ArticlePDF AvailableLiterature Review

Fertility Preservation in Cervical Cancer—Treatment Strategies and Indications

Authors:

Abstract and Figures

Cervical cancer is frequently diagnosed in women during their reproductive years, and fertility preservation is an essential part of their cancer treatment. In highly selected patients with early stage, low-risk cervical cancer and a tumor size ≤ 2 cm, several treatment strategies can be offered for patients wishing to preserve fertility, including radical/simple trachelectomy or conization with pelvic lymph node assessment. Trachelectomy can be performed through a vaginal, abdominal, or minimally invasive approach and has been shown to have an equivalent oncologic outcome compared to radical hysterectomy. All surgical approaches for radical trachelectomy seem to have excellent survival with comparable oncologic outcomes. Nevertheless, patients undergoing vaginal trachelectomy have better obstetric outcomes compared to the other routes. In patients with larger tumors (2–4 cm), neoadjuvant chemotherapy followed by fertility-sparing surgery is an alternative option. Several chemotherapy regimens have been used for this indication, with a pathologic complete response rate of 17–73%. For locally advanced diseases that require radical hysterectomy or primary chemoradiation, fertility preservation can be performed using oocyte, embryo, or ovarian tissue cryopreservation, as well as ovarian transposition. For these patients, future pregnancy is possible through surrogacy. In addition to fertility preservation, ovarian transposition, where the ovaries are repositioned outside of the radiation field, is performed to maintain ovarian hormonal function and prevent premature ovarian failure. In summary, fertility-preservation treatment strategies for patients with early stage cervical cancer are continuously evolving, and less radical surgeries are becoming more acceptable. Additional and ongoing evidence is helping determine the impact of conservative procedures on oncologic and obstetric outcomes in these patients.
Content may be subject to copyright.
Citation: Salman, L.; Covens, A.
Fertility Preservation in Cervical
Cancer—Treatment Strategies and
Indications. Curr. Oncol. 2024,31,
296–306. https://doi.org/
10.3390/curroncol31010019
Received: 26 November 2023
Revised: 28 December 2023
Accepted: 2 January 2024
Published: 4 January 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Review
Fertility Preservation in Cervical Cancer—Treatment Strategies
and Indications
Lina Salman 1and Allan Covens 1, 2, *
1Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, University of Toronto,
Toronto, ON M5G 2M9, Canada; lina.salman@uhn.ca
2Division of Gynecologic Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre,
Toronto, ON M4N 3M5, Canada
*Correspondence: al.covens@sunnybrook.ca
Abstract: Cervical cancer is frequently diagnosed in women during their reproductive years, and
fertility preservation is an essential part of their cancer treatment. In highly selected patients with
early stage, low-risk cervical cancer and a tumor size
2 cm, several treatment strategies can be
offered for patients wishing to preserve fertility, including radical/simple trachelectomy or conization
with pelvic lymph node assessment. Trachelectomy can be performed through a vaginal, abdominal,
or minimally invasive approach and has been shown to have an equivalent oncologic outcome
compared to radical hysterectomy. All surgical approaches for radical trachelectomy seem to have
excellent survival with comparable oncologic outcomes. Nevertheless, patients undergoing vaginal
trachelectomy have better obstetric outcomes compared to the other routes. In patients with larger
tumors (2–4 cm), neoadjuvant chemotherapy followed by fertility-sparing surgery is an alternative
option. Several chemotherapy regimens have been used for this indication, with a pathologic complete
response rate of 17–73%. For locally advanced diseases that require radical hysterectomy or primary
chemoradiation, fertility preservation can be performed using oocyte, embryo, or ovarian tissue
cryopreservation, as well as ovarian transposition. For these patients, future pregnancy is possible
through surrogacy. In addition to fertility preservation, ovarian transposition, where the ovaries are
repositioned outside of the radiation field, is performed to maintain ovarian hormonal function and
prevent premature ovarian failure. In summary, fertility-preservation treatment strategies for patients
with early stage cervical cancer are continuously evolving, and less radical surgeries are becoming
more acceptable. Additional and ongoing evidence is helping determine the impact of conservative
procedures on oncologic and obstetric outcomes in these patients.
Keywords: cervical cancer; fertility preservation; trachelectomy; ovarian transposition
1. Introduction
Cervical cancer is the fourth most common cancer in women worldwide [1].
It is frequently diagnosed in women aged 35–44, and it is the second leading cause of
cancer death in women aged 20–39 [
2
]. Treatment of cervical cancer is based on the stage of
the disease [
3
]. The standard treatment for patients with early stage disease (stage IA2-IB1)
is/has been radical hysterectomy with pelvic lymph node dissection, while patients with
locally advanced and metastatic disease are treated with primary radiation therapy (RT)
+/
systemic treatment [
3
,
4
]. As 37% of patients with newly diagnosed cervical cancer are
under the age of 45 [
5
], fertility preservation treatment options are often desired. Surgical
treatment modalities include radical and simple trachelectomy as well as cervical conization.
In certain cases of locally advanced disease where uterine preservation is not an option,
fertility preservation can be maintained through assisted-reproduction technologies (ART)
and ovarian transposition, which also have the advantage of preserving ovarian hormonal
function [
6
8
]. In this review, we present fertility preservation treatment strategies for early
Curr. Oncol. 2024,31, 296–306. https://doi.org/10.3390/curroncol31010019 https://www.mdpi.com/journal/curroncol
Curr. Oncol. 2024,31 297
stage cervical cancer, including indications, oncologic, and obstetric outcomes. In addition,
we present evidence supporting conservative management of these patients.
2. Radical Trachelectomy with Lymph Node Assessment
Selected patients with early stage cervical cancer wishing to preserve fertility might
be eligible for trachelectomy [
9
]. Radical trachelectomy was first described by Dargent
in 1987 [
10
], and it consists of the removal of the cervix, vaginal margins, and parame-
tria. Historical indications for trachelectomy included stage IA1 with lymphovascular
space invasion (LVSI), stage IA2-IB1 (tumor size
2 cm), negative nodal metastasis, and
the absence of deep stromal invasion [
3
,
11
]. Excluding nodal disease prior to fertility
preservation treatment is crucial, as positive lymph nodes are a poor prognostic factor
and can determine the appropriate treatment [
12
]. The feasibility of sentinel lymph node
mapping led to a paradigm shift in lymph node assessment in cervical cancer [
3
]. Ac-
cording to the NCCN guidelines, sentinel lymph node biopsy can replace pelvic lymph
node dissection in FIGO 2018 stage IA1 with LVSI and stage IA2-IB1 [
3
,
13
], whereas in
the 2023 ESGO guidelines, pelvic lymph node dissection should be performed in stage
IB1 with negative sentinel nodes frozen [
4
]. Although data from the prospective SENTI-
COL I and SENTICOL II studies showed that omitting full pelvic lymphadenectomy for
patients with stage IA-IIA with bilateral negative sentinel lymph nodes does not seem to
be associated with an increased recurrence rate, evidence on the accuracy and oncologic
safety of sentinel lymph node biopsy in cervical cancer is still evolving, and additional
prospective data are needed [
14
]. Nica et al. evaluated the outcome of patients with early
stage cervical cancer (tumor size
2 cm) undergoing cervical conization with laparoscopic
sentinel lymph node assessment [
15
]. Of the 44 patients included in the analysis, 93%
had negative sentinel nodes and did not require further nodal procedures, while 6.8% of
patients had micrometastases detected in the sentinel nodes and underwent ipsilateral
lymphadenectomy. All the remaining non-sentinel lymph nodes were negative. After a
median follow-up of 44 months, no recurrences were documented. Another retrospective
study included 36 patients with stage IA2-IBI cervical cancer [
16
]. All patients underwent
FSS and laparoscopic sentinel lymph node dissections. Of those, 50% underwent sentinel
lymph node dissection alone, and 50% underwent sentinel lymph node followed by full
pelvic lymph node dissection. In total, there were four recurrences: two in the sentinel and
two in the pelvic lymphadenectomy groups. The results of these studies are promising,
but larger prospective studies are required to assess the accuracy of sentinel lymph node
dissection in early stage cervical cancer and to evaluate the safety of minimally invasive
surgeries for lymph node assessment.
Radical trachelectomy originally combined vaginal approaches. Since then, the proce-
dure has been modified and is currently performed via a vaginal, abdominal, or minimally
invasive approach, based on the surgeon’s preference and experience [
17
]. Several studies
evaluated the oncologic outcome of radical trachelectomy and found it to have comparable
survival and recurrence rates compared to radical hysterectomy [
18
20
], making it a safe
alternative for patients wishing to preserve fertility.
The question of whether one surgical approach is superior to the other has been eval-
uated in several retrospective studies. A systematic review by Smith et al. compared the
surgical, oncologic, and obstetric outcomes of radical trachelectomy according to the surgi-
cal route: vaginal, abdominal, and laparoscopic [
17
]. Out of 2566 patients included in the
analysis, 75% had stage IB1 (tumor size < 2 cm). The majority of patients underwent vaginal
radical trachelectomy (58%), 37% had abdominal procedures, and only 4.7% underwent
laparoscopic trachelectomy. The vaginal approach had a shorter median operative time
compared to the abdominal and laparoscopic routes, and had lower rates of positive mar-
gins. The post-operative pregnancy rate was found to be highest in the vaginal approach
(38%) compared to abdominal and laparoscopy (10% and 9%, respectively), in addition
to lower rates of preterm delivery. It should be noted that the group undergoing vaginal
trachelectomy had a longer follow-up time, which might explain the higher pregnancies
Curr. Oncol. 2024,31 298
reported. While this review included a large number of patients, it is based on published
retrospective data; therefore, selection and publication biases should be taken into account
when interpreting these findings.
Since the publication of the Laparoscopic Approach to Cervical Cancer (LACC)
trial [
21
], the safety of minimally invasive surgery (MIS) for radical trachelectomy has
raised some concerns. The LACC trial was a randomized controlled trial that evaluated
disease-free survival in patients undergoing MIS versus open radical hysterectomy for
early stage cervical cancer. The results demonstrated increased recurrence and death rates
in patients undergoing radical hysterectomy via the MIS approach in patients with stage
IB2, as these comprised the majority of the study cohort. Prospective data on MIS versus
open surgeries for stage IA2/IB1 cervical cancer are limited, as is the safety of MIS for
lymph node assessment in these patients. A retrospective study using the National Cancer
Database evaluated the trends, characteristics, and survival outcomes of patients with stage
IA2-IB cervical cancer undergoing radical trachelectomy via MIS versus laparotomy [
22
].
Of the 246 people included in the study, 144 underwent surgery via the MIS approach.
Patients undergoing vaginal trachelectomy were excluded. The authors found a significant
increase in using the MIS approach throughout the years (increasing from 29% in 2010
to 75% in 2015). Death events were 7.6% in the laparotomy group compared to 3.5% in
the MIS (p= 0.025). The absolute number of events in the study was very low, and the
authors conclude that although no survival difference was found, the effect of MIS radical
trachelectomy on survival remains unknown.
A further comparison of MIS versus open radical trachelectomy was published by
the International Radical Trachelectomy Assessment (IRTA) [
23
]. In this retrospective
study,
646 patients
were included in the analysis. 358 underwent open surgery, and
288 underwent
MIS. At 4.5 years, 4.8% had a recurrence in the open surgery and 6.3%
in the MIS, but this was not statistically significant. In addition, there was no difference in
overall survival between groups (99.2% in open surgery vs. 99.0% in MIS). As this is not a
common procedure, the feasibility of performing a randomized clinical trial to evaluate
survival outcomes in MIS versus open trachelectomy is unlikely, and clinical practice will
be based on the best available evidence.
Regardless of the surgical approach, the radicality of the procedure is continuously
being refined for more conservative surgeries. Based on tumor factors, patients may
undergo simple trachelectomy or cervical conization rather than radical trachelectomy with
similar oncologic outcomes [24].
3. Simple Trachelectomy and Cone Biopsy
The rationale for performing more conservative surgeries is supported by the re-
sults of several studies evaluating the risk of parametrial involvement in patients with
early stage cervical cancer who have favorable pathologic features (tumor
2 cm, depth
of
invasion 10 mm
, and negative pelvic nodes) [
25
,
26
]. In this well-defined cohort of
patients, the risk of parametrial involvement is lower than <1%, putting the benefit of
removing the parametria in doubt. While cervical conization is typically performed to
treat high-grade premalignant lesions of the cervix [
27
], the above evidence led to studies
evaluating the performance of cone biopsy as a treatment for cervical cancer. The ConCerv
trial was the first prospective study to evaluate the feasibility and oncologic outcomes of
conization alone or simple hysterectomy in early stage low-risk cervical cancer [
28
]. In this
study, they included patients with FIGO 2009 stage IA2-IB1 cervical cancer [
29
] who meet
the following criteria: squamous cell or adenocarcinoma, tumor size
2 cm, no LVSI, depth
of invasion
10 mm, negative imaging for metastatic disease, and negative conization
margins. Patients were allowed to undergo a repeated cone if the first one had positive
margins. Patients desiring fertility underwent a second conization with pelvic lymph node
assessment, and those not desiring fertility preservation underwent a simple hysterectomy
with pelvic lymph node assessment. In total, 44 patients were enrolled in the fertility
preservation arm. Of those, two patients had positive lymph nodes, and one patient had
Curr. Oncol. 2024,31 299
recurrent disease. The one patient with recurrent disease had a stromal invasion of 13 mm
on the first conization with positive margins. Repeated conization was negative for cancer,
but margins were positive for high-grade dysplasia. This had led the investigators to
amend the protocol and exclude patients with positive margins not only for invasive cancer
but also for intra-epithelial neoplasia. Data from Bogani et al. on 32 patients with FIGO
2018 stage IA2, IB1, and IB2 undergoing conization with pelvic lymph node assessment
showed 5-year disease-free survival and overall survival to be 94% and 97%, respectively.
Another published study by Plante et al. evaluated the obstetric and oncologic outcomes
of simple vaginal trachelectomy/conization in patients with low-risk, early stage cervical
cancer. The 5-year progression-free survival and overall survival were 97.9% and 97.6%,
respectively [30].
A systematic review looking into obstetrics and oncologic outcomes following fertility
preservation treatment for early cervical cancer included 347 cases that underwent coniza-
tion [
31
]. In this group, the recurrence rate was 0.4%, and the pregnancy rate was 36.1%
with no death events. A more recent systematic review published by Nezhat et al. evaluated
reproductive and oncologic outcomes after fertility-sparing surgery (FSS) for stage IA1-IB1
cervical outcomes [
32
]. They included patients who underwent conization/simple trach-
electomy, or radical trachelectomy via different surgical approaches. Of the 3044 patients
included, the pregnancy rate was 55.4% in patients attempting to conceive, with the highest
clinical pregnancy rate after vaginal trachelectomy (67.5%). After a median follow-up of
39.7 months, the mean cancer recurrence rate was 3.2%, and the cancer death rate was
0.6%. These data highlight the excellent oncologic outcome and safety of performing these
procedures in these patients.
Another aspect that should be considered when choosing a treatment modality is the
impact of such treatment on quality of life. The Gynecologic Oncology Group (GOG)-0278
is a phase I/II study evaluating physical function and quality of life in patients with cervical
cancer stage IA1 with LVSI and IA2-IB1 (
2 cm) who underwent simple hysterectomy or
cone biopsy with pelvic lymphadenectomy (ClinicalTrials.gov/NCT01649089). This study
will look into urinary, gastrointestinal, and sexual function following non-radical surgery.
The study completed accrual, and results are anticipated to be presented in early 2024.
4. Oncologic Safety of Fertility-Preservation Surgeries in Different Histologic Types
Squamous cell carcinoma (SCC) is the most common histological type of cervical cancer,
followed by adenocarcinoma, which accounts for approximately 25% of all cases [
33
]. The
incidence of adenocarcinoma in developed countries has increased in the last decade,
whereas the incidence of SCC has been decreasing [
34
]. Studies on patients with early stage
cervical cancer undergoing definitive treatment showed conflicting results regarding the
oncologic outcome of adenocarcinoma compared to SCC [
35
37
]. Concerns were raised
regarding the oncologic safety of FSS in patients with adenocarcinoma or adenosquamous.
Zusterzeel et al. looked at recurrence risk following radical vaginal trachelectomy in early
stage cervical cancer [
38
]. In this retrospective study of 132 patients, 72% had SCC, 24.2%
had adenocarcinoma, and 3.8% had adenosquamous. The majority of patients in this study
had FIGO 2009 stage IB1 disease. The overall recurrence rate was 6.8%, with a median
time to recurrence of 21 months. Adenosquamous carcinoma had the highest recurrence
rate of 20%, followed by 12.5% for adenocarcinoma and 4.2% for SCC. Additional studies
failed to show a difference in oncologic outcome in adenocarcinoma compared to SCC for
patients undergoing radical trachelectomy [
39
,
40
]. A recently published multicenter study
evaluated risk factors for recurrence in patients with early stage cervical cancer treated
with FSS [
41
]. They included 733 patients who underwent any type of FSS. The majority
of patients (70%) had SCC, and 24% had adenocarcinoma. A total of 49% of patients had
FIGO 2018 stage IB1 disease. After a median follow-up of 72 months, histologic subtype
was not found to be a risk factor for recurrence. In fact, the only significant risk factor was
tumor size >2 cm.
Curr. Oncol. 2024,31 300
While some histological types have a worse prognosis than SCC, it is not evident
that FSS increases the risk of recurrence. Although additional adjuvant therapy might
be indicated based on pathology to decrease the risk of recurrence, FSS should not be a
contraindication to its administration.
5. Neoadjuvant Chemotherapy
Neoadjuvant chemotherapy (NACT) is an alternative option for patients with bulky
cervical cancer (tumor size > 2 cm) wishing to preserve fertility. The rationale for administer-
ing chemotherapy is to shrink the tumor and make an FSS feasible. Different chemotherapy
regimens were studied in small case series, including paclitaxel/cisplatin/ifosphamide
(TIP) [
42
,
43
], paclitaxel/cisplatin [
44
], and carboplatin/paclitaxel [
45
], with varying num-
bers of cycles (Table 1).
Table 1. Summary of several studies evaluating neoadjuvant chemotherapy followed by fertility-
sparing surgery for cervical cancer.
Study # of Patients Stage of Disease aChemotherapy Regimen # of Chemo
Cycles
Complete
Pathologic
Response to NACT
FSS after NACT
Maneo et al., 2008
[43]21 IB1 (tumor size
< 3 cm)
Cisplatin 75 mg/m2, paclitaxel
175 mg/m2, and ifosfamide
5 g/m2
3 24% PLND + conization
Vercellino et al.,
2012 [46]18 IB1-IB2 (tumor size
2–5 cm)
Ifosphamide 5 g/m2, cisplatin
100 mg/m2, and paclitaxel
200 mg/m2
2–3 17% PLND + RVT
Lanowska et al.,
2014 [47]18 IB1-IB2 (tumor size
2–5 cm)
Ifosphamide 5 g/m2, cisplatin
100 mg/m2, and paclitaxel
200 mg/m2
2–3 50% PLND + RVT
Robova et al., 2014
[48]28 IB1-IB2 (tumor size
1.5–4 cm)
1.Cisplatin 75 mg/m2and
ifosfamide (2 g/m2)
OR
2. Cisplatin (75 mg/m2) and
doxorubicine (35 mg/m2)
3 21% PLND + SVT
Salihi et al., 2015
[49]11 IB1-IB2 (tumor size
1.2–5.2 cm)
1.Paclitaxel 90 mg/m2and
carboplatin AUC 4
OR
2. Ifosphamide 5 g/m2, cisplatin
75 mg/m2, and paclitaxel
175 mg/m2
OR
3. Dose-dense paclitaxel
60 mg/m2
and carboplatin AUC 2
3–9 73% Conization
Tesfai et al., 2020
[45]19 IB1-IIA (tumor size
3.5–6 cm)
Weekly cisplatin 70 mg/m2and
paclitaxel 70 mg/m26 47% ART
Zusterzeel et al.,
2020 [50]18 IB2 b(tumor size
2.2–4 cm)
Weekly cisplatin 70 mg/m2and
paclitaxel 70 mg/m26 39% RVT
a
According to 2009 FIGO staging;
b
according to 2018 FIGO staging. #—Number; NACT—Neoadjuvant
Chemotherapy; FSS—Fertility-Sparing Surgery; PLND—Pelvic Lymph Node Dissection; RVT—Radical Vaginal
Trachelectomy; SVT—Simple Vaginal Trachelectomy; and ART—Abdominal Radical Trachelectomy.
The results of several case series investigating the oncologic and obstetric outcomes
of NACT followed by fertility preservation surgery in patients with tumor sizes 2–4 cm
have been summarized in a systematic review by Gwacham et al. [
51
]. All patients in-
cluded in this review (n = 114) had FIGO 2018 stage IB2 cervical cancer. The most common
chemotherapy regimen was TIP (89.5% of patients). Pelvic lymphadenectomy was per-
formed in 49% of patients prior to starting NACT, whereas 51% underwent NACT without
nodal assessment. FSS was performed on 99.1% of patients. The most common procedure
performed was radical vaginal trachelectomy (40.7%). The response to treatment was high,
with a complete pathologic response reported to be 39.5% and a partial response of 45.6%.
As for obstetric outcomes, 69.4% had full-term delivery, 9.7% had preterm delivery, and
16.1% had miscarriages. Although these data are obtained from small retrospective studies,
these findings are promising. As mentioned earlier, different chemotherapy regimens were
used, and it is not clear whether one regimen is superior to the other. In addition, there
Curr. Oncol. 2024,31 301
was no consistency between studies in the timing of performing lymph node dissection.
While some studies performed lymph node dissection after NACT, one would argue that it
should be performed prior to starting NACT, as positive nodes are associated with a poor
prognosis and these patients would require adjuvant treatment with chemotherapy and
radiation [52].
The outcome of NACT, followed by FSS, is currently being investigated by the CoN-
tESSA trial. This is a prospective multi-center trial evaluating NACT followed by FSS for
premenopausal patients with cervical cancer, FIGO 2018 stage IB2, wishing to preserve
fertility [
53
]. Participants are treated with NACT, consisting of platinum-based chemother-
apy (cisplatin or carboplatin) with paclitaxel. Those with a complete/partial response will
undergo fertility-sparing surgery. The primary end point of this study is to assess the rate
of functional uterus defined as successful fertility-sparing surgery and no adjuvant therapy.
The study is recruiting, and results are expected in 2025.
6. Ovarian Transposition
In patients with locally advanced cervical cancer, treatment includes external beam
radiation (EBRT) +/
brachytherapy +/
chemotherapy [
3
,
54
]. The standard dose used
in EBRT for cervical cancer is lethal to the ovaries and leads to ovarian failure [
55
]. In
premenopausal patients, this can result in a post-menopausal state with its associated symp-
toms and manifestations such as vasomotor symptoms, urogenital atrophy, osteoporosis,
and long-term cardiovascular complications [
56
,
57
]. In patients receiving EBRT, ovarian
transposition (OT) can be offered prior to treatment initiation to preserve ovarian function
in addition to fertility preservation. In this procedure, the ovaries are transposed laterally,
well above the pelvic brim, avoiding tension or torsion of the gonadal vessels [
58
,
59
]. When
OT is considered, it should be performed as soon as possible and with a minimally invasive
approach to enhance recovery, as a longer duration of time from diagnosis to treatment
negatively affects prognosis [
60
]. In a systemic review by Buonomo et al. [
61
], looking into
the outcomes of 1377 patients with cervical cancer undergoing OT followed by RT, it was
found that ovarian function was preserved in 61.7% (range 16.6–100%). Several factors
could explain the low rate of ovarian function preservation noted in that study: First, there
are surgical techniques, as the transposed ovaries might not be situated far enough from the
radiation borders, therefore exposing the ovaries to significant amounts of scatter radiation.
Second, the patient’s age at the time of the procedure plays an important factor. Although
this procedure is performed in premenopausal patients, the more advanced the patient’s
age, the higher the likelihood of ovarian failure [
62
]. Smaller doses of exposure to radiation
in these patients can lead to ovarian insufficiency as the effective sterilizing radiation dose
decreases with increasing age [63].
In addition to preserving hormonal function, ovarian transposition plays a role in
fertility preservation, and there have been reports of successful pregnancies following
this procedure [
6
]. In patients with preserved ovarian function, ovulation induction and
transabdominal oocyte retrieval can be performed, with successful pregnancies reported
through surrogacy [61,64].
7. Cryopreservation of Oocytes, Embryos and Ovarian Tissue
For young patients who are not eligible for any of the fertility preservation options
discussed above, it is important to refer them to fertility specialists to discuss other options
using ART. Fertility preservation can be performed through mature oocyte cryopreserva-
tion, embryo cryopreservation, or, in cases where chemotherapy cannot be delayed, ovarian
tissue cryopreservation (OTC) [
7
] (Table 2). Controlled ovarian hyperstimulation with
gonadotropins, followed by oocyte retrieval and cryopreservation of oocytes or embryos,
can be performed before initiating gonadotoxic treatments [
8
]. The stimulation can have a
“random start” regardless of the phase of the menstrual cycle, which facilitates the process
without impacting the quality or number of retrieved oocytes [
65
]. A study looking at
the long-term reproductive outcome of controlled ovarian stimulation in patients with
Curr. Oncol. 2024,31 302
gynecologic malignancies found that 17 out of 68 patients (25%) returned to the clinic
to claim their oocytes/embryos in a median time of 36 months. Out of this sample, the
successful livebirth rate was 58.8% [
66
]. While oocyte and embryo cryopreservation are
well-established techniques for fertility preservation, OTC is considered an innovative
technique [
67
]. In OTC, the cortex of harvested ovarian tissue is separated and cryopre-
served. Once gonadotoxic treatment is completed, the ovarian tissue can be thawed and
transplanted back to the patient to regain ovarian hormonal function and fertility [
68
].
The site of transplant can be on the remaining ovary, pelvic side walls, subcutaneously, or
intramuscularly [
67
]. In a systematic review and individual patient data meta-analysis of
ovarian tissue transplants, 87 studies and 735 women were included [
68
]. In this review,
most patients underwent ovarian transplant via laparoscopy, either to the remaining ovary
or pelvic side wall/peritoneum. Pooled rates for pregnancy were 37%, and the live birth
rate was 28%. The median time of graft function was 2.5 years (range 0.7–5 years). In the
cohort included in this study, it was not surprising but worth noting that no pregnancies
were achieved in patients with cervical cancer. This has been demonstrated in other studies
showing that patients with cervical cancer have a lower chance of pregnancy compared to
other types of cancer [69]. This is secondary to radiation-induced uterine fibrosis [70].
Table 2. Comparison of different artificial reproductive technologies used in fertility preservation for
cervical cancer.
Artificial Reproductive
Technology
Indication in Cervical
Cancer Advantages Reported Pregnancy
Rate
Reported Livebirth
Rate
Ovarian transposition
Prior to pelvic radiation
Preserves ovarian
hormonal function 75–89% [59] -
Oocyte and embryo
cryopreservation
Prior to systemic
chemotherapy
Well-established
technique with high
success rate
- 58.8% [66]
Ovarian tissue
cryopreservation
Prior to systemic
chemotherapy
Can be performed in
any patient irrespective
of age, without delays
in cancer treatment
37% [68] 28% [68]
While this seems to be a promising option for fertility preservation, there are some
concerns regarding this approach, mainly the risk of recurrence. Ovarian tissue from
cancer patients may have microscopic disease, and auto-transplantation of this tissue can
theoretically lead to cancer recurrence [
71
]. Although the harvested tissue is examined for
cancer cells, the strip of tissue examined is not used for cryopreservation, and one could
argue that ovarian tissue that is actually cryopreserved has cancer cells. More research is
required in this field, as there is no consensus regarding the size of ovarian tissue used for
auto-transplants or the ideal site of transplantation [68].
8. Summary
Several fertility preservation modalities are available for patients with early stage cer-
vical cancer. Radical trachelectomy has an excellent survival outcome with low recurrence
rates when compared to radical hysterectomy, regardless of the surgical approach. The low
risk of parametrial involvement in early stage disease justifies the performance of less radi-
cal procedures such as simple trachelectomy and cone biopsy for highly selected patients.
For patients with large tumors not eligible for primary surgery, NACT is associated
with a good response, making an FSS more feasible.
In cases where uterine preservation is not possible, fertility preservation can be per-
formed using cryopreservation of oocytes, embryos, and ovarian tissues, as well as ovar-
ian transposition. These methods enable patients to have future fertility through ART
and surrogacy.
Curr. Oncol. 2024,31 303
Great advances have been made in treating cervical cancer; however, evidence on
treatment options for fertility preservation should be interpreted with caution as it is extrap-
olated from retrospective and prospective single-arm studies. Since randomized clinical
trials are unlikely to be feasible due to the rarity of these procedures, multi-institutional
collaboration is needed to continuously evaluate patients’ outcomes, especially in an era of
less radicality.
Author Contributions: L.S.—conceptualization; methodology; resources; writing—original draft
preparation; writing—review and editing; and visualization. A.C.—conceptualization; methodology;
resources; writing—review and editing; visualization; and supervision. 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 conflicts of interest.
References
1.
Arbyn, M.; Weiderpass, E.; Bruni, L.; de Sanjose, S.; Saraiya, M.; Ferlay, J.; Bray, F. Estimates of incidence and mortality of cervical
cancer in 2018: A worldwide analysis. Lancet Glob. Health 2020,8, e191–e203. [CrossRef] [PubMed]
2. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023,73, 17–48. [CrossRef] [PubMed]
3.
Abu-Rustum, N.R.; Yashar, C.M.; Bean, S.; Bradley, K.; Campos, S.M.; Chon, H.S.; Chu, C.; Cohn, D.; Crispens, M.A.; Damast, S.;
et al. NCCN Guidelines Insights: Cervical Cancer, Version 1.2020. J. Natl. Compr. Cancer Netw. 2020,18, 660–666. [CrossRef]
[PubMed]
4.
Cibula, D.; Rosaria Raspollini, M.; Planchamp, F.; Centeno, C.; Chargari, C.; Felix, A.; Fischerova, D.; Jahnn-Kuch, D.; Joly, F.;
Kohler, C.; et al. ESGO/ESTRO/ESP Guidelines for the management of patients with cervical cancer—Update 2023. Radiother.
Oncol. 2023,184, 109682. [CrossRef]
5. Viale, P.H. The American Cancer Society’s Facts & es: 2020 Edition. J. Adv. Pract. Oncol. 2020,11, 135–136. [CrossRef]
6.
Terenziani, M.; Piva, L.; Meazza, C.; Gandola, L.; Cefalo, G.; Merola, M. Oophoropexy: A relevant role in preservation of ovarian
function after pelvic irradiation. Fertil. Steril. 2009,91, 935.e15–935.e16. [CrossRef]
7.
Donnez, J.; Dolmans, M.M.; Diaz, C.; Pellicer, A. Ovarian cortex transplantation: Time to move on from experimental studies to
open clinical application. Fertil. Steril. 2015,104, 1097–1098. [CrossRef]
8. Taylan, E.; Oktay, K. Fertility preservation in gynecologic cancers. Gynecol. Oncol. 2019,155, 522–529. [CrossRef]
9.
Zaccarini, F.; Sanson, C.; Maulard, A.; Scherier, S.; Leary, A.; Pautier, P.; Chargari, C.; Genestie, C.; Gouy, S.; Morice, P. Cervical
Cancer and Fertility-Sparing Treatment. J. Clin. Med. 2021,10, 4825. [CrossRef]
10.
Dargent, D.; Martin, X.; Sacchetoni, A.; Mathevet, P. Laparoscopic vaginal radical trachelectomy: A treatment to preserve the
fertility of cervical carcinoma patients. Cancer 2000,88, 1877–1882. [CrossRef]
11.
Machida, H.; Iwata, T.; Okugawa, K.; Matsuo, K.; Saito, T.; Tanaka, K.; Morishige, K.; Kobayashi, H.; Yoshino, K.; Tokunaga, H.;
et al. Fertility-sparing trachelectomy for early-stage cervical cancer: A proposal of an ideal candidate. Gynecol. Oncol. 2020,156,
341–348. [CrossRef] [PubMed]
12.
Ho, C.M.; Chien, T.Y.; Huang, S.H.; Wu, C.J.; Shih, B.Y.; Chang, S.C. Multivariate analysis of the prognostic factors and outcomes
in early cervical cancer patients undergoing radical hysterectomy. Gynecol. Oncol. 2004,93, 458–464. [CrossRef] [PubMed]
13.
Koh, W.J.; Abu-Rustum, N.R.; Bean, S.; Bradley, K.; Campos, S.M.; Cho, K.R.; Chon, H.S.; Chu, C.; Clark, R.; Cohn, D.; et al.
Cervical Cancer, Version 3.2019, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2019,17, 64–84.
[CrossRef] [PubMed]
14.
Balaya, V.; Guani, B.; Morice, P.; Querleu, D.; Fourchotte, V.; Leblanc, E.; Darai, E.; Baron, M.; Marret, H.; Leveque, J.; et al.
Long-term oncological safety of sentinel lymph node biopsy in early-stage cervical cancer: A post-hoc analysis of SENTICOL I
and SENTICOL II cohorts. Gynecol. Oncol. 2022,164, 53–61. [CrossRef] [PubMed]
15.
Nica, A.; Marchocki, Z.; Gien, L.T.; Kupets, R.; Vicus, D.; Covens, A. Cervical conization and lymph node assessment for early
stage low-risk cervical cancer. Int. J. Gynecol. Cancer 2021,31, 447–451. [CrossRef] [PubMed]
16.
Gil-Ibanez, B.; Glickman, A.; Del Pino, M.; Boada, D.; Fuste, P.; Diaz-Feijoo, B.; Pahisa, J.; Torne, A. Vaginal fertility-sparing
surgery and laparoscopic sentinel lymph node detection in early cervical cancer. Retrospective study with 15 years of follow-up.
Eur. J. Obstet. Gynecol. Reprod. Biol. 2020,251, 23–27. [CrossRef] [PubMed]
17.
Smith, E.S.; Moon, A.S.; O’Hanlon, R.; Leitao, M.M., Jr.; Sonoda, Y.; Abu-Rustum, N.R.; Mueller, J.J. Radical Trachelectomy for the
Treatment of Early-Stage Cervical Cancer: A Systematic Review. Obstet. Gynecol. 2020,136, 533–542. [CrossRef]
18.
Guo, J.; Zhang, Y.; Chen, X.; Sun, L.; Chen, K.; Sheng, X. Surgical and Oncologic Outcomes of Radical Abdominal Trachelectomy
Versus Hysterectomy for Stage IA2-IB1 Cervical Cancer. J. Minim. Invasive Gynecol. 2019,26, 484–491. [CrossRef]
19.
Prodromidou, A.; Iavazzo, C.; Fotiou, A.; Psomiadou, V.; Douligeris, A.; Vorgias, G.; Kalinoglou, N. Short- and long term
outcomes after abdominal radical trachelectomy versus radical hysterectomy for early stage cervical cancer: A systematic review
of the literature and meta-analysis. Arch. Gynecol. Obstet. 2019,300, 25–31. [CrossRef]
Curr. Oncol. 2024,31 304
20.
Beiner, M.E.; Hauspy, J.; Rosen, B.; Murphy, J.; Laframboise, S.; Nofech-Mozes, S.; Ismiil, N.; Rasty, G.; Khalifa, M.A.; Covens,
A. Radical vaginal trachelectomy vs. radical hysterectomy for small early stage cervical cancer: A matched case-control study.
Gynecol. Oncol. 2008,110, 168–171. [CrossRef]
21.
Ramirez, P.T.; Frumovitz, M.; Pareja, R.; Lopez, A.; Vieira, M.; Ribeiro, R.; Buda, A.; Yan, X.; Shuzhong, Y.; Chetty, N.; et al.
Minimally Invasive versus Abdominal Radical Hysterectomy for Cervical Cancer. N. Engl. J. Med. 2018,379, 1895–1904. [CrossRef]
[PubMed]
22.
Matsuo, K.; Chen, L.; Mandelbaum, R.S.; Melamed, A.; Roman, L.D.; Wright, J.D. Trachelectomy for reproductive-aged women
with early-stage cervical cancer: Minimally invasive surgery versus laparotomy. Am. J. Obstet. Gynecol. 2019,220, 469.e1–469.e13.
[CrossRef] [PubMed]
23.
Salvo, G.; Ramirez, P.T.; Leitao, M.M.; Cibula, D.; Wu, X.; Falconer, H.; Persson, J.; Perrotta, M.; Mosgaard, B.J.; Kucukmetin,
A.; et al. Open vs minimally invasive radical trachelectomy in early-stage cervical cancer: International Radical Trachelectomy
Assessment Study. Am. J. Obstet. Gynecol. 2022,226, 97.e1–97.e16. [CrossRef] [PubMed]
24.
Tseng, J.H.; Aloisi, A.; Sonoda, Y.; Gardner, G.J.; Zivanovic, O.; Abu-Rustum, N.R.; Leitao, M.M., Jr. Less versus more radical
surgery in stage IB1 cervical cancer: A population-based study of long-term survival. Gynecol. Oncol. 2018,150, 44–49. [CrossRef]
[PubMed]
25.
Covens, A.; Rosen, B.; Murphy, J.; Laframboise, S.; DePetrillo, A.D.; Lickrish, G.; Colgan, T.; Chapman, W.; Shaw, P. How
important is removal of the parametrium at surgery for carcinoma of the cervix? Gynecol. Oncol. 2002,84, 145–149. [CrossRef]
[PubMed]
26.
Stegeman, M.; Louwen, M.; van der Velden, J.; ten Kate, F.J.; den Bakker, M.A.; Burger, C.W.; Ansink, A.C. The incidence of
parametrial tumor involvement in select patients with early cervix cancer is too low to justify parametrectomy. Gynecol. Oncol.
2007,105, 475–480. [CrossRef]
27.
Mosseri, J.; Hocquemiller, R.; Mergui, J.L.; Uzan, C.; Canlorbe, G. Laser conization for cervical intraepithelial neoplasia:
Effectiveness and obstetric outcomes. J. Gynecol. Obstet. Hum. Reprod. 2022,51, 102341. [CrossRef] [PubMed]
28.
Schmeler, K.M.; Pareja, R.; Lopez Blanco, A.; Humberto Fregnani, J.; Lopes, A.; Perrotta, M.; Tsunoda, A.T.; Cantu-de-Leon, D.F.;
Ramondetta, L.M.; Manchana, T.; et al. ConCerv: A prospective trial of conservative surgery for low-risk early-stage cervical
cancer. Int. J. Gynecol. Cancer 2021,31, 1317–1325. [CrossRef]
29.
Pecorelli, S. Revised FIGO staging for carcinoma of the vulva, cervix, and endometrium. Int. J. Gynaecol. Obstet. 2009,105, 103–104.
[CrossRef]
30.
Plante, M.; Renaud, M.C.; Sebastianelli, A.; Gregoire, J. Simple vaginal trachelectomy in women with early-stage low-risk cervical
cancer who wish to preserve fertility: The new standard of care? Int. J. Gynecol. Cancer 2020,30, 981–986. [CrossRef]
31.
Zhang, Q.; Li, W.; Kanis, M.J.; Qi, G.; Li, M.; Yang, X.; Kong, B. Oncologic and obstetrical outcomes with fertility-sparing treatment
of cervical cancer: A systematic review and meta-analysis. Oncotarget 2017,8, 46580–46592. [CrossRef] [PubMed]
32.
Nezhat, C.; Roman, R.A.; Rambhatla, A.; Nezhat, F. Reproductive and oncologic outcomes after fertility-sparing surgery for early
stage cervical cancer: A systematic review. Fertil. Steril. 2020,113, 685–703. [CrossRef] [PubMed]
33.
Williams, N.L.; Werner, T.L.; Jarboe, E.A.; Gaffney, D.K. Adenocarcinoma of the cervix: Should we treat it differently? Curr. Oncol.
Rep. 2015,17, 17. [CrossRef] [PubMed]
34. Williams, M. The Art of Coding and Thematic Exploration in Qualitative Research. Int. Manag. Rev. 2019,15, 45–55.
35.
Ruengkhachorn, I.; Hanamornroongruang, S.; Leelaphatanadit, C.; Sangkarat, S. Does Microinvasive Adenocarcinoma of Cervix
Have Poorer Treatment Outcomes than Microinvasive Squamous Cell Carcinoma? Asian Pac. J. Cancer Prev. 2016,17, 4013–4017.
36.
Noh, J.M.; Park, W.; Kim, Y.S.; Kim, J.Y.; Kim, H.J.; Kim, J.; Kim, J.H.; Yoon, M.S.; Choi, J.H.; Yoon, W.S.; et al. Comparison
of clinical outcomes of adenocarcinoma and adenosquamous carcinoma in uterine cervical cancer patients receiving surgical
resection followed by radiotherapy: A multicenter retrospective study (KROG 13-10). Gynecol. Oncol. 2014,132, 618–623.
[CrossRef]
37.
Gadducci, A.; Guerrieri, M.E.; Cosio, S. Adenocarcinoma of the uterine cervix: Pathologic features, treatment options, clinical
outcome and prognostic variables. Crit. Rev. Oncol. Hematol. 2019,135, 103–114. [CrossRef]
38.
Zusterzeel, P.L.; Pol, F.J.; van Ham, M.; Zweemer, R.P.; Bekkers, R.L.; Massuger, L.F.; Verheijen, R.H. Vaginal Radical Trachelectomy
for Early-Stage Cervical Cancer: Increased Recurrence Risk for Adenocarcinoma. Int. J. Gynecol. Cancer 2016,26, 1293–1299.
[CrossRef]
39.
Helpman, L.; Grisaru, D.; Covens, A. Early adenocarcinoma of the cervix: Is radical vaginal trachelectomy safe? Gynecol. Oncol.
2011,123, 95–98. [CrossRef]
40.
Winer, I.; Alvarado-Cabrero, I.; Hassan, O.; Ahmed, Q.F.; Alosh, B.; Bandyopadhyay, S.; Thomas, S.; Albayrak, S.; Talukdar, S.;
Al-Wahab, Z.; et al. The prognostic significance of histologic type in early stage cervical cancer—A multi-institutional study.
Gynecol. Oncol. 2015,137, 474–478. [CrossRef]
41.
Slama, J.; Runnebaum, I.B.; Scambia, G.; Angeles, M.A.; Bahrehmand, K.; Kommoss, S.; Fagotti, A.; Narducci, F.; Matylevich, O.;
Holly, J.; et al. Analysis of risk factors for recurrence in cervical cancer patients after fertility-sparing treatment: The FERTIlity
Sparing Surgery retrospective multicenter study. Am. J. Obstet. Gynecol. 2023,228, 443.e1–443.e10. [CrossRef]
42.
Plante, M.; Lau, S.; Brydon, L.; Swenerton, K.; LeBlanc, R.; Roy, M. Neoadjuvant chemotherapy followed by vaginal radical
trachelectomy in bulky stage IB1 cervical cancer: Case report. Gynecol. Oncol. 2006,101, 367–370. [CrossRef]
Curr. Oncol. 2024,31 305
43.
Maneo, A.; Chiari, S.; Bonazzi, C.; Mangioni, C. Neoadjuvant chemotherapy and conservative surgery for stage IB1 cervical
cancer. Gynecol. Oncol. 2008,111, 438–443. [CrossRef]
44.
van Gent, M.D.; van den Haak, L.W.; Gaarenstroom, K.N.; Peters, A.A.; van Poelgeest, M.I.; Trimbos, J.B.; de Kroon, C.D.
Nerve-sparing radical abdominal trachelectomy versus nerve-sparing radical hysterectomy in early-stage (FIGO IA2-IB) cervical
cancer: A comparative study on feasibility and outcome. Int. J. Gynecol. Cancer 2014,24, 735–743. [CrossRef]
45.
Tesfai, F.M.; Kroep, J.R.; Gaarenstroom, K.; De Kroon, C.; Van Loenhout, R.; Smit, V.; Trimbos, B.; Nout, R.A.; van Poelgeest,
M.I.E.; Beltman, J.J. Fertility-sparing surgery of cervical cancer > 2 cm (International Federation of Gynecology and Obstetrics
2009 stage IB1-IIA) after neoadjuvant chemotherapy. Int. J. Gynecol. Cancer 2020,30, 115–121. [CrossRef]
46.
Vercellino, G.F.; Piek, J.M.; Schneider, A.; Kohler, C.; Mangler, M.; Speiser, D.; Chiantera, V. Laparoscopic lymph node dissection
should be performed before fertility preserving treatment of patients with cervical cancer. Gynecol. Oncol. 2012,126, 325–329.
[CrossRef]
47.
Lanowska, M.; Mangler, M.; Speiser, D.; Bockholdt, C.; Schneider, A.; Kohler, C.; Vasiljeva, J.; Al-Hakeem, M.; Vercellino, G.F.
Radical vaginal trachelectomy after laparoscopic staging and neoadjuvant chemotherapy in women with early-stage cervical
cancer over 2 cm: Oncologic, fertility, and neonatal outcome in a series of 20 patients. Int. J. Gynecol. Cancer 2014,24, 586–593.
[CrossRef]
48.
Robova, H.; Halaska, M.J.; Pluta, M.; Skapa, P.; Matecha, J.; Lisy, J.; Rob, L. Oncological and pregnancy outcomes after high-dose
density neoadjuvant chemotherapy and fertility-sparing surgery in cervical cancer. Gynecol. Oncol. 2014,135, 213–216. [CrossRef]
49.
Salihi, R.; Leunen, K.; Van Limbergen, E.; Moerman, P.; Neven, P.; Vergote, I. Neoadjuvant chemotherapy followed by large cone
resection as fertility-sparing therapy in stage IB cervical cancer. Gynecol. Oncol. 2015,139, 447–451. [CrossRef] [PubMed]
50.
Zusterzeel, P.L.M.; Aarts, J.W.M.; Pol, F.J.M.; Ottevanger, P.B.; van Ham, M. Neoadjuvant Chemotherapy Followed by Vaginal
Radical Trachelectomy as Fertility-Preserving Treatment for Patients with FIGO 2018 Stage 1B2 Cervical Cancer. Oncologist 2020,
25, e1051–e1059. [CrossRef] [PubMed]
51.
Gwacham, N.I.; McKenzie, N.D.; Fitzgerald, E.R.; Ahmad, S.; Holloway, R.W. Neoadjuvant chemotherapy followed by fertility
sparing surgery in cervical cancers size 2-4 cm; emerging data and future perspectives. Gynecol. Oncol. 2021,162, 809–815.
[CrossRef]
52.
Peters, W.A., 3rd; Liu, P.Y.; Barrett, R.J., 2nd; Stock, R.J.; Monk, B.J.; Berek, J.S.; Souhami, L.; Grigsby, P.; Gordon, W., Jr.; Alberts,
D.S. Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy
after radical surgery in high-risk early-stage cancer of the cervix. J. Clin. Oncol. 2000,18, 1606–1613. [CrossRef]
53.
Plante, M.; van Trommel, N.; Lheureux, S.; Oza, A.M.; Wang, L.; Sikorska, K.; Ferguson, S.E.; Han, K.; Amant, F. FIGO 2018
stage IB2 (2-4 cm) Cervical cancer treated with Neo-adjuvant chemotherapy followed by fertility Sparing Surgery (CONTESSA);
Neo-Adjuvant Chemotherapy and Conservative Surgery in Cervical Cancer to Preserve Fertility (NEOCON-F). A PMHC, DGOG,
GCIG/CCRN and multicenter study. Int. J. Gynecol. Cancer 2019,29, 969–975. [CrossRef]
54.
Cibula, D.; Potter, R.; Planchamp, F.; Avall-Lundqvist, E.; Fischerova, D.; Haie Meder, C.; Kohler, C.; Landoni, F.; Lax, S.; Linde-
gaard, J.C.; et al. The European Society of Gynaecological Oncology/European Society for Radiotherapy and Oncology/European
Society of Pathology Guidelines for the Management of Patients with Cervical Cancer. Int. J. Gynecol. Cancer 2018,28, 641–655.
[CrossRef]
55. Wallace, W.H.; Thomson, A.B.; Saran, F.; Kelsey, T.W. Predicting age of ovarian failure after radiation to a field that includes the
ovaries. Int. J. Radiat. Oncol. Biol. Phys. 2005,62, 738–744. [CrossRef]
56.
Winarto, H.; Febia, E.; Purwoto, G.; Nuranna, L. The need for laparoscopic ovarian transposition in young patients with cervical
cancer undergoing radiotherapy. Int. J. Reprod. Med. 2013,2013, 173568. [CrossRef]
57. Dalal, P.K.; Agarwal, M. Postmenopausal syndrome. Indian J. Psychiatry 2015,57, S222–S232. [CrossRef]
58.
Marchocki, Z.; May, T. High laparoscopic bilateral ovarian transposition to the upper abdomen prior to pelvic radiotherapy. Int. J.
Gynecol. Cancer 2021,31, 1384–1385. [CrossRef] [PubMed]
59.
Laios, A.; Otify, M.; Papadopoulou, A.; Gallos, I.D.; Ind, T. Outcomes of ovarian transposition in cervical cancer; an updated
meta-analysis. BMC Womens Health 2022,22, 305. [CrossRef] [PubMed]
60.
Song, S.; Rudra, S.; Hasselle, M.D.; Dorn, P.L.; Mell, L.K.; Mundt, A.J.; Yamada, S.D.; Lee, N.K.; Hasan, Y. The effect of treatment
time in locally advanced cervical cancer in the era of concurrent chemoradiotherapy. Cancer 2013,119, 325–331. [CrossRef]
[PubMed]
61.
Buonomo, B.; Multinu, F.; Casarin, J.; Betella, I.; Zanagnolo, V.; Aletti, G.; Peccatori, F. Ovarian transposition in patients with
cervical cancer prior to pelvic radiotherapy: A systematic review. Int. J. Gynecol. Cancer 2021,31, 360–370. [CrossRef] [PubMed]
62.
Findeklee, S.; Lotz, L.; Heusinger, K.; Hoffmann, I.; Dittrich, R.; Beckmann, M.W. Fertility Protection in Female Oncology Patients:
How Should Patients Be Counseled? Geburtshilfe Frauenheilkd. 2015,75, 1243–1249. [CrossRef] [PubMed]
63.
Wo, J.Y.; Viswanathan, A.N. Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients. Int.
J. Radiat. Oncol. Biol. Phys. 2009,73, 1304–1312. [CrossRef] [PubMed]
64.
Moawad, N.S.; Santamaria, E.; Rhoton-Vlasak, A.; Lightsey, J.L. Laparoscopic Ovarian Transposition Before Pelvic Cancer
Treatment: Ovarian Function and Fertility Preservation. J. Minim. Invasive Gynecol. 2017,24, 28–35. [CrossRef] [PubMed]
65.
Sonmezer, M.; Turkcuoglu, I.; Coskun, U.; Oktay, K. Random-start controlled ovarian hyperstimulation for emergency fertility
preservation in letrozole cycles. Fertil. Steril. 2011,95, 2125.e9–2125.e11. [CrossRef] [PubMed]
Curr. Oncol. 2024,31 306
66.
Tsonis, O.; Kopeika, J. Fertility preservation in patients with gynaecologic malignancy: Response to ovarian stimulation and
long-term outcomes. Eur. J. Obstet. Gynecol. Reprod. Biol. 2023,290, 93–100. [CrossRef] [PubMed]
67.
Fraison, E.; Huberlant, S.; Labrune, E.; Cavalieri, M.; Montagut, M.; Brugnon, F.; Courbiere, B. Live birth rate after female fertility
preservation for cancer or haematopoietic stem cell transplantation: A systematic review and meta-analysis of the three main
techniques; embryo, oocyte and ovarian tissue cryopreservation. Hum. Reprod. 2023,38, 489–502. [CrossRef]
68.
Khattak, H.; Malhas, R.; Craciunas, L.; Afifi, Y.; Amorim, C.A.; Fishel, S.; Silber, S.; Gook, D.; Demeestere, I.; Bystrova, O.; et al.
Fresh and cryopreserved ovarian tissue transplantation for preserving reproductive and endocrine function: A systematic review
and individual patient data meta-analysis. Hum. Reprod. Update 2022,28, 400–416. [CrossRef]
69.
Anderson, R.A.; Brewster, D.H.; Wood, R.; Nowell, S.; Fischbacher, C.; Kelsey, T.W.; Wallace, W.H.B. The impact of cancer on
subsequent chance of pregnancy: A population-based analysis. Hum. Reprod. 2018,33, 1281–1290. [CrossRef]
70.
Teh, W.T.; Stern, C.; Chander, S.; Hickey, M. The impact of uterine radiation on subsequent fertility and pregnancy outcomes.
Biomed. Res. Int. 2014,2014, 482968. [CrossRef]
71.
Meirow, D.; Ben Yehuda, D.; Prus, D.; Poliack, A.; Schenker, J.G.; Rachmilewitz, E.A.; Lewin, A. Ovarian tissue banking in patients
with Hodgkin’s disease: Is it safe? Fertil. Steril. 1998,69, 996–998. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... Literature data support the safety and efficacy of fertility preservation methods in early stages of gynecologic cancers [122][123][124], but even for more advanced cases, new assisted reproduction techniques can provide tailored options for obtaining a viable pregnancy. ...
Article
Full-text available
Simple Summary Gynecological cancers affect many women of reproductive age, necessitating the development of fertility preservation methods to fulfill family planning. Consequently, providing treatment options that preserve fertility in women diagnosed with gynecological cancers has become a crucial component of care for survivors. This leads to improved quality of life, allowing survivors to become mothers even in the seemingly adverse circumstances posed by cancers. However, although there are guidelines on fertility preservation in the context of neoplasms, physicians do not routinely consider it and do not discuss these options with their patients. It is important for patients to be informed about the available options for fertility preservation and to be encouraged to make informed decisions in collaboration with their medical team. Standardized guideline recommendations for onco-fertility should be considered in the future. We explore possible methods that can be employed for obtaining a pregnancy after gynecologic cancer treatment, including more exotic ones like uterine transplant. Abstract (1) Background: Currently, an increasing number of women postpone pregnancy beyond the age of 35. Gynecological cancers affect a significant proportion of women of reproductive age, necessitating the development of fertility preservation methods to fulfill family planning. Consequently, providing treatment options that preserve fertility in women diagnosed with gynecological cancers has become a crucial component of care for survivors. (2) Methods: We conducted an extensive search of relevant scientific publications in PubMed and Embase databases and performed a narrative review, including high-quality peer-reviewed research on fertility after being treated for gynecologic cancers, reporting pregnancy rates, birth rates, and pregnancy outcomes in cancer survivors as well as therapeutic options which partially preserve fertility and methods for obtaining a pregnancy in survivors. (3) Discussion: The medicine practiced today is focused on both treating the neoplasm and preserving the quality of life of the patients, with fertility preservation being an important element of this quality. This leads to an improved quality of life, allowing these women to become mothers even in the seemingly adverse circumstances posed by such a pathology. However, although there are guidelines on female fertility preservation in the context of neoplasms, an analysis shows that physicians do not routinely consider it and do not discuss these options with their patients. (4) Conclusions: Advancements in medicine have led to a better understanding and management of gynecological neoplasms, resulting in increased survival rates. Once the battle against these neoplasms is won, the issue of preserving the quality of life for these women arises, with fertility preservation being an important aspect for women who have not yet fulfilled their family planning desires at the time of diagnosis. It is important for patients to be informed about the available options for fertility preservation and to be encouraged to make informed decisions in collaboration with their medical team. Standardized recommendations for onco-fertility into guidelines should be taken into consideration in the future.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths in the United States and compiles the most recent data on population‐based cancer occurrence and outcomes using incidence data collected by central cancer registries and mortality data collected by the National Center for Health Statistics. In 2023, 1,958,310 new cancer cases and 609,820 cancer deaths are projected to occur in the United States. Cancer incidence increased for prostate cancer by 3% annually from 2014 through 2019 after two decades of decline, translating to an additional 99,000 new cases; otherwise, however, incidence trends were more favorable in men compared to women. For example, lung cancer in women decreased at one half the pace of men (1.1% vs. 2.6% annually) from 2015 through 2019, and breast and uterine corpus cancers continued to increase, as did liver cancer and melanoma, both of which stabilized in men aged 50 years and older and declined in younger men. However, a 65% drop in cervical cancer incidence during 2012 through 2019 among women in their early 20s, the first cohort to receive the human papillomavirus vaccine, foreshadows steep reductions in the burden of human papillomavirus‐associated cancers, the majority of which occur in women. Despite the pandemic, and in contrast with other leading causes of death, the cancer death rate continued to decline from 2019 to 2020 (by 1.5%), contributing to a 33% overall reduction since 1991 and an estimated 3.8 million deaths averted. This progress increasingly reflects advances in treatment, which are particularly evident in the rapid declines in mortality (approximately 2% annually during 2016 through 2020) for leukemia, melanoma, and kidney cancer, despite stable/increasing incidence, and accelerated declines for lung cancer. In summary, although cancer mortality rates continue to decline, future progress may be attenuated by rising incidence for breast, prostate, and uterine corpus cancers, which also happen to have the largest racial disparities in mortality.
Article
Full-text available
STUDY QUESTION What are the chances of achieving a live birth after embryo, oocyte and ovarian tissue cryopreservation (OTC) in female cancer survivors? SUMMARY ANSWER The live birth rates (LBRs) following embryo and oocyte cryopreservation are 41% and 32%, respectively, while for IVF and spontaneous LBR after tissue cryopreservation and transplantation, these rates are 21% and 33%, respectively. WHAT IS KNOWN ALREADY Currently, fertility preservation (FP) has become a major public health issue as diagnostic and therapeutic progress has made it possible to achieve an 80% survival rate in children, adolescents and young adults with cancer. In the latest ESHRE guidelines, only oocyte and embryo cryopreservation are considered as established options for FP. OTC is still considered to be an innovative method, while it is an acceptable FP technique in the American Society for Reproductive Medicine guidelines. However, given the lack of studies on long-term outcomes after FP, it is still unclear which technique offers the best chance to achieve a live birth. STUDY DESIGN, SIZE, DURATION We performed a systematic review and meta-analysis of published controlled studies. Searches were conducted from January 2004 to May 2021 in Medline, Embase and the Cochrane Library using the following search terms: cancer, stem cell transplantation, FP, embryo cryopreservation, oocyte vitrification, OTC and reproductive outcome. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 126 full-text articles were preselected from 1436 references based on the title and abstract and assessed via the Newcastle–Ottawa Quality Assessment Scale. The studies were selected, and their data were extracted by two independent reviewers according to the Cochrane methods. A fixed-effect meta-analysis was performed for outcomes with high heterogeneity. MAIN RESULTS AND THE ROLE OF CHANCE Data from 34 studies were used for this meta-analysis. Regarding cryopreserved embryos, the LBR after IVF was 41% (95% CI: 34–48, I2: 0%, fixed effect). Concerning vitrified oocytes, the LBR was 32% (95% CI: 26–39, I2: 0%, fixed effect). Finally, the LBR after IVF and the spontaneous LBR after ovarian tissue transplantation were 21% (95% CI: 15–26, I2: 0%, fixed-effect) and 33% (95% CI: 25–42, I2: 46.1%, random-effect), respectively. For all outcomes, in the sensitivity analyses, the maximum variation in the estimated percentage was 1%. LIMITATIONS, REASONS FOR CAUTION The heterogeneity of the literature prevents us from comparing these three techniques. This meta-analysis provides limited data which may help clinicians when counselling patients. WIDER IMPLICATIONS OF THE FINDINGS This study highlights the need for long-term follow-up registries to assess return rates, as well as spontaneous pregnancy rates and birth rates after FP. STUDY FUNDING/COMPETING INTEREST(S) This work was sponsored by an unrestricted grant from GEDEON RICHTER France. The authors have no competing interests to declare. REGISTRATION NUMBER CRD42021264042.
Article
Full-text available
Background Cervical cancer is the most common indication for ovarian transposition in reproductive-age women. Ovarian transposition should be performed in premenopausal women undergoing pelvic irradiation to preserve ovarian function, and prevent early menopause. As women become more knowledgeable about their fertility options, it is still unclear who will benefit from the intervention. We updated our previous meta-analysis of ovarian function preservation, symptomatic ovarian cysts, and metastases to the transposed ovaries following ovarian transposition in cervical cancer patients to further guide current clinical practice. Methods A systematic search of Medline, Embase, Web of Science, and The Cochrane Library databases, dating from January 1980 to July 2021, was conducted. We computed the summary proportions of women who had ovarian function preservation, non-ovarian cyst formation and metastases to the transposed ovaries following ovarian transposition by random-effects meta-analysis and we explored study heterogeneity by type of radiotherapy. Results There were 29 publications reporting on 1160 women with cervical cancer who underwent ovarian transposition. In the group that underwent surgery alone, 91% of the women had preserved ovarian function (95% CI 83–100), 89% (95% CI 80–99) of women who did not develop ovarian cysts, and 99% (95% CI 1–5) of women who did not suffer metastases to the transposed ovaries. In the surgery ± brachytherapy (BR) group, the proportion of women with the preserved ovarian function was 93% (95% CI 76–113), 84% (95% CI 69–103) of women who did not develop ovarian cysts, and 99% (95% CI 82–120) of women who did not suffer metastases to the transposed ovaries. In the external beam pelvic radiotherapy (EBRT) ± BR ± surgery group, the proportion of women with the preserved ovarian function was 61% (95% CI 55–69), and 95% (95% CI 85–107) of women who developed ovarian cysts. There were no metastases to the transposed ovaries in that group. Conclusions In women with cervical cancer, ovarian transposition offers a significant preservation of the ovarian function. Despite an expected incidence of ovarian cyst formation, it carries almost no risk for metastases to the transposed ovaries.
Article
Full-text available
Background: Ovarian tissue cryopreservation involves freezing and storing of surgically retrieved ovarian tissue in liquid or vapour nitrogen below -190°C. The tissue can be thawed and transplanted back with the aim of restoring fertility or ovarian endocrine function. The techniques for human ovarian tissue freezing and transplantation have evolved over the last 20 years, particularly in the context of fertility preservation in pre-pubertal cancer patients. Fresh ovarian tissue transplantation, using an autograft or donor tissue, is a more recent development; it has the potential to preserve fertility and hormonal function in women who have their ovaries removed for benign gynaecological conditions. The techniques of ovarian tissue cryopreservation and transplantation have progressed rapidly since inception; however, the evidence on the success of this intervention is largely based on case reports and case series. Objective and rationale: The aim of this study was to systematically review the current evidence by incorporating study-level and individual patient-level meta-analyses of women who received ovarian transplants, including frozen-thawed transplant, fresh or donor graft. Search methods: The review protocol was registered with PROSPERO (CRD42018115233). A comprehensive literature search was performed using MEDLINE, EMBASE, CINAHL and Cochrane Central Register of Controlled Trials from database inception to October 2020. Authors were also contacted for individual patient data if relevant outcomes were not reported in the published manuscripts. Meta-analysis was performed using inverse-variance weighting to calculate summary estimates using a fixed-effects model. Outcomes: The review included 87 studies (735 women). Twenty studies reported on ≥5 cases of ovarian transplants and were included in the meta-analysis (568 women). Fertility outcomes included pregnancy, live birth and miscarriage rates, and endocrine outcomes included oestrogen, FSH and LH levels. The pooled rates were 37% (95% CI: 32-43%) for pregnancy, 28% (95% CI: 24-34%) for live birth and 37% (95% CI: 30-46%) for miscarriage following frozen ovarian tissue transplantation. Pooled mean for pre-transplant oestrogen was 101.6 pmol/l (95% CI: 47.9-155.3), which increased post-transplant to 522.4 pmol/l (95% CI: 315.4-729; mean difference: 228.24; 95% CI: 180.5-276). Pooled mean of pre-transplant FSH was 66.4 IU/l (95% CI: 52.8-84), which decreased post-transplant to 14.1 IU/l (95% CI: 10.9-17.3; mean difference 61.8; 95% CI: 57-66.6). The median time to return of FSH to a value <25 IU/l was 19 weeks (interquartile range: 15-26 weeks; range: 0.4-208 weeks). The median duration of graft function was 2.5 years (interquartile range: 1.4-3.4 years; range: 0.7-5 years). The analysis demonstrated that ovarian tissue cryopreservation and transplantation could restore reproductive and hormonal functions in women. Further studies with larger samples of well-characterized populations are required to define the optimal retrieval, cryopreservation and transplantation processes. Wider implications: Ovarian tissue cryopreservation and transplantation may not only be effective in restoring fertility but also the return of reproductive endocrine function. Although this technology was developed as a fertility preservation option, it may have the scope to be considered for endocrine function preservation.
Article
Full-text available
Radical hysterectomy with pelvic node dissection is the standard treatment for early-stage cervical cancer. However, the latter can be diagnosed at a young age when patients have not yet achieved their pregnancy plans. Dargent first described the vaginal radical trachelectomy for patients with tumors <2 cm. It has since been described a population of low risk of recurrence: patients with tumors <2 cm, without deep stromal infiltration, without lymphovascular invasion (LVSI), and with negative lymph nodes. These patients can benefit from a less radical surgery such as conization or simple trachelectomy with the evaluation of the pelvic node status. Tumors larger than 2 cm have a higher risk of recurrence and their treatment is a challenge. There are currently two options for these patients: abdominal radical trachelectomy or neoadjuvant chemotherapy (NACT), followed by fertility-sparing surgery. All patients who wish to preserve their fertility must be referred to expert centers.
Article
Full-text available
Objectives To compare oncologic outcomes of patients with early-stage cervical cancer and negative nodes who underwent sentinel lymph node biopsy alone (SLNB) versus pelvic lymphadenectomy (PL). Methods An ancillary analysis of two prospective multicentric trials on SLN biopsy for cervical cancer (SENTICOL I and II) was conducted. Only patients with early-stage cervical cancer (IA to IIA FIGO stage), bilateral detection of SLN, negative SLN after ultrastaging and negative non-SLN after final pathologic examination were included. Risk-factors of recurrence and disease-specific mortality were determined by Cox proportional hazard models. Results Between January 2005 and July 2012, 259 node-negative patients were analyzed: 87 in the SLNB group and 172 in the PL group. The median follow-up was 47 months [4–127]. During the follow-up, 21 patients (8.1%) experienced recurrences, including 4 nodal recurrences (1.9%), and 9 patients (3.5%) died of cervical cancer. Disease-free survival (DFS) and disease-specific survival (DSS) were similar between SLNB and PL groups, 85.1% vs. 80.4%, p = 0.24 and 90.8% vs. 97.2%, p = 0.22 respectively. By Cox multivariate analysis, SLNB compared to PL was not associated with DFS (HR = 1.78, 95%CI = [0.71–4.46], p = 0.22) neither with DSS (HR = 3.02, 95%CI = [0.69–13.18], p = 0.14). Only pathologic risk level according to the Sedlis criteria was an independent predictor of DFS and DSS. Conclusions Omitting full pelvic lymphadenectomy for patients with bilateral negative SLN does not seem to be associated with an increased risk of recurrence in this series. Survival non-inferiority needs to be confirmed by prospective trials.
Article
Objective To the best of our knowledge, the available evidence on the effect and efficacy of controlled ovarian stimulation (COS) in this group of patients remains poorly reported. Concerns related to the impact of stimulation to cancer progression and recurrence, as well as the risk of disease dissemination during egg collection, might explain the aforementioned trend. Methods Overall, our FP Service received 192 gynaecological referrals, between 2005 and 2021, regarding gynaecologic conditions mainly cancer related. A total of 68 (35.4%) patients underwent COS. These patients were diagnosed with the following gynaecologic pathologies: 33 cases (48,5%) of cervical cancer were noted (stage 1b1-2b), 25 ovarian pathology (36.7%), 9 cases (13.2%) of endometrial cancer, and a single case of vaginal cancer (1.5%). Results The mean age of patients attending the fertility preservation service was 31.5 (std 5.8). The patients presenting to their initial appointment with a mean BMI 24.5 (IQR 6.9) and a median AFC of 12 (IQR 13). The mean duration of COS was 11 days (IQR 3), and the median dose of gonadotrophins was calculated at 300 IU (IQR 75 IU). In 95.4% of the cases, GnRH agonist was used as a trigger for final maturation. The median number of follicles measuring more than 14 mm at the time of trigger was 11 (IQR 8), whereas the median number of oocytes collected was 11 (IQR 9). The complication rate was reported at less than 2%. So far, one in four women of this FP group (17/68, 25% of the overall group) returned to our service to claim their cryopreserved eggs/embryos and successful livebirths were reported in 58.8% of this sample (10/17 cases). The mean time to return to use their oocytes/embryos was 36 months (min value 16 months – max value 85 months). There was no significant difference in mortality rate between patients who received FP vs those who did not (hazard ratio of mortality was estimated at 0.91 (p-value 0.88)). Conclusion Based on our findings, ovarian stimulation for patients presenting with gynaecologic malignancy is a safe and efficient method of fertility preservation. Undoubtedly, the sample size is limited, however our results are reassuring and highlight the efficacy of COS for the purpose of FP based on data coming from the largest Assisted Conception Unit of the South-East of the UK.
Article
In 2018, the European Society of Gynecological Oncology (ESGO) jointly with the European Society for Radiotherapy and Oncology (ESTRO) and the European Society of Pathology (ESP) published evidence-based guidelines for the management of patients with cervical cancer. Given the large body of new evidence addressing the management of cervical cancer, the three sister societies jointly decided to update these evidence-based guidelines. The update includes new topics to provide comprehensive guidelines on all relevant issues of diagnosis and treatment in cervical cancer. To serve on the expert panel (27 experts across Europe) ESGO/ESTRO/ESP nominated practicing clinicians who are involved in managing patients with cervical cancer and have demonstrated leadership through their expertise in clinical care and research, national and international engagement, profile, and dedication to the topics addressed. To ensure the statements were evidence based, new data identified from a systematic search was reviewed and critically appraised. In the absence of any clear scientific evidence, judgment was based on the professional experience and consensus of the international development group. Before publication, the guidelines were reviewed by 155 independent international practitioners in cancer care delivery and patient representatives. These updated guidelines are comprehensive and cover staging, management, follow-up, long-term survivorship, quality of life and palliative care. Management includes fertility sparing treatment, early and locally advanced cervical cancer, invasive cervical cancer diagnosed on a simple hysterectomy specimen, cervical cancer in pregnancy, rare tumors, recurrent and metastatic diseases. The management algorithms and the principles of radiotherapy and pathological evaluation are also defined.
Article
BACKGROUND Fertility-sparing treatment in patients with cervical cancer should, in principle, follow identical algorithms to that in patients without future reproductive plans. In recent years, a trend towards non-radical procedures such as conization or simple trachelectomy has become apparent in medical literature, because of their associations with better pregnancy outcomes. However, the published reports included small numbers of patients and heterogenous treatment strategies to ascertain safety of such approaches. OBJECTIVE Our aims were to collect multi-institutional data regarding the oncological outcomes after fertility-sparing treatment in cervical cancer patients and to identify prognostic risk factors, including the influence of the radicality of individual cervical procedures. METHODS Patients aged 18–40 years with FIGO 2018 stage IA1 + lymphovascular space invasion or ≥IA2 cervical cancer who underwent any type of fertility-sparing procedure were eligible for this retrospective observational study, regardless of their histotype, tumor grade, and history of neoadjuvant chemotherapy. Associations between disease- and treatment-related characteristics with the risk of recurrence were analyzed. RESULTS A total of 733 patients from 44 institutions across 13 countries were included in this study. Almost half of the patients had stage IB1 cervical cancer (49%), and two-thirds were nulliparous (66%). After a median follow-up of 72 months, 51 patients (7%) experienced recurrence of whom 19 (2.6%) died due to the disease. The most common sites of recurrence were the cervix (53%) and pelvic nodes (22%). The risk of recurrence was three times higher in patients with tumors >2 cm in size than in patients with smaller tumors, irrespective of the treatment radicality (19.4% vs. 5.7%; hazard ratio 2.982, 95% CI 1.383–6.431, P = .005). The recurrence risk in patients with tumors ≤2 cm in size did not differ between patients who underwent radical trachelectomy or non-radical (conization and simple trachelectomy) cervical procedures (P = .957), regardless of tumor size subcategory (<1 or 1–2 cm) or lymphovascular space invasion. CONCLUSIONS Fertility-sparing non-radical cervical procedures were not associated with increased risk of recurrence compared with radical procedures in patients with tumors ≤2 cm in size in this large, multicenter retrospective study. The risk of recurrence after any type of fertility-sparing procedure was significantly greater in patients with tumors >2 cm in size.
Article
Purpose To evaluate the effectiveness and obstetric outcomes after laser conization for cervical intraepithelial neoplasia Methods This retrospective study included 757 patients who underwent laser conization between 2014 and 2020. Patients with a diagnosis of invasive lesions or adenocarcinoma in situ were excluded. Histological data from the conization specimen, pre and postoperative histological and virological data (Human PapillomaVirus (HPV) test) and obstetric outcomes were collected from the medical record. The primary endpoint was the negative surgical margin rates after laser conization. The secondary endpoint were the size of the operative specimen, the postoperative virological test results (3 to 6 months after surgery), factors associated with negative or positive margin and postoperative obstetric outcomes (prematurity). Patient characteristics and outcomes were compared using Student's t test, χ² test, or Fisher exact test. Values of p ≤0.05 were considered significant. Results This study included 757 patients. Negative surgical margins were obtained in 76.1% of the cases and were associated with more negative HPV tests at 6 months (64.9% vs. 52.5%, p=0.006) and fewer repeat surgeries (0.2% vs. 2.2%, p=0.013) than for patients with positive margins. Among the patients under 43 years at the time of conization, 71 achieved a pregnancy with a term >22 weeks, and of these 66 (93%) delivered at term (≥37 weeks). Conclusion Laser conization appears to be an effective technique for the management of cervical intraepithelial neoplasia both in terms of the quality of the resection margins and the obstetric prognosis.