Outlook Truths and myths of oocyte sensitivity to controlled rate freezing

Size: px
Start display at page:

Download "Outlook Truths and myths of oocyte sensitivity to controlled rate freezing"

Transcription

1 RBMOnline - Vol 15. No Reproductive BioMedicine Online; on web 22 May 2007 Outlook Truths and myths of oocyte sensitivity to controlled rate freezing Giovanni Coticchio was awarded a PhD in cell and developmental biology from the University of Palermo, Italy in In , he specialized in human IVF, achieving a Master of Medical Sciences Degree in assisted reproduction technology at the University of Nottingham, UK. Whilst there ( ), he developed his research and clinical embryology interests as post-doctoral fellow. In 1998 he joined Tecnobios Procreazione, Bologna, Italy, where he has been appointed research supervisor. In 1999 he was visiting scientist at the Department of Obstetrics and Gynaecology, University of Sydney, Australia. His major areas of interests are oocyte cryopreservation and oocyte maturation. Dr Giovanni Coticchio G Coticchio 1, MA Bonu, R Sciajno, E Sereni, V Bianchi, A Borini Tecnobios Procreazione, via Dante 15, Bologna, Italy 1 Correspondence: Tel: ; Fax: ; coticchio@tecnobiosprocreazione.it Abstract The mammalian oocyte is especially sensitive to cryopreservation. Because of its size and physiology, it can easily undergo cell death or sub-lethal damage as a consequence of intracellular ice formation, increase in the concentration of solutes and other undesired effects during the conversion of extracellular water into ice. This has generated the belief that oocyte storage cannot be achieved with the necessary efficiency and safety. However, many concerns raised by oocyte freezing are the result of unproven hypotheses or observations conducted under sometimes inappropriate conditions. For instance, spindle organization can undergo damage under certain freezing conditions but not with other protocols. The controversial suggestion that cryopreservation induces cortical granule discharge and zona pellucida hardening somehow questions the routine use of sperm microinjection. Damage to mouse oocytes caused by solute concentration is well documented but, in the human, there is no solid evidence that modifications of freezing mixtures, to prevent this problem, provide an actual advantage. The hope of developing oocyte cryopreservation as a major IVF option is becoming increasingly realistic, but major efforts are still required to clarify the authentic implications of oocyte cryopreservation at the cellular level and identify freezing conditions compatible with the preservation of viability and developmental ability. Keywords: calcium, cryopreservation, cryoprotectant, intracellular ice formation, oocyte, solute concentration Introduction 24 Over the last three decades, IVF has become an established procedure for the treatment of almost all types of human infertility. In this respect, cryopreservation has played an important role, allowing the postponed use of surplus embryos and thereby considerably increasing the chances of achieving a pregnancy per cycle of stimulation (Tiitinen et al., 2004). Nevertheless, embryo storage implies well-recognized legal and ethical complications that make it a less than ideal form of fertility preservation. In principle, oocyte cryopreservation would offer a better option, circumventing the major objections raised against embryo freezing, giving women increased control of their reproductive function and making oocyte donation safer and simpler. In 1977, Whittingham (1977) pioneered the field of oocyte cryopreservation, obtaining in the mouse relatively high postthaw survival rates (60 70%). Whittingham described a reduced fertilization ability of frozen thawed oocytes. However, he also reported that embryos from cryopreserved oocytes displayed high implantation and developmental ability after transfer to pseudopregnant recipients. The first attempts at introducing oocyte cryopreservation into the clinical routine date back to as early as 1986 (Chen, 1986; Al-Hasani et al., 1987). Having been performed with inadequate protocols and methodologies, it should not have been surprising that these studies did not result in significant progress. Human oocyte cryopreservation was not explored for the following decade, having been acritically assumed to be too difficult to achieve. More recently, viable pregnancies obtained from frozen oocytes have stimulated renewed interest. However, in general the IVF community still appears reluctant to replace more orthodox cryopreservation procedures. Reasonably, having been tested only in limited cases, oocyte storage should 2007 Published by Reproductive Healthcare Ltd, Duck End Farm, Dry Drayton, Cambridge CB3 8DB, UK

2 still be considered experimental. For a number of reasons, the mature oocyte is undoubtedly more difficult to preserve than the cleavage-stage embryo. Notwithstanding, the assumption that the oocyte is especially sensitive to low temperature storage has often been based on mere prejudice. For example, concerns on possible damage to the meiotic spindle have been disproved by recent findings based on oocytes cryopreserved with slow cooling protocols, as discussed below. Recently, on the other hand, vitrification has stirred considerable enthusiasm (Kuwayama et al., 2005). The notion that this approach causes less harm to the oocyte and, as consequence, is able to generate a superior clinical outcome is quickly spreading among IVF specialists. In fact, at the moment the evidence in support of these opinions remains largely insufficient or absent. Clearly, the oocyte soft spots are diverse. Studies aiming at assessing possible cell damage after low temperature storage should comprehend objective and appropriate analyses of all the major oocyte attributes, to liberate oocyte cryopreservation from preconceptions or unproven arguments. This paper reviews some of the most significant issues that pertain to the sensitivity of the mature oocyte to the conditions occurring during the thermal transitions between physiological and liquid nitrogen temperatures according to the controlled-rate freezing (slow cooling) procedure. Vitrification, the alternative methodology for the low temperature preservation of cells and tissues, described for the first time by Rall and Fahy (1985), will not be discussed specifically in this review. Births from vitrified oocytes have been described by Kuleshova et al. (1999) and Katayama et al. (2003). Yoon et al. (2003) reported other births from vitrified oocytes, but the overall implantation rate per thawed oocyte (1.7%) was not superior to the one achieved with slow cooling protocols. Recently, Kuwayama et al. (2005) vitrified 64 human oocytes, obtaining very impressive survival, fertilization and blastocyst rates rate (91%, 90% and 62%, respectively). In this study, the pregnancy rate was 43%, an outcome that, if it were possible to reproduce, would represent a landmark progress in oocyte storage. Recently, oocyte cryopreservation with the cryotop vitrification technique has been described to generate high survival (87.2%), fertilization (89.2%), and pregnancy rates (56%), although it should be noted that the majority of pregnancies (9/13) were obtained from oocyte donation cycles (Lucena et al., 2006). Similar data were reported also by Antinori et al. (2007). Oocyte size During controlled rate freezing, the surface-to-volume ratio is a critical factor, influencing the rate of dehydration and therefore the risk of intracellular ice formation. This factor has been deemed to be responsible for the failures of the early attempts of freezing mature oocytes, in the light of the spherical shape and very large size of these cells. In fact, the oocyte geometry is unlikely to be the major reason that makes it not amenable to freezing. The conventional slow cooling protocol, which was originally designed for cleavage-stage embryos, is unable to ensure survival rates in excess of 40 45% when applied to human mature oocytes. Yet interestingly, with the same protocols, it is possible to achieve survival rates ranging around 80% after the freezing and thawing of fully grown germinal vesicle-stage oocytes (Sereni et al., 2000) or pronuclear-stage eggs (Senn et al., 2000). Clearly, during the journey from the breakdown of the germinal vesicle to the formation of pronuclei, some key attributes of the oocyte undergo important changes that can account for such different responses to the same cryopreservation conditions. In line with this hypothesis, Ford et al. (2000) showed that, in rat oocytes, the oolemma becomes less permeable to water during maturation. This is concomitant with the disappearance of the aquaporin-9 transcript. This mrna encodes for a member of the family of membrane-bound proteins described as a broad selective channels, responsible for solute and water transfer. Dynamic changes of the oocyte permeability are also suggested by previous studies in which it was observed that the permeability of mouse oocytes increases during fertilization (Jackowski et al., 1980). This change occurs rather rapidly, within 3 h from sperm penetration. So, although the purely physical characteristics of the oocyte definitely play a role in the exchange of water and cryoprotectants between the intra- and extracellular compartments, other more specific factors, such as membrane permeability, concur more decisively to determine cell survival after thawing. Influence of cryoprotectants on oocyte function Intracellular ice formation is perhaps the major, but surely not the exclusive, source of injury during cryopreservation. As a result of osmotic stress and/or chemical toxicity, cryoprotectants can inflict significant damage, for example, to the delicate mechanism of fertilization. Shaw and Trounson (1989) reported that, in mouse oocytes, exposure at 27 C for 5 min to 1.5 M propane-1,2-diol (PrOH), but not dimethyl sulphoxide (DMSO), causes oocyte activation at a rate (over 80%) comparable to that typically achieved with the classical activating agent ethanol. The difference in the effects of the two cryoprotectants is paradoxical considering that DMSO is generally believed to be more cytotoxic. Besides, the effect of PrOH should be considered in the light of the fact the oocytes used in that study were treated at 15 h after human chorionic gonadotrophin injection, i.e. at a stage in which ageing may have enhanced their sensitivity to activating conditions. Full activation caused by PrOH treatment does not occur when oocytes are treated at an earlier time (13 h) after induction (Stachecki et al., 1998a). Concerning human oocytes, Gook et al. (1995) were the first to conclude that culture and exposure to PrOH during the dehydration and rehydration procedure do not induce parthenogenetic activation. Such a conclusion is confirmed by more recent data, where the observed rates of one-pronucleate oocytes following cryopreservation and microinjection are not higher than the percentages normally obtained with fresh oocytes (Borini et al., 2006; Levi Setti et al., 2006). Nevertheless, the hypothesis that PrOH may trigger, at least to some extent, the activation process remains a matter of debate. One of the early cellular manifestations that occurs during fertilization, and is induced illegitimately during activation, is the release of cortical granules. Under physiological conditions, the enzymatic contents of the granules play the important role of modification of the zona pellucida (zona hardening) and prevention of polyspermic fertilization. In their studies, Gook et al. (1995) showed that fluorescence derived from specific cortical granule staining was comparable in fresh and 25

3 26 frozen oocytes, while it was massively reduced in oocytes that were activated deliberately. This evidence has been recently challenged by fluorescence and electron microscopy studies. By testing the same protocol, Ghetler et al. (2006) found that mere exposure to PrOH leads to a significant reduction in cortical granule staining in human oocytes. Ultrastructural analysis confirmed that freezing and thawing induces extensive loss of cortical granules from the cortex of mature oocytes. Transmission electron microscopy data converge on the hypothesis of a perturbing effect of cryopreservation on the mechanism of cortical granule release. Particularly observed was that the population of sub-cortical granules undergoes a decrease in oocytes frozen with PrOH (Nottola et al., 2006). The magnitude of this phenomenon appears to be influenced by the type of protocol. In particular, a low concentration (0.1 mol/l) of the extracellular cryoprotectant sucrose in the freezing solution is associated with a more conspicuous loss of cortical granules in comparison with a higher concentration (0.3 mol/l) of the same cryoprotectant. Data generated in the mouse model indicate that zona hardening as a consequence of cortical granule release may occur after cryopreservation. Carroll et al. (1990) showed that the low rate of fertilization (48.8%) observed in frozen thawed oocytes could be restored to a normal value (87.8%) after zona drilling. Collectively, this evidence lends credit to the notion that the insemination of cryopreserved human oocytes should be pursued via sperm microinjection rather than standard IVF. In effect, almost all the clinical and experimental experiences gained so far have implied the use of intracytoplasmic sperm injection (ICSI). Exception to this rule is represented by a recent report (Li et al., 2005), which suggests that, in oocytes frozen with a slow cooling method, high fertilization rates (in excess of 80%) can be achieved irrespective of the use of ICSI or standard IVF. Consistent with this, in the same study it is described that immunostaining of frozen thawed oocytes does not reveal evidence of the premature release of cortical granules. This work cannot be considered conclusive, especially because of the small number of oocytes subjected to analysis. Yet, it has the merit to challenge the credence that cortical granules are lost after cryopreservation, thereby stimulating novel studies on this matter. The release of cortical granules is one of the manifestations of the more comprehensive mechanism of fertilization. Central to this process is the regulation via intracellular calcium signalling. As soon as gamete fusion occurs, the sperm-derived factor phospholipase C ζ triggers, via the formation of inositol trisphosphate, a periodical release and uptake of Ca 2+ from the smooth endoplasmic reticulum. The oscillations of cytosolic [Ca 2+ ] thereby generated initiate a sequence of events including cortical granule release, exit from metaphase II (MII) arrest mediated by a decrease in maturation promoting factor and microtubule-associated protein kinase activities, extrusion of the polar body II, pronuclear formation and recruitment of maternal mrnas (Ducibella et al., 2002). In such a perspective, the hypothesis that cryopreservation may cause cortical granule release bears the important implication that [Ca 2+ ] i regulation may also be perturbed. This may not only influence the conversion of an oocyte into a zygote as discussed above but also, in principle, affect the expression of the normal pattern of fetal development. In particular it has been observed in the rabbit that amplitude and frequency of Ca 2+ oscillations experimentally applied to pre-ovulatory oocytes, apart from inducing egg activation, influence the morphology and size of the ensuing parthenogenetically developed fetuses (Ozil and Huneau, 2001). Recent evidence provides arguments for the possibility that the oocyte Ca 2+ release mechanism may be affected by cryopreservation. Measuring the relative fluorescence intensity emitted by the Ca 2+ -specific dye Fluo-3, Jones et al. (2004) found that the ability of cryopreserved human oocytes to release Ca 2+ from the intracellular stores after treatment with the Ca 2+ ionophore A23187 was significantly reduced in comparison to unfrozen controls. The authors speculated that such a defect in the Ca 2+ signalling could alter the normal course of fertilization and have downstream consequences for normal development. However, it should be noticed that the slow cooling protocol applied by Jones et al. appear rather inappropriate. In particular, dehydration was conducted by sequential exposure to 1.5 mol/l PrOH for 20 min and 1.5 mol/l PrOH and 0.2 mol/l sucrose for 10 min, i.e. for time periods far in excess of those actually needed. In fact, Paynter et al. (2001) showed that, after 10 min, the dehydration effect of 1.5 mol/l PrOH is completed and further incubation is unnecessary or perhaps detrimental. In a successive study (Paynter et al., 2005), it was also concluded that the dehydration step achieved with 1.5 mol/l PrOH and 0.2 ml/ sucrose does not require more than 5 min. In addition, as far as is known, the post-thaw rehydration procedure, including the two steps in the presence of sucrose alone applied by Jones et al., are rather arbitrary and do not necessarily reflect conditions aimed at alleviating possible osmotic stress. So it may be entirely possible that the conditions tested compromise the oocyte ability to release Ca 2+ from the intracellular store. Nevertheless, the same conditions diverge significantly from those that should be applied in order to minimize potential damage from cryostorage. More recently, Larman et al. (2006) also investigated possible detrimental effects of cryoprotectants and cryopreservation on the physiology of mouse oocytes. By using fluorescence microscopy, intracellular [Ca 2+ ] changes were monitored using the signal emitted by the Ca 2+ -sensitive dye Indo-1 AM. They observed that treatment with DMSO, and to lesser extent ethylene glycol, caused an important and prolonged rise in intracellular [Ca 2+ ] in association with hardening of the zona. According to the authors, this could explain the poor efficiency of cryopreservation, considering the central role of intracellular [Ca 2+ ] i in the fertilization process as well as in much more advanced developmental stages, as discussed above. Yet, similar to the case of the study of Jones et al., the conditions under which the DMSO effects were tested by Larman et al. do not coincide with those identified as more appropriate for mouse oocyte cryopreservation. In particular, these authors monitored changes in [Ca 2+ ] i caused by DMSO exposure at room temperature but, in fact, slow-cooling storage of mouse oocytes with this cryoprotectant requires incubation at 0 4 C. Over two decades ago, Carroll et al. (1993) successfully cryopreserved mouse oocytes with a slow-cooling method involving exposure to DMSO pre-cooled at 0 C. After thawing, these oocytes underwent fertilization after standard IVF and pre- and post-implantation development with rates comparable to those of non-frozen controls, suggesting that, under the conditions of the study, crucial events that are regulated by the [Ca 2+ ] i signalling system, such as cortical granule release and expression of epigenetic information, are not perturbed.

4 In conclusion, while it is conceivable that cryopreservation conditions may influence the mechanism of [Ca 2+ ] i regulation, it remains to be established whether, in all cases, this effect may compromise oocyte physiology, either neutrally or paradoxically beneficial. The experiences conducted by Jones et al. (2004) and Larman et al. (2006) are certainly interesting and stimulating. However, they provide information of limited importance because they are derived from experimental conditions that do not reflect those applied in the most advanced and efficient protocols. On the other hand, the work conducted on the mouse (Carroll et al., 1993) bears the important lesson that, given appropriate conditions, cryopreserved oocytes can be fertilized successfully with standard IVF in the absence of any sort of micromanipulation-assisted insemination. There is no reason to believe that the human oocyte is governed by a different physiology of fertilization, an assumption that should encourage novel studies aiming at adopting standard IVF as the method of choice and circumventing the potentially negative implications of ICSI. Cytoskeletal sensitivity to cryopreservation The effects of low temperatures on the oocyte cytoskeletal organization (Pickering and Johnson, 1987; Pickering et al., 1990; Wang et al., 2001) and the physical stresses to which the whole cell is subjected during the dehydration rehydration and phase transitions of extracellular water (Leibo et al., 1978) are well known. Direct observation of the freezing process has unveiled that the growing ice front distorts the oocyte, an effect that is independent from the osmotic stress imposed by the presence of cryoprotectants in the freezing solution and remains until thawing. This predisposes the oocyte to blebbing of the oolemma and physical damage of intracellular structures (Ashwood-Smith et al., 1988). For such reasons, preservation of the MII spindle in frozen thawed oocytes has raised concerns since the very early days of oocyte cryopreservation. This matter has remained contended over a number of years, with several studies for and against the hypothesis of spindle disruption after cryopreservation (Coticchio et al., 2005). Recent evidence extends the dispute. Mullen et al. (2004) exposed mature human oocytes to anisosmotic conditions generated by using buffer solutions including different concentrations of sucrose, with the aim of simulating the osmotic stress occurring during slow cooling and ascertaining possible consequences to the MII spindle structure. The authors found that both hypo- and hyperosmotic conditions caused a decrease in the proportion of oocytes with a normal spindle, in extreme cases with a total loss of a recognizable microtubular structure. However, these authors did not include penetrating cryoprotectants in their treatments, which may influence microtubule dynamics (Pickering and Johnson, 1987). Recently, evidence from the authors study centre was reported that argues against the loss of normal spindle organization after cryostorage (Coticchio et al., 2006). In fact, by using confocal microscopy, partial disruption of the MII spindle and associated chromosomes was found only in coincidence with slow-cooling conditions that were showed to be inadequate for oocyte storage (Borini et al., 2004). However, it is known that a protocol adopting a higher sucrose concentration in the freezing solution (Fabbri et al., 2001) promotes the retention of an intact chromosome segregation apparatus comparable in incidence to freshly collected oocytes. Ultrastructural analysis (Nottola et al., 2006), on oocytes frozen with the same high sucrose concentration protocol, established also that the MII spindle was regularly barrel-shaped, anastral, with slightly pointed poles and properly positioned in the ooplasm of both fresh and frozen thawed oocytes. The suggestion that the structure of the MII spindle can remain unaltered after cryopreservation given appropriate conditions converges with clinical experience. Should the MII spindle be significantly affected by the freezing procedure, flawed chromosome segregation at meiosis II would occur at high frequency and the aneuploidy condition produced in the ensuing embryo would doubtless increase the proportion of early pregnancy loss. In fact, the same cryopreservation conditions can generate pregnancies whose abortion rate is comparable to the one normally observed in pregnancies from fresh cycles (Borini et al., 2006). Low abortion rates in pregnancies from frozen oocytes have been confirmed in relation to the use of another slow-cooling protocol (Bianchi et al., 2007), whose effects on the MII spindle are currently under investigation. Another important component of the oocyte cytoskeletal apparatus is the cortical meshwork of actin filaments, whose integrity is essential for the cytokinesis events, including polar body II extrusion and the first cleavage after fertilization. Evidence of possible effects of cryostorage on actin filaments is virtually lacking. However, the study of Younis et al. (1996) that described a loss of polymeric actin in rhesus mature oocytes after exposure to glycerol at 20 C should be considered with some concern, although this particular cryoprotectant is not normally used for oocyte cryopreservation. Should cryopreservation give rise to disruption to cortical actin, the predictable consequences would correspond to retention of the polar body II and inhibition of cleavage after fertilization. In effect, an increased frequency in three-pronuclear fertilization in frozen thawed mouse oocytes (Carroll et al., 1989) and a rise in cleavage arrest in fertilized human eggs (Hunter et al., 1995) have been reported after storage with inappropriate protocols. L De Santis and others (personal communication) using fluorescence microscopy and phalloidin staining, have observed that, in oocytes cryopreserved with a high sucrose concentration protocol (Fabbri et al., 2001), the sub-cortical actin meshwork was unchanged in comparison to control oocytes. Also, data on the developmental performance of frozen thawed oocytes are in agreement with the conservation of a fully functional subcortical actin ring after cryopreservation. In two separate series of treatments with frozen oocytes (Borini et al., 2006; Bianchi et al., 2007), a low incidence of one- or three-pronuclear fertilization (5.8% and 6.3% in the two studies, respectively) and a high cleavage rate (90.2% and 93.7%, respectively) have been reported. These frequencies are indistinguishable from those normally generated with fresh oocytes. Sensitivity to high solute concentration during slow cooling During slow cooling, intracellular ice formation is essentially prevented by cell dehydration. This can be achieved in a controlled fashion by the strength of the osmotic gradient 27

5 28 imposed by the concentration of cryoprotectant in the freezing solution. Another, not less important, phase of dehydration occurs during the progressive conversion of liquid water into ice, a process that leads to the concentration of solutes, mainly sodium, in the unfrozen fraction of the system, a condition that draws further water out of the cell. Sufficient dehydration at this stage is guaranteed, provided that the cooling rate and consequent ice formation are sufficiently slow. However, apart from such a beneficial effect, high solute concentration may also be the source of biochemical toxicity. In particular, the stability of intracellular membranes and the plasmalemma are believed to be particularly sensitive to high solute concentration. It should be noticed, nevertheless, that cleavage-stage embryos can be safely cryopreserved with sodium-rich solutions. Because sodium is the most represented solute in freezing mixtures, it has been suggested that replacing this element with the cation choline, which does not diffuse through the plasmalemma, could significantly reduce the detrimental effects of high solute concentration on oocyte viability. On this basis, in a slow-cooling study conducted on mouse oocytes, Stachecki et al. (1998b) investigated the effects of replacing increasing proportions of sodium with equimolar amounts of choline. The authors found that the rates of survival, fertilization and development were very poor in the presence of high sodium concentration, but were maximal after replacing sodium with choline. The association between the use of choline-based media and high post-thaw survival has been subsequently expanded and confirmed with other experiments in the mouse (Stachecki and Willadsen, 2000). Choline-based media proved themselves objectively superior to conventional freezing formulation. Nevertheless, the real importance of the solution effect in oocyte cryopreservation is not entirely clarified. In particular, it is interesting to note that, by using sodium-rich media for cryopreserving mouse oocytes, Carroll et al. (1993) obtained remarkable rates of survival, fertilization and pre- and postimplantation, although overall moderately lower than those achieved by Stachecki et al. (2002) with choline-based freezing solutions. This suggests that the detrimental effects of high sodium concentration during slow cooling do not occur systematically but rather depend on, or are enhanced by, other conditions. For instance, a major difference in the protocols employed by Carroll et al. (1993) and Stachecki et al. (1998b) is the type of penetrating cryoprotectant, DMSO in the former case and PrOH in the latter. Therefore, while the solution effect is a well-recognized drawback of the slow-cooling strategy, nevertheless its relative importance in determining post-thaw cell viability may vary. Recently, low-sodium media have been tested by Stachecki et al. (2006) on human oocytes, with results that are difficult to assess and that also raise some questions. The author reported that media based on choline and 0.2 mol/l sucrose can guarantee survival rates as high as 90%. However, it is difficult to appreciate such an outcome, considering that the treatment groups of this study included no more than oocytes. In addition, no control groups frozen with sodium-rich media were included. Therefore, it cannot be concluded that cholinebased media have a genuinely beneficial effect on the viability of human oocytes. Lastly, after thawing and fertilization, even the choline-based freezing mixture that produced the highest survival rate generated a low rate of cleavage (78%). On the contrary, as discussed above, various conventional sodium-rich freezing media ensure cleavage rates (90 93%) that appear unchanged compared with unfrozen control (Borini et al., 2006; Levi Setti et al., 2006). Sodium-depleted media have also been recently applied in clinical IVF (Boldt et al., 2006). Survival rates were suboptimal (61% and 59%), irrespective of the adoption of phosphate - buffered saline (PBS) or HEPES as ph buffers in separate treatment groups. In the PBS group, fertilization after ICSI was also inadequate (55.8%), while cleavage rates were not reported for either group. Based on implantation rates of 10.6% and 15.9% obtained with the PBS and HEPES groups, respectively, the authors concluded that sodium-depleted media is beneficial to the cryopreservation of human oocytes. It should be considered, however, that the freezing solution formulation adopted by Boldt et al. (2006) was not the same as the one successfully developed with mouse oocytes by Stachecki et al. (2002). In fact, the cryopreservation method applied by Boldt et al. (2006) included other changes in comparison with more traditional slow-cooling protocols. Firstly, dehydration was performed in a single step with a freezing mixture containing PrOH and sucrose. Secondly, post-thaw rehydration was carried out with a single solution containing a high sucrose concentration (0.5 mol/l), but no decreasing concentrations of PrOH. Individually or in association, these two protocol modifications, rather than the use of choline, may have produced the observed outcome. Thawing conditions So far, attempts to improve oocyte cryopreservation protocols have involved changes mainly limited to the dehydration steps required before freezing or the composition of the freezing mixtures. Limited attention has been given to the optimization of post-thaw rehydration conditions (Tao et al., 1995; Stachecki and Willadsen, 2000). In reality, rehydration is likely to be as important as other steps of the entire procedure, or perhaps even more. Removal of the intracellular cryoprotectant and reestablishment of the original water content is an obvious source of osmotic stress. This occurs after exposure of the oocyte to the very challenging conditions of freezing and thawing. Such treatments could sensitize the cell and make it particularly vulnerable. Nothing is known on the magnitude and dynamics of the oocyte volume excursions imposed by the diverse thawing solutions. Currently, applied dehydration conditions for oocyte freezing are generally derived from protocols designed for cleavage-stage embryos and are likely to be rather inappropriate. It is not a coincidence that an important proportion of oocytes that appear intact immediately after thawing and release from the storage device afterwards degenerate during rehydration. Therefore, rehydration conditions should be put under rigorous scrutiny in a fashion similar to the investigations conducted on the pre-freezing dehydration phase (Paynter et al., 2001, 2005). Conclusions For many years, oocyte cryopreservation has been considered with little enthusiasm and even scepticism. The oocyte characteristics are surely unique, making this cell not amenable to freezing. Critical changes in the plasmalemma occurring during the various functional stages regulate the passage of water and small molecules (including cryoprotectants) between the intra- and extracellular compartment, influencing the rate

6 of survival after freezing and thawing in final analysis. Under particular conditions, cryoprotectants make storage possible, but are themselves a potential source of cell damage. Their toxicity may depend on the species considered, making findings obtained in animal models not always applicable to the human. For example, PrOH causes substantial activation in aged mouse oocytes, but can activate aged human oocytes only to a rather limited extent. This should be taken into account to appreciate the relative importance and implications of activation-related events, such as [Ca 2+ ] i changes, which will most likely occur in all mammalian oocytes following cryoprotectant treatment but whose outcome may differ between species. Cortical granules are not always released, indicating that insemination via microinjection could be avoided. Observations made here argue against a high risk of chromosome and meiotic spindle disorganization, under appropriate storage conditions. This should clear the ground from one of the major arguments that has fuelled concerns about an increased risk of aneuploidy in frozen oocytes. Cryodamage has been postulated to be derived from the increase in solute concentration during freezing. This appears entirely plausible and efforts aiming at clarifying such a possibility should be encouraged. However, the experiences reported in the human so far have not demonstrated that the solution effect has a major impact on oocyte survival and viability. Protocol optimization should be intended as an extensive endeavour. It is unlikely that decisive improvements will originate from individual changes. Rather, multiple approaches should be undertaken to establish better overall freezing conditions. In such a perspective, rehydration after thawing should be assessed with particular interest. References Al-Hasani S, Diedrich K, van der Ven H et al Cryopreservation of human oocytes. Human Reproduction 2, Antinori M, Licata E, Dani G et al Cryotop vitrification of human oocytes results in high survival rate and healthy deliveries. Reproductive BioMedicine Online 14, Ashwood-Smith MJ, Morris GW, Fowler R et al Physical factors are involved in the destruction of embryos and oocytes during freezing and thawing procedures. Human Reproduction 3, Bianchi V, Coticchio G, Distratis V et al Differential sucrose concentration during dehydration (0.2 mol/l) and rehydration (0.3 mol/l) increases the implantation rate of frozen human oocytes. Reproductive BioMedicine Online 14, Boldt J, Tidswell N, Sayers A et al Human oocyte cryopreservation: 5-year experience with a sodium-depleted slow freezing method. Reproductive BioMedicine Online 13, Borini A, Sciajno R, Bianchi V et al Clinical outcome of oocyte cryopreservation after slow cooling with a protocol utilizing a high sucrose concentration. Human Reproduction 21, Borini A, Bonu MA, Coticchio G et al Pregnancies and births after oocyte cryopreservation. Fertility and Sterility 82, Carroll J, Wood MJ, Whittingham DG 1993 Normal fertilization and development of frozen thawed mouse oocytes: protective action of certain macromolecules. Biology of Reproduction 48, Carroll J, Depypere H, Matthews CD 1990 Freeze thaw-induced changes of the zona pellucida explains decreased rates of fertilization in frozen thawed mouse oocytes. Journal of Reproduction and Fertility 90, Carroll J, Warnes GM, Matthews CD 1989 Increase in digyny explains polyploidy after in-vitro fertilization of frozen thawed mouse oocytes. Journal of Reproduction and Fertility 85, Chen C 1986 Pregnancy after human oocyte cryopreservation. Lancet 1, Coticchio G, De Santis L, Rossi G et al Sucrose concentration influences the rate of human oocytes with normal spindle and chromosome configurations after slow-cooling cryopreservation. Human Reproduction 21, Coticchio G, Bonu MA, Bianchi V et al Criteria to assess human oocyte quality after cryopreservation. Reproductive BioMedicine Online 11, Ducibella T, Huneau D, Angelichio E et al Egg-to-embryo transition is driven by differential responses to Ca 2+ oscillation number. Developmental Biology 250, Fabbri R, Porcu E, Marsella T et al Human oocyte cryopreservation: new perspectives regarding oocyte survival. Human Reproduction 16, Ford P, Merot J, Jawerbaum A et al Water permeability in rat oocytes at different maturity stages: aquaporin-9 expression. Journal of Membrane Biology 176, Ghetler Y, Skutelsky E, Ben Nun I et al Human oocyte cryopreservation and the fate of cortical granules. Fertility and Sterility 86, Gook DA, Osborn SM, Johnston WI 1995 Parthenogenetic activation of human oocytes following cryopreservation using 1,2-propanediol. Human Reproduction 10, Hunter JE, Fuller BJ, Bernard A et al Vitrification of human oocytes following minimal exposure to cryoprotectants; initial studies on fertilization and embryonic development. Human Reproduction 10, Jackowski S, Leibo SP, Maxur P 1980 Glycerol permeabilities of fertilized and infertilized mouse ova. Journal of Experimental Zoology 212, Jones A, Van Blerkom J, Davis P et al Cryopreservation of metaphase II human oocytes effects mitochondrial membrane potential: implications for developmental competence. Human Reproduction 19, Katayama KP, Stehlik J, Kuwayama M et al High survival rate of vitrified human oocytes results in clinical pregnancy. Fertility and Sterility 80, Kuleshova L, Gianaroli L, Magli C et al Birth following vitrification of a small number of human oocytes: case report. Human Reproduction 14, Kuwayama M, Vajta G, Kato O et al Highly efficient vitrification method for cryopreservation of human oocytes. Reproductive BioMedicine Online 11, Larman MG, Sheehan CB, Gardner DK 2006 Calcium-free vitrification reduces cryoprotectant-induced zona pellucida hardening and increases fertilization rates in mouse oocytes. Reproduction 131, Leibo SP, McGrath JJ, Cravalho EG 1978 Microscopic observation of intracellular ice formation in unfertilized mouse ova as a function of cooling rate. Cryobiology 15, Levi Setti PE, Albani E, Novara PV et al Cryopreservation of supernumerary oocytes in IVF/ICSI cycles. Human Reproduction 21, Li XH, Chen SU, Zhang X et al Cryopreserved oocytes of infertile couples undergoing assisted reproductive technology could be an important source of oocyte donation: a clinical report of successful pregnancies. Human Reproduction 20, Lucena E, Bernal DP, Lucena C et al Successful ongoing pregnancies after vitrification of oocytes. Fertility and Sterility 85, Mullen SF, Agca Y, Broermann DC et al The effect of osmotic stress on the metaphase II spindle of human oocytes, and the relevance to cryopreservation. Human Reproduction 19, Nottola SA, Macchiarelli G, Coticchio G et al Ultrastructure of human mature oocytes after slow cooling cryopreservation using different sucrose concentrations. Human Reproduction 22, Ozil JP, Huneau D 2001 Activation of rabbit oocytes: the impact of the Ca2+ signal regime on development. Development 128, Paynter SJ, Borini A, Bianchi V et al Volume changes of mature 29

7 human oocytes on exposure to cryoprotectant solutions used in slow cooling procedures. Human Reproduction 20, Paynter SJ, O Neil L, Fuller BJ et al Membrane permeability of human oocytes in the presence of the cryoprotectant propane-1,2- diol. Fertility and Sterility 75, Pickering SJ, Johnson MH 1987 The influence of cooling on the organization of the meiotic spindle of the mouse oocyte. Human Reproduction 2, Pickering SJ, Braude PR, Johnson MH et al Transient cooling to room temperature can cause irreversible disruption of the meiotic spindle in the human oocyte. Fertility and Sterility 54, Rall WF, Fahy GM 1985 Ice-free cryopreservation of mouse embryos at 196 degrees C by vitrification. Nature 313, Senn A, Vozzi C, Chanson A et al Prospective randomized study of two cryopreservation policies avoiding embryo selection: the pronucleate stage leads to a higher cumulative delivery rate than the early cleavage stage. Fertility and Sterility 74, Sereni E, Bonu MA, Borini A et al High survival rate after cryopreservation of human prophase I oocytes. Fertility and Sterility 74 (Suppl. 1), S161. Shaw JM, Trounson AO 1989 Parthenogenetic activation of unfertilized mouse oocytes by exposure to 1,2-propanediol is influenced by temperature, oocyte age, and cumulus removal. Gamete Research 24, Stachecki JJ, Willadsen SM 2000 Cryopreservation of mouse oocytes using a medium with low sodium content: effect of plunge temperature. Cryobiology 40, Stachecki JJ, Cohen J, Garrisi J et al Cryopreservation of unfertilized human oocytes. Reproductive BioMedicine Online 13, Stachecki JJ, Cohen J, Schimmel T et al Fetal development of mouse oocytes and zygotes cryopreserved in a nonconventional freezing medium. Cryobiology 44, Stachecki JJ, Cohen J, Willadsen SM 1998a Cryopreservation of unfertilized mouse oocytes: the effect of replacing sodium with choline in the freezing medium. Cryobiology 37, Stachecki JJ, Cohen J, Willadsen S 1998b Detrimental effects of sodium during mouse oocyte cryopreservation. Biology of Reproduction 59, Tao J, Du J, Kleinhans FW et al The effect of collection temperature, cooling rate and warming rate on chilling injury and cryopreservation of mouse spermatozoa. Journal of Reproduction and Fertility 104, Tiitinen A, Hyden-Granskog C, Gissler M 2004 What is the most relevant standard of success in assisted reproduction?: The value of cryopreservation on cumulative pregnancy rates per single oocyte retrieval should not be forgotten. Human Reproduction 19, Wang WH, Meng L, Hackett RJ et al Limited recovery of meiotic spindles in living human oocytes after cooling rewarming observed using polarized light microscopy. Human Reproduction 16, Whittingham DG 1977 Fertilization in vitro and development to term of unfertilized mouse oocytes previously stored at 196 degrees C. Journal of Reproduction and Fertility 49, Yoon TK, Kim TJ, Park SE et al Live births after vitrification of oocytes in a stimulated in-vitro fertilization embryo transfer program. Fertility and Sterility 79, Younis AI, Toner M, Albertini DF et al Cryobiology of non-human primate oocytes. Human Reproduction 11, Paper based on contribution presented at the Tecnobios Procreazione Symposium 2006 and 2nd International Conference on the Cryopreservation of the Human Oocyte in Bologna, Italy, 5 7 October Received 19 February 2007; refereed 15 March 2007; accepted 30 April

RBMOnline - Vol 14. No Reproductive BioMedicine Online; on web 14 November 2006

RBMOnline - Vol 14. No Reproductive BioMedicine Online;   on web 14 November 2006 RBMOnline - Vol 14. No 1. 2007 64-71 Reproductive BioMedicine Online; www.rbmonline.com/article/2455 on web 14 November 2006 Novel protocols have increased survival and fertilization rates of cryopreserved

More information

RBMOnline - Vol 15. No Reproductive BioMedicine Online; on web 19 July 2007

RBMOnline - Vol 15. No Reproductive BioMedicine Online;   on web 19 July 2007 RBMOnline - Vol 15. No 3. 2007 338-345 Reproductive BioMedicine Online; www.rbmonline.com/article/2889 on web 19 July 2007 Recent studies of fundamental cryobiology, empirical observations and more systematic

More information

Cryopreservation of human oocytes with slow freezing techniques

Cryopreservation of human oocytes with slow freezing techniques ESHRE Campus Symposium Cryobiology and cryopreservation of human gametes and embryos Athens, Greece 25-26 September 2009 Cryopreservation of human oocytes with slow freezing techniques Giovanni Coticchio

More information

Dr. Andrea Borini Clinical and Scientific Director Tecnobios Procreazione Bologna, Italy

Dr. Andrea Borini Clinical and Scientific Director Tecnobios Procreazione Bologna, Italy Oocyte Cryopreservation: Beating the Clock Dr. Andrea Borini Clinical and Scientific Director Tecnobios Procreazione Bologna, Italy Question Why do we need to cryopreserve oocytes? Answers For women suffering

More information

Article Cryopreservation of immature and in-vitro matured human oocytes by vitrification

Article Cryopreservation of immature and in-vitro matured human oocytes by vitrification RBMOnline - Vol 19 No 3. 2009 369-373 Reproductive BioMedicine Online; www.rbmonline.com/article//3704 on web 8 July 2009 Article Cryopreservation of immature and in-vitro matured human oocytes by vitrification

More information

RapiDVIT & rapidwarm oocyte. Specialised media for oocyte vitrification.

RapiDVIT & rapidwarm oocyte. Specialised media for oocyte vitrification. RapiDVIT & rapidwarm oocyte Specialised media for oocyte vitrification. Special media for A unique cell Cryopreservation of oocytes requires care. Some preservation techniques cause premature oocyte activation

More information

Article A rational approach to oocyte cryopreservation

Article A rational approach to oocyte cryopreservation RBMOnline - Vol 10. No 5. 2005 578 586 Reproductive BioMedicine Online; www.rbmonline.com/article/1657 on web 28 February 2005 Article A rational approach to oocyte cryopreservation Dr Paynter was awarded

More information

Slow freezing of mouse embryos Slow freezing of domestic animal embryos Slow freezing of human embryos 1972 1973/74 1983 Slow freezing of human embryos Slow freezing of human oocytes 1985 1989 1993 1996

More information

Theoretical and experimental basis of slow freezing

Theoretical and experimental basis of slow freezing Reproductive BioMedicine Online (2011) 22, 125 132 www.sciencedirect.com www.rbmonline.com REVIEW Theoretical and experimental basis of slow freezing Lucia De Santis a, Giovanni Coticchio b, * a IVF Unit,

More information

A critical appraisal of cryopreservation (slow cooling versus vitrification) of human oocytes and embryos

A critical appraisal of cryopreservation (slow cooling versus vitrification) of human oocytes and embryos Human Reproduction Update Advance Access published April 25, 2012 Human Reproduction Update, Vol.0, No.0 pp. 1 19, 2012 doi:10.1093/humupd/dms016 A critical appraisal of cryopreservation (slow cooling

More information

Cryotop Vitrification Affects Oocyte Quality and Embryo Developmental Potential

Cryotop Vitrification Affects Oocyte Quality and Embryo Developmental Potential Cronicon OPEN ACCESS Ling Jia*, Bo Xu*, Yu-sheng Liu and Xian-hong Tong Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, China *These authors contributed

More information

Comparison of survival and embryonic development in human oocytes cryopreserved by slow-freezing and vitrification

Comparison of survival and embryonic development in human oocytes cryopreserved by slow-freezing and vitrification Comparison of survival and embryonic development in human oocytes cryopreserved by slow-freezing and vitrification Yun-Xia Cao, M.D., Ph.D., Qiong Xing, M.D., Li Li, M.D., Lin Cong, M.D., Zhi-Guo Zhang,

More information

Abstract. Introduction. RBMOnline - Vol 19. No Reproductive BioMedicine Online; on web 21 August 2009

Abstract. Introduction. RBMOnline - Vol 19. No Reproductive BioMedicine Online;   on web 21 August 2009 RBMOnline - Vol 19. No 4. 2009 521 525 Reproductive BioMedicine Online; www.rbmonline.com/article/4153 on web 21 August 2009 Article Increasing dehydration of human cleavagestage embryos prior to slow

More information

Preimplantation genetic diagnosis: polar body and embryo biopsy

Preimplantation genetic diagnosis: polar body and embryo biopsy Human Reproduction, Vol. 15, (Suppl. 4), pp. 69-75, 2000 Preimplantation genetic diagnosis: polar body and embryo biopsy Luca Gianaroli SISMER, Via Mazzini 12, 40138 Bologna, Italy Scientific Director

More information

Sucrose concentration influences the rate of human oocytes with normal spindle and chromosome configurations after slow-cooling cryopreservation*

Sucrose concentration influences the rate of human oocytes with normal spindle and chromosome configurations after slow-cooling cryopreservation* Human Reproduction Vol.21, No.7 pp. 1771 1776, 2006 Advance Access publication March 20, 2006. doi:10.1093/humrep/del073 Sucrose concentration influences the rate of human oocytes with normal spindle and

More information

Limited recovery of meiotic spindles in living human oocytes after cooling rewarming observed using polarized light microscopy

Limited recovery of meiotic spindles in living human oocytes after cooling rewarming observed using polarized light microscopy Human Reproduction Vol.16, No.11 pp. 2374 2378, 2001 Limited recovery of meiotic spindles in living human oocytes after cooling rewarming observed using polarized light microscopy Wei-Hua Wang 1,2,4, Li

More information

FERTILITY PRESERVATION. Juergen Eisermann, M.D., F.A.C.O.G South Florida Institute for Reproductive Medicine South Miami Florida

FERTILITY PRESERVATION. Juergen Eisermann, M.D., F.A.C.O.G South Florida Institute for Reproductive Medicine South Miami Florida FERTILITY PRESERVATION Juergen Eisermann, M.D., F.A.C.O.G South Florida Institute for Reproductive Medicine South Miami Florida 1 2 3 4 Oocyte Cryopreservation Experimental option Offer to single cancer

More information

Permeability of human oocytes to ethylene glycol and their survival and spindle configurations after slow cooling cryopreservation

Permeability of human oocytes to ethylene glycol and their survival and spindle configurations after slow cooling cryopreservation Human Reproduction Vol.22, No.10 pp. 2776 2783, 2007 Advance Access publication on August 3, 2007 doi:10.1093/humrep/dem240 Permeability of human oocytes to ethylene glycol and their survival and spindle

More information

Oocyte slow freezing using a M sucrose concentration protocol: is it really the time to trash the cryopreservation machine?

Oocyte slow freezing using a M sucrose concentration protocol: is it really the time to trash the cryopreservation machine? Oocyte slow freezing using a 0.2 0.3 M sucrose concentration protocol: is it really the time to trash the cryopreservation machine? Veronica Bianchi, Ph.D., Michela Lappi, B.Sc., Maria Antonietta Bonu,

More information

Interspecies Challenges

Interspecies Challenges Cryobiological Challenges of Banking Reproductive Cells, and Tissues Interspecies Challenges Mammals Domestic species Lab animal species Endangered Species Humans (Reproductive Med) Birds Domestic species

More information

Impact of oocyte cryopreservation on embryo development

Impact of oocyte cryopreservation on embryo development Impact of oocyte cryopreservation on embryo development M. Cristina Magli, M.Sc., Michela Lappi, B.Sc., Anna P. Ferraretti, M.D., Alessandra Capoti, B.Sc., Alessandra Ruberti, B.Sc., and Luca Gianaroli,

More information

Article Cryotop vitrification of human oocytes results in high survival rate and healthy deliveries

Article Cryotop vitrification of human oocytes results in high survival rate and healthy deliveries RBMOnline - Vol 14 No 1. 2007 72-79 Reproductive BioMedicine Online; www.rbmonline.com/article/2548 on web 23 November 2006 Article Cryotop vitrification of human oocytes results in high survival rate

More information

Vitrification of Oocytes: Biological Lessons Learned From Mice, Applied to Women

Vitrification of Oocytes: Biological Lessons Learned From Mice, Applied to Women ESHRE Cryobiology Mtg Athens, Greece 9/26/09 Vitrification of Oocytes: Biological Lessons Learned From Mice, Applied to Women Gary D. Smith Ph.D., HCLD Associate Professor Director of Reproductive Sciences

More information

Pregnancies and births after oocyte cryopreservation

Pregnancies and births after oocyte cryopreservation FERTILITY AND STERILITY VOL. 82, NO. 3, SEPTEMBER 2004 Copyright 2004 American Society for Reproductive Medicine Published by Elsevier Inc. Printed on acid-free paper in U.S.A. Pregnancies and births after

More information

Abstract. Introduction. RBMOnline - Vol 8. No Reproductive BioMedicine Online; on web 15 December 2003

Abstract. Introduction. RBMOnline - Vol 8. No Reproductive BioMedicine Online;   on web 15 December 2003 RBMOnline - Vol 8. No 2. 207-211 Reproductive BioMedicine Online; www.rbmonline.com/article/1023 on web 15 December 2003 Article Determining the most optimal stage for embryo cryopreservation Anthony Anderson

More information

Article Human oocyte vitrification: in-vivo and in-vitro maturation outcomes

Article Human oocyte vitrification: in-vivo and in-vitro maturation outcomes RBMOnline - Vol 17. No 5. 2008 684-688 Reproductive BioMedicine Online; www.rbmonline.com/article/3484 on web 30 September 2008 Article Human oocyte vitrification: in-vivo and in-vitro maturation outcomes

More information

VERGE 3 Lundeberg 1. Dependence of fertilization in sea urchins, Strongylocentrotus purpuratus, on microfilament

VERGE 3 Lundeberg 1. Dependence of fertilization in sea urchins, Strongylocentrotus purpuratus, on microfilament VERGE 3 Lundeberg 1 Dependence of fertilization in sea urchins, Strongylocentrotus purpuratus, on microfilament formation and internal calcium concentration Megan Lundeberg Amy Ruggerio and Amy Isaacson

More information

Assessment of a ne w cryoloop vitrif ication protocol in the cryopreservation of mouse mature oocytes

Assessment of a ne w cryoloop vitrif ication protocol in the cryopreservation of mouse mature oocytes 38 2008 2 17 1, 3,, (,,, 100730) ED15 (15 %ethylene glycol, EG + 15 % dimethylsulphoxide, DMSO),,, ( EG) (DMSO) 0 1 2 h, ;, 98. 2 %, 0 1 2 h (87. 0 % 90. 9 % 90. 3 % vs 95 %, P > 0. 05 ;91. 3 % 95. 4 %

More information

Oocyte cryopreservation: slow freezing and vitrification. Laura Rienzi, Rome, Italy Senior Clinical Embryologist

Oocyte cryopreservation: slow freezing and vitrification. Laura Rienzi, Rome, Italy Senior Clinical Embryologist Consensus meeting on fertility preservation Barcelona, June 6 th -7 th 2011 Oocyte cryopreservation: slow freezing and vitrification Laura Rienzi, Rome, Italy Senior Clinical Embryologist Cryopreservation

More information

OVERVIEW AND FACTS: CRYOPRESERVATION

OVERVIEW AND FACTS: CRYOPRESERVATION OVERVIEW AND FACTS: CRYOPRESERVATION Imprint Published in August, 2013 By Victory A.R.T. Laboratory Phils, Inc. This ebook was created by http://www.ivfvictoryphilippines.com/ in hopes of helping bring

More information

Understanding eggs, sperm and embryos. Marta Jansa Perez Wolfson Fertility Centre

Understanding eggs, sperm and embryos. Marta Jansa Perez Wolfson Fertility Centre Understanding eggs, sperm and embryos Marta Jansa Perez Wolfson Fertility Centre What does embryology involve? Aims of the embryology laboratory Creation of a large number of embryos and supporting their

More information

The Cytotoxic Effect of Cryoprotective Agents on in vitro Fertilization Rates of Mammalian Oocytes

The Cytotoxic Effect of Cryoprotective Agents on in vitro Fertilization Rates of Mammalian Oocytes Cean A. et al./scientific Papers: Animal Science and Biotechnologies, 2013, 46 (2) The Cytotoxic Effect of Cryoprotective Agents on in vitro Fertilization Rates of Mammalian Oocytes Ada Cean 1,2,*, Ivan

More information

Oocyte Cryopreservation

Oocyte Cryopreservation J. Mamm. Ova Res. Vol. 24, 2 7, 2007 2 Mini Review Oocyte Cryopreservation Masashige Kuwayama 1 * 1 Kato Ladies Clinic, Advanced Medical Research Institute of Fertility, 7-20-3, Nishishinjuku, Shinjuku,

More information

ASSISTED REPRODUCTIVE TECHNOLOGIES (ART)

ASSISTED REPRODUCTIVE TECHNOLOGIES (ART) ASSISTED REPRODUCTIVE TECHNOLOGIES (ART) Dr. Herve Lucas, MD, PhD, Biologist, Andrologist Dr. Taher Elbarbary, MD Gynecologist-Obstetrician Geneva Foundation for Medical Education and research Definitions

More information

The 5th World congress of the INTERNATIONAL SOCIETY FOR FERTILITY PRESERVATION Vienna, Austria November 16-18, 2017

The 5th World congress of the INTERNATIONAL SOCIETY FOR FERTILITY PRESERVATION Vienna, Austria November 16-18, 2017 Vitrification: "Robots" versus Human Comparing automated vitrification outcomes The 5th World congress of the INTERNATIONAL SOCIETY FOR FERTILITY PRESERVATION Vienna, Austria November 16-18, 2017 Zsolt

More information

Abstract. Introduction. Materials and methods. Patients and methods

Abstract. Introduction. Materials and methods. Patients and methods RBMOnline - Vol 8. No 3. 344-348 Reproductive BioMedicine Online; www.rbmonline.com/article/1178 on web 20 January 2004 Article Cumulative live birth rates after transfer of cryopreserved ICSI embryos

More information

Problems in the cryopreservation of unfertilized eggs by slow cooling in dimethyl sulfoxide*

Problems in the cryopreservation of unfertilized eggs by slow cooling in dimethyl sulfoxide* FERTILITY AND STERILITY Vol. 52, No.5, November 1989 Copyright" 1989 The American Fertility Society Printed on acid-free paper in U.S.A. Problems in the cryopreservation of unfertilized eggs by slow cooling

More information

Effect of Warming on the Survivability and Fertilizability of Vitrified Matured Bovine Oocytes

Effect of Warming on the Survivability and Fertilizability of Vitrified Matured Bovine Oocytes International Journal of Agricultural Technology 2014 Vol. 10(1):49-58 Available online http://www.ijat-aatsea.com ISSN 2630-0192 (Online) Fungal Diversity Effect of Warming on the Survivability and Fertilizability

More information

Rapid- Vitrification System. Closed system for simple and successful vitrification.

Rapid- Vitrification System. Closed system for simple and successful vitrification. Rapid- Vitrification System Closed system for simple and successful vitrification. 3 working together for you Media Method Device & accessories Rapid-i Vitrification System puts you in control. The method,

More information

Tammie Roy Genea Biomedx Sydney, Australia. Declared to be stakeholder in Genea Biomedx

Tammie Roy Genea Biomedx Sydney, Australia. Declared to be stakeholder in Genea Biomedx Tammie Roy Genea Biomedx Sydney, Australia Declared to be stakeholder in Genea Biomedx 1 24-25 September 2015 Madrid and Alicante, Spain Importance of cryopreservation in Assisted Reproductive Technology

More information

Planning, design and coordination of research programs - All laboratory activities relevant to human IVF

Planning, design and coordination of research programs - All laboratory activities relevant to human IVF E U R O P E A N C U R R I C U L U M V I T A E F O R M A T PERSONAL INFORMATION Name Address Telephone Fax E-mail GIOVANNI COTICCHIO Nationality Italian Date of birth 12 April 1962 WORK EXPERIENCE Dates

More information

Ultrarapid freezing of early cleavage stage human embryos and eight-cell mouse embryos*

Ultrarapid freezing of early cleavage stage human embryos and eight-cell mouse embryos* FERTILITY AND STERILITY Copyright 1988 The American Fertility Society Printed in U.S.A. Ultrarapid freezing of early cleavage stage human embryos and eight-cell mouse embryos* Alan Trounson, Ph.D.t:!:

More information

Reducing multiple pregnancies is a concern

Reducing multiple pregnancies is a concern S 3 Vitrifcation System: A Novel Approach To Blastocyst Freezing James J. Stachecki, Ph.D. Jacques Cohen, Ph.D. Tyho-Galileo Research Laboratories, 3 Regent Street, Suite 301, Livingston, NJ 07039 Email:

More information

In vitro Culture, Storage and Transfer of Goat Embryos

In vitro Culture, Storage and Transfer of Goat Embryos Aust. J. Bio!. Sci., 1976,29, 125-9 In vitro Culture, Storage and Transfer of Goat Embryos R. J. Bilton and N. W. Moore Department of Animal Husbandry, University of Sydney, Camden, N.S.W. 2570. Abstract

More information

Page 1 of 5 Egg Freezing Informed Consent Form version 2018 Main Line Fertility Center. Egg Freezing. Informed Consent Form

Page 1 of 5 Egg Freezing Informed Consent Form version 2018 Main Line Fertility Center. Egg Freezing. Informed Consent Form Page 1 of 5 Egg Freezing Informed Consent Form version 2018 Egg Freezing Informed Consent Form Embryos and sperm have been frozen and thawed with good results for many years. Egg (oocyte) freezing is a

More information

Egg banking in the United States: current status of commercially available cryopreserved oocytes

Egg banking in the United States: current status of commercially available cryopreserved oocytes Egg banking in the United States: current status of commercially available cryopreserved oocytes Alexander M. Quaas, M.D., Ph.D., Alexander Melamed, M.D., M.P.H., Karine Chung, M.D., Kristin A. Bendikson,

More information

The first human birth from a frozen oocyte

The first human birth from a frozen oocyte FERTILITY Egg freezing is no longer deemed experimental. Here are current protocols, fertility expectations, and safety outcomes as well as ethical considerations for oocyte cryopreservation. Mary E. Abusief,

More information

This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and

This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and education use, including for instruction at the authors institution

More information

Vitrification: "Robots" versus Human Comparing automated vitrification outcomes

Vitrification: Robots versus Human Comparing automated vitrification outcomes Vitrification: "Robots" versus Human Comparing automated vitrification outcomes The 5th World congress of the INTERNATIONAL SOCIETY FOR FERTILITY PRESERVATION Vienna, Austria November 16-18, 2017 Zsolt

More information

Vitrification of reproductive cells: The next breakthrough in ART? Department of Obstetrics and Gynaecology University of Aberdeen

Vitrification of reproductive cells: The next breakthrough in ART? Department of Obstetrics and Gynaecology University of Aberdeen Vitrification of reproductive cells: The next breakthrough in ART? Maureen J Wood PhD Department of Obstetrics and Gynaecology University of Aberdeen breakthrough Some signal achievement in scientific

More information

E.S.C. Siqueira-Pyles 1, F.C. Landim-Alvarenga 1,2

E.S.C. Siqueira-Pyles 1, F.C. Landim-Alvarenga 1,2 Anim. Reprod., v.2. n.1, p.63-69, Jan./March 2005 Effect of meiotic stage on viability and developmental competence of bovine oocytes vitrified in TCM-199 and bovine follicular fluid E.S.C. Siqueira-Pyles

More information

Impact of phase transition on the mouse oocyte spindle during vitrification

Impact of phase transition on the mouse oocyte spindle during vitrification Reproductive BioMedicine Online (2011) 22, 184 191 www.sciencedirect.com www.rbmonline.com ARTICLE Impact of phase transition on the mouse oocyte spindle during vitrification Ching-Chien Chang a, Chih-Jen

More information

AAB/CRB 2017 Houston, Texas

AAB/CRB 2017 Houston, Texas AAB/CRB 2017 Houston, Texas Advanced Current & Future Cryogenic Technologies for ART James J. Stachecki Ph.D. Innovative Cryo Enterprises LLC Disclosures Founder of Innovative Cryo Enterprises LLC We focus

More information

Fertilization depends on mechanisms that help sperm meet eggs of the same species.

Fertilization depends on mechanisms that help sperm meet eggs of the same species. Fertilization depends on mechanisms that help sperm meet eggs of the same species. www.uchsc.edu/ltc/fertilization.html Fertilization union of sperm and egg Is a chain of events. Interruption of any step

More information

Effect of sucrose and propylene glycol on the vitrification of sheep oocytes

Effect of sucrose and propylene glycol on the vitrification of sheep oocytes Journal of Cell and Animal Biology Vol. 7 (3), pp. 25-30, March 2013 Available online at http://www.academicjournals.org/jcab DOI: 10.5897/JCAB12.033 ISSN 1996-0867 2013 Academic Journals Full Length Research

More information

FRESH OR FROZEN EMBYOS WHAT IS THE LATEST EVIDENCE? DR. ASMA MOMANI CLEVELAND CLINIC, ANDROLOGY LAB TRAINEE 2018

FRESH OR FROZEN EMBYOS WHAT IS THE LATEST EVIDENCE? DR. ASMA MOMANI CLEVELAND CLINIC, ANDROLOGY LAB TRAINEE 2018 FRESH OR FROZEN EMBYOS WHAT IS THE LATEST EVIDENCE? DR. ASMA MOMANI CLEVELAND CLINIC, ANDROLOGY LAB TRAINEE 2018 OBJECTIVES Hisory Indication of freezing embryos Slow freezing versus vitrification Advantages

More information

Cleavage Stage Embryo Cryopreservation Slow Freezing Versus Vitrification

Cleavage Stage Embryo Cryopreservation Slow Freezing Versus Vitrification Cleavage Stage Embryo Cryopreservation Slow Freezing Versus Vitrification Basak Balaban VKF American Hospital of Istanbul Assisted Reproduction Unit Head of IVF Laboratory Turkish Society of Clinical Embryologists

More information

Review Current trends, biological foundations and future prospects of oocyte and embryo cryopreservation

Review Current trends, biological foundations and future prospects of oocyte and embryo cryopreservation RBMOnline - Vol 19. No 1. 2009 126-140 Reproductive BioMedicine Online; www.rbmonline.com/article/3857 on web 8 May 2009 Review Current trends, biological foundations and future prospects of oocyte and

More information

Outcome of 518 salvage oocyte-cryopreservation cycles performed as a routine procedure in an in vitro fertilization program

Outcome of 518 salvage oocyte-cryopreservation cycles performed as a routine procedure in an in vitro fertilization program OOCYTE CRYOPRESERVATION Outcome of 518 salvage oocyte-cryopreservation cycles performed as a routine procedure in an in vitro fertilization program Giovanni B. La Sala, M.D., a Alessia Nicoli, B.Sc., a

More information

University Hospital of Ghent, Ghent, Belgium

University Hospital of Ghent, Ghent, Belgium FERTILITY AND STERILITY VOL. 74, NO. 3, SEPTEMBER 2000 Copyright 2000 American Society for Reproductive Medicine Published by Elsevier Science Inc. Printed on acid-free paper in U.S.A. Cryopreservation

More information

Optimization of Cryoprotectant Loading into Murine and Human Oocytes

Optimization of Cryoprotectant Loading into Murine and Human Oocytes Optimization of Cryoprotectant Loading into Murine and Human Oocytes Karlsson, J. O. M., Szurek, E. A., Higgins, A. Z., Lee, S. R., & Eroglu, A. (2014). Optimization of cryoprotectant loading into murine

More information

Female Patient Name: Social Security # Male Patient Name: Social Security #

Female Patient Name: Social Security # Male Patient Name: Social Security # Female Patient Name: Social Security # Male Patient Name: Social Security # THE CENTER FOR HUMAN REPRODUCTION (CHR) ILLINOIS/NEW YORK CITY * ASSISTED REPRODUCTIVE TECHNOLOGIES PROGRAM (A.R.T.) CRYOPRESERVATION

More information

Oocyte morphology does not affect post-warming survival rate in an egg-cryobanking donation program

Oocyte morphology does not affect post-warming survival rate in an egg-cryobanking donation program J Assist Reprod Genet (2011) 28:1177 1181 DOI 10.1007/s10815-011-9677-7 GAMETE BIOLOGY Oocyte morphology does not affect post-warming survival rate in an egg-cryobanking donation program Amanda Souza Setti

More information

IVF: PAST, PRESENT AND FUTURE

IVF: PAST, PRESENT AND FUTURE IVF: PAST, PRESENT AND FUTURE Mark Larman Chief Scientific Officer 1 HISTORY OF IVF IVF first achieved with rabbits in 1959 IVF with human gametes - pioneered by Robert Edwards and Patrick Steptoe during

More information

Colorado Center for Reproductive Medicine, Englewood, Colorado

Colorado Center for Reproductive Medicine, Englewood, Colorado FERTILITY AND STERILITY VOL. 72, NO. 6, DECEMBER 1999 Copyright 1999 American Society for Reproductive Medicine Published by Elsevier Science Inc. Printed on acid-free paper in U.S.A. REPRODUCTIVE BIOLOGY

More information

Preimplantation Genetic Testing (PGT) Fresh and Frozen Embryos Process, Risk, and Consent

Preimplantation Genetic Testing (PGT) Fresh and Frozen Embryos Process, Risk, and Consent Preimplantation Genetic Testing (PGT) Fresh and Frozen Embryos Process, Risk, and Consent PGT analysis is offered to patients that seek to identify a chromosomal abnormality in their embryos prior to initiating

More information

CONSENT TO CRYOPRESERVATION AND STORAGE OF HUMAN EMBRYOS

CONSENT TO CRYOPRESERVATION AND STORAGE OF HUMAN EMBRYOS 1. Name(s) of Party/Parties A. Party/parties requesting freezing of embryos a. Couple We, and of County, City of in the state of are (married or domestic partners) and are over the age of twenty-one years.

More information

Original Article. Abstract

Original Article. Abstract Original Article Cryopreservation of embryos by vitrification at a private sector reproductive medicine facility in Karachi Majida Khan, Shaheen Zafar, Serajuddaula Syed Sindh Institute of Reproductive

More information

Overview. Solution Comparison. 1 of 7 7/6/ :10 PM. October 2005 (see also "M22 Implementation" from Alcor News, Oct.

Overview. Solution Comparison. 1 of 7 7/6/ :10 PM. October 2005 (see also M22 Implementation from Alcor News, Oct. EXHIBIT CC 1 of 7 7/6/2013 11:10 PM October 2005 (see also "M22 Implementation" from Alcor News, Oct. 13, 2005) Overview Reversible suspended animation requires successful preservation and recovery of

More information

Article Comparison of open and closed methods for vitrification of human embryos and the elimination of potential contamination

Article Comparison of open and closed methods for vitrification of human embryos and the elimination of potential contamination RBMOnline - Vol 11. No 5. 2005 608 614 Reproductive BioMedicine Online; www.rbmonline.com/article/1925 on web 26 September 2005 Article Comparison of open and closed methods for vitrification of human

More information

Abstract. Introduction. RBMOnline - Vol 17. No Reproductive BioMedicine Online; on web 3 July 2008

Abstract. Introduction. RBMOnline - Vol 17. No Reproductive BioMedicine Online;   on web 3 July 2008 RBMOnline - Vol 17. No 3. 2008 378-384 Reproductive BioMedicine Online; www.rbmonline.com/article/3216 on web 3 July 2008 Article Vitrification with DMSO protects embryo membrane integrity better than

More information

Alterations of the cytoskeleton and polyploidy induced by cryopreservation of metaphase II mouse oocytes

Alterations of the cytoskeleton and polyploidy induced by cryopreservation of metaphase II mouse oocytes FERTILITY AND STERILITY VOL. 69, NO. 5, MAY 1998 Copyright 1998 American Society for Reproductive Medicine Published by Elsevier Science Inc. Printed on acid-free paper in U.S.A. Alterations of the cytoskeleton

More information

Steven F. Mullen Ph.D. Scientific Director The World Egg Bank

Steven F. Mullen Ph.D. Scientific Director The World Egg Bank Steven F. Mullen Ph.D. Scientific Director The World Egg Bank Steven F. Mullen Ph.D. Scientific Director The World Egg Bank Disclosure Scientific Director for The World Egg Bank A For-Profit Company providing

More information

Maturation and Freezing of Bovine Oocytes

Maturation and Freezing of Bovine Oocytes Maturation and Freezing of Bovine Oocytes D. Mapes and M. E. Wells Story in Brief Immature bovine oocytes were aspirated from small to medium size follicles of bovine ovaries by needle and syringe. The

More information

The storage of cow eggs at room temperature and at low temperatures

The storage of cow eggs at room temperature and at low temperatures The storage of cow eggs at room temperature and at low temperatures A. O. Trounson, S. M. Willadsen, L. E. A. Rowson and R. Newcomb A.R.C. Unit of Reproductive Physiology and Biochemistry, Cambridge, U.K.*

More information

A comparison of the effects of estrus cow. nuclear maturation of bovine oocytes

A comparison of the effects of estrus cow. nuclear maturation of bovine oocytes A comparison of the effects of estrus cow serum and fetal calf serum on in vitro nuclear maturation of bovine oocytes J Spiropoulos, SE Long University of Bristol, School of Veterinary Science, Department

More information

Improved human oocyte development after vitrification: a comparison of thawing methods

Improved human oocyte development after vitrification: a comparison of thawing methods FERTILITY AND STERILITY VOL. 72, NO. 1, JULY 1999 Copyright 1999 American Society for Reproductive Medicine Published by Elsevier Science Inc. Printed on acid-free paper in U.S.A. Improved human oocyte

More information

Cryopreservation of supernumerary oocytes in IVF/ICSI cycles

Cryopreservation of supernumerary oocytes in IVF/ICSI cycles Human Reproduction Vol.21, No.2 pp. 370 375, 2006 Advance Access publication October 20, 2005. doi:10.1093/humrep/dei347 Cryopreservation of supernumerary oocytes in IVF/ICSI cycles P.E.Levi Setti 1,2,

More information

Department of Obstetrics and Gynecology, NYU Fertility Center, NYU School of Medicine, New York 10016, USA; 2

Department of Obstetrics and Gynecology, NYU Fertility Center, NYU School of Medicine, New York 10016, USA; 2 RBMOnline - Vol 18 No 6. 2009 769-776 Reproductive BioMedicine Online; www.rbmonline.com/article/3971 on web 8 April 2009 Article Over 900 oocyte cryopreservation babies born with no apparent increase

More information

Leicester City, East Leicestershire and Rutland & West Leicestershire Collaborative Commissioning Policy Gamete/Embryo cryopreservation

Leicester City, East Leicestershire and Rutland & West Leicestershire Collaborative Commissioning Policy Gamete/Embryo cryopreservation Leicester City Clinical Commissioning Group West Leicestershire Clinical Commissioning Group East Leicestershire and Rutland Clinical Commissioning Group Leicester City, East Leicestershire and Rutland

More information

Article Polar body morphology and spindle imaging as predictors of oocyte quality

Article Polar body morphology and spindle imaging as predictors of oocyte quality RBMOnline - Vol 11. No 1. 2005 36 42 Reproductive BioMedicine Online; www.rbmonline.com/article/1744 on web 25 April 2005 Article Polar body morphology and spindle imaging as predictors of oocyte quality

More information

The Viability Of Human Embryos After Transport In A Dry Shipper Between Assisted Conception Laboratories

The Viability Of Human Embryos After Transport In A Dry Shipper Between Assisted Conception Laboratories Article ID: WMC001104 2046-1690 The Viability Of Human Embryos After Transport In A Dry Shipper Between Assisted Conception Laboratories Corresponding Author: Ms. Clare Pinkus, Scientist, Hollywood Fertility

More information

Male Fertility: Your Questions Answered

Male Fertility: Your Questions Answered Male Fertility: Your Questions Answered Michael S. Neal Scientific Director, ONE Fertility, 3210 Harvester Rd. Burlington, Ontario www.onefertility.com mneal@onefertility.com Outline Assisted Conception

More information

Keywords elective oocyte cryopreservation, germinal vesicle, in-vitro maturation, metaphase I and survival

Keywords elective oocyte cryopreservation, germinal vesicle, in-vitro maturation, metaphase I and survival REVIEW C URRENT OPINION In-vitro maturation of germinal vesicle and metaphase I eggs prior to cryopreservation optimizes reproductive potential in patients undergoing fertility preservation Joseph A. Lee

More information

CIC Edizioni Internazionali

CIC Edizioni Internazionali Original article Oocyte vitrification/storage/handling/transportation/warming, effect on survival and clinical results in donation programmes Lodovico Parmegiani Antonio Manuel Maccarini Azzurra Rastellini

More information

Report of four donor-recipient oocyte cryopreservation cycles resulting in high pregnancy and implantation rates

Report of four donor-recipient oocyte cryopreservation cycles resulting in high pregnancy and implantation rates Report of four donor-recipient oocyte cryopreservation cycles resulting in high pregnancy and implantation rates Jason Barritt, Ph.D., Martha Luna, M.D., Marlena Duke, M.Sc., Lawrence Grunfeld, M.D., Tanmoy

More information

Basic principles of cryopreservation

Basic principles of cryopreservation Basic principles of cryopreservation Henri Woelders Centre for Genetic Resources, The Netherlands (CGN) Animal Sciences Group of Wageningen UR Lelystad, The Netherlands Centre for Genetic Resources, the

More information

Adoption and Foster Care

Adoption and Foster Care GLOSSARY Family building via Adoption and Foster Care October 2018 www.familyequality.org/resources A Anonymous Donor: A person who donated sperm or eggs with the intention of never meeting resulting children.

More information

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists 3,500 108,000 1.7 M Open access books available International authors and editors Downloads Our

More information

LOW RESPONDERS. Poor Ovarian Response, Por

LOW RESPONDERS. Poor Ovarian Response, Por LOW RESPONDERS Poor Ovarian Response, Por Patients with a low number of retrieved oocytes despite adequate ovarian stimulation during fertility treatment. Diagnosis Female About Low responders In patients

More information

THE EFFECT OF VITRIFICATION OF OOCYTES FOR MICE AND EXAMINING SURVIVED OOCYTE PERCENTAGE

THE EFFECT OF VITRIFICATION OF OOCYTES FOR MICE AND EXAMINING SURVIVED OOCYTE PERCENTAGE : 376-390 ISSN: 2277 4998 THE EFFECT OF VITRIFICATION OF OOCYTES FOR MICE AND EXAMINING SURVIVED OOCYTE PERCENTAGE FATEMEH FAZELI SHORAKI Fazali101.f@gmail.com MOHAMMAD MEHDI AKHONDI Akhondi@avicenna.ac.ir

More information

Retrospective analysis of outcomes following transfer of previously cryopreserved oocytes, pronuclear zygotes and supernumerary blastocysts

Retrospective analysis of outcomes following transfer of previously cryopreserved oocytes, pronuclear zygotes and supernumerary blastocysts Reproductive BioMedicine Online (2011) 23, 118 123 www.sciencedirect.com www.rbmonline.com ARTICLE Retrospective analysis of outcomes following transfer of previously cryopreserved oocytes, pronuclear

More information

Article Comparison of embryo quality between sibling embryos originating from frozen or fresh oocytes

Article Comparison of embryo quality between sibling embryos originating from frozen or fresh oocytes RBMOnline - Vol 13 No 4. 2006 497-503 Reproductive BioMedicine Online; www.rbmonline.com/article/2284 on web 15 August 2006 Article Comparison of embryo quality between sibling embryos originating from

More information

Vitrification of oocytes

Vitrification of oocytes DOI: 10.1111/j.1744-4667.2011.00078.x The Obstetrician & Gynaecologist http://onlinetog.org 2012;14:45 49 SAC review Vitrification of oocytes This article was commissioned by the Scientific Advisory Committee

More information

Provisional Patent Application

Provisional Patent Application PPA: Method of Cryopreservation of Whole Brains Proprietary Information: Property of Cryonics Institute Provisional Patent Application Title: Method of Cryopreservation of Whole Brains Inventor: Yuriy

More information

Theoretical and experimental basis of oocyte vitrification

Theoretical and experimental basis of oocyte vitrification Reproductive BioMedicine Online (2011) 23, 298 306 www.sciencedirect.com www.rbmonline.com SYMPOSIUM: OOCYTE CRYOPRESERVATION REVIEW Theoretical and experimental basis of oocyte vitrification Gary D Smith

More information

Center, University of Bologna, Bologna, Italy. Keywords: human oocyte cryopreservation; slow freezing; high sucrose concentration; embryo quality

Center, University of Bologna, Bologna, Italy. Keywords: human oocyte cryopreservation; slow freezing; high sucrose concentration; embryo quality Human Reproduction Vol.23, No.8 pp. 1771 1777, 2008 Advance Access publication on May 12, 2008 doi:10.1093/humrep/den119 Freezing within 2 h from oocyte retrieval increases the efficiency of human oocyte

More information

Article Highly efficient vitrification method for cryopreservation of human oocytes

Article Highly efficient vitrification method for cryopreservation of human oocytes RBMOnline - Vol 11. No 3. 2005 300-308 Reproductive BioMedicine Online; www.rbmonline.com/article/1827 on web 15 July 2005 Article Highly efficient vitrification method for cryopreservation of human oocytes

More information

Failed fertilization after clinical intracytoplasmic sperm injection

Failed fertilization after clinical intracytoplasmic sperm injection Failed fertilization after clinical intracytoplasmic sperm injection Reproductive BioMedicine Online 2009 Vol. 19 No.2. 216 220 Present by R4 郭恬妮 Introduction intracytoplasmic sperm injection (ICSI) choice

More information