US20050208651A1 - Vitro maturation of immature human oocytes - Google Patents

Vitro maturation of immature human oocytes Download PDF

Info

Publication number
US20050208651A1
US20050208651A1 US10/506,713 US50671305A US2005208651A1 US 20050208651 A1 US20050208651 A1 US 20050208651A1 US 50671305 A US50671305 A US 50671305A US 2005208651 A1 US2005208651 A1 US 2005208651A1
Authority
US
United States
Prior art keywords
culture medium
oocyte
growth factor
oocytes
human
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/506,713
Inventor
Ri-Cheng Chian
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
McGill University
Original Assignee
McGill University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by McGill University filed Critical McGill University
Priority to US10/506,713 priority Critical patent/US20050208651A1/en
Assigned to MCGILL UNIVERSITY reassignment MCGILL UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHIAN, RI-CHANG
Publication of US20050208651A1 publication Critical patent/US20050208651A1/en
Priority to US11/969,522 priority patent/US7790459B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0608Germ cells
    • C12N5/0609Oocytes, oogonia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/31Pituitary sex hormones, e.g. follicle-stimulating hormone [FSH], luteinising hormone [LH]; Chorionic gonadotropins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2517/00Cells related to new breeds of animals
    • C12N2517/10Conditioning of cells for in vitro fecondation or nuclear transfer

Definitions

  • the present invention relates to the field of assisted reproductive technology (ART), particularly to methods for culturing immature human oocytes so that they mature to metaphase-II stage capable of being fertilized and forming a viable embryo.
  • ART assisted reproductive technology
  • HCG human chorionic gonadotropin
  • the harvested oocyte is then fertilized with the sperm.
  • fertilization is effected by intracytoplasmic sperm injection (ICSI).
  • ICSI intracytoplasmic sperm injection
  • M-II metaphase-II
  • a M-II oocyte is ready to accept the sperm and be fertilized.
  • Metaphase-II is characterized by exclusion of one polar body from the cytoplasm, and is typically detected by visual identification under a microscope.
  • the harvested oocyte remains surrounded by cumulus cells that support and promote the final maturation and development of the oocyte.
  • the cumulus cells interfere with ICSI and obscure microscopic observation of the oocyte. Therefore, prior to ICSI, it is necessary to denude the oocyte of cumulus cells to permit determination of the level of oocyte maturity. This is generally accomplished by enzymatic and mechanical stripping of the cumulus cells.
  • PCOS polycystic ovarian syndrome
  • IVM media and methods have involved the use of human follicular fluid, in an effort to mimic the conditions within the follicle. In view of the risk of transmission of disease, allergic reaction, etc., use of biological fluids in IVM techniques is prohibited in most countries. Instead, the use of a “chemically-defined medium”, wherein the chemical composition of all of the ingredients is known, is preferred.
  • TCM-199 tissue culture medium 199
  • TCM-199 is a complex medium that contains many components, and was designed for in vitro culture of somatic cells, not germ cells. Notably, TCM-199 does not contain any growth factors.
  • TCM-199 was first used for sheep oocyte IVM (Moor & Trounson, 1977), and has since been used routinely for bovine oocyte IVM (Brackett & Zuelke, 1993).
  • the present invention provides in vitro maturation methods that are useful for culturing immature human oocytes to maturity.
  • the invention provides a method for in vitro maturation of immature human oocytes, comprising the steps of:
  • step (a) the subject has not been treated with a gonadotrophin releasing hormone agonist, human menopausal gonadotrophin, HCG, or follicle stimulating hormone (FSH).
  • a gonadotrophin releasing hormone agonist human menopausal gonadotrophin, HCG, or follicle stimulating hormone (FSH).
  • FSH follicle stimulating hormone
  • step (a) comprises administering to the subject HCG or luteinizing hormone (LH), or both.
  • HCG is administered in an amount of about 5000 to about 20,000 IU, more preferably about 10,000 IU.
  • the immature human oocyte is preferably an M-I stage or GV stage ooctye.
  • the oocyte preferably is cultured until it reaches M-II.
  • the immature human oocyte is essentially free of cumulus cells and is cultured in a culture medium comprising:
  • At least one growth factor is at least one growth factor.
  • the growth factor is fibroblast growth factor or epidermal growth factor, or both.
  • the culture medium further comprises at least one hormone, e.g. insulin.
  • the culture medium comprises from 0.5 mg/L to 50 mg/L insulin.
  • the culture medium further comprises human transferrin, preferably from 5 mg/L to 500 mg/L human transferrin.
  • the culture medium comprises from 0.0001 mg/L to 0.001 mg/L fibroblast growth factor.
  • the culture medium comprises from 0.0001 to 0.01 mg/L epidermal growth factor.
  • the culture medium comprises one or more vitamins, preferably biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal-HCl, riboflavin, and thiamine-HCl.
  • vitamins preferably biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal-HCl, riboflavin, and thiamine-HCl.
  • the culture medium may further comprise hydrocortisone or selenite, or both.
  • Preferred inorganic salts include CaCl 2 , KCl, MgSO 4 , NaCl, NaHCO 3 and NaH 2 PO4-H 2 O.
  • Preferred energy sources include D-glucose and sodium pyruvate, or both D-glucose and sodium pyruvate.
  • the amino acids comprise alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • the culture medium comprises the inorganic salts, amino acids, vitamins and other components as set forth in Table 1, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ⁇ 50% (weight/volume), more preferably ⁇ 10% (weight/volume), of the amount specified in Table 1.
  • the oocyte has a cumulus that is intact or at least partially intact, and/or the oocyte is cultured in the presence of cumulus cells.
  • the oocyte may be cultured in a culture medium that is essentially free of one or more of epidermal growth factor, fibroblast growth factor, human transferrin, insulin, selenite, and hydrocortisone.
  • the oocyte is cultured in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ⁇ 50% (weight/volume), more preferably ⁇ 10% (weight/volume) of the amount specified in Table 2.
  • the culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components identified in Table 2.
  • the culture medium consists of the inorganic salts, amino acids, vitamins and other components identified in Table 2.
  • the invention provides a method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising:
  • At least one growth factor is at least one growth factor.
  • This embodiment is particularly useful when the immature human oocyte is essentially free of cumulus cells.
  • the invention provides a method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
  • This embodiment is particularly useful when the immature human oocyte is cultured in the presence of cumulus cells.
  • the culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
  • the culture medium consists of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
  • each inorganic salt, amino acid, vitamin and other component is present in an amount of ⁇ 50%, more preferably ⁇ 10% (weight/volume), of the amount specified in Table 2.
  • certain of the components listed in the “other components” sections of Table 1 and 2 may be absent from the medium.
  • Components that may be absent include, without limitation, phenol red, penicillin G, streptomycin, and human serum albumin. This may apply wherever reference is made herein to Table 1 or 2.
  • FIG. 1 shows fertilization rates of M-I stage oocytes derived from stimulation and ICSI cycles following 6 h, 24 h or 48 h of maturation, respectively.
  • FIG. 2 shows cleavage rates of M-I stage oocytes derived from stimulation and ICSI cycles following 6 h, 24 h or 48 h of maturation, respectively.
  • FIGS. 3A and 3B show embryos developing to the 8-cell stage (A) and the blastocyst stage (B) following 6 h, 24 h or 48 h of maturation of M-I stage oocytes derived from stimulation and ICSI cycles.
  • FIG. 4 shows fertilization rates of GV stage oocytes derived from stimulation and ICSI cycles following 24 h or 48 h of maturation.
  • FIG. 5 shows cleavage rates of GV stage oocytes derived from stimulation and ICSI cycles following 24 h or 48 h of maturation.
  • FIGS. 6A and 6B show embryos developing to the 8-cell stage (A) and the blastocyst stage (B) following 24 h or 48 h of maturation of GV stage oocytes derived from stimulation and ICSI cycles.
  • FIG. 7 shows a blastocyst derived from GV stage oocytes following 24 h of maturation in vitro and illustrates that there is no difference in morphology from oocytes matured in vivo.
  • the scale bar indicates 5 ⁇ m length.
  • any female human subject who possesses viable oocytes is a candidate for IVM therapy.
  • the subject will suffer from some form of infertility.
  • the subject may experience normal oocyte production but have an impediment to fertilization, as in, e.g. PCOS or PCOS-like ovaries.
  • IVM is also useful in women infertility with endometriosis, blockage of either or both fallopian tubes, etc.
  • numerous follicles mature simultaneously in the ovaries, without the appearance of a dominant follicle.
  • the subject's menstrual cycle may be regular, irregular or non-existent.
  • IVM may be especially useful in women who are susceptible to or suffer from OHSS and are therefore not suitable candidates for traditional in vitro fertilization techniques involving an ovarian stimulation protocol.
  • the subject may be an individual who does not suffer from infertility or have any reproductive impediment whatsoever, but for whom an in vitro fertilization method nevertheless remains desirable, as in, e.g. cases of male factor infertility.
  • IVM in accordance with the invention does not necessarily involve an ovarian stimulation protocol.
  • One of two “ovarian stimulation protocols” is usually used in conventional IVF, a “long protocol” or a “short protocol.”
  • the long protocol has two steps. The first step involves pre-treating the subject for two or three weeks with GnRHa to down-regulate pituitary activity. Once pituitary suppression has been achieved, in the second step, HMG or FSH is administered to induce the development of multiple follicles.
  • the short protocol involves only the ovarian stimulation step, but not the down-regulation step.
  • Ovarian stimulation is required in conventional IVF techniques to permit the oocytes to mature to the M-II stage prior to harvest.
  • ovarian stimulation need not be used. That is, the subject need not be pre-treated with GnRHa's, HMG and/or FSH.
  • LH Luteinizing Hormone
  • an increase in endogenous levels of LH is induced in the subject (also described herein as “priming”). This may be accomplished by e.g. administering to the subject an effective amount of human chorionic gonadotropin (HCG) (Profasi Serono, Oakville, Ontario, Canada) or LH, either of which stimulate endogenous LH production.
  • HCG human chorionic gonadotropin
  • Baseline concentration of LH in the plasma of premenopausal women is usually about 2-4 mIU/ml, and at midcycle peak may be up to 25-35 mIU/ml.
  • the expression “inducing in a female human subject an increase in endogenous luteinizing hormone levels” means increasing the plasma LH concentration above the baseline concentration.
  • plasma LH concentration is increased at least 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or 2000% above baseline concentration.
  • HCG or LH may vary depending on the patient and the circumstances and may be determined by those of skill in the art.
  • Subcutaneous injection is a preferred mode of administration although other modes of administration may be used.
  • a single injection of about 5000 to about 20,000 IU of HCG is generally sufficient, with a single dose of about 10,000 IU of HCG being preferred.
  • commencement of menstrual bleeding In the case of a subject having a regular menstrual cycle, the time for inducement may be determined relative to the commencement of menstrual bleeding, which is considered “day 0”.
  • commencement of menstrual bleeding i.e. the initiation of “day 0”
  • progesterone available from e.g. Prometrium; Schering, Pointe-Claire, Quebec, Canada
  • intravaginally in an amount of approximately 300 mg/day for 10 days.
  • the ovaries are examined by ultrasound to assess the size and the number of follicles.
  • the dominant follicle reaches a diameter of at least about 16 mm by day 10-14.
  • a dominant follicle is present (e.g. as in normal ovarian function, as in the case of a healthy subject or a subject suffering from e.g. endometriosis or tubal blockage)
  • the increase in endogenous LH activity is induced (e.g. by administration of HCG) when the dominant follicle reaches a diameter of about 15-17 mm, preferably at least about 16 mm.
  • inducement of an increase in endogenous LH levels preferably occurs when the follicles are less than about 12 mm in diameter.
  • Oocytes are generally retrieved from about 32 to about 40 hours after priming, more preferably about 34 to 38 hours after priming and even more preferably about 36 hours after priming.
  • Means for retrieving oocytes are known in the art.
  • transvaginal ultrasonographically-guided oocyte collection is done using a 17-20 gauge, preferably 19 gauge single-lumen aspiration needle at an aspiration pressure of from about 5 to about 10 kPa, preferably about 7.5 kPa.
  • Immature human oocyte means a human oocyte that has not yet reached metaphase-II (M-II). As discussed previously, metaphase-II is characterized by exclusion of one polar body from the cytoplasm. Immature human oocytes used in the invention are typically at the germinal vesicle (GV) or metaphase-I (M-I) stage.
  • GV germinal vesicle
  • M-I metaphase-I
  • Oocytes may be cultured with their cumulus intact, in a form known as a “cumulus-oocyte-complex” (COC), or oocytes may be partially or entirely denuded from cumulus cells.
  • COC can be stripped with 85 IU/ml hyaluronidase in HEPES buffered medium and mechanically pipetted until oocytes are denuded.
  • An oocyte that is “essentially free of cumulus cells” is an oocyte that is associated with sufficiently few cumulus cells that the cumulus cells have no detectable physiological effect on the oocyte.
  • Culture conditions for human oocytes are known in the art and are not critical to the invention. Suitable culture conditions include e.g. culturing the oocytes at 37° C. in an atmosphere of 95% air and 5% CO 2 at high humidity, e.g. 100% humidity. A “triple gas” atmosphere of 5% O 2 , 5% CO 2 and 90% N 2 may also be used. Mineral oil may be overlaid on the medium to control evaporation and/or temperature. Oocytes are typically cultured in a well containing 1 ml of culture medium or more, or may be cultured in 10 ⁇ l of culture medium or less in a droplet in a culture dish.
  • Immature oocytes may be cultured for e.g. about 24 to about 48 hours. Because about 60% of oocytes typically reach maturity (M-II) after 24 hours of culture, when oocytes are cultured with an intact or partially intact cumulus, they may be denuded at about 24 hours and observed under a microscope to assess oocyte maturity. The physiological influence of the cumulus cells on the oocyte persists for only about the first 12 hours in culture, so denuding the oocyte after about 24 hours in culture has little or no negative effect.
  • oocytes Due to the influence of cumulus cells on the maturation of oocytes, somewhat different culture media may be used when the oocytes are cultured for the first 12-24 hours in the presence of cumulus cells. This can occur e.g. when the oocytes are cultured as COCs or partially denuded or when denuded oocytes are co-cultured in the presence of cumulus cells.
  • the inorganic salts, essential and non-essential amino acids, and energy source in the IVM medium are not critical to the invention. Suitable salts, amino acids and energy sources are known in the art.
  • Inorganic salts are provided to buffer the pH of the medium within a range preferably of about 7.2-7.4 and to maintain correct osmolarity of the medium with the oocytes.
  • Suitable inorganic salts and concentrations thereof as used in culture media are known in the art.
  • Typical inorganic salts include CaCl 2 , KCl, MgSO 4 , NaCl, NaHCO 3 , NaH 2 PO4.H 2 O, FE(NO 3 ) 3 .9H 2 O, KH 2 PO 4 , Na.acetate, Na 2 H 2 PO 4 , etc.
  • the IVM medium contains at least one amino acid or source thereof.
  • at least the essential amino acids, or sources thereof are included in IVM medium.
  • “Essential” amino acids are those amino acids not synthesized in the oocyte and that are essential for protein synthesis.
  • the essential amino acids are generally considered to be isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine.
  • a commercial amino acid mixture, containing the eight essential amino acids as well as some or all of the remaining non-essential amino acids may conveniently be used.
  • Non-naturally occuring amino acids or amino acid derivatives as are known in the art may also be included in the IVM medium.
  • the IVM medium of the invention comprises alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • the IVM medium preferably contains vitamins as are known in the art for inclusion in culture media.
  • Vitamins that may be included in the media include, without limitation, vitamins A1 (retinol), A2 (an alternative form of retinol), B1 (thiamine), B2 (riboflavin), B6 (pyridoxine), B9 (folic acid), B12 (cyanocobalamin), B17, C (ascorbic acid), D, D2 (calciferol), D3 (cholecalciferol), E (tocopherol), H (biotin), K, K1 (phylloquinone), K2, K3 (menadione), P, etc.
  • a particularly preferred combination of vitamins comprises biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal HCl, riboflavin, and thiamine-HCl.
  • the IVM medium contains at least one growth factor (GF).
  • growth factors are proteins that bind to receptors on the cell surface, with the primary result of activating cellular proliferation and/or differentiation. Many growth factors are quite versatile, stimulating cellular division in numerous different cell types, while others are specific to a particular cell-type. Growth factors may act on membranes of oocytes to positively stimulate oocyte maturation (Maruo et al., 1993; Goud et al., 1998).
  • Useful growth factors in the context of the present invention include those selected from the following growth factor superfamilies: epidermal growth factor (EGF) family; platelet derived growth factor (PDGF) family; insulin-like growth factor (IGF) family; nerve growth factor (NGF) family; transforming growth factor (TGF) family; fibroblast growth factor (FGF) family; hepatocyte growth factor (HGF) family; hemapoietic growth factors; and cytokines.
  • EGF epidermal growth factor
  • PDGF platelet derived growth factor
  • IGF insulin-like growth factor
  • NGF nerve growth factor
  • TGF transforming growth factor
  • FGF fibroblast growth factor
  • HGF hepatocyte growth factor
  • hemapoietic growth factors and cytokines.
  • the growth factors in IVM medium comprise fibroblast growth factor (FGF) and epidermal growth factor (EGF).
  • FGF and EGF are readily available from commercial sources, and may be purchased together in a cell growth supplement.
  • the IVM medium preferably contains from 0.0001 mg/L to 0.001 mg/L FGF, more preferably from 0.0002 mg/L to 0.001 mg/L FGF, and even more preferably about 0.0005 mg/L FGF.
  • the IVM medium preferably contains from 0.0001 to 0.01 mg/L EGF, more preferably from 0.0005 mg/L to 0.002 mg/L EGF, and even more preferably about 0.001 mg/L EGF.
  • the IVM medium preferably also contains at least one hormone.
  • Preferred hormones include insulin, estradiol, follicle-stimulating hormone (FSH) and luteinizing hormone (LH).
  • FSH follicle-stimulating hormone
  • LH luteinizing hormone
  • Human menopausal gonadotropin (HMG) can generally be substituted for FSH
  • human chorionic gonadotropin (HCG) can generally be substituted for LH.
  • the IVM medium comprises insulin. Normally, insulin is involved in energy metabolism and amino acid transportation into cells.
  • the IVM medium preferably contains from 0.5 mg/L to 50 mg/L insulin, more preferably 0.25 mg/L to 10 mg/L insulin, and even more preferably about 5 mg/L insulin.
  • the IVM medium further comprises estradiol, LH and FSH.
  • the amount of estradiol is preferably about 1 ⁇ g/ml ⁇ 10%, ⁇ 20%, ⁇ 30%, ⁇ 40% or ⁇ 50%.
  • the amount of each of FSH and LH is preferably about 0.08 IU/ml ⁇ 10%, ⁇ 20%, ⁇ 30%, ⁇ 40% or ⁇ 50%.
  • the IVM medium also contains human transferrin (TF).
  • TF is a 75 kDa glycoprotein containing 679 amino acids and two glycan chains. For each, the last residue is a sialic (N-acetyl neuraminic) acid, which can be hydrolyzed by neuraminidase. TF not only transports iron in all extracellular fluid but also exerts many additional properties, including cell growth stimulation (Gross-Weege et al., 1986).
  • the IVM medium preferably contains from from 5 mg/L to 500 mg/L TF, more preferably from 25 mg/L to 100 mg/L TF, and even more preferably about 50 mg/L TF.
  • growth factors, hormones and transferrins discussed above may be naturally occurring, synthetic or recombinant, and encompass biologically active fragments, variants, derivatives and homologs of these substances that retain at least some of the biological activity of the naturally-occurring, synthetic or recombinantly-produced substances.
  • a protein may include one or more amino acid insertions, deletions, substitutions or modifications without substantially reducing its biological activity.
  • energy source is not critical to the invention, and may be e.g. glucose, sodium pyruvate, lactate, or a mixture of some or all of these energy sources.
  • the IVM medium may additional contain additional components such as selenite or selenium and hydrocortisone.
  • Buffers for controlling the pH of the medium may be added, such as Hepes, as may be pH indicators such as phenol red.
  • Antibiotics such as penicillin G and streptomycin may be added to the IVM medium to prevent contamination.
  • Synthetic Serum Supplement may be included in the IVM medium as a source of protein for the oocyte and to prevent cells from adhering to glassware during in vitro culture.
  • SSS has the advantage of having received regulatory approval for human IVF, and is commercially available.
  • the IVM medium includes the components set forth in Table 1.
  • the absolute and relative quantities of each component can vary substantially, for example by up to ⁇ 10%, ⁇ 20%, ⁇ 30%, ⁇ 40%, ⁇ 50% weight by volume.
  • Culture media as described above are also useful for culturing immature oocytes that have a cumulus that is intact or at least partially intact, and/or that are co-cultured with cumulus cells. But the presence of the cumulus cells in the first 12-24 hours of culture eliminates the need for growth factors, so the culture medium need not contain growth factors such as fibroblast growth factor, epidermal growth factor, etc.
  • the culture medium described in the preceding section preferably contains human transferrin, insulin, selenite and hydrocortisone, these are not of substantial benefit when culturing oocytes in the presence of cumulus cells, and are therefore generally absent from the culture medium.
  • complex culture media such as tissue culture media (e.g. TCM-199)
  • tissue culture media e.g. TCM-199
  • complex culture media such as TCM-199
  • oocytes that are essentially free of cumulus cells are cultured in media that consists of, or consists essentially of the components listed in Table 1
  • oocytes that are cultured in the presence of cumulus cells are cultured in media that consists of or consists essentially of the components listed in Table 2.
  • fertilization is accomplished by intracytoplasmic sperm injection (ICSI).
  • ICSI intracytoplasmic sperm injection
  • spermatozoa can be prepared by gradient separation by centrifugation or by “swim-up” followed by washing with a suitable medium such as HTF supplemented with 10% SSS.
  • a single spermatozoon may be injected into an M-II oocyte.
  • the oocyte may be transferred to e.g. fertilization medium supplemented with 10% SSS in a tissue culture dish. Fertilization can be detected by the appearance of two distinct pronuclei and two polar bodies approximately 16-18 hours after ICSI.
  • fertilized oocytes may be cultured in fertilization media until about 72 hours after ICSI and then transferred to e.g. embryo developmental medium (Vitrolife, Göteborg, Sweden) in a tissue culture dish under mineral oil for an additional 48 hours.
  • embryo developmental medium e.g. embryo developmental medium (Vitrolife, Göteborg, Sweden) in a tissue culture dish under mineral oil for an additional 48 hours.
  • a total of 521 immature oocytes (213 oocytes at GV stage and 308 oocytes at M-I stage) were collected from 183 women (age range between 28 to 40 years) who underwent controlled ovarian stimulation and ICSI cycles under informed consent.
  • Ovarian stimulation was performed with GnRHa, HMG or FSH and HCG using standard protocols.
  • oocyte retrieval was carried out using an ultrasound-guided trans-vaginal probe.
  • the collected COC were cultured in human tubal fluid medium (HTF; Irvine Scientific) supplemented with 10% synthetic serum substitute (SSS; Irvine Scientific) and placed in a 5% CO 2 incubator (37° C. with high humidity) for at least 1 h before they were stripped from their cumulus cells.
  • HIF human tubal fluid medium
  • SSS synthetic serum substitute
  • COC were stripped with 85 IU/ml hyaluronidase in HTF medium and mechanical pipetting. All oocytes were completely denuded from their cumulus cells. Denuded oocytes were observed under an inverted microscope to assess for oocyte maturity. The mature oocytes were determined by the presence of a first polar body extrusion (1PB). The oocytes with a visual polar body (M-II) were provided for clinical application to perform the ICSI procedure immediately. Immature oocytes were defined by the absence of 1PB and then identified as GV or M-I stages depending upon whether the oocytes contained a visible GV in the cytoplasm under microscope. Immature oocytes without a GV in the cytoplasm were considered as M-I stage. These immature oocytes were used for in vitro maturation.
  • PB first polar body extrusion
  • M-II visual polar body
  • the immature oocytes were cultured in HTF medium supplemented with 10% SSS (Irvine Scientific) placed in a 37° C. and 5% CO 2 incubator before transferred to the maturation medium.
  • the time of the immature oocytes were transferred to maturation medium is defined as the starting point for maturation in culture. Normally, it takes approximately 3-4 h from oocyte retrieval to the start of oocyte maturation.
  • the immature oocytes were washed at least twice in maturation medium.
  • the immature oocytes were cultured in an organ tissue culture dish (60 ⁇ 15 mm; Falcon) containing 1 ml of maturation medium (TCM-199 or IVM-medium) at 37° C. in an atmosphere of 5% CO 2 and 95% air with high humidity.
  • TCM-199 or IVM-medium 1 ml of maturation medium
  • a maximum of 5 oocytes (1 to 5) were placed in each organ tissue culture dish.
  • the maturity of the oocytes was determined under the microscope at 6 h for M-I stage oocytes and then at 24 h and 48 h for both M-I and GV stage oocytes.
  • ICSI Intracytoplasmic Sperm Injection
  • sperm for ICSI were prepared by PureSperm (Nidacon, Sweden) gradient separation (45%, 70% and 90%) at 560 g for 20 min. Following gradient separation, the sperm pellet was washed twice (200 g) with 2 ml of HTF medium (Irvine Scientific) supplemented with 10% SSS.
  • each oocyte was transferred into a 20 ⁇ l droplet of HTF medium supplemented with 10% SSS in a tissue culture dish (35 ⁇ 10 mm; Falcon) under mineral oil (Sigma).
  • Fertilization was assessed 16-18 h after ICSI for the appearance of two distinct pronuclei and two polar bodies. Since the oocytes were not matured at the same time following culture, the sperm samples were kept in 5 ml Falcon tubes (tight tube cap) containing 0.5-1.0 ml HTF medium supplemented with 10% SSS at room temperature for further insemination by ICSI at 6 h, 24 h or 48 h, respectively.
  • Fertilized oocytes (with two pronuclei) were cultured in HTF medium supplemented with 10% SSS until 72 h after ICSI, and then each embryo was transferred into a 20 ⁇ l droplet of G2.2 medium (VitroLife, Sweden) in a tissue culture dish (35 ⁇ 10 mm; Falcon) under mineral oil (Sigma) further culture for 48 h. Embryo development was recorded at 24 h intervals following culture until day 5 (120 h). All embryos were destroyed on day 5 following consent instruction from the patients.
  • FIG. 1 shows the fertilization rates of the oocytes matured in the IVM-medium or TCM-199 at 6 h (93.8% vs. 90.1%), 24 h (89.2% vs. 84.9%) and 48 h (50.0% vs. 0.0%), respectively.
  • the treatment cycles was initiated by the administration of intra-vaginal progesterone (Prometrium; Schering, Pointe-Clair, Quebec, Canada) in a dose of 300 mg daily for 10 days. Withdrawal bleeding occurred within 3 days after the last dose.
  • intra-vaginal progesterone Prometrium; Schering, Pointe-Clair, Quebec, Canada
  • Oocyte retrieval was performed on day 10 to 14 of the cycle. Before oocyte collection 36 hours, the patients were given s.c. 10,000 IU of human chorionic gonadotropin (HCG) for priming.
  • HCG human chorionic gonadotropin
  • Transvaginal ultrasound-guided oocyte collection was performed using a specially designed 19 G single-lumen aspiration needle (Cook, Australia) with an aspiration pressure of 7.5 kPa. Aspiration of all small follicles was performed under local anaesthesia. Oocytes were collected in 10 ml culture tubes (Falcon, USA) containing 2.0 ml warm 0.9% saline contained with 2 IU/ml heparin (Baxter, Toronto, Ontario, Canada).
  • COCs cumulus-oocyte complexes
  • the mature oocytes (metaphase-II stage) were inseminated by ICSI. Fertilization was assessed 18 h after ICSI for the appearance of two distinct pronuclei and two polar bodies. Embryo transfer was performed on day 2 or 3 after ICSI. Since the oocytes were inseminated either 24 or 48 hours following culture, the developmental stages of embryos were variable.
  • estradiol Estace; Roberts Pharmaceutical, Mississauga, Canada
  • estradiol Estace; Roberts Pharmaceutical, Mississauga, Canada
  • the endometrial thickness on the day of oocyte retrieval was ⁇ 4 mm, a 10 mg dose was administered; if it was >6 mm, a 6 mg dose was given.
  • Luteal support was provided by 400 mg intravaginal progesterone (Prometrium) twice daily for 16 days starting from the day after oocyte collection.
  • Inorganic Salt CaCl 2 200.0000 KCl 400.0000 MgSO 4 98.0000 NaCl 6800.0000 NaHCO 3 1250.0000 NaH 2 PO 4 .H 2 O 125.0000 Amino Acids: L-Alanine 8.9000 L-Arginine 126.4000 L-Asparagine 13.2000 L-Aspartic Acid 13.3000 L-Cystine 24.0000 L-Glutamic Acid 14.7000 L-Glutamine 292.0000 Glycine 7.5000 L-Histidine.HCI.H 2 O 42.0000 L-Isoleucine 52.4000 L-Leucine 52.4000 L-Lysine.HCI 72.5000 L-Methionine 15.1000 L-Phenylalanine 33.0000 L-Proline 11.5000 L-Serine 10.5000 L-Threonine 47.6000 L-Tryptophan 10.2000 L-Tyrosine 36.0000 L-Valine 46.8000 Vitamins: Biotin 0.0100 D-Ca Pant
  • Pincus G, Enzmann E V The comparative behaviour of mammalian eggs in vivo and in vitro. 1. The activation of ovarian eggs. J Exp Med 1935; 62:665-675.

Abstract

The invention provides methods for in vitro maturation of immature human oocytes. In one aspect, the invention provides a method for in vitro maturation of immature human oocytes, comprising the steps of: (a) inducing in a female human subject an increase in endogenous luteinizing hormone levels, said subject having not undergone an ovarian stimulation protocol prior to said inducing step; (b) obtaining from said subject an immature oocyte; and (c) culturing said oocyte until maturity.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. provisional patent application No. 60/362,395, filed Mar. 8, 2002, which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to the field of assisted reproductive technology (ART), particularly to methods for culturing immature human oocytes so that they mature to metaphase-II stage capable of being fertilized and forming a viable embryo.
  • BACKGROUND OF THE INVENTION
  • Since the first successful human pregnancy by way of in vitro fertilization (IVF) was achieved in 1978, ART has helped thousands of thousands of women to overcome infertility problem
  • Normally, during the follicular phase of a woman's reproductive cycle, only a single follicle grows to the preovulatory stage and releases its oocyte for potential fertilization. However, current IVF treatment requires that multiple oocytes be harvested. Therefore, women are normally pre-treated for approximately two or three weeks with gonadotropin releasing hormone agonists (GnRHa). After pituitary suppression has been achieved, human menopausal gonadotropin (HMG) or purified follicle-stimulating hormone (FSH) is administered to induce development of multiple follicles. Once two leading follicles have reached a diameter of at least about 18-20 mm, the patient is typically given 5,000 to 10,000 IU of human chorionic gonadotropin (HCG) to trigger final oocyte maturation. Approximately 36 hours later, a large number of oocytes (10 on average) are collected.
  • The harvested oocyte is then fertilized with the sperm. In many instances of human infertility, fertilization is effected by intracytoplasmic sperm injection (ICSI). Successful IVF generally requires that the oocyte have reached the metaphase-II (M-II) stage before fertilization is attempted. A M-II oocyte is ready to accept the sperm and be fertilized. Metaphase-II is characterized by exclusion of one polar body from the cytoplasm, and is typically detected by visual identification under a microscope.
  • The harvested oocyte remains surrounded by cumulus cells that support and promote the final maturation and development of the oocyte. The cumulus cells interfere with ICSI and obscure microscopic observation of the oocyte. Therefore, prior to ICSI, it is necessary to denude the oocyte of cumulus cells to permit determination of the level of oocyte maturity. This is generally accomplished by enzymatic and mechanical stripping of the cumulus cells.
  • Unfortunately, not all of the oocytes have matured to the M-II stage when they are harvested in ovarian stimulation. As a result of follicular asynchrony, in general, at least 10-15% of oocytes are still immature, and are at the germinal vesicle (GV) or metaphase-I (M-I) stage, at the time of harvest. These immature oocytes, denuded of cumulus cells, are discarded at most IVF clinics.
  • It would be desirable if these immature GV and M-I stage oocytes, presently wasted, could be brought to maturity, in vitro, and then successfully fertilized. Moreover, it would be desirable to be able to harvest immature oocytes from unstimulated ovaries and mature them to the M-II stage by way of in vitro maturation (IVM), thereby avoiding the entire regimen of treatment of women with GnRHa and stimulation of ovaries with gonadotropin. These procedures of ovarian stimulation require frequent blood sampling and ultrasound monitoring, at considerable cost. Moreover, current IVF treatment causes substantial discomfort and, in some cases, results in ovarian hyperstimulation syndrome (OHSS) and/or even death. There is also anxiety that the long-term effects of repeated ovarian stimulation may increase the risk of ovarian, endometrial and breast cancer.
  • Thus, there is substantial interest in developing IVF techniques that rely upon in vitro maturation of oocytes, rather than harvesting mature oocytes by ovarian stimulation. But only a few live births have been obtained from cumulus-denuded oocytes matured in vitro (Nagy et al., 996; Jaroudi et al., 1997; Edirisinghe et al., 1997). Clearly then, there is an opportunity for improving the maturational and developmental competence of cumulus-denuded immature oocytes matured in vitro.
  • Recovery of immature oocytes followed by IVM of these immature oocytes is a potentially useful treatment for women with polycystic ovarian syndrome (PCOS) related infertility. PCOS is one of the most common reproductive disorders in women of childbearing age. It has a heterogeneous presentation, which is clinically characterized by anovulation and hyperandrogenism, and on pelvic ultrasound examination shows numerous antral follicles within the ovaries (Adams et al., 1986). There is a significantly higher risk of OHSS in these group of women compared with normal ovaries (MacDougall et al., 1993). Although the first pregnancy and live birth from immature oocytes retrieved from women with PCOS followed by IVM has been reported, the success rates are still low because oocyte maturation rates are relatively low.
  • Some attempts at developing IVM media and methods have involved the use of human follicular fluid, in an effort to mimic the conditions within the follicle. In view of the risk of transmission of disease, allergic reaction, etc., use of biological fluids in IVM techniques is prohibited in most countries. Instead, the use of a “chemically-defined medium”, wherein the chemical composition of all of the ingredients is known, is preferred.
  • Mammalian immature oocytes removed from antral follicles will resume meiosis spontaneously without hormonal stimulation when cultured in a simple medium (Pincus & Enamann, 1935). However, oocyte maturation in vitro is profoundly affected by culture conditions.
  • Most attempts at human IVM have employed tissue culture medium 199 (TCM-199) as the maturation medium for immature oocytes (Trounson et al., 1994; Barnes et al., 1995; Chian et al., 1999a, 1999b, 2000, 2001). TCM-199 is a complex medium that contains many components, and was designed for in vitro culture of somatic cells, not germ cells. Notably, TCM-199 does not contain any growth factors. TCM-199 was first used for sheep oocyte IVM (Moor & Trounson, 1977), and has since been used routinely for bovine oocyte IVM (Brackett & Zuelke, 1993).
  • Although different culture media have been used to mature human oocytes (Shea et al., 1975; Cha et al., 1991; Trounson et al., 1994; Cha & Chian, 1998; Chian et al., 1999a, 1999b, 2000), it has been indicated recently that there is no apparent benefit for oocyte maturation, fertilization and embryonic development from use of these culture media (Trounson et al., 1998; 2001).
  • Thus, there remains a need for methods for maturing human oocytes in vitro.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides in vitro maturation methods that are useful for culturing immature human oocytes to maturity.
  • In a broad aspect, the invention provides a method for in vitro maturation of immature human oocytes, comprising the steps of:
    • (a) inducing in a female human subject an increase in endogenous luteinizing hormone levels, the subject having not undergone an ovarian stimulation protocol prior to the inducing step;
    • (b) obtaining from the subject an immature oocyte; and
    • (c) culturing the oocyte until maturity.
  • Preferably, prior to step (a), the subject has not been treated with a gonadotrophin releasing hormone agonist, human menopausal gonadotrophin, HCG, or follicle stimulating hormone (FSH).
  • In one embodiment, step (a) comprises administering to the subject HCG or luteinizing hormone (LH), or both. In a preferred embodiment, HCG is administered in an amount of about 5000 to about 20,000 IU, more preferably about 10,000 IU.
  • The immature human oocyte is preferably an M-I stage or GV stage ooctye. The oocyte preferably is cultured until it reaches M-II.
  • In one embodiment, the immature human oocyte is essentially free of cumulus cells and is cultured in a culture medium comprising:
  • at least one inorganic salt;
  • essential amino acids or a source thereof;
  • an energy source; and
  • at least one growth factor.
  • In one embodiment, the growth factor is fibroblast growth factor or epidermal growth factor, or both.
  • In one embodiment, the culture medium further comprises at least one hormone, e.g. insulin. In a preferred embodiment, the culture medium comprises from 0.5 mg/L to 50 mg/L insulin.
  • In one embodiment, the culture medium further comprises human transferrin, preferably from 5 mg/L to 500 mg/L human transferrin.
  • In one embodiment, the culture medium comprises from 0.0001 mg/L to 0.001 mg/L fibroblast growth factor.
  • In one embodiment, the culture medium comprises from 0.0001 to 0.01 mg/L epidermal growth factor.
  • In one embodiment, the culture medium comprises one or more vitamins, preferably biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal-HCl, riboflavin, and thiamine-HCl.
  • The culture medium may further comprise hydrocortisone or selenite, or both.
  • Preferred inorganic salts include CaCl2, KCl, MgSO4, NaCl, NaHCO3 and NaH2PO4-H2O.
  • Preferred energy sources include D-glucose and sodium pyruvate, or both D-glucose and sodium pyruvate.
  • In one embodiment, the amino acids comprise alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • In one embodiment, the culture medium comprises the inorganic salts, amino acids, vitamins and other components as set forth in Table 1, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume), more preferably ±10% (weight/volume), of the amount specified in Table 1.
  • In another embodiment, the oocyte has a cumulus that is intact or at least partially intact, and/or the oocyte is cultured in the presence of cumulus cells.
  • In this embodiment, the oocyte may be cultured in a culture medium that is essentially free of one or more of epidermal growth factor, fibroblast growth factor, human transferrin, insulin, selenite, and hydrocortisone.
  • In one embodiment the oocyte is cultured in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume), more preferably ±10% (weight/volume) of the amount specified in Table 2.
  • In one embodiment, the culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components identified in Table 2.
  • In another embodiment, the culture medium consists of the inorganic salts, amino acids, vitamins and other components identified in Table 2.
  • In another embodiment, the invention provides a method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising:
  • at least one inorganic salt;
  • essential amino acids or a source thereof;
  • an energy source; and
  • at least one growth factor.
  • This embodiment is particularly useful when the immature human oocyte is essentially free of cumulus cells.
  • In another embodiment, the invention provides a method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2. This embodiment is particularly useful when the immature human oocyte is cultured in the presence of cumulus cells.
  • In a preferred embodiment, the culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
  • In another preferred embodiment, the culture medium consists of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
  • In particularly preferred embodiments, each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50%, more preferably ±10% (weight/volume), of the amount specified in Table 2.
  • In another embodiment, certain of the components listed in the “other components” sections of Table 1 and 2 may be absent from the medium. Components that may be absent include, without limitation, phenol red, penicillin G, streptomycin, and human serum albumin. This may apply wherever reference is made herein to Table 1 or 2.
  • By the methods of the invention, increases may be obtained in one or more of:
    • (a) total number of oocytes reaching M-II stage;
    • (b) total number of oocytes fertilized;
    • (c) total number of embryos developing to the 8-cell stage; and
    • (d) total number of blastocysts formed; as measured at 6 hours, 12 hours, 24 hours or 48 hours after commencement of maturation in IVM media in accordance with the invention. Increases in one or more of (a) to (d) of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 300%, 400% or 500% may be advantageously obtained.
    BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows fertilization rates of M-I stage oocytes derived from stimulation and ICSI cycles following 6 h, 24 h or 48 h of maturation, respectively.
  • FIG. 2 shows cleavage rates of M-I stage oocytes derived from stimulation and ICSI cycles following 6 h, 24 h or 48 h of maturation, respectively.
  • FIGS. 3A and 3B show embryos developing to the 8-cell stage (A) and the blastocyst stage (B) following 6 h, 24 h or 48 h of maturation of M-I stage oocytes derived from stimulation and ICSI cycles.
  • FIG. 4 shows fertilization rates of GV stage oocytes derived from stimulation and ICSI cycles following 24 h or 48 h of maturation.
  • FIG. 5 shows cleavage rates of GV stage oocytes derived from stimulation and ICSI cycles following 24 h or 48 h of maturation.
  • FIGS. 6A and 6B show embryos developing to the 8-cell stage (A) and the blastocyst stage (B) following 24 h or 48 h of maturation of GV stage oocytes derived from stimulation and ICSI cycles.
  • FIG. 7 shows a blastocyst derived from GV stage oocytes following 24 h of maturation in vitro and illustrates that there is no difference in morphology from oocytes matured in vivo. The scale bar indicates 5 μm length.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Candidates for IVM
  • Any female human subject who possesses viable oocytes is a candidate for IVM therapy. Typically, the subject will suffer from some form of infertility. For instance, the subject may experience normal oocyte production but have an impediment to fertilization, as in, e.g. PCOS or PCOS-like ovaries. IVM is also useful in women infertility with endometriosis, blockage of either or both fallopian tubes, etc. In typical PCOS, numerous follicles mature simultaneously in the ovaries, without the appearance of a dominant follicle. The subject's menstrual cycle may be regular, irregular or non-existent. IVM may be especially useful in women who are susceptible to or suffer from OHSS and are therefore not suitable candidates for traditional in vitro fertilization techniques involving an ovarian stimulation protocol.
  • Alternatively, the subject may be an individual who does not suffer from infertility or have any reproductive impediment whatsoever, but for whom an in vitro fertilization method nevertheless remains desirable, as in, e.g. cases of male factor infertility.
  • Absence of Ovarian Stimulation Protocol
  • Unlike in conventional in vitro fertilization methods, IVM in accordance with the invention does not necessarily involve an ovarian stimulation protocol. One of two “ovarian stimulation protocols” is usually used in conventional IVF, a “long protocol” or a “short protocol.” The long protocol has two steps. The first step involves pre-treating the subject for two or three weeks with GnRHa to down-regulate pituitary activity. Once pituitary suppression has been achieved, in the second step, HMG or FSH is administered to induce the development of multiple follicles. The short protocol involves only the ovarian stimulation step, but not the down-regulation step.
  • Ovarian stimulation is required in conventional IVF techniques to permit the oocytes to mature to the M-II stage prior to harvest. In the present invention, wherein oocytes are harvested at an immature stage, i.e. prior to reaching the M-II stage, ovarian stimulation need not be used. That is, the subject need not be pre-treated with GnRHa's, HMG and/or FSH.
  • Increasing Endogenous Levels of Luteinizing Hormone (LH)
  • Prior to retrieving immature oocytes, an increase in endogenous levels of LH is induced in the subject (also described herein as “priming”). This may be accomplished by e.g. administering to the subject an effective amount of human chorionic gonadotropin (HCG) (Profasi Serono, Oakville, Ontario, Canada) or LH, either of which stimulate endogenous LH production. Baseline concentration of LH in the plasma of premenopausal women is usually about 2-4 mIU/ml, and at midcycle peak may be up to 25-35 mIU/ml. The expression “inducing in a female human subject an increase in endogenous luteinizing hormone levels” means increasing the plasma LH concentration above the baseline concentration. Preferably plasma LH concentration is increased at least 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1000%, or 2000% above baseline concentration.
  • The dosage and mode of administration of HCG or LH may vary depending on the patient and the circumstances and may be determined by those of skill in the art. Subcutaneous injection is a preferred mode of administration although other modes of administration may be used. A single injection of about 5000 to about 20,000 IU of HCG is generally sufficient, with a single dose of about 10,000 IU of HCG being preferred.
  • In the case of a subject having a regular menstrual cycle, the time for inducement may be determined relative to the commencement of menstrual bleeding, which is considered “day 0”. In a subject having an irregular or absent menstrual cycle, commencement of menstrual bleeding (i.e. the initiation of “day 0”) can be achieved by procedures known in the art, e.g. by the administration of progesterone (available from e.g. Prometrium; Schering, Pointe-Claire, Quebec, Canada), e.g. intravaginally, in an amount of approximately 300 mg/day for 10 days.
  • Typically, on day three and day eight, the ovaries are examined by ultrasound to assess the size and the number of follicles. Usually, the dominant follicle reaches a diameter of at least about 16 mm by day 10-14.
  • In instances where a dominant follicle is present (e.g. as in normal ovarian function, as in the case of a healthy subject or a subject suffering from e.g. endometriosis or tubal blockage) the increase in endogenous LH activity is induced (e.g. by administration of HCG) when the dominant follicle reaches a diameter of about 15-17 mm, preferably at least about 16 mm.
  • If a dominant follicle is not present, as in cases of e.g. PCOS, inducement of an increase in endogenous LH levels preferably occurs when the follicles are less than about 12 mm in diameter.
  • Retrieval of Immature Oocytes
  • Oocytes are generally retrieved from about 32 to about 40 hours after priming, more preferably about 34 to 38 hours after priming and even more preferably about 36 hours after priming. Means for retrieving oocytes are known in the art. In one embodiment, transvaginal ultrasonographically-guided oocyte collection is done using a 17-20 gauge, preferably 19 gauge single-lumen aspiration needle at an aspiration pressure of from about 5 to about 10 kPa, preferably about 7.5 kPa.
  • Culture of Immature Oocytes
  • As used herein, the term “immature human oocyte” means a human oocyte that has not yet reached metaphase-II (M-II). As discussed previously, metaphase-II is characterized by exclusion of one polar body from the cytoplasm. Immature human oocytes used in the invention are typically at the germinal vesicle (GV) or metaphase-I (M-I) stage.
  • Oocytes may be cultured with their cumulus intact, in a form known as a “cumulus-oocyte-complex” (COC), or oocytes may be partially or entirely denuded from cumulus cells. COC can be stripped with 85 IU/ml hyaluronidase in HEPES buffered medium and mechanically pipetted until oocytes are denuded. An oocyte that is “essentially free of cumulus cells” is an oocyte that is associated with sufficiently few cumulus cells that the cumulus cells have no detectable physiological effect on the oocyte.
  • Culture conditions for human oocytes are known in the art and are not critical to the invention. Suitable culture conditions include e.g. culturing the oocytes at 37° C. in an atmosphere of 95% air and 5% CO2 at high humidity, e.g. 100% humidity. A “triple gas” atmosphere of 5% O2, 5% CO2 and 90% N2 may also be used. Mineral oil may be overlaid on the medium to control evaporation and/or temperature. Oocytes are typically cultured in a well containing 1 ml of culture medium or more, or may be cultured in 10 μl of culture medium or less in a droplet in a culture dish.
  • Immature oocytes may be cultured for e.g. about 24 to about 48 hours. Because about 60% of oocytes typically reach maturity (M-II) after 24 hours of culture, when oocytes are cultured with an intact or partially intact cumulus, they may be denuded at about 24 hours and observed under a microscope to assess oocyte maturity. The physiological influence of the cumulus cells on the oocyte persists for only about the first 12 hours in culture, so denuding the oocyte after about 24 hours in culture has little or no negative effect.
  • Culture Medium for Oocytes Essentially Free of Cumulus Cells
  • Due to the influence of cumulus cells on the maturation of oocytes, somewhat different culture media may be used when the oocytes are cultured for the first 12-24 hours in the presence of cumulus cells. This can occur e.g. when the oocytes are cultured as COCs or partially denuded or when denuded oocytes are co-cultured in the presence of cumulus cells.
  • The inorganic salts, essential and non-essential amino acids, and energy source in the IVM medium are not critical to the invention. Suitable salts, amino acids and energy sources are known in the art.
  • Inorganic salts are provided to buffer the pH of the medium within a range preferably of about 7.2-7.4 and to maintain correct osmolarity of the medium with the oocytes. Suitable inorganic salts and concentrations thereof as used in culture media are known in the art. Typical inorganic salts include CaCl2, KCl, MgSO4, NaCl, NaHCO3, NaH2PO4.H2O, FE(NO3)3.9H2O, KH2PO4, Na.acetate, Na2H2PO4, etc.
  • The IVM medium contains at least one amino acid or source thereof. Preferably, at least the essential amino acids, or sources thereof, are included in IVM medium. “Essential” amino acids are those amino acids not synthesized in the oocyte and that are essential for protein synthesis. The essential amino acids are generally considered to be isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine. A commercial amino acid mixture, containing the eight essential amino acids as well as some or all of the remaining non-essential amino acids may conveniently be used. Non-naturally occuring amino acids or amino acid derivatives as are known in the art may also be included in the IVM medium. In a particularly preferred embodiment, the IVM medium of the invention comprises alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glutamine, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • The IVM medium preferably contains vitamins as are known in the art for inclusion in culture media. Vitamins that may be included in the media include, without limitation, vitamins A1 (retinol), A2 (an alternative form of retinol), B1 (thiamine), B2 (riboflavin), B6 (pyridoxine), B9 (folic acid), B12 (cyanocobalamin), B17, C (ascorbic acid), D, D2 (calciferol), D3 (cholecalciferol), E (tocopherol), H (biotin), K, K1 (phylloquinone), K2, K3 (menadione), P, etc. A particularly preferred combination of vitamins comprises biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal HCl, riboflavin, and thiamine-HCl.
  • The IVM medium contains at least one growth factor (GF). Broadly speaking, growth factors are proteins that bind to receptors on the cell surface, with the primary result of activating cellular proliferation and/or differentiation. Many growth factors are quite versatile, stimulating cellular division in numerous different cell types, while others are specific to a particular cell-type. Growth factors may act on membranes of oocytes to positively stimulate oocyte maturation (Maruo et al., 1993; Goud et al., 1998). Useful growth factors in the context of the present invention include those selected from the following growth factor superfamilies: epidermal growth factor (EGF) family; platelet derived growth factor (PDGF) family; insulin-like growth factor (IGF) family; nerve growth factor (NGF) family; transforming growth factor (TGF) family; fibroblast growth factor (FGF) family; hepatocyte growth factor (HGF) family; hemapoietic growth factors; and cytokines.
  • In a preferred embodiment, the growth factors in IVM medium comprise fibroblast growth factor (FGF) and epidermal growth factor (EGF). FGF and EGF are readily available from commercial sources, and may be purchased together in a cell growth supplement. The IVM medium preferably contains from 0.0001 mg/L to 0.001 mg/L FGF, more preferably from 0.0002 mg/L to 0.001 mg/L FGF, and even more preferably about 0.0005 mg/L FGF. The IVM medium preferably contains from 0.0001 to 0.01 mg/L EGF, more preferably from 0.0005 mg/L to 0.002 mg/L EGF, and even more preferably about 0.001 mg/L EGF.
  • The IVM medium preferably also contains at least one hormone. Preferred hormones include insulin, estradiol, follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Human menopausal gonadotropin (HMG) can generally be substituted for FSH, and human chorionic gonadotropin (HCG) can generally be substituted for LH. In a preferred embodiment, the IVM medium comprises insulin. Normally, insulin is involved in energy metabolism and amino acid transportation into cells. The IVM medium preferably contains from 0.5 mg/L to 50 mg/L insulin, more preferably 0.25 mg/L to 10 mg/L insulin, and even more preferably about 5 mg/L insulin. In a particularly preferred embodiment, the IVM medium further comprises estradiol, LH and FSH. The amount of estradiol is preferably about 1 μg/ml ±10%, ±20%, ±30%, ±40% or ±50%. The amount of each of FSH and LH is preferably about 0.08 IU/ml ±10%, ±20%, ±30%, ±40% or ±50%.
  • In a preferred embodiment, the IVM medium also contains human transferrin (TF). TF is a 75 kDa glycoprotein containing 679 amino acids and two glycan chains. For each, the last residue is a sialic (N-acetyl neuraminic) acid, which can be hydrolyzed by neuraminidase. TF not only transports iron in all extracellular fluid but also exerts many additional properties, including cell growth stimulation (Gross-Weege et al., 1986). The IVM medium preferably contains from from 5 mg/L to 500 mg/L TF, more preferably from 25 mg/L to 100 mg/L TF, and even more preferably about 50 mg/L TF.
  • The growth factors, hormones and transferrins discussed above may be naturally occurring, synthetic or recombinant, and encompass biologically active fragments, variants, derivatives and homologs of these substances that retain at least some of the biological activity of the naturally-occurring, synthetic or recombinantly-produced substances. By way of a non-limiting example, a protein may include one or more amino acid insertions, deletions, substitutions or modifications without substantially reducing its biological activity.
  • The choice of energy source is not critical to the invention, and may be e.g. glucose, sodium pyruvate, lactate, or a mixture of some or all of these energy sources.
  • The IVM medium may additional contain additional components such as selenite or selenium and hydrocortisone.
  • Buffers for controlling the pH of the medium may be added, such as Hepes, as may be pH indicators such as phenol red.
  • Antibiotics such as penicillin G and streptomycin may be added to the IVM medium to prevent contamination.
  • Synthetic Serum Supplement (SSS) may be included in the IVM medium as a source of protein for the oocyte and to prevent cells from adhering to glassware during in vitro culture. SSS has the advantage of having received regulatory approval for human IVF, and is commercially available.
  • In a particularly preferred embodiment, the IVM medium includes the components set forth in Table 1. The absolute and relative quantities of each component can vary substantially, for example by up to ±10%, ±20%, ±30%, ±40%, ±50% weight by volume.
  • Culture Medium for Oocytes in the Presence of Cumulus Cells
  • Culture media as described above are also useful for culturing immature oocytes that have a cumulus that is intact or at least partially intact, and/or that are co-cultured with cumulus cells. But the presence of the cumulus cells in the first 12-24 hours of culture eliminates the need for growth factors, so the culture medium need not contain growth factors such as fibroblast growth factor, epidermal growth factor, etc.
  • Also, whereas the culture medium described in the preceding section preferably contains human transferrin, insulin, selenite and hydrocortisone, these are not of substantial benefit when culturing oocytes in the presence of cumulus cells, and are therefore generally absent from the culture medium.
  • More generally, complex culture media, such as tissue culture media (e.g. TCM-199), contain many components that are designed specifically for somatic cell culture in vitro rather than for oocyte culture. The use of complex culture media such as TCM-199 provides poorer results than if less complex media as described herein are used. Thus, in particularly preferred embodiments, oocytes that are essentially free of cumulus cells are cultured in media that consists of, or consists essentially of the components listed in Table 1; and oocytes that are cultured in the presence of cumulus cells are cultured in media that consists of or consists essentially of the components listed in Table 2.
  • The term “comprising” is intended to be inclusive or open-ended and does not exclude additional, unrecited elements or method steps. The term “consisting of” is intended to exclude any element, step, or ingredient that is not specified, but for impurities ordinarily associated with specified ingredients or materials. The term “consisting essentially of” is intended to occupy a middle ground between “comprising” and “consisting of” and to exclude other than the specified materials or steps and those that do not materially affect the basic and novel characteristics of the invention herein.
  • Fertilization of Oocytes and Culture of Embryos
  • Techniques for fertilizing oocytes and culturing embryos are known in the art.
  • In one embodiment, fertilization is accomplished by intracytoplasmic sperm injection (ICSI). Generally, spermatozoa can be prepared by gradient separation by centrifugation or by “swim-up” followed by washing with a suitable medium such as HTF supplemented with 10% SSS. A single spermatozoon may be injected into an M-II oocyte. Following ICSI, the oocyte may be transferred to e.g. fertilization medium supplemented with 10% SSS in a tissue culture dish. Fertilization can be detected by the appearance of two distinct pronuclei and two polar bodies approximately 16-18 hours after ICSI.
  • In one embodiment, fertilized oocytes may be cultured in fertilization media until about 72 hours after ICSI and then transferred to e.g. embryo developmental medium (Vitrolife, Göteborg, Sweden) in a tissue culture dish under mineral oil for an additional 48 hours.
  • As used herein, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Unless defined otherwise all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs.
  • The invention is further illustrated by reference to the following non-limiting examples.
  • EXAMPLE 1
  • Source of Immature Oocytes
  • A total of 521 immature oocytes (213 oocytes at GV stage and 308 oocytes at M-I stage) were collected from 183 women (age range between 28 to 40 years) who underwent controlled ovarian stimulation and ICSI cycles under informed consent. Ovarian stimulation was performed with GnRHa, HMG or FSH and HCG using standard protocols.
  • 36 hours after HCG injection, oocyte retrieval was carried out using an ultrasound-guided trans-vaginal probe. The collected COC were cultured in human tubal fluid medium (HTF; Irvine Scientific) supplemented with 10% synthetic serum substitute (SSS; Irvine Scientific) and placed in a 5% CO2 incubator (37° C. with high humidity) for at least 1 h before they were stripped from their cumulus cells.
  • COC were stripped with 85 IU/ml hyaluronidase in HTF medium and mechanical pipetting. All oocytes were completely denuded from their cumulus cells. Denuded oocytes were observed under an inverted microscope to assess for oocyte maturity. The mature oocytes were determined by the presence of a first polar body extrusion (1PB). The oocytes with a visual polar body (M-II) were provided for clinical application to perform the ICSI procedure immediately. Immature oocytes were defined by the absence of 1PB and then identified as GV or M-I stages depending upon whether the oocytes contained a visible GV in the cytoplasm under microscope. Immature oocytes without a GV in the cytoplasm were considered as M-I stage. These immature oocytes were used for in vitro maturation.
  • The immature oocytes were cultured in HTF medium supplemented with 10% SSS (Irvine Scientific) placed in a 37° C. and 5% CO2 incubator before transferred to the maturation medium. The time of the immature oocytes were transferred to maturation medium is defined as the starting point for maturation in culture. Normally, it takes approximately 3-4 h from oocyte retrieval to the start of oocyte maturation.
  • In vitro Maturation of the Immature Oocytes
  • Immature oocytes were divided into two groups. One was the GV stage oocytes (n=213), another was the M-I stage oocytes (n=308). Each group of oocytes was randomly cultured in one of two maturation media respectively: (1) TCM-199 (GIBCO, Catalogue No. 11153) or, (2) IVM-medium (Table 1). Both TCM-199 and IVM-medium were supplemented with 10% SSS (Irvine Scientific), 0.075 IU/ml FSH +LH (Humegon, Ontario, Canada) and, 1.0 μg/ml estradiol (Sigma). Additional supplements to TCM-199 included 0.25 mM sodium pyruvate (Sigma), 0.05 unit/ml penicillin G and streptomycin (GIBCO).
  • Before culture, the immature oocytes were washed at least twice in maturation medium. The immature oocytes were cultured in an organ tissue culture dish (60×15 mm; Falcon) containing 1 ml of maturation medium (TCM-199 or IVM-medium) at 37° C. in an atmosphere of 5% CO2 and 95% air with high humidity. A maximum of 5 oocytes (1 to 5) were placed in each organ tissue culture dish.
  • Following culture, the maturity of the oocytes was determined under the microscope at 6 h for M-I stage oocytes and then at 24 h and 48 h for both M-I and GV stage oocytes.
  • Intracytoplasmic Sperm Injection (ICSI)
  • In vitro matured oocytes were inseminated by ICSI with sperm of the patient's husband. All semen samples were obtained from the ejaculate for clinical utilization. The remaining samples were donated for this experiment under informed consent.
  • Sperm for ICSI were prepared by PureSperm (Nidacon, Sweden) gradient separation (45%, 70% and 90%) at 560 g for 20 min. Following gradient separation, the sperm pellet was washed twice (200 g) with 2 ml of HTF medium (Irvine Scientific) supplemented with 10% SSS.
  • A single sperm was injected into each M-II oocyte. Following ICSI, each oocyte was transferred into a 20 μl droplet of HTF medium supplemented with 10% SSS in a tissue culture dish (35×10 mm; Falcon) under mineral oil (Sigma).
  • Fertilization was assessed 16-18 h after ICSI for the appearance of two distinct pronuclei and two polar bodies. Since the oocytes were not matured at the same time following culture, the sperm samples were kept in 5 ml Falcon tubes (tight tube cap) containing 0.5-1.0 ml HTF medium supplemented with 10% SSS at room temperature for further insemination by ICSI at 6 h, 24 h or 48 h, respectively.
  • Culture of Embryos
  • Fertilized oocytes (with two pronuclei) were cultured in HTF medium supplemented with 10% SSS until 72 h after ICSI, and then each embryo was transferred into a 20 μl droplet of G2.2 medium (VitroLife, Sweden) in a tissue culture dish (35×10 mm; Falcon) under mineral oil (Sigma) further culture for 48 h. Embryo development was recorded at 24 h intervals following culture until day 5 (120 h). All embryos were destroyed on day 5 following consent instruction from the patients.
  • Statistical Analysis
  • Statistical significant differences in the percentages of oocyte maturation, fertilization and development between the groups were determined and compared by Student-Newman-Keuls' test (Steel & Torrie, 1980). A P-value of <0.05 was considered statistically significant.
  • Results
  • As shown in Table 3, 52.0% and 46.1% of M-I stage oocytes were mature 6 h after culture in the IVM medium and TCM-199, respectively. Following further culture for 24 h and 48 h, there were no differences in oocyte maturation rates between IVM-medium (76.0% and 78.6%) and TCM-199 (67.5% and 70.8%).
  • As shown in Table 4, the final fertilization rates also were not different between these two media (90.0% vs. 84.4%).
  • FIG. 1 shows the fertilization rates of the oocytes matured in the IVM-medium or TCM-199 at 6 h (93.8% vs. 90.1%), 24 h (89.2% vs. 84.9%) and 48 h (50.0% vs. 0.0%), respectively.
  • The final cleavage rates of the fertilized oocytes were not different between the oocytes matured in these two media (Table 4; 97.3% vs. 98.9%), and there were also no differences in these two media when the oocytes were matured 6 h (100.0% vs. 98.4%) and 24 h (93.9% vs. 100.0%) respectively (FIG. 2).
  • There were no significant differences in the embryos that developed to the 8-cell stage between the oocytes matured in IVM-medium (63.7%) and TCM-199 (57.1%), and the percentages of embryos that developed to the 8-cell stage were also not different when the oocytes matured 6 h (66.7% vs. 60.3%), 24 h (54.6% vs. 50.0%) and 48 h (0.0% vs. 0.0%) in these two media, respectively (FIG. 3 a).
  • However, total blastocyst formation rates were significantly different (P<0.05) between the IVM-medium and TC-199 respectively (Table 4; 19.6% vs. 7.7%). Furthermore, the percentage of blastocyst formation was different (P<0.05) for these two media when the oocytes matured 6 h (24.0% vs. 7.9%) after maturation in culture (FIG. 3 b).
  • However, there were no differences between these two media when the oocytes matured 24 h (9.7% vs. 7.1%) and 48 h (0.0% vs. 0.0%), respectively.
  • As shown in Table 5, when GV stage oocytes were matured in vitro, there were significant differences (P<0.01) in the maturation rates between the IVM-medium and TCM-199 at 24 h (50.1% vs. 34.0%) and 48 h (75.7% vs. 55.7%) respectively.
  • However, as shown in Table 6, the final fertilization rates were not different between the IVM-medium (86.4%) and TCM-199 (78.0%).
  • There were also no differences in the fertilization rates between these two media when the oocytes were matured 24 h (88.9% vs. 83.3%) and 48 h (81.5% vs. 69.6%) (FIG. 4).
  • Final cleavage rates were not different between these two media (Table 6, 100.0% vs. 95.6%). The cleavage rates of the fertilized oocytes were also not different between these two media when the oocytes matured 24 h (100.0% vs. 96.7%) and 48 h (100.0% vs. 93.8%) (FIG. 5).
  • However, there were significant differences (P<0.05) in embryos that developed to the 8-cell stage and blastocyst stage between the IVM-medium and TCM-199 (Table 6, 58.6% vs. 40.9% and 12.9% vs. 0.0%, respectively).
  • When oocytes were matured for 24 h, percentages of embryos that developed to the 8-cell stage (FIG. 6 a; 75.0% vs. 51.7%) and the blastocyst stage (FIG. 6 b; 18.8% vs. 0.0%) were significantly different (P<0.05) between the IVM-medium and TCM-199, respectively.
  • However, there were no differences in the embryos that developed to the 8-cell stage (FIG. 6 a; 22.7% vs. 20.0%) and blastocyst (FIG. 6 b; 0.0% vs. 0.0%) between these two media when the oocytes were matured for 48 h.
  • EXAMPLE 2
  • A total of 40 women with PCOS underwent 40 completed IVM treatment cycles. All patients were under 40 years of age (mean 32.3±3.9) and some patients presented irregular menstrual cycles or anovulation. All patients had a minimum 2-year history of infertility.
  • If the patients had irregular menstrual cycles, the treatment cycles was initiated by the administration of intra-vaginal progesterone (Prometrium; Schering, Pointe-Clair, Quebec, Canada) in a dose of 300 mg daily for 10 days. Withdrawal bleeding occurred within 3 days after the last dose.
  • On day 2 or 3 following the onset of menstrual bleeding, the patients underwent a baseline ultrasound scan to ensure that no ovarian cysts were present. Transvaginal ultrasound scans were repeated on day 8 to exclude the development of a dominant follicle. The size of all follicles on ultrasound scan had to be <10 mm in diameter on day 8 of the cycle.
  • Oocyte retrieval was performed on day 10 to 14 of the cycle. Before oocyte collection 36 hours, the patients were given s.c. 10,000 IU of human chorionic gonadotropin (HCG) for priming.
  • Transvaginal ultrasound-guided oocyte collection was performed using a specially designed 19 G single-lumen aspiration needle (Cook, Australia) with an aspiration pressure of 7.5 kPa. Aspiration of all small follicles was performed under local anaesthesia. Oocytes were collected in 10 ml culture tubes (Falcon, USA) containing 2.0 ml warm 0.9% saline contained with 2 IU/ml heparin (Baxter, Toronto, Ontario, Canada).
  • Following oocyte collection, cumulus-oocyte complexes (COCs) were cultured in an organ tissue culture dish (60×15 mm; Falcon) containing 1 ml of modified IVM-medium (Table 2) at 37° C. in an atmosphere of 5% CO2 and 95% air with high humidity.
  • Following culture of 24 and 48 hours, the mature oocytes (metaphase-II stage) were inseminated by ICSI. Fertilization was assessed 18 h after ICSI for the appearance of two distinct pronuclei and two polar bodies. Embryo transfer was performed on day 2 or 3 after ICSI. Since the oocytes were inseminated either 24 or 48 hours following culture, the developmental stages of embryos were variable.
  • For the preparation of the endometrium, the patients were given estradiol (Estace; Roberts Pharmaceutical, Mississauga, Canada) depending on the endometrial thickness on the day of oocyte retrieval in divided doses, starting on the day of oocyte retrieval. If the endometrial thickness on the day of oocyte retrieval was <4 mm, a 10 mg dose was administered; if it was >6 mm, a 6 mg dose was given. Luteal support was provided by 400 mg intravaginal progesterone (Prometrium) twice daily for 16 days starting from the day after oocyte collection.
  • A total of 653 immature oocytes were collected from 40 women with PCOS who underwent IVM treatment. Following culture of 48 hours, 81.5% (532/653) of oocytes became mature. Following embryo transfer, 13 patients became pregnant (32.5%=13/40). Results are tabulated in Table 7.
    TABLE 1
    Composition of oocyte in vitro maturation (IVM) medium.
    mg/L
    Inorganic Salt:
    CaCl2 200.0000
    KCl 400.0000
    MgSO4 98.0000
    NaCl 6800.0000
    NaHCO3 1250.0000
    NaH2PO4.H2O 125.0000
    Amino Acids:
    L-Alanine 8.9000
    L-Arginine 126.4000
    L-Asparagine 13.2000
    L-Aspartic Acid 13.3000
    L-Cystine 24.0000
    L-Glutamic Acid 14.7000
    L-Glutamine 292.0000
    Glycine 7.5000
    L-Histidine.HCI.H2O 42.0000
    L-Isoleucine 52.4000
    L-Leucine 52.4000
    L-Lysine.HCI 72.5000
    L-Methionine 15.1000
    L-Phenylalanine 33.0000
    L-Proline 11.5000
    L-Serine 10.5000
    L-Threonine 47.6000
    L-Tryptophan 10.2000
    L-Tyrosine 36.0000
    L-Valine 46.8000
    Vitamins:
    Biotin 0.0100
    D-Ca Pantothenate 1.0000
    Choline Chloride 1.0000
    Folic Acid 1.0000
    i-Inositol 2.0000
    Nicotinamide 1.0000
    Pyridoxal.HCI 1.0000
    Riboflavin 0.1000
    Thiamine.HCI 1.0000
    Other Components:
    D-Glucose 1000.0000
    Sodium Pyruvate 110.0000
    Insulin 5.0000
    Human Transferrin 50.0000
    Selenite 0.0052
    Hydrocortisone 0.0036
    FGF 0.0005
    EGF 0.0010
    Phenol Red 5.0000
    Penicillin G 50.0000 units
    Streptomycin 50.0000 units
    FSH 0.075 IU/ml
    LH 0.075 IU/ml
    estradiol 1.0 μg/ml
  • TABLE 2
    Modified Composition of oocyte in vitro maturation (IVM) medium.
    mg/L
    Inorganic Salt:
    CaCl2 200.0000
    KCl 400.0000
    MgSO4 98.0000
    NaCl 6800.0000
    NaHCO3 1250.0000
    NaH2PO4.H2O 125.0000
    Amino Acids:
    L-Alanine 8.9000
    L-Arginine 126.4000
    L-Asparagine 13.2000
    L-Aspartic Acid 13.3000
    L-Cystine 24.0000
    L-Glutamic Acid 14.7000
    L-Glutamine 292.0000
    Glycine 7.5000
    L-Histidine.HCl.H2O 42.0000
    L-Isoleucine 52.4000
    L-Leucine 52.4000
    L-Lysine.HCl 72.5000
    L-Methionine 15.1000
    L-Phenylalanine 33.0000
    L-Proline 11.5000
    L-Serine 10.5000
    L-Threonine 47.6000
    L-Tryptophan 10.2000
    L-Tyrosine 36.0000
    L-Valine 46.8000
    Vitamins:
    D-Ca Pantothenate 1.0000
    Choline Chloride 1.0000
    Folic Acid 1.0000
    i-Inositol 2.0000
    Nicotinamide 1.0000
    Pyridoxal.HCl 1.0000
    Riboflavin 0.1000
    Thiamine.HCl 1.0000
    Other Components:
    D-Glucose 1000.0000
    Sodium Pyruvate 110.0000
    Estradiol 1.0000
    Phenol Red 5.0000
    FSH 75.0000 IU
    LH 75.0000 IU
    Human serum albumin 1000.0000
    Penicillin G 50.0000 units
    Streptomycin 50.0000 μg
  • TABLE 3
    Maturation of immature human oocytes (metaphase-I
    stage) cultured 6 h, 24 h or 48 h in the IVM-medium or
    TCM-199.
    Culture No. of oocytes No. of oocytes matured at (%):
    media examined 6 h 24 h 48 h
    IVM medium 154 80 (52.0) 117 (76.0) 121 (78.6)
    TCM-199 154 71 (46.1) 104 (67.5) 109 (70.8)
  • TABLE 4
    Fertilization and embryonic development of immature
    human oocytes (metaphase-I stage)
    matured in the IVM-medium or TCM-199.
    No. of No. of No. of embryos
    oocytes oocytes No. of oocytes developed to (%):
    Culture media inseminated fertilized (%) cleaved (%) 8-cell stage Blastocyst
    IVM medium 121 110 (90.0) 107 (97.3) 68 (63.6) 21 (19.6)a
    TCM-199 109  92 (84.4)  91 (98.9) 52 (57.1)  7 (7.7)b

    abDifferent letters indicate significant differences within columns (P < 0.05).
  • TABLE 5
    Maturation of immature human oocytes (germinal vesicle
    stage) cultured 24 h or 48 h in the IVM-medium or TCM-199.
    No. of oocytes No. of oocytes
    Culture No. of oocytes matured at 24 h matured at 48 h
    media examined (%) (%)
    IVM medium 107 54 (50.1)a 81 (75.7)a
    TCM-199 106 36 (34.0)b 59 (55.7)b

    abDifferent letters indicate significant differences within columns (P < 0.05).
  • TABLE 6
    Fertilization and embryonic development of immature human oocytes (germinal
    vesicle stage) matured in the IVM-medium or TCM-199.
    No. of No. of No. of embryos
    Culture oocytes oocytes No. of oocytes developed to (%):
    media inseminated fertilized (%) cleaved (%) 8-cell stage Blastocyst
    IVM medium 81 70 (86.4) 70 (100.0) 41 (58.6)a 9 (12.9)a
    TCM-199 59 46 (78.0) 44 (95.6) 18 (40.9)b 0 (0.0)b

    abDifferent letters indicate significant differences within columns (p < 0.05).
  • TABLE 7
    Results of in vitro maturation and fertilization of oocytes using
    modified IVM-medium followed by embryo transfer in women with
    polycystic ovaries (PCO) or polycystic overy syndrome (PCOS)
    (from January 2002 to September 2002 at McGill Reproductive Center).*
    Cycles  40
    Age - yrs 32.3 ± 3.9
    No. of oocytes collected
    Total 653
    Mean 15.9 ± 5.9
    No. of oocytes matured (%) 532 (81.5)
    No. of oocytes fertilized (%) 336 (63.2)
    No. of oocytes cleaved (%) 329 (97.9)
    No. of embryos transferred
    Total 154
    Mean  3.9 ± 0.6
    No. of clinical pregnancies (%)  13 (32.5)
    No. of implantation (%)  17 (11.0)

    *Plus-minus values are mean ± SD.
  • References
  • Adams J, Polson D, Frank S. Prevalence of polycystic ovaries in women with anovulation or idiopathic hirsutism. Br Med J 1986; 293:355-359.
  • Barnes F L, Crombie A, Gardner D K, Kausche A, Lacham-Kaplan O, Suikkari A A, Tiglias J, Wood C, Trounson A O. Blastocyst development and birth after in vitro maturation of human primary oocytes, intracytoplasmic sperm injection and assisted hatching. Hum Reprod 1995; 10:3243-3247.
  • Brackett B G, Zuelke K A. Analysis of factors involved in the in vitro production of bovine embryos. Theriogenology 1993; 39:43-64.
  • Cha K Y, Koo J J, Ko J J, Choi D H, Han S Y, Yoon T K. 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 1991; 55:109-113.
  • Cha K Y, Chian R C. Maturation in vitro of immature human oocytes for clinical use. Hum Reprod Update; 4:103-120.
  • Chian R C, Niwa K, Sirard M A. Effects of cumulus cells on male pronuclear formation and subsequent early development of bovine oocytes in vitro. Theriogenology 1994; 41:1499-1508.
  • Chian R C, Sirard M A. Effects of cumulus cells and follicle-stimulating hormone during in vitro maturation on parthenogenetic activation of bovine oocytes. Mol Reprod Dev 1995; 42:425-431.
  • Chian R C, Park S E, Park E H, Son W Y, Chung H M, Lim J G, Ko J J, Cha K Y. Molecular and structural characteristics between immature human oocytes retrieved from stimulated and unstimulated ovaries. Gormel V, Leung P C K (eds), In Vitro Fertilization and Assisted Reproduction; Monduzzi, Bologna, pp 315-319.
  • Chian R C, Buckett W M, Too L L, Tan S L. Pregnancies resulting from in vitro matured oocytes retrieved from patients with polycystic ovary syndrome after priming with human chorionic gonadotropin. Fertil Steril 1999; 72:639-642.
  • Chian R C, Gulekli B, Buckett W M, Tan S L. Priming with human chorionic gonadotropin before retrieval of immature oocytes in women with infertility due to the polycystic ovary syndrome. New Eng J Med 1999; 341:1624-1626.
  • Chian R C, Ao A, Clarke H J, Tulandi T, Tan S L. Production of steroids from human cumulus cells treated with different concentrations of gonadotropins during culture in vitro. Fertil Steril 1999; 71:61-66.
  • Chian R C, Buckett W M, Tulandi T, Tan S L. Prospective randomized study of human chorionic gonadotrophin priming before immature oocyte retrieval from unstimulated women with polycystic ovarian syndrome. Hum Reprod 2000; 15:165-170.
  • Chian R C, Gulekli B, Buckett W M, Tan S L. Pregnancy and delivery after cryopreservation of zygotes produced by in-vitro matured oocytes retrieved from a woman with polycystic ovarian syndrome. Hum Reprod 2001; 16:1700-1702.
  • Edirisinghe W R, Junk S M, Matson P L, Yovich J L. Birth from cryopreserved embryos following in vitro maturation of oocytes and intracytoplasmic sperm injection. Hum Reprod 1997; 12:1056-1058.
  • Goud P T, Goud A P, Qian C, Laverge H, Van der Elst J, De Sutter P, Dhont M. In-vitro maturation of human germinal vesicle stage oocytes: role of cumulus cells and epidermal growth factor in the culture medium. Hum Reprod 1998; 13:1638-1644.
  • Gross-Weege W, Theobald K, Konig W. Inhibition of histamine release from rat peritoneal mast cells by a factor from human serum: identification as transferring. Agents Actions 1986; 19:10-17.
  • Jaroudi K A, Hollanders J M G, Elnour A M, Roca G L, Atared A M, Coskun S. Embryo development and pregnancies from in vitro matured and fertilized human oocytes. Hum Reprod 1999; 14:1749-1751.
  • MacDougal M J, Tan S L, Balen A, et al. A controlled study comparing patients with and without polycystic ovaries undergoing in vitro fertilization. Hum Reprod 1993; 8:233-237.
  • Maruo T, Ladines-Llave C A, Samoto T, Matsuo H, Manalo A S, Ito H, Mochizuki M. Expression of epidermal growth factor and its receptor in the human ovary during follicular growth and regression. Endocrinology 1993; 132:924-931.
  • Moor R M, Trounson A O. Hormonal and follicular factors affecting maturation of sheep oocytes in vitro and their subsequent developmental capacity. J Reprod Fertil 1977; 49:101-109.
  • Nagy Z P, Cecile J, Liu J, Loccuifer A, Devroey P, Van Steirteghem A. Pregnancy and birth after intracytoplasmic sperm injection of in vitro matured germinal vesicle stage oocytes: case report. Fertil Steril 1996; 65:1047-1050.
  • Pincus G, Enzmann E V. The comparative behaviour of mammalian eggs in vivo and in vitro. 1. The activation of ovarian eggs. J Exp Med 1935; 62:665-675.
  • Shea B F, Baker R D, Latour J P. Human follicular oocytes and their maturation in vitro. Fertil Steril 1975; 26:1075-1082.
  • Steel R G D, Torrie J H. Principles and procedures of statistics: a biometrical approach. New York, McGraw-Hill, 1980; ppl72-194.
  • Trounson A, Wood C, Kausche A. In vitro maturation and fertilization and developmental competence of oocytes recovered from untreated polycystic ovarian patients. Fertil Steril 1994; 62:353-362.
  • Trounson A O, Anderiesz C, Jones G M, Kausche A, Lolatgis N, Wood C. Oocyte maturation. Hum Reprod 1998; 13:52-62.
  • Trounson A, Anderiesz C, Jones G. Maturation of human oocytes in vitro and their developmental competence. Reproduction 2001; 121:51-75.
  • All references cited in this specification are herein incorporated by reference as if each individual reference were specifically and individually indicated to be incorporated by reference. The citation of any reference is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such reference by virtue of prior invention.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims (68)

1. A method for in vitro maturation of immature human oocytes, comprising the steps of:
(a) inducing in a female human subject an increase in endogenous luteinizing hormone levels, said subject having not undergone an ovarian stimulation protocol prior to said inducing step;
(b) obtaining from said subject an immature oocyte; and
(c) culturing said oocyte until maturity.
2. The method according to claim 1, wherein step (a) comprises administering to said subject human chorionic gonadotrophin or luteinizing hormone, or both.
3. The method according to claim 1, wherein step (a) comprises administering to said subject human chorionic gonadotropin in an amount of about 5000 to about 20,000 IU.
4. The method according to claim 1, wherein said immature human oocyte comprises an M-I stage ooctye.
5. The method according to claim 1, wherein said immature human oocyte comprises a GV stage oocyte.
6. The method according to claim 1, wherein said immature human oocyte is cultured until it reaches M-II.
7. The method according to claim 1, wherein, in step (c), said immature human oocyte is essentially free of cumulus cells and is cultured in a culture medium comprising:
at least one inorganic salt;
essential amino acids or a source thereof;
an energy source; and
at least one growth factor.
8. The method according to claim 7, wherein said at least one growth factor is selected from the group consisting of fibroblast growth factor and epidermal growth factor.
9. The method according to claim 7, wherein said culture medium comprises both fibroblast growth factor and epidermal growth factor.
10. The method according to claim 7, wherein said culture medium further comprises at least one hormone.
11. The method according to claim 10, wherein said hormone comprises insulin.
12. The method according to claim 11, wherein said culture medium comprises from 0.5 mg/L to 50 mg/L insulin.
13. The method according to claim 7, wherein said culture medium further comprises human transferrin.
14. The method according to claim 13, wherein said culture medium comprises from 5 mg/L to 500 mg/L human transferrin.
15. The method according to claim 7, wherein said culture medium comprises from 0.0001 mg/L to 0.001 mg/L fibroblast growth factor.
16. The method according to claim 7, wherein said culture medium comprises from 0.0001 to 0.01 mg/L epidermal growth factor.
17. The method according to claim 7, wherein said culture medium comprises one or more vitamins.
18. The method according to claim 17, wherein said one or more vitamins comprise biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal.HCl, riboflavin, and thiamine-HCl.
19. The method according to claim 7, wherein said culture medium comprises hydrocortisone.
20. The method according to claim 7, wherein said culture medium comprises selenite.
21. The method according to claim 7, wherein said inorganic salts comprise CaCl2, KCl, MgSO4, NaCl, NaHCO3, NaH2PO4.H20.
22. The method according to claim 7, wherein said energy source comprises D-glucose, or sodium pyruvate, or both D-glucose and sodium pyruvate.
23. The method according to claim 7, wherein said amino acids comprise alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
24. The method according to claim 7, wherein said culture medium comprises the inorganic salts, amino acids, vitamins and other components as set forth in Table 1, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume) of the amount specified in Table 1.
25. The method according to claim 24, wherein each inorganic salt, amino acid, vitamin and other component is present in said culture medium in an amount of ±10% (weight/volume) of the amount specified in Table 1.
26. The method according to claim 1, wherein, in step (c), said oocyte has a cumulus that is intact or at least partially intact, and/or said oocyte is cultured in the presence of cumulus cells.
27. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of epidermal growth factor.
28. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of fibroblast growth factor.
29. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of human transferrin.
30. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of insulin.
31. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of selenite.
32. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of hydrocortisone.
33. The method according to claim 26, wherein said oocyte is cultured in a culture medium that is essentially free of epidermal growth factor, fibroblast growth factor, human transferrin, insulin, selenite, and hydrocortisone.
34. The method according to claim 26, wherein said oocyte is cultured in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume) of the amount specified in Table 2.
35. The method according to claim 34, wherein each inorganic salt, amino acid, vitamin and other component is present in said culture medium in an amount of ±10% (weight/volume) of the amount specified in Table 2.
36. The method according to claim 26, wherein said oocyte is cultured in a culture medium consisting essentially of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
37. The method according to claim 26, wherein said oocyte is cultured in a culture medium consisting of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
38. The method according to claim 1, wherein, prior to step (a), said subject has not been treated with a gonadotrophin releasing hormone agonist, human menopausal gonadotrophin or follicle stimulating hormone.
39. A method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising:
at least one inorganic salt;
essential amino acids or a source thereof;
an energy source; and
at least one growth factor.
40. The method according to claim 39, wherein said at least one growth factor is selected from the group consisting of fibroblast growth factor and epidermal growth factor.
41. The method according to claim 39, wherein said culture medium comprises both fibroblast growth factor and epidermal growth factor.
42. The method according to claim 39, wherein said culture medium further comprises at least one hormone.
43. The method according to claim 42, wherein said hormone comprises insulin.
44. The method according to claim 43, wherein said culture medium comprises from 0.5 mg/L to 50 mg/L insulin.
45. The method according to claim 39, wherein said culture medium further comprises human transferrin.
46. The method according to claim 45, wherein said culture medium comprises from 5 mg/L to 500 mg/L human transferrin.
47. The method according to claim 39, wherein said culture medium comprises from 0.0001 mg/L to 0.001 mg/L fibroblast growth factor.
48. The method according to claim 39, wherein said culture medium comprises from 0.0001 to 0.01 mg/L epidermal growth factor.
49. The method according to claim 39, wherein said culture medium comprises one or more vitamins.
50. The method according to claim 49, wherein said one or more vitamins comprise biotin, D-Ca pantothenate, choline chloride, folic acid, i-inositol, nicotinamide, pyroxidal HCl, riboflavin, and thiamine-HCl.
51. The method according to claim 39, wherein said culture medium comprises hydrocortisone.
52. The method according to claim 39, wherein said culture medium comprises selenite.
53. The method according to claim 39, wherein said inorganic salts comprise CaCl2, KCl, MgSO4, NaCl, NaHCO3, NaH2PO4 .H2O.
54. The method according to claim 39, wherein said energy source comprises D-glucose, or sodium pyruvate, or both D-glucose and sodium pyruvate.
55. The method according to claim 39, wherein said amino acids comprise alanine, arginine, asparagine, aspartic acid, cystine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
56. The method according to claim 39, wherein said culture medium comprises the inorganic salts, amino acids, vitamins and other components as set forth in Table 1, and wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume) of the amount specified in Table 1.
57. The method according to claim 55, wherein each inorganic salt, amino acid, vitamin and other component is present in said culture medium in an amount of ±10% (weight/volume) of the amount specified in Table 1.
58. The method according to claim 39, wherein said culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components as set forth in Table 1.
59. The method according to claim 39, wherein said culture medium consists of the inorganic salts, amino acids, vitamins and other components as set forth in Table 1.
60. The method according to claim 57 or 58, wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume) of the amount specified in Table 1.
61. The method according to claim 57 or 58, wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±10% (weight/volume) of the amount specified in Table 1.
62. The method according to claim 39, wherein said immature human oocyte is essentially free of cumulus cells.
63. A method for in vitro maturation of immature human oocytes, comprising culturing an immature human oocyte in a culture medium comprising the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
64. The method according to claim 62, wherein said culture medium consists essentially of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
65. The method according to claim 62, wherein said culture medium consists of the inorganic salts, amino acids, vitamins and other components as set forth in Table 2.
66. The method according to claim any one of claims 62-64, wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±50% (weight/volume) of the amount specified in Table 2.
67. The method according to any one of claims 62-64, wherein each inorganic salt, amino acid, vitamin and other component is present in an amount of ±10% (weight/volume) of the amount specified in Table 2.
68. The method according to claim 62, wherein said oocyte is cultured in the presence of cumulus cells
US10/506,713 2002-03-08 2003-03-07 Vitro maturation of immature human oocytes Abandoned US20050208651A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/506,713 US20050208651A1 (en) 2002-03-08 2003-03-07 Vitro maturation of immature human oocytes
US11/969,522 US7790459B2 (en) 2002-03-08 2008-01-04 In vitro maturation of immature human oocytes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36239502P 2002-03-08 2002-03-08
US10/506,713 US20050208651A1 (en) 2002-03-08 2003-03-07 Vitro maturation of immature human oocytes
PCT/CA2003/000323 WO2003076600A2 (en) 2002-03-08 2003-03-07 In vitro maturation of immature human oocytes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/969,522 Division US7790459B2 (en) 2002-03-08 2008-01-04 In vitro maturation of immature human oocytes

Publications (1)

Publication Number Publication Date
US20050208651A1 true US20050208651A1 (en) 2005-09-22

Family

ID=27805171

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/506,713 Abandoned US20050208651A1 (en) 2002-03-08 2003-03-07 Vitro maturation of immature human oocytes
US11/969,522 Expired - Lifetime US7790459B2 (en) 2002-03-08 2008-01-04 In vitro maturation of immature human oocytes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/969,522 Expired - Lifetime US7790459B2 (en) 2002-03-08 2008-01-04 In vitro maturation of immature human oocytes

Country Status (6)

Country Link
US (2) US20050208651A1 (en)
EP (1) EP1483368A2 (en)
JP (1) JP2005518823A (en)
AU (1) AU2003208232B2 (en)
CA (1) CA2477743C (en)
WO (1) WO2003076600A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110709505A (en) * 2017-04-12 2020-01-17 派乔易股份有限公司 Cytokine-free adjuvants, in particular for in vitro fertilization or for cell culture media for follicles, male germ cells or embryos
CN114149962A (en) * 2021-11-25 2022-03-08 艾尔斯(浙江)医学科技有限公司 Human oocyte in vitro fertilization pre-culture method, series culture solution and application

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1697504T3 (en) * 2003-11-26 2011-11-14 Merck Serono Sa Use of IL-6 type cytokines to mature oocytes
AU2005292362B2 (en) * 2004-09-30 2012-05-31 Merck Serono Sa Use of IL-17- for maturation of oocytes
JP2008514239A (en) * 2004-09-30 2008-05-08 アプライド リサーチ システムズ エーアールエス ホールディング ナームロゼ フェンノートシャップ Use of pregnancy-specific glycoprotein for oocyte maturation
US9566256B2 (en) 2008-09-22 2017-02-14 Biochemics, Inc. Transdermal drug delivery using an osmolyte and vasoactive agent
US9278233B2 (en) 2008-12-04 2016-03-08 Biochemics, Inc. Methods and compositions for tattoo removal
AT510810B1 (en) * 2010-12-09 2017-09-15 Gonadosan Gmbh COMBINATION PROCEDURE FOR IMPROVING FEMALE FERTILITY
BR112018068474B1 (en) * 2016-02-24 2024-01-30 Universitá Degli Studi Di Milano, Unimi FOLLICULAR SYSTEM FOR IN VITRO OOCYTE MATURATION AND KIT
FR3061208B1 (en) * 2016-12-23 2024-04-05 Patrick Choay Sas ADJUVANTS FOR CELL CULTURE MEDIA, PARTICULARLY FOR IN VITRO FERTILIZATION, OR FOR THE CULTURE OF FOLLICLES, MALE GERM CELLS OR EMBRYOS
CN110643569B (en) * 2019-11-29 2020-05-26 广州达瑞生殖技术有限公司 Granular cell stripping liquid and preparation method thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4987080A (en) * 1987-11-03 1991-01-22 Grob Howard S Method for in vitro maturation of oocytes
US5563059A (en) * 1993-02-23 1996-10-08 Genentech, Inc. Use of human inhibin and human activin to increase the number of mature primate oocytes
US5882928A (en) * 1997-03-11 1999-03-16 Oocytechs Research Corporation In vitro maturation and fertilization of mammalian oocytes
US6211429B1 (en) * 1997-06-18 2001-04-03 The Curators Of The University Of Missouri Complete oocyte activation using an oocyte-modifying agent and a reducing agent
US6281013B1 (en) * 1999-02-24 2001-08-28 Novo Nordisk A/S Treatment of Infertility
US20010028878A1 (en) * 1998-06-22 2001-10-11 Svend Lindenberg In vitro maturation of human oocytes in one day

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL94184A0 (en) 1989-05-01 1991-01-31 Granada Biosciences Inc In vitro maturation of bovine oocytes
PT723453E (en) 1993-08-03 2000-07-31 Celtrix Pharma METHOD OF TREATMENT OF REPRODUCTIVE DISORDERS
TR200101538T2 (en) 1998-11-30 2001-12-21 Ivf Sciences Colorado, Inc. In vitro fertilization system and sequential culture medium.
BR0008468A (en) 1999-02-24 2002-02-05 Novo Nordisk As Method for human in vitro fertilization, use of a hypothalamic hormone and / or pituitary hormone or an agonist or antagonist of the same or an active derivative thereof, pharmaceutical kit in single dosage form, and any new aspect or combination of aspects
JP2001017160A (en) 1999-07-07 2001-01-23 Fuso Pharmaceutical Industries Ltd Medium composition for in vitro fertilization
AU1692901A (en) 1999-11-25 2001-06-04 Novo Nordisk A/S Treatment of human infertility
AU2001248282A1 (en) 2000-04-06 2001-10-23 Novo-Nordisk A/S Synchronization of the cytoplasmatic and the nuclear maturation of oocytes in vitro

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4987080A (en) * 1987-11-03 1991-01-22 Grob Howard S Method for in vitro maturation of oocytes
US5563059A (en) * 1993-02-23 1996-10-08 Genentech, Inc. Use of human inhibin and human activin to increase the number of mature primate oocytes
US5882928A (en) * 1997-03-11 1999-03-16 Oocytechs Research Corporation In vitro maturation and fertilization of mammalian oocytes
US6211429B1 (en) * 1997-06-18 2001-04-03 The Curators Of The University Of Missouri Complete oocyte activation using an oocyte-modifying agent and a reducing agent
US20010028878A1 (en) * 1998-06-22 2001-10-11 Svend Lindenberg In vitro maturation of human oocytes in one day
US20020115211A1 (en) * 1998-06-22 2002-08-22 Medi-Cult A/S A method for in vitro maturation of human gametes
US6281013B1 (en) * 1999-02-24 2001-08-28 Novo Nordisk A/S Treatment of Infertility

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110709505A (en) * 2017-04-12 2020-01-17 派乔易股份有限公司 Cytokine-free adjuvants, in particular for in vitro fertilization or for cell culture media for follicles, male germ cells or embryos
CN114149962A (en) * 2021-11-25 2022-03-08 艾尔斯(浙江)医学科技有限公司 Human oocyte in vitro fertilization pre-culture method, series culture solution and application

Also Published As

Publication number Publication date
AU2003208232B2 (en) 2008-05-22
WO2003076600A2 (en) 2003-09-18
EP1483368A2 (en) 2004-12-08
US7790459B2 (en) 2010-09-07
JP2005518823A (en) 2005-06-30
US20080176324A1 (en) 2008-07-24
AU2003208232A1 (en) 2003-09-22
WO2003076600A3 (en) 2004-02-05
CA2477743A1 (en) 2003-09-18
CA2477743C (en) 2011-11-08

Similar Documents

Publication Publication Date Title
US7790459B2 (en) In vitro maturation of immature human oocytes
Canipari Oocyte–granulosa cell interactions
Chian et al. Maturational and developmental competence of cumulus-free immature human oocytes derived from stimulated and intracytoplasmic sperm injection cycles
Bevers et al. Regulation and modulation of oocyte maturation in the bovine
Wynn et al. Pretreatment with follicle stimulating hormone promotes the numbers of human oocytes reaching metaphase II by in-vitro maturation.
Mermillod et al. Aspects of follicular and oocyte maturation that affect the developmental potential of embryos
Kim et al. In vitro maturation, fertilization, and development of human germinal vesicle oocytes collected from stimulated cycles
Sirisathien et al. Influences of epidermal growth factor and insulin-like growth factor-I on bovine blastocyst development in vitro
Luvoni et al. Factors involved in vivo and in vitro maturation of canine oocytes
Rodrigues et al. Embryonic development of in vitro matured and in vitro fertilized dog oocytes
Jaroudi et al. Pregnancy after transfer of embryos which were generated from in-vitro matured oocytes.
Smitz et al. Oocyte in-vitro maturation and follicle culture: current clinical achievements and future directions
AU2017371402B2 (en) Compositions and methods for maturation of oocytes in vitro
Tibary et al. Update on reproductive biotechnologies in small ruminants and camelids
Salha et al. Dynamics of human follicular growth and in-vitro oocyte maturation
JP2011507530A (en) Protein-free gamete and embryo handling media products and culture media products
Chian et al. In vitro maturation of immature human oocytes for clinical application
Trounson The production of ruminant embryos in vitro
Mikkelsen et al. Maternal serum supplementation in culture medium benefits maturation of immature human oocytes
KR20190052542A (en) A follicular fluid replacement medium for in vitro Maturation of oocytes and The Use thereof
Huang et al. Biochemical compositions of follicular fluid and the effects of culture conditions on the in vitro development of pig oocytes
US20010028878A1 (en) In vitro maturation of human oocytes in one day
Jee et al. Influence of well defined protein source on in vitro maturation of human oocyte: human follicular fluid versus human serum albumin
Heng et al. Effects of granulosa coculture on in-vitro oocyte meiotic maturation within a putatively less competent murine model
Heng et al. Investigations of oocyte in vitro maturation within a mouse model

Legal Events

Date Code Title Description
AS Assignment

Owner name: MCGILL UNIVERSITY, CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHIAN, RI-CHANG;REEL/FRAME:015269/0822

Effective date: 20030407

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION