fertility preservation among the cancer patients by ovarian tissue cryopreservation, transplantation, and follicular development

نویسندگان

ali abedelahi dept. of anatomical sciences, tabriz university of medical sciences, tabriz, iran

mostafa rezaei-tavirani proteomics research center, shahid beheshti university of medical sciences, tehran, iran

daryosh mohammadnejad dept. of anatomical sciences, tabriz university of medical sciences, tabriz, iran

چکیده

abstract ovarian tissue freezing or cryopreservation might be the only acceptable method for preserving the young women fertility, before radiotherapy or chemotherapy. this technology might be used for patients with recurrent ovarian cysts or endometriosis, without ovarian stimulation. many efforts have made to improve cryopreservation conditions that should be seriously considered for cancer patients. vitrification is a process which prevents ovarian tissue from cryo damage, then preserves cell viability. both methods have used for evaluating not only the follicular development, but also the fertility after freezing and thawing. in this manuscript, we have discussed the techniques of ovarian tissue vitrification, then graft and maturation or follicular development is also mentioned. please cite this article as: abedelahi a, rezaei-tavirani m, mohammadnejad d. fertility preservation among the cancer patients by ovarian tissue cryopreservation, transplantation, and follicular development. iran j cancer prev. 2013; 6(3):123-32. references 1. tauchmanova l, selleri c, de rosa g, pagano l, orio f, lombardi g, et al. high prevalence of endocrine dysfunction in longterm survivors after allogeneic bone marrow transplantation for hematologic diseases. cancer. 2002; 95: 1076-84. 2. sonmezer m, oktay k. fertility preservation in female patients. hum reprod update. 2004; 10: 251-66. 3. mohammadnejad d, abedelahi a, soleimani-rad j, mohamadi-roshandeh a, rashtbar m, azami a. degenerative effect of cisplatin on testicular germinal epithelium. apb. 2012; 2: 173-7. 4. mohammadnejad d, abedelahi a, rashtbar m. protective role of gnrh antagonist on chemotherapyinduced spermatogenesis disorder: a morphological study. apb. 2013 (in press). 5. wallace wh, anderson ra, irvine ds. fertility preservation for young patients with cancer: who is at risk and what can be offered? lancet oncol. 2005; 6: 209- 18. 6. sanchís blanco g, andrés moreno m, gregoraci af, cortés sáez j, marco macián a, rubio rubio jm, et al. ovarian cryopreservation in girls with cancer: new challenges. cir pediatr. 2011; 24: 196-200. 7. amorim ca, goncalves pb, figueiredo jr. cryopreservation of oocytes from preantral follicles. hum reprod update. 2003; 9: 119-29. 8. ozkavukcu s, erdemli e. cryopreservation: basic knowledge and biophysical effects. j ankara med school. 2002; 24: 187-96. 9. deanesly r. immature rat ovaries grafed after freezing and thawing. j endocrinol. 1954; 11: 197-200. 10. mazur p. freezing of living cell: mechanisms and implication. am j physipol. 1984; 247: 125-42. 11. santos rr, tharasanit t, van haeften t, figueiredo jr, silva jr, van den hurk r. vitrification of goat preantral follicles enclosed in ovarian tissue by using conventional and solid-surface vitrification methods. cell tissue res. 2007; 327: 167-76. 12. fuller b, paynter s. fundamental of cryobiology in reproductive medicine. reprod biomed online. 2004; 9: 680-91. 13. migishima f, suzuki-migishima r, song sy, kuramochi t, azuma s, nishijima m, et al. successful cryopreservation of mouse ovaries by vitrification. biol reprod. 2003; 68: 881-7. 14. polge c, smith au, parkes as. revival of spermatozoa after vitrification and dehydration at low temperatures. nature. 1949; 164: 666-76. 15. polge c, lovelock je. preservation of bull sperm at - 70ºc. vet rec. 1952; 64: 296-7. 16. santos rr, rodrigues ap, costa sh, silva jr, matos mh, lucci mh, et al. histological and ultrastractural analysis of cryopreserved sheep preantral follicles. anim reprod sci. 2006; 91: 249-63. 17. yamada c, caetano hv, simoes r, nicacio ac, feitosa wb, assumpcao me, et al. immature bovine oocyte cryopreservation: comparison of different associations with ethylene glycol, glycerol and dimethylsulfoxide. anim reprod sci. 2007; 99: 384-8. 18. abedelahi a, salehnia m, allameh aa, hajizadeh h. comparison of ultrastructure and morphology of mouse ovarian follicles after conventional and direct cover vitrification using different concentrations of ethylene glycol. ijrm iran j reprod med. 2009; 7: 45-52. 19. isachenko v, isachenko e, rahimi g, krivokharchenko a, alabart jl, nawroth. cryopreservation of human ovarian tissue by direct plunging into liquid nitrogen: negative effect of disaccharides in vitrification solution. cryo letters. 2002; 23: 333-44. 20. gosden rg, baird dt, wade jc, webb r. restoration of fertility to oophorectomised sheep by ovarian autografts stored at -196°c. hum reprod. 1994; 9: 597-603. 21. oktay k, buyuk e, veeck l, zaninovic n, xu k, takeuchi t, et al. embryo development after heterotopic transplantation of cryopreserved ovarian tissue. lancet. 2004; 363: 837-40. 22. donnez j, dolmans mm, demylle d, jadoul p, pirard c, squifflet j, et al. livebirth after orthotopic transplantation of cryopreserved ovarian tissue. lancet. 2004; 364: 1405-10. 23. vajta g, holm p, kuwayama m, booth pj, jacobsen h, greve t, et al. open pulled straw (ops) vitrification: a new way to reduce cryoinjuries of bovine ova and embryos. mol reprod dev. 1998; 51: 53-8. 24. wani na, maurya sn, misra ak, saxena vb, lakhchaura bd. effect of cryoprotectants and their concentration on in vitro development of vitrified-warmed immature oocytes in buffalo (bubalus bubalis). theriogenology. 2004; 61: 831-42. 25. schiewe mc, rall wf, stuart ld, wildt de. analysis of cryoprotectant, cooling rate and in situ dilution using conventional freezing or vitrification for cryopreserving sheep embryos. theriogenology. 1991; 36: 279-93. 26. sugimoto m, maeda s, manabe n, miyanoto h. development of infantile rat ovaries autotransplanted after cryopreservation by vitrification. theriogenology. 2000; 53: 1093-103. 27. courbiere b, odagescu v, baudot a, massardier j, mazoyer c, salle b, et al. cryopreservation of the ovary by vitrification as an alternative to slow-cooling protocols. fertil steril. 2006; 86: 1243-51. 28. li yb, zhou cq, yang gf, wang q, dong y. modified vitrification method for cryopreservation of human ovarian tissues. chin med j. 2007; 120: 110-4. 29. mazoochi t, salehnia m, valojerdi mr, mowla sj. morphologic, ultrastructural, and biochemical identification of apoptosis in vitrified-warmed mouse ovarian tissue. fertil steril. 2008; 90: 1480-6. 30. tirelli m, basini g, grasselli f, bianco f, tamanini c. cryopreservation of pig granulosa cells: effect of fsh addition to freezing medium. domest anim endocrinol. 2005; 28: 17-33. 31. abedelahi a, salehnia m, allameh aa, davoodi d. sodium selenite improves the in vitro follicular development by reducing the reactive oxygen species fertility preservation among the cancer patients by ovarian tissue... vol 6, no 3, summer 2013 131 level and increasing the total antioxidant capacity and glutathione peroxide activity. hum reprod. 2010; 25: 977-85. 32. hani t, tachibe t, shingai s, kamada n, ueda o, jishage k. fertility of mice receiving vitrified adult mouse ovaries. reproduction. 2006; 131: 681-7. 33. martino a, pollard jw, leibo sp. effect of chilling bovine oocytes on their developmental competence. mol reprod dev. 1996; 45: 503-12. 34. cai xy, chen ga, lian y, zheng xy, peng hm. cryoloop vitrification of rabbit oocytes. hum reprod. 2005: 20: 1969-74. 35. chen su, lien yl, chao kh, lu hf, ho hn, yang ys. cryopreservation of mature human oocytes by vitrification with ethylene glycol in straws. fertil steril. 2000; 74; 804- 48. 36. chen su, chien cl, wu my. novel direct cover vitrification for cryopreservation of ovarian tissues increases follicle viability and pregnancy capability in mice. hum reprod. 2006; 21: 2794-800. 37. whittingham dg, leibo sp, mazur p. survival of mouse embryos frozen to -196 and -269 ºc. science. 1972; 178: 411-4. 38. el danasouri i, selman h. successful pregnancies and deliveries after a simple vitrification protocol for day 3 human embryos. fertil steril. 2001; 76: 400-2. 39. van der zwalmen p, bertin g, debauche ch, standaert v, van roosendaal e, vandervorst m, et al. births after vitrification at morula and blastocyst stages: effect of artificial reduction of the blastocoelic cavity before vitrification. hum reprod. 2002; 17: 744-51. 40. chen c. pregnancy after human oocyte cryopreservation. lancet. 1986; 1: 884-6. 41. borini a, sciajno r, bianchi v, sereni e, flamigni c, coticchio g. clinical outcome of oocyte cryopreservation after slow cooling with a protocol utilizing a high sucrose concentration. hum reprod. 2006; 21:512-7. 42. fabbri r, porcu e, marsella t, rocchetta g, venturoli s, flamigni c. human oocyte cryopreservation: new perspectives regarding oocyte survival. hum reprod. 2001; 16: 411-6. 43. mandelbaum j, anastasiou o, lévy r, guérin jf, de larouzière v, antoine jm. effects of cryopreservation on the meiotic spindle of human oocytes. eur j obstet gynecol reprod biol. 2004; 113s: s17-s23. 44. ghetler y, yavin s, shalgi r , arav a. the effect of chilling on membrane lipid phase transition in human oocytes and zygotes. hum reprod. 2005; 20: 3385-9. 45. boiso i, marti m, santalo j, ponsa m, barri pn, veiga a. a confocal microscopy analysis of the spindle and chromosome configurations of human oocytes cryopreserved at the germinal vesicle and metaphase ii stage. hum reprod. 2002; 17: 1885-91. 46. levi setti pe, albani e, novara pv, cesana a, morreale g. cryopreservation of supernumerary oocytes in ivf/icsi cycles. hum reprod. 2006; 21: 370-5. 47. sheikhi m, hultenby k, niklasson b, lundqvist m, hovatta o. clinical grade vitrification of human ovarian tissue: an ultrastrctural analysis of follicles and stroma in vitrified tissue. hum reprod. 2011; 26: 594-603. 48. labied s, delforge y, munaut c, blacher s, colige a, delcombel r, et al. ovarian tissue cryopreservation by vitrification in olive baboons (papio anubis): a pilot study. transplantation. 2013; 95: 426-33. 49. meirow d, levron j, eldar-geva t, harden i, fridman e, zalel y, et al. pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. n engl j med. 2005; 353: 318-21. 50. callejo j, salvador c, gonzález-nuñez s, almeida l, rodriguez l, marqués l, et al. live birth in a woman without ovaries after autograft of frozen-thawed ovarian tissue combined with growth factors. j ovarian res. 2013; 61: 33. 51. parkes as, smith au. regeneration of rat ovarian tissue grafted after exposure to low temperatures. proc r soc. 1953; 140: 455-70. 52. candy cj, wood mj, whittingham dg. follicular development in cryopreserved marmoset ovarian tissue after transplantation. hum reprod. 1995; 10: 2334-8. 53. zhang j, dimattina m. extracorporeal development and ultrarapid freezing of human fetal ovary. j assist reprod genet. 1995; 912: 361-8. 54. newton h, aubard y, rutherford a, sharma v, gosden r. low temperature storage and grafting of human ovarian tissue. hum reprod. 1996; 11: 1487-91. 55. banu demirci b, lornage j ,salle b, poirel mt, guerin jf, franck m. the cryopreservation of ovarian tissue: uses and indications in veterinary medicine. theriogenology. 2003; 60: 999-1010. 56. deng x, zheng h, yu x, yu h, zhang c, chao l, et al. cryopreserved ovarian tissue can maintain a long term function after heterotopic autotransplantation in rat. reproduction. 2009; 138: 519-23. 57. amorim ca, jacobs s, devireddy rv, van langendonckt a, vanacker j, jaeger j, et al. successful vitrification and autografting of baboon (papio anubis) ovarian tissue. hum reprod. 2013 apr 16. [epub ahead of print] 58. yang hy, cox sl, jenkin g, findlay j, trounson a, shaw j. graft site and gonadotrophin stimulation influences the number and quality of oocytes from murine ovarian tissue grafts. reproduction. 2006; 131: 851-9. 59. dath c, van eyck as, dolmans mm, romeu l, delle vigne l, donnez j. xenotransplantation of human ovarian tissue to nude mice: comparison between four grafting sites. hum reprod. 2010; 25:1734-43. 60. li f, tao y, zhang y, li y, fang f, liu y, et al. follicle growth and oocyte development after ovary transplantation into back muscle of immune-intact adult castrated male mice. reproduction. 2010; 140: 465-76. 61. maltaris t, kayda h, hoffmann i, mueller a, beckmann mw, dittrich r. comparison of xenografting in scid mice and live/dead assay as a predictor of the developmental potential of cryopreserved ovarian tissue. in vivo. 2006; 20: 11-6. 62. schönfeldt v, chandolia r, kiesel l, nieschlag e, schlatt s, sonntag b. advanced follicle development in enografted prepubertal ovarian tissue: the common abedelahi et al. iranian journal of cancer prevention 132 marmost as a nonhuman primate model for ovarian tissue transplantation. fertil steril. 2011; 95: 1428-34. 63. amorim ca, dolmans mm, david a, jaeger j, vanacker j, camboni a, et al. vitrification and xenografting of human ovarian tissue. fertil steril. 2012; 98: 1291-8. 64. liu l, wood ga, morikawa l, ayearst r, fleming c, mckerlie c. restoration of fertility by orthotopic transplantation of frozen adult mouse ovaries. hum reprod. 2008; 23: 122-8. 65. rahimi g, isachenko v, kreienberg r, sauer h, todorov p, tawadros s, et al. re-vascularisation in human ovarian tissue after conventional freezing or vitrification and xenotransplantation. europ j obst gyn reprod boi. 2010; 149: 63-7. 66. baird dt, webb r, campbell bk, harkness lm, gosden rg. long-term ovarian function in sheep after ovariectomy and transplantation of autografts stored at - 196°c. endocrinology. 1999; 140: 462-71. 67. dissen ga, lara he, fahrenbach wh, costa me, ojeda sr. immature rat ovaries become revascularized rapidly after autotransplantation and show a gonadotropin-dependent increase in angiogenic factor gene expression. endocrinology. 1994;134:1146-54. 68. donnez j, martinez-madrid b, jadoul p, van langendoncket a, demylle d, dolmans mm. ovarian tissue cryopreservation and transplantation: a review. hum reprod update. 2006;12:519-35. 69. demeestere i, delbaere a, gervy c, bergh m, devreker f, englert y. effect of preantral follicle isolation technique on in-vitro follicular growth, oocyte maturation and embryo development in mice. hum reprod. 2002; 17: 2152-9. 70. abedelahi a, salehnia a, allameh aa. the effects of different concentrations of sodium selenite on the in vitro maturation of preantral follicles in serum-free and serum supplemented media. j assist reprod genet. 2008; 25:483-8. 71. haidari k, salehnia m, valoujerdi mr. the effect of leukemia inhibitory factor and co-culture on the in vitro maturation and ultrastructure of vitrified and non-vitrified isolated mouse preantral follicles. fertil steril. 2008; 90: 2389-97. 72. roy sk, treacy bj. isolation and long-term culture of human preantral follicles. fertil steril. 1993; 59: 783-90. 73. nagano m, atabay ep, atabay ec, hishinuma m, katagiri s, takahashi y. effects of isolation method and pre-treatment with ethylene glycol or raffinose before vitrification on in vitro viability of mouse preantral follicles. biomed res. 2007; 28: 153-60. 74. eppig jj,o,brien mj. development in vitro of mouse oocyte from primordial follicles. biol reprod. 1996; 54: 197-207. 75. smitz jj, cortvrindt rg. the earliest stages of folliculogenesis in vitro. reproduction. 2002; 123: 185- 202. 76. hovatta o, silye r, abir r, krausz t,winston rm. extracellular matrix improves survival of both stored and fresh human primordial and primary ovarian follicles in long-term culture. hum reprod. 1997; 12: 1032-6. 77. wu j, xu b, wang w. effects of luteinizing hormone and follicle stimulating hormone on the developmental competence of porcine preantral follicle oocytes grown in vitro. j assist reprod genet. 2007; 24: 419-24. 78. cavilla jl, kennedy cr, byskov ac, hartshorne gm. human immature oocytes grow during culture for ivm. hum reprod. 2008; 23: 37-45. 79. chian rc, uzelac ps, nargund g. in vitro maturation of human immature oocytes for fertility preservation. fertil steril. 2013; 99: 1173-81. 80. blandau rj, warrick e, rumery re. in vitro cultivation of fetal mouse ovaries. fertil steril. 1965;16:705-15. 81. gougeon a. dynamics of follicular growth in the human: a model for preliminary results. hum reprod. 1986; 1: 81-7. 82. choi j, lee jk, lee e, yoon bk, bae d, choi d. cryopreservation of the mouse ovary inhibits the onset of primordial follicle development. cryobiology. 2007; 54: 55-62. 83. kim ss, yang hw, kang hg, lee hh, lee hc, ko ds, et al. quantitative assessment of ischemic tissue damage in ovarian cortical tissue with or without antioxidant (ascorbic acid) treatment. fertil steril. 2004; 82: 679-85. 84. shaw jm, oranratnachai a, trounson ao. fundamental cryobiology of mammalian oocytes and ovarian tissue. theriogenology. 2000; 53: 59-72. 85. hovatta o. cryopreservation and culture of human primordial and primary ovarian follicles. mol cell endocrinol. 2000; 169: 95-7. 86. liu j, elst jvd, broecke rvd, dumortier f, dhont m. maturation of mouse primodial follicles by combination of grafting and in vitro culture. biol repord. 2000; 62: 1218 -23. 87. rezaei‐tavirany m, dolat e, hasanzadeh h, seyyedi ss, semnani v, sobhi s. tio2 nanoparticle as a sensitizer drug in radiotherapy: in vitro study. iran j cancer prev. 2013; suppl: 37-44.

برای دانلود باید عضویت طلایی داشته باشید

برای دانلود متن کامل این مقاله و بیش از 32 میلیون مقاله دیگر ابتدا ثبت نام کنید

اگر عضو سایت هستید لطفا وارد حساب کاربری خود شوید

منابع مشابه

Fertility Preservation Among the Cancer Patients by Ovarian Tissue Cryopreservation, Transplantation, and Follicular Development

Ovarian tissue freezing or cryopreservation might be the only acceptable method for preserving the young women fertility, before radiotherapy or chemotherapy. This technology might be used for patients with recurrent ovarian cysts or endometriosis, without ovarian stimulation. Many efforts have made to improve cryopreservation conditions that should be seriously considered for cancer patients. ...

متن کامل

Ovarian tissue banking for cancer patients: fertility preservation, not just ovarian cryopreservation.

While ovarian tissue cryopreservation has commonly been equated with fertility preservation in cancer patients, there is a range of alternative options to preserve fertility. Based on the type and timing of chemotherapy, the type of cancer, the patient's age and the partner status, a different strategy of fertility preservation may be needed. If the patient has a partner or accepts donor sperm,...

متن کامل

I-22: Fertility Preservation and Ovarian Stimulation in Cancer Patients

Cancer is not uncommon and no longer considered to be an incurable disorder. 10% of cancer cases occur under the age of 45. There is a remarkable improvement in treatment and survival rates. Today women have been delaying initiation of childbearing because the incidence of most cancers increases with age. Delayed childbearing results in more female cancer survivors. As a consequence there is an...

متن کامل

Cryopreservation time does not decrease follicular viability in ovarian tissue frozen for fertility preservation

OBJECTIVE To determine the effect of storage duration on cryopreserved ovarian tissue using fresh and frozen-thawed samples. METHODS Seventeen fertile patients underwent an ovarian biopsy during elective laparoscopic tubal ligation. The tissue sample was divided into three parts: one part was processed fresh (FG), and two were slowly frozen, cryopreserved for 30 (G30) or 180 days (G180), thaw...

متن کامل

Fertility preservation in pre-pubertal girls with cancer: the role of ovarian tissue cryopreservation.

With the increasing numbers of survivors of cancer in young people, future fertility and ovarian function are important considerations that should be discussed before treatment commences. Some young people, by nature of the treatment they will receive, are at high risk of premature ovarian insufficiency and infertility. For them, ovarian tissue cryopreservation (OTC) is one approach to fertilit...

متن کامل

I-11: Cryopreservation and Utilization of Ovarian Tissue: When, Where and How?

In women, around 10% of cancers occur in those younger than 45 years old. Although aggressive chemotherapy/radiotherapy and bone marrow transplantation can cure more than 90% of girls and young women affected by disorders requiring such treatment, the ovaries are very sensitive to cytotoxic drugs, especially to alkylating agents. Therefore, these treatments can result in premature ovarian failu...

متن کامل

منابع من

با ذخیره ی این منبع در منابع من، دسترسی به آن را برای استفاده های بعدی آسان تر کنید


عنوان ژورنال:
iranian journal of cancer prevention

جلد ۶، شماره ۳، صفحات ۱۲۳-۱۳۲

میزبانی شده توسط پلتفرم ابری doprax.com

copyright © 2015-2023