oncol 2020;156:467. [ Links ]

10. Todo Y, Yamamoto R, Minobe S, et al. Risk factors for postoperative lower-extremity lymphedema in endometrial cancer survivors who had treatment including lymphadenectomy. Gynecol Oncol 2010;119:60. [ Links ]

11. Nagar H, Wietek N, Goodall RJ, et al. Sentinel node biopsy for diagnosis of lymph node involvement in endometrial cancer. Cochrane Database Syst Rev 2021;6:CD013021. [ Links ]

12. Rossi EC, Kowalski LD, Scalici J, et al. A comparison of sentinel lymph node biopsy to lymphadenectomy for endometrial cancer staging (FIRES trial): a multicentre, prospective, cohort study. Lancet Oncol 2017;18:384. [ Links ]

13. Royal College of Obstetricians and Gynecologists. Sentinel Lymph Node Biopsy in Endometrial Cancer. RCOG Scientific Impact Paper No.51. July, 2016. https://www.rcog.org.uk/en/guidelines-research-services/guidelines/sip51/Links ]

14. Tanner E, Puechl A, Levinson K, et al. Use of a novel sentinel lymph node mapping algorithm reduces the need for pelvic lymphadenectomy in low-grade endometrial cancer. Gynecol Oncol 2017;147:535. [ Links ]

15. Holloway RW, Abu-Rustum NR, Backes FJ, et al. Sentinel lymph node mapping and staging in endometrial cancer: A Society of Gynecologic Oncology literature review with consensus recommendations. Gynecol Oncol 2017;146:405. [ Links ]

16. Cormier B, Rozenholc AT, Gotlieb W, et al. Sentinel lymph node procedure in endometrial cancer: A systematic review and proposal for standardization of future research. Gynecol Oncol 2015; 138:478. [ Links ]

17. Persson J, Geppert B, Lönnerfors C , Bollino M, Måsbäck. Description of a reproducible anatomically based surgical algorithm for detection of pelvic sentinel lymph nodes in endometrial cancer. Gynecol Oncol 2017;147(1):120-125. [ Links ]

18. Rossi EC, Ivanova A, Boggess JF. Robotically assisted fluorescence-guided lymph node mapping with ICG for gynecologic malignancies: a feasibility study. Gynecol Oncol 2012;124:78. [ Links ]

19. Zapardiel I, Alvarez J, Barahona M, et al. Utility of Intraoperative Fluorescence Imaging in Gynecologic Surgery: Systematic Review and Consensus Statement. Ann Surg Oncol 2021;28:3266. [ Links ]

20. Kim CH, Soslow RA, Park KJ, et al. Pathologic ultrastaging improves micrometastasis detection in sentinel lymph nodes during endometrial cancer staging. Int J Gynecol Cancer 2013;23:964. [ Links ]

21. Baiocchi G, Mantoan H, Kumagai LY, et al. The Impact of Sentinel Node-Mapping in Staging High-Risk Endometrial Cancer. Ann Surg Oncol 2017;24:3981. [ Links ]

22. Ducie JA, Eriksson AGZ, Ali N, et al. Comparison of a sentinel lymph node mapping algorithm and comprehensive lymphadenectomy in the detection of stage IIIC endometrial carcinoma at higher risk for nodal disease. Gynecol Oncol 2017;147:541. [ Links ]

23. Cusimano MC, Vicus D, Pulman K, et al. Assessment of Sentinel Lymph Node Biopsy vs Lymphadenectomy for Intermediate- and High-Grade Endometrial Cancer Staging. JAMA Surg 2021; 156:157. [ Links ]

24. Jan Persson J, Salehi S, Bollino M, Lönnerfors C et al. Pelvic Sentinel lymph node detection in High-Risk Endometrial Cancer (SHREC-trial) the final step towards a paradigm shift in surgical staging. European Journal of Cancer, 116 (2019) 77e85. [ Links ]

25. Nasioudis D, Byrne M, Ko EM, et al. The impact of sentinel lymph node sampling versus traditional lymphadenectomy on the survival of patients with stage IIIC endometrial cancer. Int J Gynecol Cancer 2021;31:840-845. [ Links ]

26. Papadia a, MD, Imboden S, Siegenthaler F et al. Laparoscopic Indocyanine Green Sentinel Lymph Node Mapping in Endometrial Cancer. Ann Surg Oncol 2016; 23:2206-2211. [ Links ]

27. Rocha A, Domínguez A, Lécuru F, Bourdel N. Indocyanine green and infrared fluorescence in detection of sentinel lymph nodes in endometrial and cervical cancer staging - a systematic review. Eur J Obstet Gynecol Reprod Biol 2016 Nov; 206:213-219. [ Links ]

28. Renz M, Marjon N, Devereaux K, et al. Immediate intraoperative sentinel lymph node analysis by frozen section is predictive of lymph node metastasis in endometrial cancer. Journal of Robotic Surgery 2018. [ Links ]

29. Ballester M, Dubernard G, Lécuru F, et al. Detection rate and diagnostic accuracy of sentinel-node biopsy in early stage endometrial cancer: a prospective multicentre study (SENTI-ENDO). Lancet Oncol 2011;12:469. [ Links ]

30. Xiong L, Gazyakan E, Yang W, et al. Indocyanine green fluorescence-guided sentinel node biopsy: A meta-analysis on detection rate and diagnostic performance. Journal of Cancer Society 2014; 1e7. [ Links ]

31. Gezer S, Duman Öztürk S, Hekimsoy T, et al. Cervical versus endometrial injection for sentinel lymph node detection in endometrial cancer: a randomized clinical trial. Int J Gynecol Cancer 2020;30:325. [ Links ]

32. Tanner EJ, Sinno AK, Stone RL, et al. Factors associated with successful bilateral sentinel lymph node mapping in endometrial cancer. Gynecol Oncol 2015;138:542. [ Links ]

33. Kumar S, Mariani A, Bakkum-Gamez JN, et al. Risk factors that mitigate the role of paraaortic lymphadenectomy in uterine endometrioid cancer. Gynecol Oncol 2013;130:441. [ Links ]

34. Bogani G, Dowdy S, Cliby W et al. Role of pelvic and para-aortic lymphadenectomy in endometrial cancer: Current evidence. J Obstet Gynaecol Res. 2014; 40(2):301-311. [ Links ]

35. Abu-Rustum N, Gomez J, Alektiar K et al. The incidence of isolated paraaortic nodal metastasis in surgically staged endometrial cancer patients with negative pelvic lymph nodes. Gynecologic Oncology 2009; 236-238. [ Links ]

Received: November 25, 2021; Accepted: February 23, 2023

Correspondence to: Sara Sousa Sales E-mail: saramatilde.sousa@gmail.com

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License