Abstract

The review covers current options for ovarian tissue cryopreservation and transplantation and provides a systematic review of the existing literature from the last 10 years, taking into account all previously published reviews on the subject. The different cryopreservation options available for fertility preservation in cancer patients are embryo cryopreservation, oocyte cryopreservation and ovarian tissue cryopreservation. The choice depends on various parameters: the type and timing of chemotherapy, the type of cancer, the patient’s age and the partner status. The different options and their results are discussed, as well as their putative indications and efficacy. The review concludes that advances in reproductive technology have made fertility preservation techniques a real possibility for patients whose gonadal function is threatened by premature menopause, or by treatments such as radiotherapy, chemotherapy or surgical castration.

Introduction

In 2006, 1 399 790 new cancer cases are expected in the United States, of which 679 540 will be women (Jemal et al., 2006). According to previous reports, 8% of these women will be under the age of 40. By 2010, it is estimated that one in every 250 people in the adult population will be childhood cancer survivors (Blatt, 1999). Advances in the diagnosis and treatment of childhood, adolescent and adult cancer have greatly increased the life expectancy of premenopausal women with cancer but have resulted in a growing population of adolescent and adult long-term survivors of childhood malignancies (Blatt, 1999), who may experience infertility problems due to induced premature ovarian failure (POF).

Aggressive chemotherapy and radiotherapy, and bone marrow transplantation (BMT), can cure >90% of girls and young women affected by such disorders (Ries et al., 1999). However, the ovaries are very sensitive to cytotoxic treatment, especially to alkylating agents, which are classified as high risk for gonadal dysfunction (e.g. cyclophosphamide, busulfan, melphalan, chlorambucil, dacarbazine, procarbazine, ifosfamide, thiotepa and nitrogen mustard) (Warne et al., 1973; Koyama et al., 1977; Fisher et al., 1979; Viviani et al., 1985; Mackie et al., 1996; Teinturier et al., 1998; Legault and Bonny, 1999; Meirow et al., 1999; Blumenfeld et al., 2000; Kenney et al., 2001; Tauchmanova et al., 2002). Doxorubicin and the alkylating-like agents cisplatin and carboplatin fall into the medium-risk category (Hortobagyi et al., 1986; Wallace et al., 1989; Maneschi et al., 1994; Tangir et al., 2003), whereas methotrexate, bleomycin, 5-fluorouracil, actinomycin-D, mercaptopurine and vincristine are considered as low risk for gonadal dysfunction (Van Thiel et al., 1970; Shamberger et al., 1981; Stillman et al., 1981; Sudman et al., 1992; Bines et al., 1996; Bower et al., 1998; Sonmezer and Oktay, 2004; Wallace et al., 2005a) (Table I). The type and dose of chemotherapeutic agent influence the progression to ovarian failure, with alkylating agents increasing the risk of POF by a factor of 9 (Byrne et al., 1992).

Table I.

Cytotoxic agents according to the degree of gonadotoxicity

High riskIntermediate riskLow/no risk
CyclophosphamideDoxorubicinMethotrexate
BusulfanCisplatinBleomycin
MelphalanCarboplatin5-Fluorouracil
ChlorambucilActinomycin-D
DacarbazineMercaptopurine
ProcarbazineVincristine
Ifosfamide
Thiotepa
Nitrogen mustard
High riskIntermediate riskLow/no risk
CyclophosphamideDoxorubicinMethotrexate
BusulfanCisplatinBleomycin
MelphalanCarboplatin5-Fluorouracil
ChlorambucilActinomycin-D
DacarbazineMercaptopurine
ProcarbazineVincristine
Ifosfamide
Thiotepa
Nitrogen mustard
Table I.

Cytotoxic agents according to the degree of gonadotoxicity

High riskIntermediate riskLow/no risk
CyclophosphamideDoxorubicinMethotrexate
BusulfanCisplatinBleomycin
MelphalanCarboplatin5-Fluorouracil
ChlorambucilActinomycin-D
DacarbazineMercaptopurine
ProcarbazineVincristine
Ifosfamide
Thiotepa
Nitrogen mustard
High riskIntermediate riskLow/no risk
CyclophosphamideDoxorubicinMethotrexate
BusulfanCisplatinBleomycin
MelphalanCarboplatin5-Fluorouracil
ChlorambucilActinomycin-D
DacarbazineMercaptopurine
ProcarbazineVincristine
Ifosfamide
Thiotepa
Nitrogen mustard

Cyclophosphamide is the agent most commonly implicated in causing damage to oocytes and granulosa cells in a dose-dependent manner (Warne et al., 1973; Sanders et al., 1996; Meirow et al., 1999; Kenney et al., 2001). Since Meirow et al. (1999) demonstrated primordial follicular depletion in mice treated with cyclophosphamide, tremendous progress has been made towards elucidating the cellular and molecular mechanisms responsible for mediating oocyte death and follicular depletion under normal and pathological conditions. Perez et al. (2000) demonstrated that doxorubicin induces apoptosis in pregranulosa cells, and Takai et al. (2003) showed that bax and caspase-2 and caspase-3 are functionally important mediators of vinylcyclohexene diepoxide-induced ootoxicity in mice.

This follicular destruction generally results in the loss of both endocrine and reproductive functions, depending on the dose and the age of the patient. Indeed, Larsen et al. (2003) reported a four-fold increased risk of POF in teenagers treated for cancer, and a risk increased by a factor of 27 in women between 21 and 25 years of age. Complete amenorrhoea was reported after a dose of 5 g of cyclophosphamide in women over 40 years of age, and after doses of 9 and 20 g in women of 30–40 and 20–30 years of age, respectively (Shalet, 1980). A combination of various chemotherapeutic agents further increases gonadal toxicity. After chemotherapy with MOPP/ABV (chlorethamine, vincristine, procarbazine, prednisone, doxorubicin, bleomycin, vinblastine) hybrid chemotherapy, Schilsky et al. (1981) found that amenorrhoea developed in 89 and 20% of patients >25 and <25 years of age at the time of treatment, respectively. The median age of patients who became amenorrhoeic after therapy was significantly higher than that of patients who maintained normal menses (26 versus 20 years; P = 0.008).

Abdominal ionizing radiation associated with alkylating agents often induces POF, rendering patients infertile in almost 100% of cases. Indeed, for radiotherapy, it has been stated that a dose of 5–20 Gy administered to the ovary is sufficient to completely impair gonadal function (Wallace et al., 2005b), whatever the age of the patient. The dose of radiation required to destroy 50% of the oocyte reserve has been found to be <2 Gy (Wallace et al., 2003). Moreover, uterine irradiation at a young age reduces adult uterine volume (Larsen et al., 2004). Radiation doses between 14 and 30 Gy have been reported to result in uterine dysfunction (Critchley et al., 1992; Bath et al., 1999; Critchley and Wallace, 2005). The practitioner should be aware of this effect of radiotherapy on the uterus, which could interfere with the implantation capacity of embryos.

In the field of oncological indications for ovarian tissue cryopreservation, there have been no major modifications since our review published in 1998 (Donnez and Bassil, 1998) (Table II). In countries where surrogacy is not legal, a conservative fertility approach in case of gynaecological malignancy is only valuable if the uterus can be spared during surgery. This includes cases of early cervical carcinoma (Dargent et al., 2000; Burnett et al., 2003; Tanguay et al., 2004), early vaginal carcinoma (Hicks and Piver, 1992), early endometrial adenocarcinoma (Jadoul and Donnez, 2003), ovarian tumours of low malignancy (Donnez et al., 2003; Boran et al., 2005; Fauvet et al., 2005; Tinelli et al., 2005) and some selected cases of unilateral ovarian carcinoma (stage IA) (Donnez and Bassil, 1998; Kleine, 1996). The choice of a possible conservative surgical approach in these patients and the question of implementing such treatment alone remain controversial, as all the published results were obtained on the basis of retrospective studies and/or case reports. The fertility outcome is conditioned by the adjuvant therapy, i.e. local radiotherapy and/or chemotherapy.

Table II.

Indications for ovarian tissue cryopreservation in case of malignant and non-malignant diseases

MalignantNon-malignant
Extrapelvic diseasesUni/bilateral oophorectomy
    Bone cancer (osteosarcoma—Ewing’s sarcoma)    Benign ovarian tumours
    Breast cancer    Severe and recurrent endometriosis
    Melanoma    BRCA-1 or BRCA-2 mutation carriers
    Neuroblastoma
    Bowel malignancy
Pelvic diseasesRisk of premature menopause
    Non-gynaecological malignancy    Turner’s syndrome
        Pelvic sarcoma    Family history
        Rhabdomyosarcoma Sacral tumours Rectosigmoid tumours    Benign diseases requiring chemotherapy: autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, Behçet’s disease and Wegener’s disease)
    Gynaecological malignancy
        Early cervical carcinoma
        Early vaginal carcinoma
        Early vulvar carcinoma
        Selected cases of ovarian carcinoma (stage IA)
        Ovarian borderline tumours
Systemic diseasesBone marrow transplantation
    Hodgkin’s disease    Benign haematological diseases: sickle cell anaemia, thalassaemia major and aplastic anaemia
    Non-Hodgkin’s lymphoma    Autoimmune diseases unresponsive to immunosuppressive therapy
    Leukaemia
    Medulloblastoma
MalignantNon-malignant
Extrapelvic diseasesUni/bilateral oophorectomy
    Bone cancer (osteosarcoma—Ewing’s sarcoma)    Benign ovarian tumours
    Breast cancer    Severe and recurrent endometriosis
    Melanoma    BRCA-1 or BRCA-2 mutation carriers
    Neuroblastoma
    Bowel malignancy
Pelvic diseasesRisk of premature menopause
    Non-gynaecological malignancy    Turner’s syndrome
        Pelvic sarcoma    Family history
        Rhabdomyosarcoma Sacral tumours Rectosigmoid tumours    Benign diseases requiring chemotherapy: autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, Behçet’s disease and Wegener’s disease)
    Gynaecological malignancy
        Early cervical carcinoma
        Early vaginal carcinoma
        Early vulvar carcinoma
        Selected cases of ovarian carcinoma (stage IA)
        Ovarian borderline tumours
Systemic diseasesBone marrow transplantation
    Hodgkin’s disease    Benign haematological diseases: sickle cell anaemia, thalassaemia major and aplastic anaemia
    Non-Hodgkin’s lymphoma    Autoimmune diseases unresponsive to immunosuppressive therapy
    Leukaemia
    Medulloblastoma
Table II.

Indications for ovarian tissue cryopreservation in case of malignant and non-malignant diseases

MalignantNon-malignant
Extrapelvic diseasesUni/bilateral oophorectomy
    Bone cancer (osteosarcoma—Ewing’s sarcoma)    Benign ovarian tumours
    Breast cancer    Severe and recurrent endometriosis
    Melanoma    BRCA-1 or BRCA-2 mutation carriers
    Neuroblastoma
    Bowel malignancy
Pelvic diseasesRisk of premature menopause
    Non-gynaecological malignancy    Turner’s syndrome
        Pelvic sarcoma    Family history
        Rhabdomyosarcoma Sacral tumours Rectosigmoid tumours    Benign diseases requiring chemotherapy: autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, Behçet’s disease and Wegener’s disease)
    Gynaecological malignancy
        Early cervical carcinoma
        Early vaginal carcinoma
        Early vulvar carcinoma
        Selected cases of ovarian carcinoma (stage IA)
        Ovarian borderline tumours
Systemic diseasesBone marrow transplantation
    Hodgkin’s disease    Benign haematological diseases: sickle cell anaemia, thalassaemia major and aplastic anaemia
    Non-Hodgkin’s lymphoma    Autoimmune diseases unresponsive to immunosuppressive therapy
    Leukaemia
    Medulloblastoma
MalignantNon-malignant
Extrapelvic diseasesUni/bilateral oophorectomy
    Bone cancer (osteosarcoma—Ewing’s sarcoma)    Benign ovarian tumours
    Breast cancer    Severe and recurrent endometriosis
    Melanoma    BRCA-1 or BRCA-2 mutation carriers
    Neuroblastoma
    Bowel malignancy
Pelvic diseasesRisk of premature menopause
    Non-gynaecological malignancy    Turner’s syndrome
        Pelvic sarcoma    Family history
        Rhabdomyosarcoma Sacral tumours Rectosigmoid tumours    Benign diseases requiring chemotherapy: autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, Behçet’s disease and Wegener’s disease)
    Gynaecological malignancy
        Early cervical carcinoma
        Early vaginal carcinoma
        Early vulvar carcinoma
        Selected cases of ovarian carcinoma (stage IA)
        Ovarian borderline tumours
Systemic diseasesBone marrow transplantation
    Hodgkin’s disease    Benign haematological diseases: sickle cell anaemia, thalassaemia major and aplastic anaemia
    Non-Hodgkin’s lymphoma    Autoimmune diseases unresponsive to immunosuppressive therapy
    Leukaemia
    Medulloblastoma

Intensive chemotherapy and/or total body irradiation required before BMT constitute the treatment combination presenting the greatest risk of POF. Indeed, such high doses of chemotherapy (commonly using the highly cytotoxic cyclophosphamide/busulfan regimen) and/or radiotherapy lead to subsequent ovarian failure in almost all cases, children and adults alike (Sanders et al., 1996; Meirow and Nugent, 2001; Lobo, 2005). The risk of POF was estimated to be 92% in the study by Meirow and Nugent (2001), while Teinturier et al. (1998) actually reported 0% of ovarian recovery after busulfan treatment before BMT. A large retrospective survey of pregnancy outcomes after haematopoietic stem cell transplantation (HSCT) (peripheral blood or BMT) involving 37 362 patients revealed that only 0.6% of patients conceived after autologous or allogenic SCT (Salooja et al., 2001; Lutchman Singh et al., 2005). It is thus obvious that high doses of alkylating agents, irradiation and advancing age increase the risk of gonadal damage.

Cryopreservation should not be reserved solely for women with malignant disease (Donnez et al., 2006). Indeed, HSCT has been increasingly used in recent decades for non-cancerous diseases, such as benign haematological disease (sickle cell anaemia, thalassaemia major and aplastic anaemia) and autoimmune diseases previously unresponsive to immunosuppressive therapy (systemic lupus erythematosus and autoimmune thrombocytopenia) (Slavin et al., 2001; Mattle et al., 2005; Wolner-Hanssen et al., 2005; Donnez et al., 2006). Other benign diseases, such as recurrent ovarian endometriosis or recurrent ovarian mucinous cysts, are also indications for ovarian cryopreservation. Patients undergoing oophorectomy for prophylaxis may potentially benefit from ovarian cryopreservation too. The indications for cryopreservation of ovarian tissue in case of non-malignant disease are summarized in Table II.

Fertility preservation in cancer patients: different cryopreservation options

Several options are currently available to preserve fertility in cancer patients and give them the opportunity to become mothers when they have overcome their disease: embryo cryopreservation, oocyte cryopreservation or ovarian tissue cryopreservation (Gosden et al., 1997; Donnez and Bassil, 1998; Oktay et al., 1998a, 2004; Donnez et al., 2000, 2005; Oktay and Karlikaya, 2000; Meirow and Nugent, 2001; Torrents et al., 2003; Sonmezer and Oktay, 2004; Gosden, 2005; Lobo, 2005; Meirow et al., 2005; Kim, 2006). The choice of the most suitable strategy for preserving fertility depends on different parameters: the type and timing of chemotherapy, the type of cancer, the patient’s age and the partner status.

The only established method of fertility preservation is embryo cryopreservation, according to the Ethics Committee of the American Society for Reproductive Medicine (2005), but this option requires the patient to be of pubertal age, have a partner or use donor sperm, and be able to undergo a cycle of ovarian stimulation, which is not possible when the chemotherapy has to be initiated immediately or when stimulation is contraindicated according to the type of cancer.

Cryopreservation of oocytes can be performed in single women who can undergo a stimulation cycle, although the effectiveness of this technique is very low, with pregnancy and delivery rates ranging from 1 to 5% per frozen oocyte (Stachecki and Cohen, 2004; Borini et al., 2006; Levi Setti et al., 2006).

Cryopreservation of ovarian tissue is the only option available for prepubertal girls, and for woman who cannot delay the start of chemotherapy. Ovarian tissue can theoretically be frozen using three different approaches: as fragments of ovarian cortex, as entire ovary with its vascular pedicle or as isolated follicles. Human ovarian cryopreservation and transplantation procedures have so far been almost exclusively limited to avascular cortical fragments, both in experimental and in clinical studies, and, for now, this is the only procedure that has yielded live births in humans after autologous transplantation (Donnez et al., 2004; Meirow et al., 2005).

Embryo cryopreservation

Embryo cryopreservation has become a routine technique in all IVF centres and has proven its efficacy in terms of pregnancy and ‘take-home-baby’ rates. Although this method has already been used for young cancer patients (Winkel and Fossum, 1993), there are significant drawbacks to its use.

First, medical reasons might impede its application (a) if the beginning of cancer treatment cannot be delayed and there is no time to complete ovarian stimulation or (b) when the stimulation procedure may be theoretically harmful to patients with hormone-sensitive tumours, such as breast cancer. Even if IVF can theoretically be undertaken on the basis of a spontaneous ovarian cycle (Brown et al., 1996), the small number of obtainable oocytes (and subsequently viable embryos for transfer) makes it extremely unlikely that any live births will be achieved in these conditions.

Ginsburg et al. (2001) showed that women undergoing IVF after chemotherapy had a poor response to gonadotrophins and, more recently, Dolmans et al. (2005) clearly demonstrated the poor results of IVF and embryo cryopreservation, even if the attempt is carried out after only one or two regimens of chemotherapy. Recently, tamoxifen and letrozole were employed to stimulate the ovaries for IVF and embryo cryopreservation with some success, whilst possibly providing a safe alternative to traditional ovarian stimulation methods in these patients (Oktay et al., 2005).

The partner status of the patient may also impede embryo cryopreservation. If the patient has no partner or is an adolescent, the only available solution is using donor sperm to ensure fertilization of her oocytes. Finally, this technique is inappropriate for children, who have not reached puberty.

In conclusion, embryo cryopreservation is an efficient technique, but only an option for patients from whom mature oocytes can be collected and who have a partner (or are willing to use donor sperm).

Oocyte cryopreservation

Oocyte cryopreservation is an alternative option for patients with the same characteristics as those described above for embryo cryopreservation but who are not with a partner and do not wish to use donated sperm. In this case, IVF of their oocytes to produce embryos to be frozen for future implantation is not possible. Thus, the oocytes themselves must be cryopreserved either as mature or as immature oocytes. Human oocyte cryopreservation has been rapidly incorporated into clinical practice in several centres (Porcu et al., 1997, 2004; Fabbri et al., 2001; Porcu, 2005).

Mature oocyte cryopreservation

Mature oocyte freezing appears, at least in theory, to be the most logical way of storing female germ cells, comparable to the routinely performed sperm banking. It is an attractive option for women without a partner, if they have time to complete ovarian stimulation before cancer therapy. However, ovarian stimulation and oocyte collection are not applicable for children (Oktay et al., 1998a; Torrents et al., 2003; Akar and Oktay, 2005).

Still, this procedure has proved largely disappointing. Since the first report of a live birth from a frozen oocyte (Chen, 1986), the results of this procedure worldwide have been variable, with a reported success rate of <2% per thawed oocyte, despite an improved success rate when combined with ICSI (Porcu et al., 1997, 2004; Fabbri et al., 2001; Lutchman Singh et al., 2005; Porcu, 2005). Techniques developed in recent years to improve survival rates of mature oocytes after slow-freezing protocols include increased sucrose concentrations (Fabbri et al., 2001), longer pre-equilibration and thawing times (Yang et al., 2002) and the use of sodium-depleted media (Boldt et al., 2003; Stachecki and Cohen, 2004). Data on frozen-thawed mature oocytes from 21 studies in peer-reviewed journals were examined by Sonmezer and Oktay (2004), who reported a mean survival rate of 47%, a mean fertilization rate of 52.5% and a mean pregnancy rate per thawed oocyte of 1.52%. Very recent data indicate that, although the combination of slow cooling and high sucrose concentrations ensures high rates of oocyte survival, it is not sufficient to guarantee a high standard of clinical efficiency (Borini et al., 2006). Indeed, 18 clinical pregnancies were obtained from 927 oocytes in Borini’s study. In another very recent study, 2900 oocytes were cryopreserved, of which 1087 were subsequently thawed. In this series, 18 pregnancies were also obtained (Levi Setti et al., 2006). Even in the very latest studies, the mean pregnancy rate per thawed oocyte does not exceed 1.8% (Borini et al., 2006; Levi Setti et al., 2006).

The metaphase II (MII) oocyte is a large and highly specialized cell that is extremely fragile. Oocyte freezing is accompanied by various types of cell injury, which may explain the low survival rate (Van der Elst, 2003). There are two main reasons for these poor results. First, the zona pellucida hardens during the freezing process, probably as a consequence of premature exocytosis of the cortical granules. It could then act as a fence, impairing sperm penetration and normal fertilization, although micromanipulation techniques (ICSI) can, to a certain extent, bypass this problem (Porcu et al., 1997; Fabbri et al., 2001; Gook et al., 1993, 1995). Second, in the mature oocyte, the metaphase chromosomes are lined up by the meiotic spindle along the equatorial plate, but the spindle apparatus is easily damaged by intracellular ice formation during the freezing or thawing process (Pickering et al., 1990) (for review, see Mandelbaum et al., 2004). The cellular cooling process induces depolymerization of the meiotic spindle, which is a dynamic structure (microtubules being continually assembled at one of its ends and separated at the other). The cell is thus at risk of losing chromosomes and suffering aneuploidy.

There are two main steps in the process of cryopreservation: (i) chilling, i.e. lowering the temperature from the physiological temperature to the point of freezing and (ii) freezing, i.e. further reducing the temperature to the storage temperature (liquid nitrogen at –196°C). Chilling injury can modify the structure of membranes and therefore their integrity (Ghetler et al., 2005). It also affects oocyte microtubules (Albertini and Eppig, 1995), cytoskeletal organization (Overstrom et al., 1990) and the zona pellucida (Vajta et al., 1998).

Chilling injury is temperature dependent; it is caused by changes in membrane properties and integrity and is responsible for the extensive cell damage that occurs during the process of cryopreservation. Inappropriate handling of oocytes at room temperature appears to pose the greatest threat (Mandelbaum et al., 2004).

In conclusion, mature oocyte banking is still limited by its low success rate; oocytes are sensitive to chilling, often fail to survive freeze-thawing processes, and are susceptible to cytoskeletal damage and aneuploidy. Currently, even the most optimistic success rates offer patients only a slim chance of pregnancy, if few oocytes are available (Gosden, 2005). Ultrarapid freezing with vitrification may offer advantages over conventional slow cooling protocols by improving post-thawing survival rates but needs to be investigated further. Despite the few promising studies on vitrification (Yoon et al., 2000; Katayama et al., 2003), even less is known about the potentially detrimental effects of this process compared with conventional cryopreservation techniques; to date, slow freezing and rapid thawing is the protocol of choice for freezing human oocytes (Falcone et al., 2004). Better evaluation of sucrose concentrations in the freezing medium could lead to improved results in the future (Porcu, 2005; Borini et al., 2006).

Immature oocyte cryopreservation and in vitro maturation

Oocytes at the diplotene stage of prophase I, or germinal vesicle (GV) stage, survive the cryopreservation procedure better than those frozen at the MII stage (Boiso et al., 2002). These cells have reached full size and complete meiotic competence but have not yet resumed their maturation process and initiated their second metaphase. Although the risk of hardening of the zona pellucida or damage to the cytoskeleton cannot be avoided, it is probable that the absence of a meiotic spindle and the presence of a nuclear membrane protecting the chromatin guarantees the absence of cytogenetic anomalies during further cellular divisions.

Freezing immature oocytes followed by in vitro maturation thus offers practical and theoretical advantages (Gosden, 2005), but this method is still suboptimal. Frozen–thawed immature oocytes have to follow a process of in vitro maturation before they are ready to be fertilized. Oocyte maturation is considered as the reinitiation and completion of the first meiotic division from the GV stage to the MII stage, and the accompanying cytoplasmic maturation phase for fertilization and early embryonic development (Cha and Chian, 1998). The co-ordination of nuclear and cytoplasmic maturation in vitro has proved very difficult to achieve.

Although there are several reports of pregnancies achieved after in vitro maturation of fresh GV-stage oocytes (Cha et al., 1991; Trounson et al., 1994), only one live birth has resulted from an immature oocyte cryopreserved at the GV stage, with subsequent in vitro maturation (Tucker et al., 1998). Two other teams have described non-ongoing pregnancies. Kan et al. (2004) reported one pregnancy from an immature frozen oocyte which ended in a blighted ovum at 12 weeks, and Wu et al. (2001) reported one biochemical pregnancy. According to a recent review by Kim, the cryopreservation of GV-stage oocytes will not be a practicable strategy until in vitro maturation of these oocytes becomes more reliable (Kim, 2006).

Ovarian tissue cryopreservation

For patients who need immediate chemotherapy, ovarian tissue cryopreservation is the only possible alternative (Gosden et al., 1994; Donnez and Bassil, 1998; Meirow et al., 1998; Oktay et al., 1998a; Donnez et al., 2000, 2005). The main aim of this strategy is to reimplant cortical ovarian tissue into the pelvic cavity (orthotopic site) or a heterotopic site like the forearm or the abdominal wall once treatment is completed and the patient is disease-free (Donnez and Bassil, 1998; Oktay et al., 1998a, 2004; Oktay and Karlikaya, 2000; Radford et al., 2001; Donnez et al., 2004, 2005, 2006; Kim et al., 2004a; Meirow et al., 2005; Schmidt et al., 2005; Demeestere et al., 2006) (Table III).

Table III.

Autotransplantation of cryopreserved human ovarian tissue

ReferenceAge before freezingChemo before freezingIndicationGraft siteGraft sizeRecovery of ovarian functionOutcome
Oktay and Karlikaya (2000)29NoIntractable menometrorrhagiaOrthotopic: pelvic peritoneum8 pc (5–10 × 2 mm)16 weeks↑ E2, FD after stimulation, ovulation, menses
Radford et al. (2001)36YesHodgkin’s lymphomaOrthotopic: ovarian2 pc (10 × 5 × 1 mm)8 months↓ FSH and LH, ↑ E2, FD, ovulation, menses (1 cycle)
Callejo et al. (2001)47NoUterine leiomyomaHeterotopic: rectus abdominis muscle40–45 pc (2–3 mm3)3–4 months↑ E2, FD after stimulation (follicle of 20 mm)
Kim et al. (2004a)37NoSquamous cell cervical carcinoma (Ib)Heterotopic: breast [between breast tissue and pectoralis muscle (a)] + abdominal [rectus muscle (b)]40 pc (5 × 5 × 1–2 mm): 20 (a) + 20 (b)14 weeks↑ E2, ↓ FSH, FD (follicle of 11–16 mm) only in (b), ovulation
Oktay et al. (2004)30NoBreast cancerHeterotopic: s.c. abdominal wall15 pc (from 5 × 5 × 1 to 15 × 5 × 2 mm)3 monthsFD after gonadotrophin stimulation, oocyte retrieval, 20 oocytes, 8 IVF–ICSI, 4-cell embryo transfer + 1 aneuploidic embryo
Donnez et al. (2004)25NoHodgkin’s lymphomaOrthotopic: ovarian fossa peritoneum1 pc (12 × 4 × 1 mm) + 67 (35 + 32) pc (2 × 2 × 1 mm)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 22 mm), regular cycles (± 5 weeks), pregnancy, live birth
Meirow et al. (2005)28YesNon-Hodgkin’s lymphomaOrthotopic: ovarian3 pc (15 × 5 × 1–2 mm) + tiny fragments injected8 months↓ FSH, ↑AMH and inhibin B, FD, ovulation, menses, modified natural cycle, oocyte retrieval, 1 metaphase II oocyte IVF–ICSI, 4-cell embryo transfer, pregnancy and live birth
Schmidt et al. (2005)28NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)19/22 weeks↑ E2, ↓ FSH, FD (19 weeks), menses (22 weeks), no stimulation, empty follicle (of 20 mm)
25NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)18/25 weeks↑ E2 ↓ FSH, FD (18 weeks), menses (25 weeks), 2 IVF cycles, 2 metaphase II oocytes, 2-cell embryo
32NoNon-Hodgkin’s lymphomaOrthotopic: ovarian6 pc8/14 weeks↑ E2, ↓ FSH, FD (8 weeks), menses (14 weeks), 1 IVF cycle, 1 metaphase II + 1 GV oocyte, 4-cell embryo
Wolner-Hanssen et al. (2005)30NoPure red cell aplasia + BMTHeterotopic: s.c. forearm10 pc (1–2 × 1–2 × 0.5–1 mm)18 weeksFD (12.6 mm follicle and 6.7 mm follicle) after local gonadotrophin stimulation
Donnez et al. (2006)21NoSickle cell anaemia + BMTOrthotopic: ovarian (a) + ovarian fossa peritoneum (b)69 pc (2 × 2 × 1 mm): 45 (a) + 24 (b)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 20 mm), menses
Oktay (2006)29YesHodgkin’s lymphomaHeterotopic: s.c. abdominal wallLeft ovary in pieces2 months↑ E2, FD, ovulation, 6-week pregnancy and miscarriage, and pregnancy with live birth from native ovary
Demeestere et al. (2006)24YesHodgkin’s lymphomaOrthotopic: ovarian (a) + ovarian fossa peritoneum (b) and heterotopic: s.c. abdominal wall (c)18 pc (5 × 5 × 2 mm): 3 (a) + 9 (b) + 6 (c)4 months↑ inhibin B and E2, ↓ FSH, FD in all sites: (a) large follicles in ovarian site, (b) only one dominant follicle and (c) follicles <13 mm, 6 ovulations, natural conception, pregnancy, miscarriage at 7 weeks (aneuploidy)
Donnez et al. (unpublished data)24YesNon-Hodgkin’s lymphomaOrthotopic: ovarian5 pc (10 × 4–5 × 1 mm)5 months↑ E2, ↓ FSH and LH, FD (follicle of 21 mm), menses
ReferenceAge before freezingChemo before freezingIndicationGraft siteGraft sizeRecovery of ovarian functionOutcome
Oktay and Karlikaya (2000)29NoIntractable menometrorrhagiaOrthotopic: pelvic peritoneum8 pc (5–10 × 2 mm)16 weeks↑ E2, FD after stimulation, ovulation, menses
Radford et al. (2001)36YesHodgkin’s lymphomaOrthotopic: ovarian2 pc (10 × 5 × 1 mm)8 months↓ FSH and LH, ↑ E2, FD, ovulation, menses (1 cycle)
Callejo et al. (2001)47NoUterine leiomyomaHeterotopic: rectus abdominis muscle40–45 pc (2–3 mm3)3–4 months↑ E2, FD after stimulation (follicle of 20 mm)
Kim et al. (2004a)37NoSquamous cell cervical carcinoma (Ib)Heterotopic: breast [between breast tissue and pectoralis muscle (a)] + abdominal [rectus muscle (b)]40 pc (5 × 5 × 1–2 mm): 20 (a) + 20 (b)14 weeks↑ E2, ↓ FSH, FD (follicle of 11–16 mm) only in (b), ovulation
Oktay et al. (2004)30NoBreast cancerHeterotopic: s.c. abdominal wall15 pc (from 5 × 5 × 1 to 15 × 5 × 2 mm)3 monthsFD after gonadotrophin stimulation, oocyte retrieval, 20 oocytes, 8 IVF–ICSI, 4-cell embryo transfer + 1 aneuploidic embryo
Donnez et al. (2004)25NoHodgkin’s lymphomaOrthotopic: ovarian fossa peritoneum1 pc (12 × 4 × 1 mm) + 67 (35 + 32) pc (2 × 2 × 1 mm)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 22 mm), regular cycles (± 5 weeks), pregnancy, live birth
Meirow et al. (2005)28YesNon-Hodgkin’s lymphomaOrthotopic: ovarian3 pc (15 × 5 × 1–2 mm) + tiny fragments injected8 months↓ FSH, ↑AMH and inhibin B, FD, ovulation, menses, modified natural cycle, oocyte retrieval, 1 metaphase II oocyte IVF–ICSI, 4-cell embryo transfer, pregnancy and live birth
Schmidt et al. (2005)28NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)19/22 weeks↑ E2, ↓ FSH, FD (19 weeks), menses (22 weeks), no stimulation, empty follicle (of 20 mm)
25NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)18/25 weeks↑ E2 ↓ FSH, FD (18 weeks), menses (25 weeks), 2 IVF cycles, 2 metaphase II oocytes, 2-cell embryo
32NoNon-Hodgkin’s lymphomaOrthotopic: ovarian6 pc8/14 weeks↑ E2, ↓ FSH, FD (8 weeks), menses (14 weeks), 1 IVF cycle, 1 metaphase II + 1 GV oocyte, 4-cell embryo
Wolner-Hanssen et al. (2005)30NoPure red cell aplasia + BMTHeterotopic: s.c. forearm10 pc (1–2 × 1–2 × 0.5–1 mm)18 weeksFD (12.6 mm follicle and 6.7 mm follicle) after local gonadotrophin stimulation
Donnez et al. (2006)21NoSickle cell anaemia + BMTOrthotopic: ovarian (a) + ovarian fossa peritoneum (b)69 pc (2 × 2 × 1 mm): 45 (a) + 24 (b)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 20 mm), menses
Oktay (2006)29YesHodgkin’s lymphomaHeterotopic: s.c. abdominal wallLeft ovary in pieces2 months↑ E2, FD, ovulation, 6-week pregnancy and miscarriage, and pregnancy with live birth from native ovary
Demeestere et al. (2006)24YesHodgkin’s lymphomaOrthotopic: ovarian (a) + ovarian fossa peritoneum (b) and heterotopic: s.c. abdominal wall (c)18 pc (5 × 5 × 2 mm): 3 (a) + 9 (b) + 6 (c)4 months↑ inhibin B and E2, ↓ FSH, FD in all sites: (a) large follicles in ovarian site, (b) only one dominant follicle and (c) follicles <13 mm, 6 ovulations, natural conception, pregnancy, miscarriage at 7 weeks (aneuploidy)
Donnez et al. (unpublished data)24YesNon-Hodgkin’s lymphomaOrthotopic: ovarian5 pc (10 × 4–5 × 1 mm)5 months↑ E2, ↓ FSH and LH, FD (follicle of 21 mm), menses

AMH, anti-Müllerian hormones; BMT, bone marrow transplantation; E2, estradiol; FD, follicular development; GV, germinal vesicle; pc, pieces.

Table III.

Autotransplantation of cryopreserved human ovarian tissue

ReferenceAge before freezingChemo before freezingIndicationGraft siteGraft sizeRecovery of ovarian functionOutcome
Oktay and Karlikaya (2000)29NoIntractable menometrorrhagiaOrthotopic: pelvic peritoneum8 pc (5–10 × 2 mm)16 weeks↑ E2, FD after stimulation, ovulation, menses
Radford et al. (2001)36YesHodgkin’s lymphomaOrthotopic: ovarian2 pc (10 × 5 × 1 mm)8 months↓ FSH and LH, ↑ E2, FD, ovulation, menses (1 cycle)
Callejo et al. (2001)47NoUterine leiomyomaHeterotopic: rectus abdominis muscle40–45 pc (2–3 mm3)3–4 months↑ E2, FD after stimulation (follicle of 20 mm)
Kim et al. (2004a)37NoSquamous cell cervical carcinoma (Ib)Heterotopic: breast [between breast tissue and pectoralis muscle (a)] + abdominal [rectus muscle (b)]40 pc (5 × 5 × 1–2 mm): 20 (a) + 20 (b)14 weeks↑ E2, ↓ FSH, FD (follicle of 11–16 mm) only in (b), ovulation
Oktay et al. (2004)30NoBreast cancerHeterotopic: s.c. abdominal wall15 pc (from 5 × 5 × 1 to 15 × 5 × 2 mm)3 monthsFD after gonadotrophin stimulation, oocyte retrieval, 20 oocytes, 8 IVF–ICSI, 4-cell embryo transfer + 1 aneuploidic embryo
Donnez et al. (2004)25NoHodgkin’s lymphomaOrthotopic: ovarian fossa peritoneum1 pc (12 × 4 × 1 mm) + 67 (35 + 32) pc (2 × 2 × 1 mm)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 22 mm), regular cycles (± 5 weeks), pregnancy, live birth
Meirow et al. (2005)28YesNon-Hodgkin’s lymphomaOrthotopic: ovarian3 pc (15 × 5 × 1–2 mm) + tiny fragments injected8 months↓ FSH, ↑AMH and inhibin B, FD, ovulation, menses, modified natural cycle, oocyte retrieval, 1 metaphase II oocyte IVF–ICSI, 4-cell embryo transfer, pregnancy and live birth
Schmidt et al. (2005)28NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)19/22 weeks↑ E2, ↓ FSH, FD (19 weeks), menses (22 weeks), no stimulation, empty follicle (of 20 mm)
25NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)18/25 weeks↑ E2 ↓ FSH, FD (18 weeks), menses (25 weeks), 2 IVF cycles, 2 metaphase II oocytes, 2-cell embryo
32NoNon-Hodgkin’s lymphomaOrthotopic: ovarian6 pc8/14 weeks↑ E2, ↓ FSH, FD (8 weeks), menses (14 weeks), 1 IVF cycle, 1 metaphase II + 1 GV oocyte, 4-cell embryo
Wolner-Hanssen et al. (2005)30NoPure red cell aplasia + BMTHeterotopic: s.c. forearm10 pc (1–2 × 1–2 × 0.5–1 mm)18 weeksFD (12.6 mm follicle and 6.7 mm follicle) after local gonadotrophin stimulation
Donnez et al. (2006)21NoSickle cell anaemia + BMTOrthotopic: ovarian (a) + ovarian fossa peritoneum (b)69 pc (2 × 2 × 1 mm): 45 (a) + 24 (b)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 20 mm), menses
Oktay (2006)29YesHodgkin’s lymphomaHeterotopic: s.c. abdominal wallLeft ovary in pieces2 months↑ E2, FD, ovulation, 6-week pregnancy and miscarriage, and pregnancy with live birth from native ovary
Demeestere et al. (2006)24YesHodgkin’s lymphomaOrthotopic: ovarian (a) + ovarian fossa peritoneum (b) and heterotopic: s.c. abdominal wall (c)18 pc (5 × 5 × 2 mm): 3 (a) + 9 (b) + 6 (c)4 months↑ inhibin B and E2, ↓ FSH, FD in all sites: (a) large follicles in ovarian site, (b) only one dominant follicle and (c) follicles <13 mm, 6 ovulations, natural conception, pregnancy, miscarriage at 7 weeks (aneuploidy)
Donnez et al. (unpublished data)24YesNon-Hodgkin’s lymphomaOrthotopic: ovarian5 pc (10 × 4–5 × 1 mm)5 months↑ E2, ↓ FSH and LH, FD (follicle of 21 mm), menses
ReferenceAge before freezingChemo before freezingIndicationGraft siteGraft sizeRecovery of ovarian functionOutcome
Oktay and Karlikaya (2000)29NoIntractable menometrorrhagiaOrthotopic: pelvic peritoneum8 pc (5–10 × 2 mm)16 weeks↑ E2, FD after stimulation, ovulation, menses
Radford et al. (2001)36YesHodgkin’s lymphomaOrthotopic: ovarian2 pc (10 × 5 × 1 mm)8 months↓ FSH and LH, ↑ E2, FD, ovulation, menses (1 cycle)
Callejo et al. (2001)47NoUterine leiomyomaHeterotopic: rectus abdominis muscle40–45 pc (2–3 mm3)3–4 months↑ E2, FD after stimulation (follicle of 20 mm)
Kim et al. (2004a)37NoSquamous cell cervical carcinoma (Ib)Heterotopic: breast [between breast tissue and pectoralis muscle (a)] + abdominal [rectus muscle (b)]40 pc (5 × 5 × 1–2 mm): 20 (a) + 20 (b)14 weeks↑ E2, ↓ FSH, FD (follicle of 11–16 mm) only in (b), ovulation
Oktay et al. (2004)30NoBreast cancerHeterotopic: s.c. abdominal wall15 pc (from 5 × 5 × 1 to 15 × 5 × 2 mm)3 monthsFD after gonadotrophin stimulation, oocyte retrieval, 20 oocytes, 8 IVF–ICSI, 4-cell embryo transfer + 1 aneuploidic embryo
Donnez et al. (2004)25NoHodgkin’s lymphomaOrthotopic: ovarian fossa peritoneum1 pc (12 × 4 × 1 mm) + 67 (35 + 32) pc (2 × 2 × 1 mm)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 22 mm), regular cycles (± 5 weeks), pregnancy, live birth
Meirow et al. (2005)28YesNon-Hodgkin’s lymphomaOrthotopic: ovarian3 pc (15 × 5 × 1–2 mm) + tiny fragments injected8 months↓ FSH, ↑AMH and inhibin B, FD, ovulation, menses, modified natural cycle, oocyte retrieval, 1 metaphase II oocyte IVF–ICSI, 4-cell embryo transfer, pregnancy and live birth
Schmidt et al. (2005)28NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)19/22 weeks↑ E2, ↓ FSH, FD (19 weeks), menses (22 weeks), no stimulation, empty follicle (of 20 mm)
25NoHodgkin’s lymphomaOrthotopic: ovarian (a) + peritoneum (b) and heterotopic: abdominal wall (c)12 pc: 4 (a) + 4 (b) + 4 (c)18/25 weeks↑ E2 ↓ FSH, FD (18 weeks), menses (25 weeks), 2 IVF cycles, 2 metaphase II oocytes, 2-cell embryo
32NoNon-Hodgkin’s lymphomaOrthotopic: ovarian6 pc8/14 weeks↑ E2, ↓ FSH, FD (8 weeks), menses (14 weeks), 1 IVF cycle, 1 metaphase II + 1 GV oocyte, 4-cell embryo
Wolner-Hanssen et al. (2005)30NoPure red cell aplasia + BMTHeterotopic: s.c. forearm10 pc (1–2 × 1–2 × 0.5–1 mm)18 weeksFD (12.6 mm follicle and 6.7 mm follicle) after local gonadotrophin stimulation
Donnez et al. (2006)21NoSickle cell anaemia + BMTOrthotopic: ovarian (a) + ovarian fossa peritoneum (b)69 pc (2 × 2 × 1 mm): 45 (a) + 24 (b)4½ months↑ E2, ↓ FSH and LH, FD (follicle of 20 mm), menses
Oktay (2006)29YesHodgkin’s lymphomaHeterotopic: s.c. abdominal wallLeft ovary in pieces2 months↑ E2, FD, ovulation, 6-week pregnancy and miscarriage, and pregnancy with live birth from native ovary
Demeestere et al. (2006)24YesHodgkin’s lymphomaOrthotopic: ovarian (a) + ovarian fossa peritoneum (b) and heterotopic: s.c. abdominal wall (c)18 pc (5 × 5 × 2 mm): 3 (a) + 9 (b) + 6 (c)4 months↑ inhibin B and E2, ↓ FSH, FD in all sites: (a) large follicles in ovarian site, (b) only one dominant follicle and (c) follicles <13 mm, 6 ovulations, natural conception, pregnancy, miscarriage at 7 weeks (aneuploidy)
Donnez et al. (unpublished data)24YesNon-Hodgkin’s lymphomaOrthotopic: ovarian5 pc (10 × 4–5 × 1 mm)5 months↑ E2, ↓ FSH and LH, FD (follicle of 21 mm), menses

AMH, anti-Müllerian hormones; BMT, bone marrow transplantation; E2, estradiol; FD, follicular development; GV, germinal vesicle; pc, pieces.

Other strategies, such as the transplantation of isolated cryopreserved primordial follicles and the transplantation of cryopreserved whole ovary, will also be discussed here.

Fragments of cortical ovarian tissue

Lessons learned from auto- and xenografting of cryopreserved animal ovarian tissue

To date, ovarian tissue has been successfully cryopreserved and transplanted into rodents, rabbits, sheep, and marmoset monkeys (Candy et al., 1995, 2000; Salle et al., 2002; Almodin et al., 2004). Successful fertilization and pregnancy after oocyte collection from fresh transplanted ovarian tissue have been described in a primate (Lee et al., 2004); the grafted tissue functioned without any surgical connection to major blood vessels.

The generation of live young from xenografted mouse ovarian tissue into a rat recipient was described by Snow et al. (2002). Fresh mouse ovarian tissue was xenografted under the kidney capsule of rat recipients. Mature oocytes were produced from the xenografted tissue and subsequently fertilized; they developed into fertile adult mice. The authors state that xenotransplantation may thus prove to be a potentially promising method for the conservation of rare and endangered species of animals. The use of this strategy in human assisted reproduction should be considered with caution, however (Snow et al., 2002).

In a recent review of their experimental studies, Baird et al. (2004) observed a significant increase in the proportion of growing follicles from <20% in ovaries before grafting to >70% at 7, 30 and 60 days after grafting. According to the authors, the massive recruitment of primordial follicles, which also occurs in cultured fragments, suggests the removal of some inhibitory mechanisms regulating FSH. Following autotransplantation, the number of antral follicles and the secretion of inhibin A are reduced, resulting in raised basal levels of FSH, which could account for the massive recruitment, although the early stage of folliculogenesis can occur in the absence of FSH and LH.

Experimental studies have indicated that the fall in the number of primordial follicles in grafted tissue is due to hypoxia and the delay that occurs before reimplanted cortical tissue becomes revascularized. The loss of primordial follicles in cryopreserved ovarian tissue after transplantation is estimated to be 50–65% in some studies (Baird et al., 1999; Nisolle et al., 2000) and >90% in one study (Aubard et al., 1999).

A very recent study in mice concluded that the graft site affects the number and quality of oocytes produced from ovarian grafts (Yang et al., 2006). The study used a mouse ovarian grafting model to investigate whether the graft site (bursal cavity, kidney capsule or site) influences the number, fertilization rate and developmental potential of oocytes recovered from grafts. Graft retrieval and the number of oocytes found in each graft were lowest from the s.c. graft site. The number of 2-cell embryos produced was significantly higher with oocytes from grafts to the bursa, compared with the other sites.

In a recent article, Israely et al. (2004) analyzed angiogenic events following ovary xenotransplantation. Rat ovaries were transplanted into the muscle of castrated nude mice. The characterization of the neovasculature by dynamic contrast-enhanced magnetic resonance imaging (MRI) confirmed that the graft was devoid of any blood supply. Functional vessels within the graft were detected by MRI and histology from day 7 onwards. By 2–3 weeks, both the blood volume fraction and the permeability were measured using albumin-based MR contrast material and were found to be higher in the graft than in adjacent muscle. The same team recently demonstrated that transplantation into angiogenic granulation tissue created during wound healing shortened the ischaemic interval (Israely et al., 2006). This confirms the benefits of the technique of induced angiogenesis and neovascularization that was used in the first case of successful orthotopic transplantation of cryopreserved ovarian tissue (Donnez et al., 2004).

The assessment of tumour oxygenation by electro-paramagnetic resonance (EPR) was recently reviewed by Gallez et al. (2004). In this review, different methods were used to estimate the oxygenation of tumours and compare EPR oximetry with nuclear magnetic resonance and MRI. The evaluation of oximetry in human ovarian xenografts was initiated to improve the revascularization process, which remains one of the main limitations of ovarian cortical strip use (Donnez et al., 2005).

Lessons learned from xenografting cryopreserved human ovarian tissue

Human ovarian tissue can be successfully cryopreserved, showing good survival and function after thawing. Hovatta (2005) arrived at this conclusion after reviewing all relevant studies since 1996, when the first case of cryopreservation of human ovarian tissue was described. In these studies, survival and morphological and functional recovery of ovarian tissue after thawing were proved by live/dead assays, organ culture, xenotransplantation to severe combined immunodeficient (SCID) mice and normal light and transmission electron microscopy (TEM) analysis.

Adequate penetration of cryoprotectant through the stroma and granulosa cells to the oocytes is necessary, however, while at the same time avoiding possible cryoprotectant toxicity. Indeed, Newton et al. (1996, 1998) demonstrated the importance of the diffusion rate and the diffusion temperature. Ice crystal formation must also be minimized by choosing optimal freezing and thawing rates. The choice of cryoprotectant with maximum permeation capacity but minimum toxicity and ice crystal formation potential is specific to each cell and tissue type (Fuller and Paynter, 2004). Thus, in the ovary, it is a compromise between the stroma, the follicular cells and the oocytes (Hovatta, 2005). On the basis of current knowledge, the standard method for human ovarian cryopreservation is slow-programmed freezing, using human serum albumin-containing medium, and propanediol, dimethylsulphoxide or ethylene glycol as a cryoprotectant, combined or not with sucrose (Hovatta, 2005).

Kim et al. (2004b) showed that a correlation exists between ischaemic tissue damage and the duration of ischaemia. It has been demonstrated that vascular endothelial growth factor does not accelerate revascularization, either after s.c. (Schnorr et al., 2002) or peritoneal (Donnez et al., personal data) implantation of ovarian tissue. According to Kim et al. (2004b), the ovarian cortex can tolerate ischaemia for at least 3 h at 4°C. They also demonstrated that an antioxidant (ascorbic acid) reduces apoptosis in ovarian cortex by up to 24 h in case of incubation in vitro. In their study, significant ischaemic damage was evidenced by decreased oxygen consumption and increased apoptosis. Moreover, it appeared that stromal cells were more vulnerable to ischaemia than primordial follicles. The apoptosis rate of stromal cells was higher in the frozen-thawed group than in the fresh group, regardless of the duration of incubation, which may reflect a degree of freeze-thawing injury to ovarian cortex.

Several immunodeficient animal strains can be used as xenograft recipients, with SCID and non-obese diabetic-SCID mice best suited to xenografting to obviate potential rejection problems, especially if the graft is to be borne for a long period (Weissman et al., 1999; Aubard, 2003). All reports on human ovarian tissue grafting to mice (for review, see Aubard, 2003) that have studied the implantation site have shown peritoneal transplantation, either under the peritoneum (Nisolle et al., 2000) or under the kidney capsule (Abir et al., 2003; Hernandez-Fonseca et al., 2004) to be better than s.c. transplantation in terms of follicular survival and development. After xenografting cryopreserved human ovarian tissue into the kidney capsule of SCID mice, Oktay et al. (1998b) achieved follicular development up to the antral stage, and (Gook et al. 2003, 2005) obtained MII-stage oocytes. The ovulatory capacity of frozen-thawed human follicles in xenografts was evidenced by the formation of morphologically normal corpora lutea and elevated progesterone levels in immunodeficient mice (Kim et al., 2002; Gook et al., 2003).

Very few data are available on the final maturation of follicles in xenografts and therefore the quality of oocytes obtained. In a recent study, Kim et al. (2005) assessed the integrity of human oocytes obtained after s.c. xenografting of cryopreserved ovarian tissue into SCID mice and in vitro maturation of retrieved cumulus–oocyte complexes. By immunocytochemical analysis of microtubules and DNA, some oocytes showed abnormal nuclear and cytoplasmic maturation. Nevertheless, the authors could not determine whether these alterations were due to freeze–thawing injury, the lack of optimal ovarian stimulation protocols, suboptimal conditions of animal hosts for the growth of human follicles, inadequate in vitro maturation techniques or the choice of a s.c. transplantation site. It has therefore not been established whether human oocytes matured in a xenograft are ultrastructurally normal and functionally competent.

Autotransplantation of cryopreserved human ovarian tissue

Reported cases of autotransplantation of cryopreserved ovarian tissue, either to an orthotopic or to a heterotopic site, are summarized in Table III, detailing in each case the age of the patient before freezing, whether the patient received chemotherapy before freezing, the indications for cryopreservation, the graft site and size, the interval before recovery of ovarian function after grafting and the outcome of transplantation (Oktay and Karlikaya, 2000; Callejo et al., 2001; Radford et al., 2001; Kim et al., 2004a; Oktay et al., 2004; Oktay, 2006; Donnez et al., 2004, 2006; unpublished data;Meirow et al., 2005; Schmidt et al., 2005; Wolner-Hanssen et al., 2005; Demeestere et al., 2006).

Orthotopic autotransplantation of cryopreserved human ovarian tissue

In theory, natural pregnancy may be achieved via orthotopic tissue transplantation if the fallopian tubes remain intact.

In 2000, Oktay and Karlikaya (2000) reported laparoscopic transplantation of frozen–thawed ovarian tissue to the pelvic side wall in a 29-year-old patient, who had undergone bilateral oophorectomy for a non-malignant disease. Pieces of cryopreserved ovarian tissue were thawed and transplanted. The patient was stimulated by gonadotrophins once after 15 weeks and then again after 10 months. Follicular development was demonstrated by ultrasonography and ovulation occurred in response to HCG administration.

Radford et al. (2001) reported a patient with a history of Hodgkin’s disease treated by chemotherapy, whose ovarian tissue had been biopsied and cryopreserved 4 years after chemotherapy and later reimplanted. In this case, histological section of the ovarian cortical tissue revealed only a few primordial follicles because of the previous chemotherapy. Estradiol (E2) was detected and the FSH and LH levels decreased 8 months after reimplantation. The patient had one menstrual period, but 9 months after reimplantation, her LH and FSH concentrations returned definitively to menopausal levels.

We reported the first successful transplantation of cryopreserved ovarian tissue (Donnez et al., 2004) resulting in a pregnancy and live birth. In 1997, a 25-year-old woman presented with clinical stage IV Hodgkin’s lymphoma. Ovarian tissue cryopreservation was undertaken before chemotherapy. By laparoscopy, we took five cortical biopsies, about 12–15 mm long and 5 mm wide, from the left ovary. The removal of the whole ovary was not an option because one can never completely exclude recovery of ovarian function after chemotherapy.

After laparoscopy, the patient received hybrid chemotherapy from August 1997 to February 1998, followed by supradiaphragmatic radiotherapy (38 Gy).

According to Schilsky et al. (1981), the risk of POF after such a regimen in a woman of 26 years of age is >90%, whereas according to Wallace et al. (2005a) and Lobo (2005), the risk of subfertility after Hodgkin’s treatment with alkylating agents is >80%. Indeed, not only the type of drug and dose but also the age are important factors when evaluating the risk of POF after chemotherapy.

In 2003, once the patient had been declared completely disease-free, transplantation went ahead. A large strip and 35 small cubes of frozen–thawed ovarian tissue were implanted into a furrow created by the peritoneal window very close to the ovarian vessels and fimbria on the right side. Four months after transplantation, a laparoscopy was carried out to check the viability of the orthotopic graft and to reimplant the remaining 32 ovarian cortical cubes. A follicular structure was visible in the area where the tissue had been reimplanted, clearly outside the native ovary. Biopsy and analysis by vital fluorescent probe staining and histology revealed the presence of viable primordial follicles and a follicular structure with inhibin A-marked cells (Donnez et al., 2004). From 5 to 9 months after reimplantation, concentrations of FSH, E2 and progesterone showed the occurrence of ovulatory cycles. At 11 months, the patient became pregnant and subsequently delivered a healthy baby.

Several lines of evidence lend support to our assertion that the origin of the pregnancy was indeed the autotransplanted cryopreserved tissue. The possibility that the egg was derived from the native ovary is highly unlikely, because vaginal echography demonstrated the development of a follicle of 18 × 22 mm in size outside the native ovary during the cycle which led to the pregnancy. The same day, the E2 level was 156 pg/ml and progesterone 0 ng/ml. This was extensively explained in the publication itself (Donnez et al., 2004), as well as in a letter later published by Donnez and Dolmans (2004) in response to comments by Oktay and Tilly (2004). Another of our main arguments was that laparoscopy performed 4 months after reimplantation proved, by direct visualization, the development of a follicle from the grafted tissue and, on histological examination, the biopsy samples indicated not only the survival of primordial follicles in the grafted tissue but also the maturation of a follicle (granulosa cells marked by inhibin A). It was the first histological proof of follicular maturation from reimplanted cryopreserved ovarian tissue. After delivery, the patient experienced ovulatory cycles every 5 to 6 weeks. Each time, an increase in FSH levels was observed (Figure 1). Figure 1 demonstrates that the ovarian graft is still functioning almost 3 years after transplantation.

Figure 1.

Post-partum LH (mIU/ml), FSH (mIU/ml) and estradiol (pg/ml) levels in the first case of successful cryopreserved ovarian tissue transplantation, demonstrating that the graft is still active (pre-pregnancy hormone levels were published in Donnez et al., 2004).

In 2006, the restoration of ovarian function after orthotopic (intraovarian and paraovarian) transplantation of cryopreserved ovarian tissue was reported in a woman treated by BMT for a non-cancerous disease (sickle cell anaemia) (Donnez et al., 2006). Thirty-nine small cryopreserved cubes were thawed and grafted into the ovary itself (24 cubes) and a peritoneal window (15 cubes). Vaginal echography and sequential measurement of FSH, LH, E2 and progesterone concentrations revealed the onset of an ovulatory cycle 4 months after the reimplantation of ovarian tissue, demonstrating the efficacy of orthotopic transplantation and confirming, once again, the time interval between reimplantation and the onset of ovulation. The patient experienced 3 cycles in total, evidenced by the development of a follicle and raised E2 levels. It should be noted that the E2 peaks never exceeded 55 pg/ml and that FSH rose to 40 mIU/ml between the cycles. However, after these 3 cycles, LH and FSH concentrations returned to castrated levels. We then decided to reimplant the 30 remaining cubes into the ovary, the patient being under GnRH agonist to decrease LH and FSH levels. She experienced a first ovulatory cycle 4 months after reimplantation. The pre-ovulatory E2 level was 120 pg/ml. The follicle measured 20 mm before the LH peak and progesterone was at 14.7 ng/ml in the mid-luteal phase (Figure 2).

Figure 2.

LH (mIU/ml), FSH (mIU/ml) and estradiol (pg/ml) levels in our second case of cryopreserved ovarian tissue transplantation. Four-and-a-half months after reimplantation, the patient experienced 3 consecutive ovulatory cycles, evidenced by the development of a follicle and raised estradiol levels (published by Donnez et al., 2006). Subsequently, FSH returned to castrated levels, and a second reimplantation was carried out. Four months later, the patient experienced a further ovulatory cycle (unpublished data).

Very recently, we applied a technique, similar to that used by Silber et al. (2005) for the transplantation of fresh ovarian cortex between monozygotic twins, in a woman who had also undergone BMT and two regimens of alkylating agents in 2000 for non-Hodgkin’s lymphoma (Table III). Ovarian tissue cryopreservation was carried out 1 year after first-line chemotherapy. One ovary was removed and biopsies of cortical ovarian tissue revealed the presence of histologically normal primordial follicles. Six ovarian cortical pieces measuring 10 × 4–5 mm were then grafted onto the remaining ovary after the cortex of this ovary had been removed (Figure 3). It was 5 months before a mature follicle (21 mm) developed and an increase in E2 levels (194 pg/ml) was noted. The patient experienced an ovulatory cycle every 5 weeks, the pre-ovulatory E2 level reaching values between 210 and 356 pg/ml.

Figure 3.

Ovarian cortical pieces measuring 4–5 mm to 1 cm in size were grafted onto the remaining ovary after the cortex of this ovary had been removed. (a) Cortex of the remaining ovary was removed. (b) Cortical pieces were sutured with 7-0 stitches.

The analysis of these cases raises some important points for discussion. First, in all three cases, it took between 4½ and 5 months after reimplantation before a follicle could be seen. The process of folliculogenesis takes ∼4–6 months, during which time the oocyte and surrounding somatic cells undergo a series of changes that eventually result in the development of a large antral follicle, capable of producing a mature oocyte (Gougeon, 1996). Thus, the appearance of the first follicle originating from the grafted tissue 5 months after reimplantation, proved by laparoscopy in one case, is totally consistent with the expected time course. This time interval between implantation of cortical tissue and the first E2 peak is also consistent with data obtained from sheep (Baird et al., 1999, 2004) and human beings (Table III), although some variations may be observed. Indeed, as summarized in Table III, the delay between transplantation and follicular development was found varying from 8 weeks to 8 months. Such a variation could be explained by a difference in follicular reserve at the time of cryopreservation.

Another very interesting finding is the persistence of relatively high FSH levels during the follicular phase. FSH levels remained as high as 25 mIU/ml during the follicular phase until ovulation and then decreased to <15 mIU/ml during the luteal phase. This may constitute an argument against the use of gonadotrophin injections. The relatively high FSH levels may be explained by the relatively low number of surviving primordial follicles in the graft. The patient should be considered a poor responder, with reduced inhibin B secretion. These results are in agreement with those obtained in sheep by Campbell et al. (2000).

A further significant observation is the return to an FSH level of >35 mIU/ml immediately after each menstrual bleed, which supports the theory suggested by Baird et al. (2004) that some inhibitory mechanisms, such as inhibin or anti-Müllerian hormones (AMH) normally produced by developing follicles in intact human ovaries, are probably almost non-existent in transplanted tissue. After transplantation, the patient would have been regarded a poor responder because, of the 500–1000 primordial follicles that would have been transplanted, >50% would have been lost owing to hypoxia (Donnez et al., 2004). This raises the question of the evaluation of the ovarian reserve. There is a lack of data on the ovarian reserve in cancer. Qu et al. (2000), Gook et al. (2005) and Schmidt et al. (2003) have all demonstrated an unequal distribution of primordial follicles in ovarian cortex.

In 2005, Meirow et al. also published a live birth after orthotopic autotransplantation of cryopreserved ovarian tissue in a patient with POF after chemotherapy. Eight months after orthotopic transplantation, the patient spontaneously menstruated. The rise in AMH and increased inhibin B levels were consistent with the presence of early growing follicles and ovulation, respectively. Nine months after transplantation, the patient experienced a second spontaneous menstrual period. After a modified natural cycle, a single mature oocyte was retrieved and fertilized. Two days later, a 4-cell embryo was transferred. The patient became pregnant from this embryo transfer and delivered a healthy infant weighing 3000 g. The possibility that the oocyte was derived from the native ovary is highly unlikely given the consistent evidence of POF after high-dose chemotherapy in this patient, from whom ovarian tissue was harvested after administration of a first-line conventional chemotherapy regimen, before second-line high-dose chemotherapy.

Schmidt et al. (2005) recently reported the results of three cases of ovarian tissue transplantation. All three patients with autotransplanted ovarian tissue regained ovarian function, as confirmed by the recovery of menses, follicles visible on ultrasonography and normal hormone levels. Two embryos were obtained from three MII oocytes and one GV oocyte, but no pregnancy resulted from embryo transfer.

Demeestere et al. (2006) very recently reported a pregnancy after natural conception in a woman who had undergone orthotopic and heterotopic transplantation of cryopreserved ovarian tissue. They observed follicular development in all three transplantation sites: large follicles in the ovarian site, only one dominant follicle in the peritoneal site and follicles <13 mm in size in the heterotopic site. Detectable HCG levels and ultrasonography confirmed the presence of a viable intrauterine pregnancy. Unfortunately, this pregnancy, obtained by natural conception, ended in miscarriage at 7 weeks due to aneuploidy. Interestingly, Demeestere et al. (2006) observed normal FSH values after orthotopic and with heterotopic transplantation of cryopreserved ovarian tissue. As stressed by the authors, this may have been due to the young age of the patient and the large number of tissue fragments transplanted (Demeestere et al., 2006), which could have yielded a rich follicular reserve in the graft.

Heterotopic autotransplantation of cryopreserved human ovarian tissue: a better option than orthotopic transplantation?

There are only six existing reports on this subject (Table III). Callejo et al. (2001) evaluated the long-term function of cryopreserved heterotopic grafts, but no conclusions could be drawn because the patient was perimenopausal at the time of ovarian biopsy for cryopreservation.

In 2004, Kim et al. (2004a) reported a case of a 37-year-old woman who underwent heterotopic (rectus and pectoralis muscle) transplantation of cryopreserved ovarian tissue. By 14 weeks of transplantation, the restoration of endocrine function was demonstrated but, ∼28 weeks after transplantation, the cessation of ovarian function was evidenced by very high FSH levels (62–99 IU/l) and very low E2 levels.

The same year, Oktay et al. (2004) reported transplantation of frozen–thawed ovarian tissue beneath the skin of the abdomen. A 4-cell embryo was obtained from 20 oocytes retrieved from an ovarian graft, but no pregnancy occurred after transfer. Oocyte quality might have been compromised by transplantation to a heterotopic site.

In 2005, Schmidt et al. reported two cases of mixed (heterotopic and orthotopic) transplantation, as did Demeestere et al. in 2006. These cases are discussed in the section on orthotopic transplantation.

Wolner-Hanssen et al. (2005) reported s.c. transplantation of frozen–thawed tissue to the forearm. Two follicles developed, but only to a maximum diameter of 12.6 and 6.7 mm, respectively, and the tissue survived 7 months. The authors suggested that the risk of graft exposure to suboptimal temperatures or mechanical stress may depend on transplantation site, and thus tissue transplanted under the skin of the forearm will probably be exposed to both higher pressure and lower temperature than ovaries in their normal location.

Very recently, Oktay (2006) reported a pregnancy after heterotopic transplantation of cryopreserved ovarian tissue, but ovulation occurred from the native ovary.

Papers describing heterotopic transplantation have all reported follicular development, but with follicles always <15 mm in size. As stressed by Wolner-Hanssen et al. (2005) and Oktay et al. (2004), differences in temperature and pressure could interfere with follicular development in heterotopic sites.

Isolated primordial follicles

The primordial follicle is resistant to cryoinjury, because the oocyte it contains has a relatively inactive metabolism, as well as a lack of meiotic spindle, zona pellucida and cortical granules. The small size of primordial follicles also greatly facilitates penetration of cryoprotectant. Oktay et al. (1997) developed an isolation technique for human primordial follicles using enzymatic digestion and microdissection and obtained high follicular viability rates with both fresh and frozen ovarian tissues.

Nevertheless, the procedure to isolate primordial follicles remains difficult (Martinez-Madrid et al., 2004a); to date, it has not been possible to grow human isolated primordial follicles in vitro to the mature oocyte stage (Torrents et al., 2003).

Although safe xenotransplantation of ovarian tissue from lymphoma patients has been reported in SCID mice (Kim et al., 2001), the possibility of reintroducing tumour cells into cancer patients by autografting of ovarian tissue cannot be excluded. To avoid transferring malignant cells, ovarian tissue culture with in vitro follicle maturation could be performed. Culturing isolated follicles from the primordial stage is another particularly attractive proposition because they represent >90% of the total follicular reserve and show high cryotolerance (Smitz and Cortvrindt, 2002). However, isolated primordial follicles do not grow properly in culture (Hovatta et al., 1999; Abir et al., 2001), and further studies are clearly needed to identify factors sustaining follicular growth and maturation in humans (Smitz and Cortvrindt, 2002) and to assess the contribution of stromal cells to these processes. Encouraging results were achieved by Hovatta (2004) when human primordial follicles were grown in organ culture. Follicle isolation, or partial follicle isolation, severely impairs follicular viability in culture and, after isolation, primordial and primary follicles degenerate within the first 24 h of culture (Hovatta et al., 1999; Abir et al., 2001). Only more advanced, multilaminar pre-antral follicular stages can survive in short-term culture, a few reaching the early antral stage (Roy and Treacy, 1993; Abir et al., 1997).

Another approach could be to transplant a suspension of isolated follicles. As the follicular basal lamina encapsulating the membrana granulosa excludes capillaries, white blood cells and nerve processes from the granulosa compartment (Motta et al., 2003; Rodgers et al., 2003), grafting fully isolated follicles could be considered safer. Moreover, this would allow the introduction of a high and known number of follicles into the host, obtaining faster angiogenesis and minimizing ischaemic and reperfusion damage (Laschke et al., 2002). The transplantation of frozen–thawed isolated primordial follicles has indeed been successfully achieved in mice (Carroll and Gosden, 1993), yielding normal offspring. For human primordial follicles, however, mechanical isolation is not possible due to their size (30–40 µm) and their fibrous and dense ovarian stroma, and therefore enzymatic digestion with collagenase or Liberase has to be used (Dolmans et al., 2006). To enhance the chances of follicular survival and reproductive function restoration, enzymatic digestion procedures for human ovarian tissue need to be optimized and standardized.

As we recently demonstrated (Dolmans et al., 2006), Liberase treatment allows the isolation of highly viable follicles with an unaltered morphology and ultrastructure (Figure 4). This purified endotoxin-free enzyme preparation is a promising alternative to collagenase preparations for the reproducible isolation of intact primordial follicles for culture and grafting purposes. Preliminary results after xenotransplantation of isolated human primordial and primary follicles are encouraging. Indeed, the survival of grafted isolated primordial follicles in nude mice was estimated to be >60%, leading us to consider the possibility of grafting isolated follicles in the future, if the risk of reintroducing malignant cells into cancer patients by autografting ovarian cortical fragments cannot be excluded.

Figure 4.

Enzymatically isolated follicles (between 30 and 110 µm) visible under an inverted fluorescence microscope after fluorescent viability staining (calcein-AM and ethidium homodimer-1). Follicles are visible on light microscopy (left) and fluorescence microscopy (right), which show all of them to be viable. (Original magnification ×400.)

Whole ovary

As previously discussed, the main drawback of ovarian tissue cryopreservation followed by avascular transplantation is that the graft is completely dependent on the establishment of neovascularization and, as a result, a large proportion of follicles are lost during the initial ischaemia occurring after transplantation (Newton et al., 1996; Candy et al., 1997; Gunasena et al., 1997; Aubard et al., 1999; Baird et al., 1999; Nisolle et al., 2000; Liu et al., 2002). Reducing the ischaemic interval between transplantation and revascularization is therefore essential to maintaining the follicular reserve and extending the life span and function of the graft. In theory, the best way to achieve this is by transplantation of intact ovary with vascular anastomosis, allowing immediate revascularization of the transplant.

Ovarian vascular transplantation has already been successfully performed using intact fresh ovaries in rats (Wang et al., 2002; Yin et al., 2003), rabbits (Winston and Browne, 1974), sheep (Goding et al., 1967; Jeremias et al., 2002), dogs (Paldi et al., 1975), monkeys (Scott et al., 1981) and humans (Leporrier et al., 1987; Hilders et al., 2004; Mhatre et al., 2005). In the last few years, attempts at freezing and grafting whole ovaries in rats (Wang et al., 2002; Yin et al., 2003), rabbits (Chen et al., 2005) and sheep (Bedaiwy et al., 2003; Arav et al., 2005; Imhof et al., 2006) have also yielded encouraging results.

The first case of restoration of fertility after whole frozen ovary transplantation was described by Wang et al. in 2002. They described successful vascular transplantation of frozen–thawed rat ovaries and reproductive tract in four of seven (57%) transplants, which survived for ≥60 days, were ovulatory and resulted in one pregnancy. Chen et al. (2005) showed that frozen–thawed rabbit ovaries remained functional for at least 7 months after microvascular transplantation in 13 of 15 (86.7%) animals.

It appears that, in large mammals and humans, cryopreserving such a large-sized intact ovary may prove more problematic than in small animals because of the difficulty of adequate diffusion of cryoprotective agents into large tissue masses and vascular injury caused by intravascular ice formation. Nevertheless, Arav et al. (2005) reported progesterone activity 36 months after vascular transplantation of frozen–thawed sheep ovaries in three of eight transplants, and retrieval of six oocytes, resulting in embryonic development up to the 8-cell stage after parthenogenic activation. Bedaiwy et al. (2003) reported the restoration of ovarian function after autotransplantation of intact frozen–thawed sheep ovaries with microvascular anastomosis, but it should be noted that 8 of 11 ovaries were lost due to thrombotic events in the reanastomosed vascular pedicle. Imhof et al. (2006) recently demonstrated that the autotransplantation of whole cryopreserved sheep ovaries with microanastomosis of the ovarian vascular pedicle could lead to pregnancy and delivery. Moreover, in this study, six of eight ovaries showed major ovarian vessels to be free of thrombosis, with the structural integrity of the ovarian stroma largely retained 18–19 months after transplantation.

Recently, Martinez-Madrid et al. described a cryopreservation protocol for intact human ovary with its vascular pedicle and proved high survival rates of follicles (75.1%) (Figure 5), small vessels and stroma, and a normal histological structure in all the ovarian components after thawing (Martinez-Madrid et al., 2004b).

Figure 5.

High survival rate of stromal cells, follicles and vessels in a cryopreserved whole ovary (viable cells stained green with calcein-AM and dead cells stained red with ethidium homodimer-1). (Original magnification ×200.)

After freeze–thawing whole human ovaries using this protocol, no induction of apoptosis was observed in any cell types, assessed by both the terminal deoxynucleotidyl transferase biotin-dUTP nick-end labelling (TUNEL) method and immunohistochemistry for active caspase-3 (Martinez-Madrid et al., 2005). TEM confirmed that the majority (96.7%) of primordial follicles were intact after cryopreservation (Camboni et al., 2005) (Figure 6). Particular attention was paid to the evaluation of the endothelial cells: TEM revealed that 96.3% of these cells had a completely normal ultrastructure, and the percentage of active caspase-3-positive endothelial cells was <1%.

Figure 6.

Primary follicle in a frozen–thawed whole ovary. The oocyte (O) is surrounded by a single layer of cuboid follicular cells (Fc) on a continuous basal membrane (bm). Note the presence of rounded mitochondria (m) with a pale matrix and peripheral cristae in the oocyte cytoplasm. Follicular cells show indented nuclei containing peripheral patches of heterochromatin and numerous rod-shaped mitochondria in the cytoplasm. N, oocyte nucleus. Original magnification: transmission electron microscopy (×4400).

Our results in humans have led us to seriously consider proposing this option to women in the future, when there is no risk of transmitting malignant cells via the graft after transplantation. So far, in our department, three whole ovaries have been cryopreserved with a view to future reimplantation (grafting) and vascular anastomosis.

Developing new cryochambers and improving protocols for whole ovary cryopreservation must therefore be considered as vital directions in ongoing research to make the transplantation of an entire ovary a feasible objective (Donnez et al., 2005; Martinez-Madrid and Donnez, 2005). Research and development of technology to cryopreserve whole organs, as well as surgical techniques for the autotransplantation of an entire ovary with its vascular pedicle, should be encouraged. This could lead to the transplantation of intact ovaries with microvascular anastomosis carried out to restore immediate vascularization and minimize post-transplantation ischaemia, responsible for the reduction in follicular density.

Safety and ethical issues

The transmission of lymphoma via grafts of ovarian tissue from diseased donor mice to healthy recipients was reported by Shaw et al. (1996). This study highlighted the risks of clinical transplantation of ovarian biopsy samples to women recovering from cancer, especially a blood-borne cancer (Shaw et al., 1996; Shaw and Trounson, 1997). However, there are certain circumstances where the risk of cancerous involvement of the ovary is absent or minimal (Meirow et al., 1998) and where autografting would present little or no danger (Gosden et al., 1997; Moomjy and Rosenwaks, 1998; Kim et al., 2001). Future experiments should help us address questions about the relevance of replacing residual malignant cells with grafted tissue in such cases. Screening methods must be developed to eliminate the risk of cancer cell transmission with reimplantation. In some diseases, other options must be considered, such as the transplantation of isolated follicles. Meanwhile, the debate rages on.

The Practice Committee of the American Society for Reproductive Medicine (2004) has summarized some important points to be taken into consideration, and Dudzinski (2004) recently underlined the need to develop policies to protect the patient’s right to self-determination with respect to her gametes. She conducted a normative analysis of ethical issues in the context of oocyte and ovarian tissue cryopreservation for adolescent cancer survivors and concluded that more research is required before adolescents can ethically be enrolled in clinical trials.

We do not fully agree with this conclusion. Indeed, approximately one-third of young women exposed to chemotherapy develop ovarian failure. In 2006, we believe it is our ethical responsibility to propose ovarian tissue cryopreservation under Institutional Review Board protocols to all adolescents and young women under having to undergo chemotherapy with alkylating agents. Indeed, is it ethical to simply accept the existing discrepancy between males and females with regard to their chances of preserving their fertility following cancer treatments? What do we then say to young women facing POF following chemotherapy, knowing that ovarian cryopreservation has been an option for >10 years? It will be too late to say ‘we should have done something—we should at least have tried’.

This is why, since 1996, we have systematically proposed cryopreservation to all women <35 years of age before chemotherapy, when there is a risk of POF. We accept that ovarian tissue cryopreservation is a more innovative and invasive procedure than sperm cryopreservation and that all possible applications in adolescents are ethically complex. But we wholeheartedly agree with Revel and Schenker (2004), who contributed to a debate published in Human Reproduction, arguing that ovarian cortex banking should be offered before chemotherapy in all cases where emergency IVF is not possible.

One of the most important ethical issues is to ensure that the intervention does not harm the patient by dangerously delaying cancer treatment and that no remnant cells are reintroduced by subsequent transplantation. Taking these points into account, we agree with Dudzinski (2004) that policies to protect the patient’s future rights to her gametes should be developed, as well as policies addressing the disposition of the gametes if the patient dies.

Although an adolescent is more vulnerable when consent is sought in the rush to begin chemotherapy, she must be mature enough to understand the risks and benefits of the procedure. Consent must then be discussed extensively, the discussion including both the adolescent patient and her parents to minimize the risk of conflict of interest or inadvertent caution (Bukovsky, 2005). Respecting the code of good practice, all patients who may become infertile have the right to receive proper consideration of their interests for future possibilities in the field of ovarian function preservation. The selection of cases should be carried out on the basis of a multidisciplinary staff discussion including oncologists, gynaecologists, biologists, psychologists and paediatricians. Counselling should be given and informed consent obtained from the patient. Cancer treatment takes priority over potential restoration of fertility, but offering the chance to preserve fertility may greatly enhance the quality of life for cancer survivors.

Conclusion

Advances in reproductive technology have made fertility preservation techniques a real possibility for patients whose gonadal function is threatened by premature menopause, or by treatments such as radiotherapy, chemotherapy or surgical castration. Decision-making in this area is particularly difficult because of the experimental nature of some of these techniques. With their continued development and optimization, however, it may one day be possible to offer an individualized approach to management, be it through embryo cryopreservation, oocyte cryopreservation or cryopreservation of ovarian tissue (isolated follicles, cortical fragments or whole ovary).

Cryopreservation of ovarian tissue should be seriously considered for any patient undergoing treatment likely to impair future fertility, the indications being pelvic, extrapelvic and/or systemic malignant diseases, as well as non-malignant diseases. The age of the patient should be taken into consideration, because the follicular reserve of the ovary is age dependent. Because a decline in fertility is now well documented after the age of 38 years, the procedure should probably be restricted to patients below this limit. In any case, irradiation and chemotherapy appear to be less harmful to the gonads of prepubertal than postpubertal patients (Haie-Meder et al., 1993; Sanders et al., 1996; Meirow and Nugent, 2001).

It has been demonstrated that cryopreserved primordial follicles can survive the thawing process. Research must now focus on investigating current options and new alternatives in the field, to identify the best way of using tissue after thawing. It is probable that the answer lies in the use of culture environments adapted to each stage of follicular development. If autografting is the aim of cryopreservation of ovarian tissue, testing for malignant cells in the tissue must be carried out using adequate techniques, especially in case of haematological malignancies.

In conclusion, live births obtained after transplantation of frozen-thawed ovarian tissue in humans give hope to young cancer patients, but there is still much work to be done. Research programs need to determine whether active angiogenesis can be induced to accelerate the process of neovascularization in grafted tissue, if isolated human follicles can be grafted, or indeed if microvascular reanastomosis of an entire cryopreserved ovary is a valuable option.

References

Abir
R
, Franks S, Mobberley MA, Moore PA, Margara RA and Winston RM (
1997
) Mechanical isolation and in vitro growth of preantral and small antral human follicles.
Fertil Steril
68
,
682
–688.

Abir
R
, Fisch B, Nitke S, Okon E, Raz A and Ben Rafael Z (
2001
) Morphological study of fully and partially isolated early human follicles.
Fertil Steril
75
,
141
–146.

Abir
R
, Orvieto R, Raanani H, Feldberg D, Nitke S and Fisch B (
2003
) Parameters affecting successful transplantation of frozen-thawed human fetal ovaries into immunodeficient mice.
Fertil Steril
80
,
421
–428.

Akar
M
and Oktay K (
2005
) Restoration of ovarian endocrine function by ovarian transplantation.
Trends Endocrinol Metab
16
,
374
–380.

Albertini
DF
and Eppig JJ (
1995
) Unusual cytoskeletal and chromatin configurations in mouse oocytes that are atypical in meiotic progression.
Dev Genet
16
,
13
–19.

Almodin
CG
, Minguetti-Câmara VC, Meister H, Ferreira J, Franco RL, Cavalcante AA, Radaelli MRM, Bahls AS, Moron AF and Murta CGV (
2004
) Recovery of fertility after grafting of cryopreserved germinative tissue in female rabbits following radiotherapy.
Hum Reprod
19
,
1287
–1293.

Arav
A
, Revel A, Nathan Y, Bor A, Gacitua H, Yavin S, Gavish Z, Uri M and Elami A (
2005
) Oocyte recovery, embryo development and ovarian function after cryopreservation and transplantation of whole sheep ovary.
Hum Reprod
20
,
3554
–3559.

Aubard
Y
(
2003
) Ovarian tissue xenografting.
Eur J Obstet Gynecol Reprod Biol
108
,
14
–18.

Aubard
Y
, Piver P, Cognié Y, Fermeaux V, Poulin N and Driancourt MA (
1999
) Orthotopic and heterotopic autografts of frozen-thawed ovarian cortex in sheep.
Hum Reprod
14
,
2149
–2154.

Baird
DT
, Webb R, Campbell BK, Harkness LM and Gosden RG (
1999
) Long-term ovarian function in sheep after ovariectomy and transplantation of autografts stored at –196°C.
Endocrinology
140
,
462
–471.

Baird
DT
, Campbell BK, Souza C and Telfer EE (
2004
) Long-term ovarian function in sheep after ovariectomy and autotransplantation of cryopreserved cortical strips.
Eur J Obstet Gynecol Reprod Biol
113
,
55
–59.

Bath
LE
, Critchley HO, Chambers SE, Anderson RA, Kelnar CJ and Wallace WH (
1999
) Ovarian and uterine characteristics after total body irradiation in childhood and adolescence: response to sex steroid replacement.
Br J Obstet Gynaecol
106
,
1265
–1272.

Bedaiwy
MA
, Jeremias E, Gurunluoglu R, Hussein MR, Siemianow M, Biscotti C and Falcone T (
2003
) Restoration of ovarian function after autotransplantation of intact frozen-thawed sheep ovaries with microvascular anastomosis.
Fertil Steril
79
,
594
–602.

Bines
J
, Oleske DM and Cobleigh MA (
1996
) Ovarian function in premenopausal women treated with adjuvant chemotherapy for breast cancer.
J Clin Oncol
14
,
1718
–1729.

Blatt
J
(
1999
) Pregnancy outcome in long-term survivors of childhood cancer.
Med Pediatr Oncol
33
,
29
–33.

Blumenfeld
Z
, Shapiro D, Shteinberg M, Avivi I and Nahir M (
2000
) Preservation of fertility and ovarian function and minimizing gonadotoxicity in young women with systemic lupus erythematosus treated by chemotherapy.
Lupus
9
,
401
–405.

Boiso
I
, Marti M, Santalo J, Ponsa M, Barri PN and Veiga A (
2002
) A confocal microscopy analysis of the spindle and chromosome configurations of human oocytes cryopreserved at the germinal vesicle and metaphase II stage.
Hum Reprod
17
,
1885
–1891.

Boldt
J
, Cline D and McLaughlin D (
2003
) Human oocyte cryopreservation as an adjunct to IVF-embryo cycles.
Hum Reprod
18
,
1250
–1255.

Boran
N
, Cil AP, Tulunay G, Ozturkoglu E, Koc S, Bulbul D and Kose MF (
2005
) Fertility and recurrence results of conservative surgery for borderline ovarian tumors.
Gynecol Oncol
97
,
845
–851.

Borini
A
, Sciajno R, Bianchi V, Sereni E, Flamigni C and Coticchio G (
2006
) Clinical outcome of oocyte cryopreservation after slow cooling with a protocol utilizing a high sucrose concentration.
Hum Reprod
21
,
512
–517.

Bower
M
, Rustin GJ, Newlands ES, Holden L, Short D, Foskett M and Bagshawe KD (
1998
) Chemotherapy for gestational trophoblastic tumours hastens menopause by 3 years.
Eur J Cancer
34
,
1204
–1207.

Brown
JR
, Modell E, Obasaju M and Ying YK (
1996
) Natural cycle in-vitro fertilisation with embryo cryopreservation prior to chemotherapy for carcinoma of the breast.
Hum Reprod
11
,
197
–199.

Bukovsky
A
(
2005
) Can ovarian infertility be treated with bone marrow- or ovary-derived germ cells?
Reprod Biol Endocrinol
3
,
36
.

Burnett
AF
, Roman LD, O’Meara AT and Morrow CP (
2003
) Radical vaginal trachelectomy and pelvic lymphadenectomy for preservation of fertility in early cervical carcinoma.
Gynecol Oncol
88
,
419
–423.

Byrne
J
, Fears TR, Gail MH, Pee D, Connelly RR, Austin DF, Holmes GF, Holmes FF, Latourette HB, Meigs JW et al. (
1992
) Early menopause in long term survivors of cancer during adolescence.
Am J Obstet Gynecol
166
,
788
–793.

Callejo
J
, Salvador C, Miralles A, Vilaseca S, Lailla JM and Balasch J (
2001
) Long-term ovarian function evaluation after autografting by implantation with fresh and frozen-thawed human ovarian tissue.
J Clin Endocrinol Metab
86
,
4489
–4494.

Camboni
A
, Martinez-Madrid B, Dolmans MM, Macchiarelli G, Donnez J and Nottola SA (
2005
) Contribution of transmission electron microscopy to the study of human ovarian tissue integrity after enzymatic isolation, cryopreservation or xenografting.
Fertil Steril
84
,S
104
: O–252.

Campbell
BK
, Telfer EE, Webb R and Baird DT (
2000
) Ovarian autografts in sheep as a model for studying folliculogenesis.
Mol Cell Endocrinol
163
,
131
–139.

Candy
CJ
, Wood MJ and Whittingham DG (
1995
) Follicular development in cryopreserved marmoset ovarian tissue after transplantation.
Hum Reprod
10
,
2334
–2338.

Candy
CJ
, Wood MJ and Whittingham DG (
1997
) Effect of cryoprotectants on the survival of follicles in frozen mouse ovaries.
J Reprod Fertil
110
,
11
–19.

Candy
CJ
, Wood MJ and Whittingham DG (
2000
) Restoration of a normal reproductive lifespan after grafting of cryopreserved mouse ovaries.
Hum Reprod
15
,
1300
–1304.

Carroll
J
and Gosden RG (
1993
) Transplantation of frozen–thawed mouse primordial follicles.
Hum Reprod
8
,
1163
–1167.

Cha
KY
and Chian RC (
1998
) Maturation in vitro of immature human oocytes for clinical use.
Hum Reprod Update
4
,
103
–120.

Cha
KY
, Koo JJ, Ko JJ, Choi DH, Han SY and Yoon TK (
1991
) Pregnancy after in vitro fertilization of human follicular oocytes collected from nonstimulated cycles, their culture in vitro and their transfer in a donor oocyte program.
Fertil Steril
55
,
109
–113.

Chen
C
(
1986
) Pregnancy after human oocyte cryopreservation.
Lancet
1
,
884
–886.

Chen
C
, Chen S, Chang F, Wu G, Liu J and Yu C (
2005
) Autologous heterotopic transplantation of intact rabbit ovary after cryopreservation.
Hum Reprod
20
(Suppl
1
),
i149
–i150.

Critchley
HO
and Wallace GH (
2005
) Impact of cancer treatment on uterine function.
J Natl Cancer Inst Monogr
34
,
64
–68.

Critchley
HO
, Wallace WH, Shalet SM, Mamtora H, Higginson J and Anderson DC (
1992
) Abdominal irradiation in childhood; the potential for pregnancy.
Br J Obstet Gynaecol
99
,
392
–394.

Dargent
D
, Martin X, Sacchetoni A and Mathevet P (
2000
) Laparoscopic vaginal radical trachelectomy: a treatment to preserve the fertility of cervical carcinoma patients.
Cancer
88
,
1877
–1882.

Demeestere
I
, Simon P, Buxant F, Robin V, Fernandez-Aguilar S, Centner J, Delbaere A and Englert Y (
2006
) Ovarian function and spontaneous pregnancy after combined heterotopic and orthotopic cryopreserved ovarian tissue transplantation in a patient previously treated with bone marrow transplantation: case report.
Hum Reprod
[
19
April
2006; Epub ahead of print].

Dolmans
MM
, Demylle D, Martinez-Madrid B and Donnez J (
2005
) Efficacy of in vitro fertilization after chemotherapy.
Fertil Steril
83
(
4
),
897
–901.

Dolmans
MM
, Michaux N, Camboni A, Martinez-Madrid B, Van Langendonckt A, Nottola S and Donnez J (
2006
) Evaluation of Liberase, a purified enzyme blend, for the isolation of human primordial and primary ovarian follicles.
Hum Reprod
21
(
2
),
413
–420.

Donnez
J
and Bassil S (
1998
) Indications for cryopreservation of ovarian tissue.
Hum Reprod Update
4
,
248
–259.

Donnez
J
and Dolmans MM (
2004
) Livebirth after orthotopic transplantation of cryopreserved ovarian tissue. Letter.
Lancet
364
,
2092
–2093.

Donnez
J
, Godin PA, Qu J and Nisolle M (
2000
) Gonadal cryopreservation in the young patient with gynaecological malignancy.
Curr Opin Obstet Gynecol
12
,
1
–9.

Donnez
J
, Munschke A, Berlière M, Pirard C, Jadoul P, Smets M and Squifflet J (
2003
) Safety of conservative management and infertile outcome in women with borderline tumors of the ovary.
Fertil Steril
79
,
1216
–1221.

Donnez
J
, Dolmans MM, Demylle D, Jadoul P, Pirard C, Squifflet J, Martinez-Madrid B and Van Langendonckt A (
2004
) Livebirth after orthotopic transplantation of cryopreserved ovarian tissue.
Lancet
364
,
1405
–1410.

Donnez
J
, Dolmans MM, Martinez-Madrid B, Demylle D and Van Langendonkt A (
2005
) The role of cryopreservation for women prior to treatment of malignancy.
Curr Opin Obstet Gynecol
17
,
333
–338.

Donnez
J
, Dolmans MM, Demylle D, Jadoul P, Pirard C, Squifflet J, Martinez-Madrid B and Van Langendonckt A (
2006
) Restoration of ovarian function after orthotopic (intraovarian and periovarian) transplantation of cryopreserved ovarian tissue in a woman treated by bone marrow transplantation for sickle cell anaemia: case report.
Hum Reprod
21
,
183
–188.

Dudzinski
DM
(
2004
) Ethical issues in fertility preservation for adolescent cancer survivors: oocyte and ovarian tissue cryopreservation.
J Pediatr Adolesc Gynecol
17
,
97
–102.

Ethics
Committee of
the American Society for Reproductive Medicine (
2005
) Fertility preservation and reproduction in cancer patients.
Fertil Steril
83
,
1622
–1628.

Fabbri
R
, Porcu E, Marsella T, Rocchetta G, Venturoli S and Flamigni C (
2001
) Human oocyte cryopreservation: new perspectives regarding oocyte survival.
Hum Reprod
16
,
411
–416.

Falcone
T
, Attaran M, Bedaiwy MA and Goldberg JM (
2004
) Ovarian function preservation in the cancer patient.
Fertil Steril
81
,
243
–257.

Fauvet
R
, Poncelet C, Boccara J, Descamps P, Fondrinier E and Daraï E (
2005
) Fertility after conservative treatment for borderline ovarian tumors: a French multicenter study.
Fertil Steril
83
,
284
–290.

Fisher
B
, Sherman B, Rockette H, Redmond C, Margolese R and Fisher ER (
1979
) 1-Phenylalanine mustard (L-PAM) in the management of premenopausal patients with primary breast cancer: lack of association of disease-free survival with depression of ovarian function. National Surgical Adjuvant Project for Breast and Bowel Cancers.
Cancer
44
,
847
–857.

Fuller
B
and Paynter S (
2004
) Fundamentals of cryobiology in reproductive medicine.
Reprod Biomed Online
9
,
680
–691.

Gallez
B
, Baudelet C and Jordan BF (
2004
) Assessment of tumor oxygenation by electron paramagnetic resonance: principles and applications.
NMR Biomed
17
,
240
–262.

Ghetler
Y
, Yavin S, Shalgi R and Arav A (
2005
) The effect of chilling on membrane lipid phase transition in human oocytes and zygotes.
Hum Reprod
20
,
3385
–3389.

Ginsburg
ES
, Yanushpolsky EH and Jackson KV (
2001
) In vitro fertilization for cancer patients and survivors.
Fertil Steril
75
,
705
–710.

Goding
JR
, McCracken JA and Baird DT (
1967
) The study of ovarian function in the ewe by means of a vascular autotransplantation technique.
J Endocrinol
39
,
37
–52.

Gook
DA
, Osborn SM and Johnston WI (
1993
) Cryopreservation of mouse and human oocytes using 1,2-propanediol and the configuration of the meiotic spindle.
Hum Reprod
8
,
1101
–1109.

Gook
DA
, Schiewe MC, Osborn SM, Asch RH, Jansen RP and Johnston WI (
1995
) Intracytoplasmic sperm injection and embryo development of human oocytes cryopreserved using 1,2-propanediol.
Hum Reprod
10
,
2637
–2641.

Gook
DA
, Edgar DH, Borg J, Archer J, Lutjen PJ and McBain JC (
2003
) Oocyte maturation, follicle rupture and luteinization in human cryopreserved ovarian tissue following xenografting.
Hum Reprod
18
,
1772
–1781.

Gook
DA
, Edgar DH, Borg J, Archer J and McBain JC (
2005
) Diagnostic assessment of the developmental potential of human cryopreserved ovarian tissue from multiple patients using xenografting.
Hum Reprod
20
,
72
–78.

Gosden
RG
(
2005
) Prospects for oocyte banking and in vitro maturation.
J Natl Cancer Inst Monogr
34
,
60
–63.

Gosden
RG
, Baird DT, Wade JC and Webb R (
1994
) Restoration of fertility to oophorectomized sheep by ovarian autografts stored at –196°C.
Hum Reprod
9
,
597
–603.

Gosden
RG
, Rutherford AJ and Norfolk DR (
1997
) Ovarian banking for cancer patients: transmission of malignant cells in ovarian grafts.
Hum Reprod
12
,
403
–405.

Gougeon
A
(
1996
) Regulation of ovarian follicular development in primates: facts and hypotheses.
Endocr Rev
17
,
121
–155.

Gunasena
KT
, Villines PM, Critser ES and Critser JK (
1997
) Live births after autologous transplant of cryopreserved mouse ovaries.
Hum Reprod
12
,
101
–106.

Haie-Meder
C
, Mlika-Cabanne N, Michel G, Briot E, Gerbaulet A, Lhomme C, Cosset JM, Sarrazin D, Flamant F and Hayat M (
1993
) Radiotherapy after ovarian transposition ovarian function and fertility preservation.
Int J Radiat Oncol Biol Phys
25
,
419
–424.

Hernandez-Fonseca
H
, Bosch P, Sirisathien S, Wininger JD, Massey JB and Brackett BG (
2004
) Effect of site of transplantation on follicular development of human ovarian tissue transplanted into intact or castrated immunodeficient mice.
Fertil Steril
81
,
888
–892.

Hicks
M
and Piver MS (
1992
) Conservative surgery plus adjuvant therapy for vulvovaginal rhabdomyosarcoma diethylstilbestrol clear cell adenocarcinoma of the vagina and unilateral germ cell tumors of the ovary.
Obstet Gynecol Clin North Am
19
,
219
–233.

Hilders
CG
, Baranski AG, Peters L, Ramkhelawan A and Trimbos JB (
2004
) Successful human ovarian autotransplantation to the upper arm.
Cancer
101
,
2771
–2778.

Hortobagyi
GN
, Buzdar AU, Marcus CE and Smith TL (
1986
) Immediate and long-term toxicity of adjuvant chemotherapy regimens containing doxorubicin in trials at M.D. Anderson Hospital and Tumor Institute.
NCI Monogr
1
,
105
–109.

Hovatta
O
(
2004
) Cryopreservation and culture of human ovarian cortical tissue containing early follicles.
Eur J Obstet Gynecol Reprod Biol
113
(Suppl
1
),
S50
–S54.

Hovatta
O
(
2005
) Methods for cryopreservation of human ovarian tissue.
Reprod Biomed Online
10
,
729
–734.

Hovatta
O
, Wright C, Krausz T, Hardy K and Winston RML (
1999
) Human primordial, primary and secondary ovarian follicles in long-term culture: effect of partial isolation.
Hum Reprod
14
,
2519
–2524.

Imhof
M
, Bergmeister H, Lipovac M, Rudas M, Hofstetter G and Huber J (
2006
) Orthotopic microvascular re-anastomosis of whole cryopreserved ovine ovaries resulting in pregnancy and life birth.
Fertil Steril
85
(Suppl
1
),
1208
–1215.

Israely
T
, Dafni H, Nevo N, Tsafriri A and Neeman M (
2004
) Angiogenesis in ectopic xenotransplantation: multiparameter characterization of the neovasculature by dynamic contrast-enhanced MRI.
Magn Reson Med
52
,
741
–750.

Israely
T
, Nevo N, Harmelin A, Neeman M and Tsafriri A (
2006
) Reducing ischaemic damage in rodent ovarian xenografts transplanted into granulation tissue.
Hum Reprod
[
3
February
2006; advance access published].

Jadoul
P
and Donnez J (
2003
) Conservative treatment
may
be beneficial for young women with atypical endometrial hyperplasia or endometrial adenocarcinoma.
Fertil Steril
80
,
1315
–1324.

Jemal
A
, Siegel R, Ward E, Murray T, Xu J, Smigal C and Thun MJ (
2006
) Cancer statistics, 2006.
CA Cancer J Clin
56
,
106
–130.

Jeremias
E
, Bedaiwy MA, Gurunluoglu R, Biscotti CV, Siemionow M and Falcone T (
2002
) Heterotopic autotransplantation of the ovary with microvascular anastomosis: a novel surgical technique.
Fertil Steril
77
,
1278
–1282.

Kan
A
, Kilani S, Tilia L, Mitchell F, Burns K and Chapman M (
2004
) Pregnancy from intracytoplasmic injection of a frozen-thawed oocyte.
Aust N Z J Obstet Gynaecol
44
,
262
–263.

Katayama
KP
, Stehlik J, Kuwayama M, Kato O and Stehlik E (
2003
) High survival rate of vitrified human oocytes results in clinical pregnancy.
Fertil Steril
80
,
223
–224.

Kenney
LB
, Laufer MR, Grant FD, Grier H and Diller L (
2001
) High risk of infertility and long term gonadal damage in males treated with high dose cyclophosphamide for sarcoma during childhood.
Cancer
91
,
613
–621.

Kim
SS
(
2006
) Fertility preservation in female cancer patients: current developments and future directions.
Fertil Steril
85
,
1
–11.

Kim
SS
, Radford J, Harris M, Varley J, Rutherford AJ, Lieberman B, Shalet S and Gosden R (
2001
) Ovarian tissue harvested from lymphoma patients to preserve fertility
may
be safe for autotransplantation.
Hum Reprod
16
,
2056
–2060.

Kim
SS
, Soules MR and Battaglia DE (
2002
) Follicular development, ovulation, and corpus luteum formation in cryopreserved human ovarian tissue after xenotransplantation.
Fertil Steril
78
,
77
–82.

Kim
SS
, Hwang I-T and Lee H-C (
2004
) Heterotopic autotransplantation of cryobanked human ovarian tissue as a strategy to restore ovarian function.
Fertil Steril
82
,
930
–932.

Kim
SS
, Yang HW, Kang HG, Lee HH, Lee HC, Ko DS and Gosden RG (
2004
) Quantitative assessment of ischemic tissue damage in ovarian cortical tissue with or without antioxidant (ascorbic acid) treatment.
Fertil Stertil
82
,
679
–685.

Kim
SS
, Kang HG, Kim NH, Lee HC and Lee H (
2005
) Assessment of the integrity of human oocytes retrieved from cryopreserved ovarian tissue after xenotransplantation.
Hum Reprod
20
,
2502
–2508.

Kleine
W
(
1996
) Results of fertility preserving operations in malignant ovarian tumors.
Zentralbl Gynakol
118
,
317
–321.

Koyama
H
, Wada T, Nishizawa Y, Iwanaga T and Aoki Y (
1977
) Cyclophosphamide-induced ovarian failure and its therapeutic significance in patients with breast cancer.
Cancer
39
,
1403
–1409.

Larsen
EC
, Muller J, Schmiegelow K, Rechnitzer C and Andersen AN (
2003
) Reduced ovarian function in long-term survivors of radiation- and chemotherapy-treated childhood cancer.
J Clin Endocrinol Metab
88
,
5307
–5314.

Larsen
EC
, Schmiegelow K, Rechnitzer C, Loft A, Muller J and Andersen AN (
2004
) Radiotherapy at a young age reduces uterine volume of childhood cancer survivors.
Acta Obstet Gynecol Scand
83
,
96
–102.

Laschke
MW
, Menger MD and Vollmar B (
2002
) Ovariectomy improves neovascularization and microcirculation of freely transplanted ovarian follicles.
J Endocrinol
172
,
535
–544.

Lee
DM
, Yeoman RR, Battaglia DE, Stouffer RL, Zelinski-Wooten B, Fanton JW and Wolf DP (
2004
) Live birth after ovarian tissue transplant.
Nature
428
,
137
–138.

Legault
L
and Bonny Y (
1999
) Endocrine complications of bone marrow transplantation in children.
Pediatr Transplant
3
,
60
–66.

Leporrier
M
, von Theobald P, Roffe JL and Muller G (
1987
) A new technique to protect ovarian function before pelvic irradiation. Heterotopic ovarian autotransplantation.
Cancer
60
,
2201
–2204.

Levi
Setti PE
, Albani E, Novara PV, Cesana A and Morreale G (
2006
) Cryopreservation of supernumerary oocytes in IVF/ICSI cycles.
Hum Reprod
21
,
370
–375.

Liu
J
, Van der Elst J, Van den Broecke R and Dhont M (
2002
) Early massive follicle loss and apoptosis in heterotopically grafted newborn mouse ovaries.
Hum Reprod
17
,
605
–611.

Lobo
RA
(
2005
) Potential options for preservation of fertility in women.
N Engl J Med
353
,
64
–73.

Lutchman
Singh K
, Davies M and Chatterjee R (
2005
) Fertility in female cancer survivors: pathophysiology, preservation and the role of ovarian reserve testing.
Hum Reprod Update
11
,
69
–89.

Mackie
EJ
, Radford M and Shalet SM (
1996
) Gonadal function following chemotherapy for childhood Hodgkin’s disease.
Med Pediatr Oncol
27
,
74
–78.

Mandelbaum
J
, Anastasiou O, Lévy R, Guérin JF, de Larouzière V and Antoine JM (
2004
) Effects of cryopreservation on the meiotic spindle of human oocytes.
Eur J Obstet Gynecol Reprod Biol
113
S,
S17
–S23.

Maneschi
F
, Benedetti-Panici P, Scambia G, Salerno MG, D’Agostino G and Mancuso S (
1994
) Menstrual and hormone patterns in women treated with high-dose cisplatin and bleomycin.
Gynecol Oncol
54
,
345
–348.

Martinez-Madrid
B
and Donnez J (
2005
) Technical challenges in freeze-thawing of human ovary.
Fertil Steril
83
,
1069
–1070.

Martinez-Madrid
B
, Dolmans MM, Van Langendonckt A, Defrère S, Van Eyck AS and Donnez J (
2004
) Ficoll density gradient method for recovery of isolated human ovarian primordial follicles.
Fertil Steril
82
,
1648
–1653.

Martinez-Madrid
B
, Dolmans MM, Van Langendonckt A, Defrère S and Donnez J (
2004
) Freeze-thawing intact human ovary with its vascular pedicle with a passive cooling device.
Fertil Steril
82
(
5
),
1390
–1394.

Martinez-Madrid
B
, Camboni A, Dolmans MM, Nottola S, Van Langendonckt A and Donnez J (
2005
) Apoptosis and ultrastructure in frozen intact human ovary.
Hum Reprod
20
(Suppl
1
),
i37
–i38.

Mattle
V
, Behringer K, Engert A and Wildt L (
2005
) Female fertility after cytotoxic therapy-protection of ovarian function during chemotherapy of malignant and non-malignant diseases.
Eur J Haematol
75
,
77
–82.

Meirow
D
and Nugent D (
2001
) The effects of radiotherapy and chemotherapy on female reproduction.
Hum Reprod Update
7
,
534
–543.

Meirow
D
, Ben Yehuda D, Prus D, Poliak A, Schenker JG, Rachmilewitz EA and Lewin A (
1998
) Ovarian tissue banking in patients with Hodgkin’s disease: is it safe?
Fertil Steril
69
,
996
–998.

Meirow
D
, Lewis H, Nugent D and Epstein N (
1999
) Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool.
Hum Reprod
14
,
1903
–1907.

Meirow
D
, Levron J, Eldar-Geva T, Hardan I, Fridman E, Zalel Y, Schiff E and Dor J (
2005
) Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy.
N Engl J Med
353
,
318
–321.

Mhatre
P
, Mhatre J and Magotra R (
2005
) Ovarian transplant: a new frontier.
Transplant Proc
37
,
1396
–1398.

Moomjy
M
and Rosenwaks Z (
1998
) Ovarian tissue cryopreservation: the time is now. Transplantation or in vitro maturation: the time awaits.
Fertil Steril
69
,
999
–1000.

Motta
PM
, Nottola SA, Familiari G, Makabe S, Stallone T and Macchiarelli G (
2003
) Morphodynamics of the follicular-luteal complex during early ovarian development and reproductive life.
Int Rev Cytol
223
,
177
–288.

Newton
H
, Aubard Y, Rutherford A, Sharma V and Gosden RG (
1996
) Low temperature storage and grafting of human ovarian tissue.
Hum Reprod
11
,
1487
–1491.

Newton
H
, Fisher J, Arnold JR, Pegg DE, Faddy MJ and Gosden RG (
1998
) Permeation of human ovarian tissue with cryoprotective agents in preparation for cryopreservation.
Hum Reprod
13
,
376
–380.

Nisolle
M
, Godin PA, Casanas-Roux F, Qu J, Motta P and Donnez J (
2000
) Histological and ultrastructural evaluation of fresh and frozen-thawed human ovarian xenografts in nude mice.
Fertil Steril
74
,
122
–129.

Oktay
K
(
2006
) Spontaneous conceptions and live birth after heterotopic ovarian transplantation: is there a germline stem cell connection?
Hum Reprod
(advance access doi: 10.1093/humrep/del007).

Oktay
K
and Karlikaya G (
2000
) Ovarian function after transplantation of frozen, banked autologous ovarian tissue.
N Engl J Med
342
,
1919
.

Oktay
K
and Tilly J (
2004
) Livebirth after cryopreserved ovarian tissue autotransplantation. Correspondence.
Lancet
364
,
2091
–2092.

Oktay
K
, Nugent D, Newton H, Salha O, Chatterjee P and Gosden RG (
1997
) Isolation and characterization of primordial follicles from fresh and cryopreserved human ovarian tissue.
Fertil Steril
67
,
481
–486.

Oktay
K
, Newton H, Aubard Y, Salha O and Gosden RG (
1998
) Cryopreservation of immature human oocytes and ovarian tissue: an emerging technology?
Fertil Steril
69
,
1
–7.

Oktay
K
, Newton H, Mullan J and Gosden RG (
1998
) Development of human primordial follicles to antral stages in SCID/hpg mice stimulated with follicle stimulating hormone.
Hum Reprod
13
,
1133
–1138.

Oktay
K
, Buyuk E, Veeck L, Zaninovic N, Xu KP, Takeuchi T, Opsahl M and Rosenwaks Z (
2004
) Embryo development after heterotopic transplantation of cryopreserved ovarian tissue.
Lancet
363
,
837
–840.

Oktay
K
, Buyuk E, Libertella N, Akar M and Rosenwaks Z (
2005
) Fertility preservation in breast cancer patients: a prospective controlled comparison of ovarian stimulation with tamoxifen and letrozole for embryo cryopreservation.
J Clin Oncol
23
,
4347
–4353.

Overstrom
EW
, Paqui-Platls D, Toner M and Cravacho EG (
1990
) Cryoprotectant and thermal effect on cytoskeletal organization and IVF rate of mouse oocytes.
Biol Reprod
42
(Suppl
1
),
175
.

Paldi
E
, Gal D, Barzilai A, Hampel N and Malberger E (
1975
) Genital organs. Auto and homotransplantation in forty dogs.
Int J Fertil
20
,
5
–12.

Perez
GI
, Maravei DV, Trbovich AM, Cidlowski JA, Tilly JL and Hughes FM Jr (
2000
) Identification of potassium-dependent and – independent components of the apoptotic machinery in mouse ovarian germ cells and granulosa cells.
Biol Reprod
63
,
1358
–1369.

Pickering
SJ
, Braude PR, Johnson MH, Cant A and Currie J (
1990
) Transient cooling to room temperature can cause irreversible disruption of the meiotic spindle in the human oocyte.
Fertil Steril
54
,
102
–108.

Porcu
E
(
2005
) Cryopreservation of oocytes: indications, risks and outcome. Abstracts of the 21st Annual Meeting of the European Society of Human Reproduction and Embryology.
Hum Reprod
20
(Suppl
1
),i
50
, O-137.

Porcu
E
, Fabbri R, Seracchioli R, Ciotti PM, Magrini O and Flamigni C (
1997
) Birth of a healthy female after intracytoplasmic sperm injection of cryopreserved human oocytes.
Fertil Steril
68
,
724
–726.

Porcu
E
, Fabbri R, Damiano G, Fratto R, Giunchi S and Venturoli S (
2004
) Oocyte cryopreservation in oncological patients.
Eur J Obstet Gynecol Reprod Biol
113
(Suppl
1
),
S14
–S16.

Practice Committee of the American Society for Reproductive Medicine (

2004
) Ovarian tissue and oocyte cryopreservation.
Fertil Steril
82
,
993
–998.

Qu
J
, Godin PA, Nisolle M and Donnez J (
2000
) Distribution and epidermal growth factor receptor expression of primordial follicles in human ovarian tissue before and after cryopreservation.
Hum Reprod
15
,
302
–310.

Radford
JA
, Lieberman BA, Brison D, Smith AR, Critchlow JD, Russell SA, Watson AJ, Clayton JA, Harris M, Gosden RG et al. (
2001
) Orthotopic reimplantation of cryopreserved ovarian cortical strips after high-dose chemotherapy for Hodgkin’s lymphoma.
Lancet
357
,
1172
–1175.

Revel
A
and Schenker J (
2004
) Ovarian tissue banking for cancer patients: is ovarian cortex cryopreservation presently justified?
Hum Reprod
19
,
14
–19.

Ries
LAG
, Percy CL and Bunin GR (
1999
) Introduction. In Ries LAG, Smith MA, Gurney JG, Linet M, Tamra T, Young JL, Bunin GR (eds)
Cancer Incidence and Survival Among Children and Adolescents
: United States SEER Program 1975–1995 [NIH Publications, no. 99–4649]. National Cancer Institute, Bethesda, MD:
1
–15.

Rodgers
RJ
, Irving-Rodgers H and Russell DL (
2003
) Extracellular matrix of the developing ovarian follicle.
Reproduction
126
,
415
–424.

Roy
SK
and Treacy BJ (
1993
) Isolation and long-term culture of human preantral follicles.
Fertil Steril
59
,
783
–790.

Salle
B
, Demirci B, Franck M, Rudigoz RF, Guerin JF and Lornage J (
2002
) Normal pregnancies and live births after autograft of frozen-thawed hemi-ovaries into ewes.
Fertil Steril
77
,
403
–408.

Salooja
N
, Szydlo RM, Socie G, Rio B, Chatterjee R, Ljungman P, Van Lint MT, Powles R, Jackson G, Hinterberger-Fischer M et al. for the Late Effects Working Party of the European Group for Blood and Marrow Transplantation (
2001
) Pregnancy outcomes after peripheral blood or bone marrow transplantation: a retrospective survey.
Lancet
358
(
9278
),
271
–276.

Sanders
J
, Hawley J, Levy W, Gooley T, Buckner CD, Deeg HJ, Doney K, Storb R, Sullivan K, Witherspoon R et al. (
1996
) Pregnancies following high-dose Cyclophosphamide with or without high-dose Busulfan or total body irradiation and bone marrow transplantation.
Blood
87
,
3045
–3052.

Schilsky
RL
, Sherins RJ, Hubbard SM, Wesley MN, Young RC and DeVita VT (
1981
) Long-term follow up of ovarian function in women treated with MOPP chemotherapy for Hodgkin’s disease.
Am J Med
71
,
552
–556.

Schmidt
KL
, Byskov AG, Nyboe Andersen A, Muller J and Andersen CY (
2003
) Density and distribution of primordial follicles in single pieces of cortex from 21 patients and in individual pieces of cortex from three entire human ovaries.
Hum Reprod
18
,
1158
–1164.

Schmidt
KL
, Yding Andersen C, Loft A, Byskov AG, Ernst E and Nyboe Andersen A (
2005
) Follow-up of ovarian function post-chemotherapy following ovarian cryopreservation and transplantation.
Hum Reprod
20
,
3539
–3546.

Schnorr
J
, Oehninger S, Toner J, Hsiu J, Lanzendorf S, Williams R and Hodgen G (
2002
) Functional studies of s.c. ovarian transplants in non-human primates: steroidogenesis, endometrial development, ovulation, menstrual patterns and gamete morphology.
Hum Reprod
17
,
612
–619.

Scott
JR
, Keye WR, Poulson AM and Reynolds WA (
1981
) Microsurgical ovarian transplantation in the primate.
Fertil Steril
36
,
512
–515.

Shalet
SM
(
1980
) Effects of cancer chemotherapy on gonadal function of patients.
Cancer Treat Rev
7
,
141
–152.

Shamberger
RC
, Rosenberg SA, Seipp CA and Sherins RJ (
1981
) Effects of high-dose methotrexate and vincristine on ovarian and testicular functions in patients undergoing postoperative adjuvant treatment of osteosarcoma.
Cancer Treat Rep
65
,
739
–746.

Shaw
JM
and Trounson AO (
1997
) Oncological implications in the replacement of ovarian tissue.
Hum Reprod
12
,
403
–405.

Shaw
JM
, Bowles S, Koopman P, Wood EC and Trounson AO (
1996
) Fresh and cryopreserved ovarian tissue samples from donors with lymphoma transmit the cancer to graft recipients.
Hum Reprod
11
,
1668
–1673.

Silber
SJ
, Lenahan KM, Levine DJ, Pineda JA, Gorman KS, Friez MJ, Crawford EC and Gosden RG (
2005
) Ovarian transplantation between monozygotic twins discordant for premature ovarian failure.
N Engl J Med
353
,
58
–63.

Slavin
S
, Nagler A, Aker M, Shapira MY, Cividalli G and Or R (
2001
) Non-myeloablative stem cell transplantation and donor lymphocyte infusion for the treatment of cancer and life-threatening non-malignant disorders.
Rev Clin Exp Hematol
5
,
135
–146.

Smitz
JE
and Cortvrindt RG (
2002
) The earliest stages of folliculogenesis in vitro.
Reproduction
123
,
185
–202.

Snow
M
, Cox SL, Jenkin G, Trounson AO and Shaw JM (
2002
) Generation of live young from xenografted mouse ovaries.
Science
297
(
5590
),2227.

Sonmezer
M
and Oktay K (
2004
) Fertility preservation in female patients.
Hum Reprod Update
10
,
251
–266.

Stachecki
JJ
and Cohen J (
2004
) An overview of oocyte cryopreservation.
Reprod Biomed Online
9
,
152
–163.

Stillman
RJ
, Schinfeld JS, Schiff I, Gelber RD, Greenberger J, Larson M, Jaffe N and Li FP (
1981
) Ovarian failure in long-term survivors of childhood malignancy.
Am J Obstet Gynecol
139
,
62
–66.

Sudman
PD
, Rutledge JC, Bishop JB and Generoso WM (
1992
) Bleomycin: female-specific dominant lethal effects in mice.
Mutat Res
296
,
143
–156.

Takai
Y
, Canning J, Perez GI, Pru JK, Schlezinger JJ, Sherr DH, Kolesnick RN, Yuan J, Flavell RA, Korsmeyer SJ et al. (
2003
) Bax, caspase-2, and caspase-3 are required for ovarian follicle loss caused by 4-vinylcyclohexene diepoxide exposure of female mice in vivo.
Endocrinology
144
,
69
–74.

Tangir
J
, Zelterman D, Ma W and Schwartz PE (
2003
) Reproductive function after conservative surgery and chemotherapy for malignant germ cell tumors of the ovary.
Obstet Gynecol
101
,
251
–257.

Tanguay
C
, Plante M, Renaud MC, Roy M and Tetu B (
2004
) Vaginal radical trachelectomy in the treatment of cervical cancer: the role of frozen section.
Int J Gynecol Pathol
23
,
170
–175.

Tauchmanova
L
, Selleri C, De Rosa G, Pagano L, Orio F, Lombardi G, Rotoli B and Colao A (
2002
) High prevalence of endocrine dysfunction in long-term survivors after allogeneic bone marrow transplantation for hematologic diseases.
Cancer
95
,
1076
–1084.

Teinturier
C
, Hartmann O, Valteau-Couanet D, Benhamou E and Bougneres PF (
1998
) Ovarian function after autologous bone marrow transplantation in childhood: high-dose busulfan is a major cause of ovarian failure.
Bone Marrow Transplant
22
,
989
–994.

Tinelli
R
, Tinelli A, Tinelli FG, Cicinelli E and Malvasi A (
2005
) Conservative surgery for borderline ovarian tumors: a review.
Gynecol Oncol
10
,
185
–191.

Torrents
E
, Boiso I, Barri PN and Veiga A (
2003
) Applications of ovarian tissue transplantation in experimental biology and medicine.
Hum Reprod Update
9
,
471
–481.

Trounson
AO
, Wood C and Kausche A (
1994
) In vitro maturation and the fertilization and developmental competence of oocytes recovered from untreated polycystic ovarian patients.
Fertil Steril
62
,
353
–362.

Tucker
MJ
, Wright G, Morton PC and Massey JB (
1998
) Birth after cryopreservation of immature oocytes with subsequent in vitro maturation.
Fertil Steril
70
,
578
–579.

Vajta
G
, Holm P, Kuwayama M, Booth PJ, Jacobsen H, Greve T and Callesen H (
1998
) Open Pulled Straw (OPS) vitrification: a new way to reduce cryoinjuries of bovine ova and embryos.
Mol Reprod Dev
51
,
53
–58.

Van der Elst
J
(
2003
) Oocyte freezing: here to stay?
Hum Reprod Update
9
,
463
–470.

Van Thiel
DH
, Ross GT and Lipsett MB (
1970
) Pregnancies after chemotherapy of trophoblastic neoplasms.
Science
169
,
1326
–1327.

Viviani
S
, Santoro A, Ragni G, Bonfante V, Bestetti O and Bonadonna G (
1985
) Gonadal toxicity after combination chemotherapy for Hodgkin’s disease. Comparative results of MOPP vs ABVD.
Eur J Cancer Clin Oncol
21
,
601
–605.

Wallace
WH
, Shalet SM, Crowne EC, Morris-Jones PH, Gattamaneni HR and Price DA (
1989
) Gonadal dysfunction due to cis-platinum.
Med Pediatr Oncol
17
,
409
–413.

Wallace
WH
, Thomson AB and Kelsey TW (
2003
) The radiosensitivity of the human oocyte.
Hum Reprod
18
,
117
–121.

Wallace
WH
, Thomson AB, Saran F and Kelsey TW (
2005
) Predicting age of ovarian failure after radiation to a field that includes the ovaries.
Int J Radiat Oncol Biol Phys
62
,
738
–744.

Wallace
WH
, Anderson RA and Irvine DS (
2005
) Fertility preservation for young patients with cancer: who is at risk and what can be offered?
Lancet Oncol
6
,
209
–218.

Wang
X
, Chen H, Yin H, Kim SS, Lin Tan S and Gosden RG (
2002
) Fertility after intact ovary transplantation.
Nature
415
,
385
.

Warne
GL
, Fairley KF, Hobbs JB and Martin FI (
1973
) Cyclophosphamide-induced ovarian failure.
N Engl J Med
289
,
1159
–1162.

Weissman
A
, Gotlieb L, Colgan T, Jurisicova A, Greenblatt EM and Casper RF (
1999
) Preliminary experience with s.c. human ovarian cortex transplantation in the NOD-SCID mouse.
Biol Reprod
60
,
1462
–1467.

Winkel
CA
and Fossum GT (
1993
) Current reproductive technology: considerations for the oncologist.
Oncology (Williston Park)
7
,
40
–45.

Winston
RM
and Browne JC (
1974
) Pregnancy following autograft transplantation of Fallopian tube and ovary in the rabbit.
Lancet
2
,
494
–495.

Wolner-Hanssen
P
, Hägglund L, Ploman F, Ramirez A, Manthrope R and Thuring A (
2005
) Autotransplantation of cryopreserved ovarian tissue to the right forearm 4½ years after autologous stem cell transplantation.
Acta Obstet Gynecol Scand
84
,
695
–698.

Wu
J
, Zhang L and Wang X (
2001
) In vitro maturation, fertilization and embryo development after ultrarapid freezing of immature human oocytes.
Reproduction
121
,
389
–393.

Yang
D
, Winslow K, Blohm P, Brown S, Nguyen K and Brubaker C (
2002
) Oocyte donation using cryopreserved donor oocytes.
Fertil Steril
78
,
S14
.

Yang
HY
, Cox SL, Jenkin G, Findlay J, Trounson A and Shaw J (
2006
) Graft site and gonadotrophin stimulation influences the number and quality of oocytes from murine ovarian tissue grafts.
Reproduction
131
(Suppl),
851
–859.

Yin
H
, Wang X, Kim SS, Chen H, Tans SL and Gosden RG (
2003
) Transplantation of intact rat gonads using vascular anastomosis: effects of cryopreservation, ischaemia and genotype.
Hum Reprod
18
,
1165
–1172.

Yoon
TK
, Chung HM, Lim JM, Han SY, Ko JJ and Cha KY (
2000
) Pregnancy and delivery of healthy infants developed from vitrified oocytes in a stimulated in vitro fertilization-embryo transfer program.
Fertil Steril
74
,
180
–181.