Fertilitatea
Fertilitatea
Fertilitatea
Jacques Donnez
Department of Gynecology and Andrology, Universite Catholique de Louvain, Brussels, Belgium
S. Samuel Kim
Division of Reproductive Endocrinology, University of Kansas, Kansas City, KS, USA
Contents
List of contributors x
Foreword by Roger G. Gosden xv
Foreword by Edward E. Wallach xvi
Preface xvii
Acknowledgements xviii
Section 1: Introduction
35
49
vii
List of contents
15 Transplantation of cryopreserved
spermatogonia 199
Jill P. Ginsberg and Ralph L. Brinster
16 Cryopreservation and transplantation
of testicular tissue 209
Christine Wyns
17 Assisted reproductive techniques and
donor sperm in cancer patients 225
Wayland Hsiao, Elizabeth Grill and Peter
N. Schlegel
viii
Section 7: Ovarian
cryopreservation and transplantation
27 General overview of ovarian
cryobanking 328
S. Samuel Kim
28 Ovarian tissue cryopreservation
Debra A. Gook and David H. Edgar
342
List of contents
421
488
Index 507
The color plate section will be found between
pages 366 and 367.
ix
Contributors
R. J. Aitken
Discipline of Biological Sciences and ARC Centre of
Excellence in Biotechnology and Development,
School of Environmental and Life Sciences,
University of Newcastle, Callaghan, NSW,
Australia
Gokhan Akkoyunlu
Department of Histology and Embryology, Faculty of
Medicine, Akdeniz University, Antalya, Turkey
David F. Albertini
Kansas University School of Medicine, Kansas City,
KS, USA
Montserrat Boada
Department of Obstetrics, Gynecology and
Reproduction, Institut Universitari Dexeus,
Barcelona, Spain
Paolo Boffetta
Mount Sinai School of Medicine, New York, NY, USA
and International Prevention Research Institute,
Lyon, France
Christiani A. Amorim
Universite Catholique de Louvain, Brussels, Belgium
Andrea Borini
Tecnobios Procreazione, Centre for Reproductive
Health, Bologna, Italy
R. A. Anderson
Division of Reproductive and Developmental Science,
University of Edinburgh, Edinburgh, UK
Baris Ata
McGill Reproductive Centre, Department of
Obstetrics and Gynecology, Division of Reproductive
Endocrinology and Infertility, McGill University,
Montreal, Quebec, Canada
Peter R. Brinsden
Bourn Hall Clinic, Bourn, Cambridge, UK
Pedro N. Barri
Department of Obstetrics, Gynecology and
Reproduction, Institut Universitari Dexeus,
Barcelona, Spain
Mohamed A. Bedaiwy
Assiut School of Medicine, Assiut, Egypt and
Department of ObstetricsGynecology, University
Hospitals Case Medical Center, Case Western Reserve
University, Cleveland, OH, USA
Rosita Bergstrom
Karolinska Institute, Stockholm, Sweden
Veronica Bianchi
Tecnobios Procreazione, Centre for Reproductive
Health, Bologna, Italy
Ralph L. Brinster
Department of Animal Biology, University of
Pennsylvania School of Veterinary Medicine,
Philadelphia, PA, USA
Jason G. Bromer
Yale University Fertility Center, New Haven, CT,
USA
A. L. Caplan
Center for Bioethics, University of Pennsylvania,
Philadelphia, PA, USA
Ri-Cheng Chian
Department of Obstetrics and Gynecology McGill
University, Montreal, Quebec, Canada
List of contributors
Ina N. Cholst
Ronald O. Perelman and Claudia Cohen Center for
Reproductive Medicine, Weill Cornell Medical
College, New York, NY, USA
A. Ciobanu
Gynecologic department, Hopital
Femme-M`ere-Enfant, Bron, France
Megan Clowse
Division of Rheumatology and Immunology, Duke
University, Durham, NC, USA
Ana Cobo
Instituto Valenciano de Infertilidad (IVI),
Valencia, Spain
Susannah C. Copland
Duke Fertility Center, Durham, NC, USA
John K. Critser
General Biotechnology LLC, Indianapolis, IN and
Comparative Medicine Center, University of
Missouri, Columbia, MO, USA
B. J. Curry
Discipline of Biological Sciences and ARC Centre of
Excellence in Biotechnology and Development,
School of Environmental and Life Sciences,
University of Newcastle, Callaghan, NSW, Australia
Giuseppe Del Priore
Indiana University School of Medicine, Department
of Obstetrics-Gynecology, Indianapolis, IN, USA
M. De Vos
Center for Reproductive Medicine, UZ Brussel
(VUB), Brussels, Belguim
Marie-Madeleine Dolmans
Department of Gynecology, Cliniques Universitaires
Saint Luc, Brussels, Belgium
Javier Domingo
IVI Las Palmas, Las Palmas de Gran Canaria, Spain
Elizabeth Grill
Ronald O. Perelman and Claudia Cohen Center for
Reproductive Medicine at New York-Presbyterian
Hospital/Weill Cornell Medical Center, New York,
NY, USA
Jacques Donnez
Department of Gynecology and Andrology,
Universite Catholique de Louvain, Brussels, Belgium
Sebastien Gouy
Service de Chirurgie Gynecologique, Institut
Gustave-Roussy, Villejuif Cedex, France
David H. Edgar
Reproductive Services, Royal Womens Hospital/
Melbourne IVF and Department of Obstetrics
Xu Han
Comparative Medicine Center, University of
Missouri, Columbia, MO, USA
xi
List of contributors
Lisa M. Harlan-Williams
University of Kansas Cancer Center, Kansas City,
KS, USA
Juergen Liebermann
Fertility Centers of Illinois, Chicago-River North IVF
Center, Chicago, Illinois, USA
Outi Hovatta MD
Division of Obstetrics and Gynecology, Huddinge
University Hospital, Huddinge, Sweden
J. Ryan Martin
Yale University Fertility Center, New Haven, CT,
USA
Wayland Hsiao
Weill Cornell Medical College, New York, NY,
USA
Elizabeth A. McGee
Department of Obstetrics and Gynecology, VCU
School of Medicine, Richmond, VA, USA
Zhongwei Huang
Nuffield Department of Obstetrics and Gynaecology,
University of Oxford, Oxford, UK
Marie McLaughlin
Centre for Integrative Physiology, University of
Edinburgh, Edinburgh, UK
E. Isachenko
Department of Obstetrics and Gynecology, University
Womens Hospital, Ulm, Germany
P. Mathevet
Hopital Femme-M`ere-Enfant, Bron, France
V. Isachenko
Department of Obstetrics and Gynecology, University
Womens Hospital, Ulm, Germany
Roy A. Jensen
University of Kansas Medical Center, Kansas City, KS,
USA
I. I. Katkov
University of San Diego in La Jolla, San Diego, CA,
USA
S. Samuel Kim
Division of Reproductive Endocrinology, University
of Kansas, Kansas City, KS, USA
Jennifer Klemp
University of Kansas Cancer Center, Kansas City, KS,
USA
Larissa A. Korde
Division of Medical Oncology, University of
Washington/Seattle Cancer Care Alliance, Seattle,
WA, USA
R. Kreienberg
Department of Obstetrics and Gynaecology,
University Womans Hospital, Ulm, Germany
Srinivasan Krishnamurthy
Department of Obstetrics and Gynecology, McGill
University, Montreal, Quebec, Canada
xii
D. Meirow
IVF Unit, Fertility Preservation Laboratory, Sheba
Medical Center, Tel-Aviv University, Israel
Philippe Morice
Service de Chirurgie Gynecologique, Institut
Gustave-Roussy, Villejuif Cedex, France
Steven F. Mullen
Reproductive Cryobiology, 21st Century Medicine,
Inc., Fontana, CA, USA
Kutluk Oktay
Division of Reproductive Medicine and Infertility and
Laboratory of Molecular Reproduction and Fertility
Preservation, Westchester Medical CenterNew York
Medical College, Valhalla, NY, USA
Pasquale Patrizio
Yale University Fertility Center, New Haven, CT,
USA
Antonio Pellicer
University of Valencia, Valencia, Spain
Pinki K. Prasad
VanderbiltIngram Cancer Center, Nashville, TN,
USA
Kenny A. Rodriguez-Wallberg
Karolinska Institute and Karolinska University
Hospital Huddinge, Fertility Unit, Department of
Obstetrics and Gynecology, Stockholm, Sweden
List of contributors
Erin Rohde
Indiana University School of Medicine, Department
of ObstetricsGynecology, Indianapolis, IN,
USA
Allison B. Rosen
Fertility Preservation Institute, Department of
Obstetrics and Gynecology, Westchester Medical
CenterNew York Medical College, Valhalla, NY, USA
Zev Rosenwaks
Ronald O. Perelman and Claudia Cohen Center for
Reproductive Medicine, Weill Cornell Medical
College, New York, NY, USA
Mara Sanchez
Gynaecology and Obstetrics at the University of
Valencia, Valencia, Spain
R. Sanchez
Center of Biotechnology in Reproduction,
Department of Basic Sciences, La Frontera University,
Temuco, Chile
Glenn L. Schattman
Ronald O. Perelman and Claudia Cohen Center for
Reproductive Medicine, Weill Cornell Medical
Center, New York, NY, USA
Peter N. Schlegel
James Buchanan Brady Foundation, Weill Cornell
Medical College, New York, NY, USA
Einat Shalom-Paz
McGill Reproductive Centre, Department of
Obstetrics and Gynecology, Division of Reproductive
Endocrinology and Infertility, McGill University,
Montreal, Quebec, Canada
Lonnie D. Shea
Department of Chemical and Biological Engineering,
McCormick School of Engineering and Applied
Science, Northwestern University, Technological
Institute, Evanston, IL, USA
Carrie A. Smith
Indiana University School of Medicine, Department
of Obstetrics-Gynecology, Indianapolis, IN, USA
J. Smitz
Center for Reproductive Medicine, UZ Brussel
(VUB), Brussels, Belgium
Miquel Sole
Department of Obstetrics, Gynecology and
Reproduction, Institut Universitari Dexeus,
Barcelona, Spain
Jean Squifflet
Universite Catholique de Louvain, Brussels, Belgium
Shane R. Stecklein
University of Kansas Medical Center, Kansas City,
KS, USA
Jerome F. Strauss, III
Virginia Commonwealth University Health System,
Sanger Hall, Richmond, VA, USA
David J. Tagler
Department of Chemical and Biological Engineering,
McCormick School of Engineering and Applied
Science, Northwestern University, Technological
Institute, Evanston, IL, USA
Seang Lin Tan
Department of Obstetrics and Gynecology at McGill
University; McGill University Health Centre, McGill
Reproductive Centre, Royal Victoria Hospital,
Montreal, Quebec, Canada
Evelyn E. Telfer
Centre for Integrative Physiology, University of
Edinburgh, Edinburgh, UK
Sreedhar Thirumala
General Biotechnology LLC, Indianapolis, IN,
USA
Gunapala Shetty
Department of Experimental Radiation Oncology,
University of Texas M. D. Anderson Cancer Center,
Houston, TX, USA
Michael J. Tucker
Shady Grove Fertility RSC, Rockville, MD, USA and
Georgia Reproductive Specialists, Atlanta, GA,
USA
Jill Simmons
Division of Pediatric Endocrinology Vanderbilt
University School of Medicine, Nashville, TN, USA
Catherine Uzan
Service de Chirurgie Gynecologique, Institut
Gustave-Roussy, Villejuif, France
xiii
List of contributors
Dagan Wells
Nuffield Department of Obstetrics and Gynaecology,
University of Oxford, Oxford, UK
Anna Veiga
Reproductive Medicine Service, Department of
Obstetrics, Gynecology and Reproduction, Institut
Universitari Dexeus, Barcelona, Spain
Teresa K. Woodruff
Department of Obstetrics and Gynecology, Feinberg
School of Medicine, Northwestern University,
Chicago; Robert H. Lurie Comprehensive Cancer
Center of Northwestern University, Chicago,
IL, USA
W. H. B. Wallace
Division of Reproductive and Developmental Science,
University of Edinburgh, Edinburgh, UK
Wenjia Wang
University of Kansas Medical Center, Kansas City,
KS, USA
Erik Woods
General Biotechnology LLC and Indiana University
School of Medicine, Department of Microbiology and
Immunology, Indianapolis, IN, USA
Brent Waters
Stead Center for Ethics and Values,
GarrettEvangelical Theological Seminary, Evanston,
IL, USA
Christine Wyns
Department of Gynecology, Universite Catholique de
Louvain, Brussels, Belgium
xiv
Foreword
xv
Foreword
xvi
Preface
xvii
Acknowledgements
xviii
tive Medicine, the University of Kansas Medical Center, for their deep and enduring dedication, and to
Carl Weiner, my department chair, for his advice and
encouragement. I am indebted to Roger Gosden, my
mentor and best friend, for his keen insight, relentless support and wisdom. My heartfelt thanks go to my
beloved family, Kris, Jean, Melissa, Derek, Monica and
my mother.
Jacques Donnez would like to thank all the
members of his clinical and research teams at the Universite Catholique de Louvain, who work tirelessly in
pursuit of scientific advancement and clinical excellence and understand that there is no life without
pressure. I am also eternally grateful to my wife, children and grandchildren for their unwavering love and
support.
Section 1
Chapter
Introduction
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
Section 1: Introduction
Figure 1.4
Lazzaro Spallanzani
(172999).
Performed the first
known
insemination of a
bitch, first in vitro
fertilization with
frogs and was the
first to successfully
freeze and thaw
sperm.
oocytes fertilized in vitro using spermatozoa capacitated in vitro [3]. Work continued, more or less successfully, over the next decade attempting to achieve
fertilization in vitro and live births of various other
mammalian species.
Some of the earliest observations on fertilization of
human oocytes were made by Robert Edwards (1925)
and published in 1965 in a landmark paper: Maturation in vitro of human ovarian oocytes [4]. It was not
possible for Edwards to progress further with efforts
to achieve IVF of human oocytes for clinical use without close collaboration with clinical colleagues, who
were able to provide a supply of human oocytes usually from patients having ovarian wedge resections for
polycystic ovary disease. It was the need for Edwards
to be able to obtain these supplies of pre-ovulatory
human oocytes that brought him and gynecologist
Patrick Steptoe (191388) together in 1968.
Patrick Steptoe became known to Robert Edwards
because he had brought laparoscopy, whereby the
female pelvic organs could be visualized by a relatively
minor operation, to England from Europe where Steptoe had studied it under both Raoul Palmer (1940
95) and Hans Frangenheim (19202001). On his
return to England, he further developed the technique
of laparoscopy, and shortly afterwards he wrote his
famous short textbook Laparoscopy in Gynaecology in
1967 [5]. His first major paper: Laparoscopy and ovulation followed in 1968 [6]. Shortly afterwards, he further developed the laparoscopic technique to enable
aspiration of oocytes from follicles under direct vision.
Robert Edwards started his career in reproductive
biology at the Institute of Animal Genetics and Embryology, Edinburgh, in 1951, having just been demobbed
Section 1: Introduction
with tubal insemination (ORTI), as they called the procedure, which was later to become known as gamete
intrafallopian transfer (GIFT).
Between the years 1968 and 1978, while they were
working closely together, Steptoe was in Oldham, Lancashire, England, working in a National Health Service hospital, and Edwards was at the University of
Cambridge. When the clinical treatment of infertile
women started, there followed a number of very difficult years in which none of the first 40 patients they
treated by IVF and embryo transfer (ET) became pregnant. In 1976 they did achieve their first pregnancy following transfer of a single blastocyst, but this subsequently turned out to be an ectopic pregnancy [10].
After 102 failed embryo transfers, including the one
ectopic pregnancy, Leslie Brown was treated and subsequently became pregnant following her first embryo
transfer. This was achieved in a natural IVF cycle,
with no stimulation; one oocyte was collected and a
single eight-cell embryo was transferred. There followed a difficult pregnancy for Mrs Brown, but her
baby, Louise Brown, was delivered by cesarian section on July 25, 1978 (Figure 1.6). Much to everyones
relief, baby Louise was found to be a perfectly normal,
fit and healthy infant. This momentous achievement
was announced with a simple publication as a letter
in the Lancet: Birth after reimplantation of a human
embryo [11]. The arrival of Louise Brown was heralded as The baby of the century. This was in spite of
considerable criticism and opposition by clinical and
scientific colleagues, the lay press and by many representatives of different religious faiths.
Work on human IVF was also being conducted
elsewhere in the world, particularly in Melbourne,
Australia, where the team of Professor Carl Wood and
Dr. Alan Trounson achieved the birth of the worlds
fourth IVF baby, Candice Reed, in June 1980 [12]. At
the same time, Drs. Howard and Georgeanna Jones
had been working in Norfolk, Virginia, USA, and Elizabeth Carr, the first US in-vitro conceived baby, was
born on December 28, 1981 [13].
In England, meanwhile, Patrick Steptoe and Robert
Edwards were unable to continue their work on human
IVF, since neither the UKs National Health Service nor
any of the Universities or the Medical Research Council were willing to provide funding to help them to continue their work. They eventually found Bourn Hall,
an old Jacobean manor house in the Cambridgeshire
countryside, where they founded the Worlds first IVF
treatment and research center Bourn Hall Clinic
which opened in September 1980 (Figure 1.7). There,
Steptoe and Edwards continued their research and,
by 1986, they had achieved 500 live births [14]. Steptoe and Edwards achieved a number of distinguished
national and international awards over the next 3
years, but, in 1988, Patrick Steptoe fell seriously ill with
prostate cancer and died on March 21, 1988. Robert
Edwards continued to work as Scientific Director of
Bourn Hall and as Editor of the newly formed journal
Human Reproduction, which he cofounded. In 1994, he
retired from working at Bourn Hall.
Section 1: Introduction
r Late 1990s2000+
GnRH antagonists + rhFSH recombinant
human luteinizing hormone (rhLH)
recombinant LH
recombinant human chorionic gonadotropin
(hCG)
fill by mass versus IU recombinant FSH
patient friendly sc injections and use of pens
for injection
As can be seen above, stimulation protocols have
undergone many changes. In the early days, IVF was
conducted in natural cycles or with clomiphene-only
stimulation. It was in 1984 that Porter et al. in London first developed the use of GnRH agonists in IVF
stimulation protocols to prevent premature LH surges
[21], which, over the following years, became the gold
standard for use in IVF stimulation protocols; indeed,
it remains so for many practitioners. Introduction of
the GnRH antagonists in stimulation protocols, first
reported by Frydman et al. in 1991 [22], increasingly
has become used, allowing a more natural cycle and
being more patient friendly, since treatment does
not last as long as do GnRH-agonist protocols. Also
from the mid 1990s, there were major developments
in the production and use of gonadotrophins. These
were produced originally from human menopausal
urine and injected intramuscularly. High purity FSH
and HMG were a great improvement and could be
injected subcutaneously by patients themselves after
training. Also from about the mid 1990s, recombinant gonadotrophins were developed, producing the
purest FSH, LH and hCG. It was in 1992 that Germond et al. [23] and Devroey et al. [24] reported
the first pregnancies using the new recombinant
FSH (rFSH).
More recently still, there has been an enthusiasm
for the concept of IVF lite. This has introduced the
concept of a milder stimulation strategy for IVF in
order to reduce the risk of complications of stimulation, particularly of ovarian hyperstimulation syndrome. It is also thought to improve the chance of
implantation by reducing interference of the development of the endometrium that may occur in some high
dose gonadotrophin stimulation protocols.
In vitro fertilization was never really successful
in the treatment of severe male factor infertility, and
techniques were developed to try to improve the outcome for men with this diagnosis, for whom the
Section 1: Introduction
References
1. Bavister BD. Early history of in vitro fertilization.
Reprod 2002; 124: 18196.
2. Clarke GN. ART and history. Hum Reprod 2006; 21:
164550.
3. Yanagimachi R and Chang MC. Fertilization of
hamster eggs in vitro. Nature 1963; 200: 2812.
4. Edwards RG. Maturation in vitro of human ovarian
oocytes. Lancet 1965; 2: 926.
5. Steptoe PC. Laparoscopy in Gynaecology. Edinburgh: E
and S Livingstone, 1967.
6. Steptoe PC. Laparoscopy and ovulation. Lancet 1968;
2: 913.
7. Edwards RG, Bavister BD and Steptoe PC. Early
stages of fertilization in vitro of human oocytes
matured in vitro. Nature 1969; 221: 6325.
8. Bavister BD, Edwards RG and Steptoe PC.
Identification of the midpiece and tail of the
spermatozoon during fertilization of human eggs in
vitro. J Reprod Fert 1969; 20: 16970.
9. Steptoe PC and Edwards RG. Laparoscopic recovery
of pre-ovulatory oocytes after priming of ovaries with
gonadotrophins. Lancet 1970; 1: 6839.
10. Steptoe PC and Edwards RG. Reimplantation of a
human embryo with subsequent tubal pregnancy.
Lancet 1976; 1: 8802.
11. Steptoe PC and Edwards RG. Birth after the
reimplantation of a human embryo. Lancet 1978; 2:
366.
12. Trounson A, Leeton J, Wood J et al. Pregnancies in
the human by fertilization in vitro and embryo
transfer in the controlled ovulatory cycle. Science 1981;
212: 6812.
13. Jones HW, Jones GS, Andrews MC et al. The program
for in vitro fertilization at Norfolk. Fertil Steril 1982;
381: 1421.
Section 1: Introduction
10
Section 1
Chapter
Introduction
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
11
Section 1: Introduction
10 000 000
1 000 000
n =325
r 2 =0.81
100 000
Observed values
WallaceKelsey model
Lower 95% Cl for model
Upper 95% Cl for model
Lower 95% prediction limit
Upper 95% prediction limit
10 000
1000
100
10
5
0
5
10
15
20
25
30
35
40
45
50
Age (in months from conception to birth; in years from birth to menopause)
55
Figure 2.1 The best model for the establishment of the non-growing follicle (NGF) population after conception and the subsequent decline
until age at menopause is described by an ADC model. The model has a correlation coefficient r2 = 0.81, fit standard error = 0.46 and
F-value = 364. The figure shows the dataset (n = 325), the model, the 95% prediction limits of the model and the 95% confidence interval (CI)
for the model. The horizontal axis denotes age in months up to birth at age 0 and age in years from birth to 51 years. Reproduced with
permission from Wallace and Kelsey [8]. See plate section for color version.
ovarian follicle decline provides no supporting evidence of neo-oogenesis in normal human physiological aging.
Clinical detection of
chemotherapy-induced
ovarian damage
Many studies use amenorrhea as a surrogate for ovarian failure, with biochemical confirmation (i.e. elevated follicle stimulating hormone [FSH] concentration) in some. While amenorrhea may of course have
other causes than ovarian failure, it would seem a reasonable surrogate for population-based studies. However, an important drawback of the use of amenorrhea or elevated FSH measurements is that these only
detect the endpoint of the decline of ovarian function. It would be of considerable utility to have a biochemical or biophysical marker of the number of follicles in the ovary, i.e. the ovarian reserve (Table 2.1
[13]) [14] to allow detection of lesser degrees of damage and earlier changes during the progress to ovarian failure. This would allow improved analysis of the
12
hormone (AMH)
r Sonography: total antral follicle count (AFC)
r Sonography: ovarian volume
r Ovarian biopsy
r Response to ovarian stimulation
Adapted from van Rooij et al. [13].
13
Section 1: Introduction
A reduced follicular reserve may result in premature ovarian failure (POF) and menopause many
years post-treatment, even in patients undergoing
chemotherapy at a very young age [24]. Significant
depletion of the primordial follicle stockpile postchemotherapy in a normally ovulating female has been
demonstrated in an animal model [30].
The risk of ovarian failure in several commonly
encountered malignancies and other disorders requiring chemotherapy and/or radiotherapy is presented
in Table 2.2. Cyclophosphamide is widely used in
combination chemotherapy regimens, and high dose
cyclophosphamide (200 mg/kg) is frequently utilized
as conditioning therapy before bone marrow transplantation (BMT), either alone, where recovery of
ovarian function is more likely, or in combination with
other chemotherapeutic agents or total body irradiation (TBI) [36].
Treatment of Hodgkins lymphoma with MOPP
(mechlorethamine, vincristine, procarbazine and
prednisolone) or ChlVPP (chlorambucil, vinblastine,
procarbazine and prednisolone) is associated with
ovarian dysfunction in 1963% of cases [37]. Amenorrhea is more commonly observed in older women,
but long-term follow-up is necessary, as a number of
young women also develop premature menopause.
The BEACOPP regimen (bleomycin, etoposide,
doxorubicin, cyclophosphamide, vincristine, procarbazine, prednisolone) results in amenorrhea in
approximately 20% of women overall, but this rises to
67% in women treated with 8 cycles of dose-escalated
BEACOPP [38] with, as discussed above, age being
an important factor. Amenorrhea was reported by
95% of women aged over 30 years compared to
51% in younger women. Treatment with an ABVD
regimen (adriamycin, bleomycin, vinblastine and
dacarbazine), which contains no alkylating agents or
procarbazine, results in significantly less gonadotoxicity, especially in patients under 25 years [39]. In a
recent cohort study [40] of 518 female 5-year survivors
of Hodgkins lymphoma aged 1440 (median age: 25
years) at treatment, the Amsterdam group explored
the incidence of POF before age 40. Alkylating agents,
especially procarbazine (hazard ratio [HR]: 8.1) and
cyclophosphamide (HR: 3.5), showed the strongest
associations. Ten years after treatment, the actuarial
risk of premature menopause was 64% after high
cumulative doses (8.4 g/m2 ), and 15% after low
doses (4.2g/m2 ), of procarbazine [29]. A small
study demonstrated normal fecundity in women
14
Study (year)
Treatment
Age (years)
Breast cancer
Lower (1999)
Premenopausal
45
35
28
Bines (1996)
Premenopausal
68
Meirow (1999)
44
50
Goodwin (1999)
Burstein (2000)
CMF
CMF
43.7 5.2
65
30
19
3039
30
CAF
3039
AC
30
Jonat (2001)
3040
0
1025
3039
13
Premenopausal
60
Hodgkins lymphoma
Howell and Shalet (1998)
Aggressive treatment
Bokemeyer (1994)
Infradiaphragmatic Rx
Brusamolino (2000)
Ovarian-sparing protocol
Behringer (2005)
Dose-escalated BEACOPP
3857
32
50
25
45
30
30
95
30
51
No. of patients
Sanders (1996)
73
38 (mean)
99
Teinturier (1998)
21
217
72
Thibaud (1998)
31
3.217
80
Meirow (1999)
63
29 (mean)
79
Grigg (2000)
19
30 (mean)
100
70
Boumpas (1993)
39 (0.51.0 g/m2 )
26
7 pulses
12
15 pulses
39
30
Blumenfeld (1996)
2030
20
Appenzeller (2007)
15
100
17.5 (12.3*)
13
0 (20*)
30
39
3040
59
15
Section 1: Introduction
Effective (
) and mean (
) sterilizing doses
22
20
18
Dose (Gy)
16
14
12
10
8
6
4
2
0
10
15
20
25
30
Age at treatment
35
Intermediate
risk
Low risk
( 20%)
Unknown
risk
(2030 Gy) during childhood [36, 48, 49]. Our understanding of the LD50 of the human oocyte has made it
possible to estimate the age at which premature ovarian failure may occur. Furthermore, we have estimated
16
40
45
50
the sterilizing dose following any given dose of radiotherapy at any given age, based upon the application of
a mathematical solution to the FaddyGosden model
for natural oocyte decline (Figure 2.2) [50, 51]. This
will help clinicians provide accurate information when
counseling women about fertility following treatment
for childhood cancer.
Gonadotropin deficiency following high dose cranial irradiation (24 Gy in the treatment of brain
tumors) manifests as delayed puberty or absent menses
and can be treated by hormone replacement therapy.
Interestingly, early puberty is often reported in females
with cranial radiation doses of 24 Gy [52]. However, we have shown a subtle decline in hypothalamic
pituitaryovarian function following low dose cranial radiotherapy (1824 Gy). This is characterized
by decreased luteinizing hormone (LH) secretion
throughout the cycle, an attenuated LH surge and short
luteal phases [53], which may compromise reproductive function. Recent data confirm lower fertility in childhood cancer survivors treated with either
hypothalamic/pituitary iradiation 30 Gy or ovarian/uterine irradiation 5 Gy [27].
Nowadays, the most commonly used fertility
preservation measure is the surgical transposition of
the ovaries outside the irradiation field before the initiation of pelvic radiation in adults with gynecological
malignancies [54]. This approach may be considered
for patients not planning to receive high dose systemic chemotherapy. Surgery is effective at protecting
the ovaries from direct irradiation damage, but fertility may be affected by scatter radiation, damage to the
Irradiation field
Right
transposed ovary
Left
transposed ovary
blood flow following sex steroid treatment, suggesting that higher doses of pelvic radiation cause greater
damage than lower doses (as in TBI), and that this
damage may be irreversible.
Testicular function
In males, testicular damage can involve the somatic
cells of the testis (Sertoli, peritubular myoid and Leydig cells) or the germ cells. Sertoli cells are responsible for nurturing developing germ cells, and Leydig
cells produce testosterone. Recent data have revealed
the essential contribution of peritubular myoid cells in
mediating the effect of testosterone on spermatogenesis. Gonadal damage in males treated for cancer can
result from either systemic chemotherapy or radiotherapy to a field that includes the testes. Cytotoxic
treatment targets rapidly dividing cells and it is therefore not surprising that spermatogenesis is impaired
after treatment for cancer. The exact mechanism of
this damage is uncertain, but it appears to be linked
to depletion of the proliferating germ cell pool and
associated stem spermatogonial cells. Although the
pre-pubertal testis does not complete spermatogenesis and produce mature spermatozoa, cytotoxic treatment given to pre-pubertal boys may impair future fertility. Importantly, the pre-pubertal testis is susceptible
to cytotoxic damage.
17
Section 1: Introduction
18
Chemoprotection
Preventing chemotherapy-induced damage to the
ovary or testes remains an elusive ideal in the field
of fertility preservation. Most attention has focused
on the potential for protecting the ovaries using the
gonadotrophin-suppressing gonadotropin-releasing
hormone (GnRH) analogues, although a clear biological basis for this approach is unclear as only
later stages of follicular growth are gonadotrophindependent. There may however be other, indirect
mechanisms on smaller follicles, or direct effects of
the GnRH analogues themselves, and the approach
is supported by animal studies in both rodents [70]
and non-human primates [71]. Initial small studies
failed to show any benefit [72] but were substantially
underpowered, and while later studies showed large
apparent benefits [73, 74], lack of randomization and
differences between treated and control groups preclude reliable interpretation. A recent randomized
controlled trial in women under the age of 40 with
breast cancer appears to support a benefit of GnRH
analogue treatment [75], but further studies are necessary to substantiate this finding as there are a number
of potential confounding methodological issues with
study design. Alternative approaches have addressed
the mechanisms of chemotherapy-induced oocyte
apoptosis using sphingolipids [32] and more recently
the c-Abl kinase inhibitor imatinib [76]. In the male,
the immunomodulator AS101 has been demonstrated
to protect against cyclophosphamide-induced sperm
damage and low fertility [77].
Summary
Whilst many children and adults diagnosed with
cancer can now realistically hope for long-term
References
1. Skinner R, Wallace WH and Levitt GA. Long-term
follow-up of people who have survived cancer during
childhood. Lancet Oncol 2006; 7: 48998.
2. Parking DM, Whelan SL, Ferlay J, Teppo L and
Thomas DB. Cancer Incidence in Five Continents, vol.
8. Lyon: IARC; 2002.
3. Meirow D and Dor J. Epidemiology and infertility in
cancer patients. In: Tulandi T and Gosden R (eds.),
Preservation of Fertility. Abingdon: Taylor and Francis,
2004: pp. 2138.
4. Oeffinger KC, Mertens AC, Sklar CA et al. Chronic
health conditions in adult survivors of childhood
cancer. N Engl J Med 2006; 355: 157282.
5. Byrne J, Fears TR, Gail MH et al. Early menopause in
long-term survivors of cancer during adolescence. Am
J Obstet Gynecol 1992; 166: 78893.
6. Thomson AB, Critchley HO, Kelnar CJ and Wallace
WH. Late reproductive sequelae following treatment of
childhood cancer and options for fertility preservation.
Best Pract Res Clin Endocrinol Metab 2002; 16:
31134.
7. Meirow D and Schiff E. Appraisal of chemotherapy
effects on reproductive outcome according to animal
studies and clinical data. J Natl Cancer Inst Monogr
2005; 34: 215.
8. Wallace WH and Kelsey TW. Human ovarian reserve
from conception to the menopause. PLoS One 2010; 5:
e8772.
9. Johnson J, Canning J, Kaneko T, Pru JK and Tilly JL.
Germline stem cells and follicular renewal in the
postnatal mammalian ovary. Nature 2004; 428:
14550.
10. Johnson J, Skaznik-Wikiel M, Lee HJ et al. Setting
the record straight on data supporting postnatal
oogenesis in female mammals. Cell Cycle 2005; 4:
14717.
11. Lee HJ, Selesniemi K, Niikura Y et al. Bone marrow
transplantation generates immature oocytes and
rescues long-term fertility in a preclinical mouse
19
Section 1: Introduction
20
21
Section 1: Introduction
22
Section 1
Chapter
Introduction
Introduction
The increasing survival rates of cancer patients [13]
have encouraged many specialists to focus on the irreversible consequences of chemotherapy and radiotherapy. Chemotherapy and radiotherapy treatment for
cancer or other pathologies has resulted in improved
survival rates, but these treatments may also lead
to sterility [4]. The increasing success of oncological
treatments means it is now even more crucial to implement procedures aimed at preserving fertility.
Similarly to cancer patients, there are some nononcological conditions currently treated with gonadotoxic agents, such as patients with autoimmune disorders or some chromosomal abnormalities that can
lead to ovarian failure. There are also other situations
where a woman may benefit from fertility preservation procedures, such as a woman with severe or recurrent endometriosis or a woman who wants to postpone
conception until her late reproductive years [5].
In addition, there are other gynecological situations regularly found in our clinical practice that were
previously difficult to deal with but which now can
be solved. Thus, oocyte or embryo vitrification can
be performed when, for any reason, we should prefer
to transfer embryos in a different cycle to the stimulated one. People with a high risk of hyperstimulation syndrome, the presence of a hydrosalpinx or
polyps during the stimulation, the absence of sperm
in the sample the day of the ovum pick-up or any
bleeding previously to embryo transfer are some situations where vitrification should be considered. As
a large number of embryos are needed, vitrification
may also be helpful for the low-responder patient with
the aim of accumulating oocytes or embryos, especially if pre-implantation genetic screening (PGS) is
planned [6].
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
23
Section 1: Introduction
Age
The ovarian cortex has a limited number of primordial
follicles that decrease with age. Previous chemotherapy
or surgery can also affect the initial status of gonads
before treatment starts. This will determine the final
impact on ovarian function.
Cumulative doses of cyclophosphamide can cause
infertility in young women [16]. Gonadotoxicity is
directly related to age: the cumulative dose needed
to cause premature ovarian failure decreases as age
24
Chemotherapy
It is known that chemotherapeutic agents can cause
mutations, DNA adducts and structural breaks, as well
as oxidative damage in somatic and germ cells. Alkylating agents such as cyclophosphamide or ifosfamide
are the most gonadotoxic agents, but also gonadotoxic
are chlorambucil, busulfan, cisplatin, melphalan, carboplatin or procarbazine [14, 15, 18].
The effect of chemotherapy on the ovary is not an
all-or-nothing phenomenon, so the number of surviving primordial follicles following chemotherapy will
depend on several factors such as age, type of agent and
doses received [16].
Chemotherapys alkylating agents join with DNA,
avoiding its replication and transcription [18]. They
are extremely gonadotoxic by acting at any phase of
the cellular cycle (cellular cycle phase independent),
causing damage to the primary follicles. Pathological
examinations of ovarian biopsies in patients treated
with cyclophosphamide show either a total absence
or a significant reduction in the number of inactive follicles, with fibrosis and no signs of follicular
maturation [19].
The mechanism of chemotherapy causing premature ovarian failure is not well known, but granulosa
cells appear to be the crucially affected cells [20]. Cellular edema of pre-granulosa cells is observed, with
queratin deposits and edema of the nucleus of the cell,
which damages the oocyte morphology [21]. Additional factors, such as vascular alterations and fibrosis
of the ovarian cortex, may contribute to the reduction
of follicles [22].
What is clear is that both the antral follicle count
and ovarian volume decrease after chemotherapy.
Radiotherapy
Similar to chemotherapy, the effect of radiotherapy on
the gonads depends on age, cumulative doses, fractioned doses and irradiation area. The average doses
needed to destroy oocytes in humans is 2 Gy [24].
Ninety-seven percent of women receiving 5.010.5 Gy
will subsequently undergo ovarian failure [25].
Especially if radiotherapy is applied during childhood, the irradiation area has been associated with
alterations of the uterine function due to the reduction of vascular flow and endometrial thickness [26].
Cranial irradiation with 3545 Gy doses can damage the hypothalamuspituitarygonadal axis but, as
gonads are not affected, they recover their function
with gonadotropin replacement.
Surgery
Repeat ovarian surgery due to endometriosis or
another benign pathology may diminish the ovarian
reserve and lead to premature ovarian failure. Furthermore, in recurrent endometriosis, normal residual
ovarian tissue may be compromised.
Tubal sterilization through electrocoagulation,
when compared to the application of mechanical clips,
also seems to have an adverse effect on ovarian reserve
in the postoperative period. Significant differences
have been detected in ovarian volumes at day 3 and
antral follicle counts at 10 months after the tubal
occlusion [27].
Fertility preservation
procedures
Several strategies have been proposed over the last few
years to protect and preserve the ovarian function in
patients with cancer or suffering from other pathologies with a high risk of premature ovarian failure. Some
25
Section 1: Introduction
26
Embryo cryopreservation
Embryo cryopreservation is a widely accepted method
that is now considered the standard practice for fertility preservation [37]. Previously, oocytes had to
be fertilized to be preserved, as thawed embryos
were considered to achieve higher survival rates than
oocytes. The female patient needed to have a partner
or sperm donor to fertilize the retrieved oocytes, creating embryos that may not have been used in the future,
which had various ethical considerations. Oocyte
cryopreservation avoids the need for sperm at the time
of oocyte retrieval and the results have been similar to
embryo cryopreservation, and thus should be considered an option.
Oocyte vitrification
transported 45 h on ice prior to freezing, demonstrating that hospitals may offer cryopreservation without
having the necessary local infrastructure.
The main disadvantage of this technique is that it
requires surgery laparoscopy to obtain the ovarian
tissue and a further re-implant with appropriate incorporation of the cryoprotectant to the tissue. Ischemic
damage and reduced follicular pool usually appear
after transplantation. The active life of the transplanted
tissue will depend on the neoangiogenesis and new
vascularization.
This approach offers great possibilities to patients
in the future, since portions of healthy tissue can be
preserved for a further use when an oophorectomy is
performed for a benign indication. The ethical basis for
performing this surgery for elective cryopreservation
has been discussed [42, 43], but a patients request for
cryopreserving small portions of ovary at the time of
any other gynecologic surgery should not be denied on
ethical grounds.
Currently, ovarian tissue cryopreservation is
only recommended as an experimental treatment in
selected patients. Research is needed to investigate the
revascularization process with the aim of reducing the
follicular loss that occurs after tissue grafting.
Ovarian tissue cryopreservation can serve as a
source of follicle for IVM. Thus, though still experimental, future fertility preservation techniques will
tend towards combining ovarian cryopreservation and
immature egg retrieval for further in-vitro oocyte maturation and vitrification. To become widely accepted,
these procedures need to be safe, easy to perform
and with favorable results. Meanwhile, a combination of ovarian cryopreserved transplantation, further
ovarian stimulation and vitrification of the retrieved
oocytes to be accumulated for an in vitro fertilization
(IVF) cycle can be used. A twin pregnancy was recently
obtained through this method [44].
Imatinib
Imatinib acts by blocking the apoptotic pathway activated by cisplatin in ovarian germ cells. Cisplatin
induces DNA damage by activating the c-AblTAp63
pathway, leading to cell death. In cell lines, c-Abl phosphorylates TAp63, which induces the activation of
proapoptotic cells. Treatment with the c-Abl kinase
inhibitor imatinib blocks these effects [53].
In-vitro maturation
27
Section 1: Introduction
Transposition of ovaries
Scatter radiotherapy can cause considerable damage
even if the gonads are outside the radiation field. The
purpose of this approach is to avoid the direct exposure of the ovaries to radiotherapy, although the indirect exposure can also cause gonadotoxicity. Thus, it
should be indicated for any pathology that requires
pelvic radiotherapy treatment. When this approach is
performed, 1690% of the patients show the ovarian
function preserved [4547].
Ovarian transposition is not suitable for nononcological patients as radiotherapy is uncommonly
used, although it is very useful for cancer patients
when they are going to receive local radiotherapy.
Autoimmune diseases
The effect of autoimmune disorders on fertility
depends most of all on a womans reproductive age.
Fertility preservation methods should be considered
in young women with severe systemic autoimmune
rheumatic diseases requiring imminent gonadotoxic
treatment.
28
Malignant
Breast cancer
Hodgkins lymphoma
Other tumors
Non-malignant
(n = 284)
170
62
52
(n = 17)
Crohn disease
Endometriosis
Nephropathies
Multiple sclerosis
Wegeners granulomatosis
The availability of new medications for the treatment of rheumatoid arthritis has dramatically changed
the prognosis for these patients, but limited data about
their reproductive or developmental toxicity are available, particularly when the medications are used for
the treatment of rheumatoid arthritis.
Cyclophosphamide is only used for refractory
rheumatoid arthritis. It seems to have a significant
beneficial effect on the clinical evolution of rheumatoid arthritis, but its administration has been limited due to its secondary aggressive effects and the
similar benefits and low risk of other anti-rheumatic
agents. Furthermore, cyclophosphamide treatment for
rheumatoid arthritis has been related to the appearance of some hematological malignant neoplasms and
bladder cancer [57, 58].
Cyclophosphamide is the elective drug of choice
in the treatment of severe manifestations of systemic
lupus erythematosus, such as proliferative nephritis,
affection of the central nervous system, pneumonitis
or severe thromobocytopenia [59].
Cyclophosphamide-treated lupus patients have
premature ovarian failure rates of up to 50% in women
below 30 years and 60% in women between 30 and 40
years of age [60]. In a manner similar to onocological
patients, as survival rates have increased ovarian failure and infertility have become important issues for
these patients.
The use of intermittent cyclophosphamide has
been proposed due to its successful application in
systemic lupus erythematosus and the observed reduction in toxicity. However, there is continued controversy over its results, and it remains unclear if secondary effects of cyclophosphamide are influenced
Table 3.2 Oocyte vitrification with non-oncological indication: Instituto Valenciano de Infertilidad (IVI), Valencia, Spain
No. patients
Age
33 3.2
20 (10 4)
Postponing childbearing
65
34 6.1
67
35.8 4.2
65
36 4.2
18
35 4.1
OHSS
12
32 4.3
Autoimmune diseases
348
39.5 3.5
28
35.5 3.3
543
36.7 3.2
Low response
ART, assisted reproductive technology; PGS, pre-implantation genetic screening; FISH, fluorescence in-situ hybridization; PCR, polymerase
chain reaction; OHSS, ovarian hyperstimulation syndrome.
As described by Oktay, letrozole stimulation consists of oral administration of 5 mg of letrozole beginning on the 2nd or 3rd day of the cycle and continuing until the day of human chorionic gonadotropin
(hCG) administration. After 2 days of letrozole administration, 150 U of gonadotropins are added. A GnRH
antagonist is administered when a follicle reaches 14
15 mm or the E2 level exceeds 250 pg/ml, and hCG
is given when the leader follicles are 20 mm. Letrozole administration is continued until the appearance
of menses [63].
29
Section 1: Introduction
30
r
r
Conclusions
For fertility clinics, advances in fertility preservation
procedures have created a new group of patients who
seek to use testicular and ovarian preservation techniques in order to prevent infertility. Specialists should
inform patients about the new approaches that will
allow them to attempt a pregnancy in the future with
their own oocytes, regardless of whether they experience ovarian failure.
References
1. National Cancer Institute, Division of Cancer
Control and Population Sciences. Surveillance,
Epidemiology and End Results Program, 19752003.
Bethesda MD: National Cancer Institute, 2006.
2. Thomson A, Critchley H and Wallace W. Fertility and
progeny. Eur J Cancer 2002; 38: 163444.
3. Linet M, Ries L, Smith MA et al. Cancer surveillance
series: recent trends in childhood cancer incidence and
mortality in the United States. J Natl Cancer Inst 1999;
91: 10518.
4. Blumenfeld Z and Eckman A. Tratamientos
oncologicos y supervivencia folicular. In: Schneider J,
Garca-Velasco JA (eds.), Cancer y reproduccion.
Cuadernos de Medicina Reproductiva. Madrid: Adalia
farma, 2005: pp. 3343.
31
Section 1: Introduction
32
33
Section 1: Introduction
34
Section 2
Chapter
Introduction
Cancer describes a group of well over 200 diseases,
each with distinct and heterogeneous molecular aberrations that result in a breakdown of cellular mechanisms that govern cell growth, death and differentiation. From the early observation of chromosomal
abnormalities in cancer cells, then the identification of
the first proto-oncogene, and now the detailed analyses of specific genes and signaling networks, we have
come to recognize cancer as an astoundingly complex
acquired genetic disease.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
35
Time
Normal
Inactivation
of APC
Dysplasia
Activation
of KRAS
Inactivation
of SMAD2/4
In situ
malignancy
Invasive
carcinoma
Inactivation
of TP53
Genetic abnormalities
36
the progression of premalignant polyp to invasive adenocarcinoma (Figure 4.1) [8, 9].
Adult
stem cell
Anomalous
self-renewal
Lineage
comitted
progenitor cells
Terminally
differentiated
cells
Tumor
subtype 2
Tumor
subtype 3
Differentiation
atio
or m
nsf
Tra
Multipotent
progenitor
cell
Anaplasia
Tumor
subtype 1
Physiological
self-renewal
Tumor
subtype 4
The long-lived resident tissue stem cell, however, provides an elegant target for mutational transformation. By slowly amassing mutations throughout its
lifetime (i.e. the lifetime of the organism), this cell
may reach the threshold for tumorigenic conversion.
Once transformed, this cell is thought to continue to
undergo symmetric and asymmetric division and generate lineage-committed progeny, albeit the hierarchical nature of this differentiation is perturbed (Figure
4.2). The retention of self-renewal capacity only in the
CSC is responsible for the limited tumorigenic potential of the cells which comprise the bulk of the tumor.
Recent evidence now suggests that lineage-committed
progenitors which have gained self-renewal capacity
may also be potential sources of CSCs.
37
overexpression of the epidermal growth factor receptor tyrosine kinases EGFR and ERBB2 enables receptor dimerization and constitutive mitogenic signaling
in the absence of ligand [16]. Alternatively, tumor cells
commonly secrete soluble growth factors that act in a
paracrine manner to induce activation of membranebound growth receptors and promote entry into the
cell cycle [17].
38
B; CD
C
M
DNA
damage
p27Kip1
p53
TGF
p16INK4A
Mitogens
p21Cip1
G1
MDM2
R point
p15INK4B
D;
CDK4/6
p57Kip2
A; CDC
G2
p18INK4C
p19INK4D
pRb
p14ARF
A;
CD
K2
E; CDK2
E2F1/2/3
Myc
Figure 4.3 Schematic representation of the cell cycle and checkpoint regulatory system.
Aberrant differentiation
Mammalian organogenesis proceeds through an elegant hierarchical establishment of cellular and functional components. Each step of this process entails
an increasing degree of cellular commitment that is
driven by changes in transcriptional and/or epigenetic
programs. The end-product of this process is the formation of terminally differentiated cellular components that are specialized to perform specific biological functions. In normal tissue morphogenesis, this
process is tightly regulated by growth factors, morphogens and signals derived from cellular and noncellular components of the microenvironment. The
CSC theory of cancer operates under the premise
that cells within this hierarchical arrangement are targets of transforming mutations and that these different cellular origins account for the heterogeneity observed in human cancers. Well before the CSC
theory gained popularity, dysregulated growth pat-
39
as demonstrated by studies of patients with inherited defects in DNA-repair genes, such as xeroderma
pigmentosum (defect in nucleotide excision repair
[NER]), Nijmegen breakage syndrome (defect in NBS
and processing of double-stranded DNA [dsDNA]
breaks), ataxia telangiectasia (defect in ATM and
homology-directed repair [HDR] of dsDNA breaks)
and familial breast and ovarian cancers (defect in
BRCA1 or BRCA2 and HDR of dsDNA breaks).
DNA damage can result from endogenous biochemical processes (e.g. depurination, deamination, oxidation) and exogenous carcinogens (e.g. radiation, alkylating agents, heterocyclic amines). Cells employ a
variety of mechanisms to repair DNA damage, including mismatch repair (MMR) enzymes that recognize
normal but misincorporated nucleotides and other
enzymes that detoxify (e.g. glutathione-S-transferase)
or remove DNA adducts (e.g. O6 -methylguanine DNA
methyltransferase). Additionally, base excision repair
(BER) recognizes chemically altered bases caused by
endogenous sources, NER recognizes helix-distorting
adducts from exogenous sources, HDR of dsDNA
breaks utilizes the undamaged, homologous DNA
sequence to direct repair and the more error-prone
non-homologous endjoining (NHEJ) results in fusion
of two dsDNA ends [41]. One anti-cancer treatment
approach takes advantage of the failure of DNA damage repair. Poly (ADP-ribose) polymerase (PARP) is
an enzyme that participates in BER, a mechanism
that cancer cells with mutations in BRCA1 or BRCA2
depend on to repair their DNA damage. These cancer
cells become hypersensitive to chemotherapy and radiation when PARP inhibitors are given to prevent repair
by this mechanism as well [42].
Hallmarks of cancer
Despite the tremendous molecular heterogeneity
observed in human cancers, malignant cells exhibit a
common set of behaviors regardless of their tissue of
origin or the specific genomic insults that they have
sustained. These hallmarks of cancer [43] (Figure 4.4)
are the biological manifestations of the molecular
anomalies that exist within cancer cells.
40
Insensitivity to
anti-growth signals
Limitless
replicative
potential
Self-sufficiency
in growth signals
Genomic
instability
CANCER
Immune
evasion
Evasion
of apoptosis
Sustained
angiogenesis
Invasion and
metastasis
in human cancers and largely compromises the antigrowth effects of TGF because release of E2F1/2/3
is no longer dependent upon formation of a cyclin
D-CDK4/6 complex (Figure 4.3). Alternatively, cancer
cells have been demonstrated to mutate the negative
regulatory sequences in the Myc promoter, resulting
in constitutive expression of this gene, and to inactivate the genes encoding the Smad transcription factors
or the TGF receptors, both of which are necessary to
transduce anti-growth signals into the nucleus [43].
Evasion of apoptosis
Inactivation of the apoptotic machinery permits the
survival of cells with accumulating mutations and promotes evolution of premalignant to malignant cells.
One strategy that is frequently encountered is inactivation of the p53 pathway. This can result from
mutations in the TP53 gene itself (which normally
induces pro-apoptotic genes), by deletion or promoter
methylation of the ARF gene (which normally blocks
MDM2 action) or by overexpression of MDM2 (which
normally blocks p53 action) (Figure 4.3). Additionally, other components of the apoptotic machinery are
altered, including promoter methylation of the APAF1
gene (which normally assembles with cytochrome c
to form the apoptosome and activate caspase 9), inactivation by mutation of the pro-apoptotic BAX gene
and increased expression of the pro-survival BCL2
gene. Finally, hyperactivation of the PI3KAKT/PKB
pathway by PTEN inactivation (phosphatase which
removes the 3 phosphate group from PIP3 that was
added by the kinase PI3K), or IGF-1/2 overexpression (which activates PI3K) also allows cancer cells to
acquire resistance to apoptosis [43].
Sustained angiogenesis
A tumors ability to attract blood vessels is essential
for its continued growth, otherwise tumor cells located
further from the vasculature experience hypoxia and
may enter apoptosis or become necrotic. The Rip-Tag
transgenic mouse model of islet cell tumor progression
has provided researchers a way to study this angiogenic switch, which involves the heterotypic interactions among the premalignant islet cells, inflammatory cells and endothelial cells [50]. The islet cells produce vascular endothelial growth factor (VEGF) that
is then sequestered by the surrounding extracellular
matrix (ECM) as well as still-unidentified signals that
recruit mast cells and macrophages. The inflammatory cells produce matrix metalloproteinase (MMP) 9
that cleaves specific components of the ECM to release
VEGF for signaling. The endothelial cells then proliferate and form the tumor vasculature in response to activated VEGF. However, the tumor-associated endothelial cells tend to form a poorly organized vasculature
41
42
Genomic instability
A cell has a variety of mechanisms to ensure the structural integrity of its DNA, including enzymes that
detoxify mutagenic molecules and proteins that recognize and repair the damage. Cancer susceptibility
increases when these repair processes are affected. This
can occur by inherited or somatic mutation of key
proteins or epigenetic alterations such as promoter
methylation that leads to functional inactivation of key
proteins. For example, MMR defects prevent the detection and repair of sequence mismatches in genes, such
as the type II TGF receptor that have microsatellite
repeats in their sequences. In this case, it introduces
a nonsense mutation, which results in a truncated
receptor that can no longer function in its growthinhibitory signaling. Changes in chromosome structure as a result of translocations or fusions of unrelated chromosomes as well as changes in chromosome
Basic immunology
The immune system has two major features: immune
recognition and immune response. Immune recognition is the ability to distinguish foreign invaders from
self-components, which then leads to the immune
response in which the foreign invader (or non-self
component) is eliminated. The immune response
to a foreign invader involves both innate immunity and adaptive immunity. Innate immunity is the
bodys first line of defense against a foreign invader.
The innate immune response is rapid and fixed and
includes anatomic barriers (e.g. skin), inflammation
and soluble molecules (e.g. interferons [IFNs]). The
major cellular components of an innate immune
response include neutrophils, macrophages, dendritic
cells and natural killer (NK) cells. The adaptive
immune response is the bodys second line of defense
that develops in response to a foreign invader. The
adaptive immune response is a delayed response that
demonstrates antigen specificity, diversity, immunological memory and self/non-self recognition. The
adaptive immune response can be further divided into
humoral and cell-mediated responses. In an adaptive humoral immune response, B lymphocytes interact with their specific antigen (pieces of a foreign
invader); differentiate into plasma cells, which then
secrete antigen-specific antibodies. The secreted antibodies bind to their specific antigen and facilitate
clearance of that antigen. In an adaptive cell-mediated
immune response, T lymphocytes recognize their specific antigen, resulting in the secretion of various
cytokines. Cytokines direct numerous cellular activities, including activation of T helper (TH ) or T cytotoxic (TC ) lymphocytes, leading to cell-mediated toxicity. The major cellular components of the adaptive
immune response are T lymphocytes, B lymphocytes,
NK cells and antigen-presenting cells (e.g. dendritic
cells).
Tumor immunology
The immune systems ability to distinguish self- from
non-self components and react only to non-self components is crucial to an appropriate immune response.
Failure in the ability to distinguish self from non-self
43
44
Immunotherapy
Manipulation of the immune system is an attractive
approach for treatment of many cancers. There are
numerous immune mechanisms to exploit, including passive cellular, passive humoral, active specific
and non-specific mechanisms. The goals would be
to stimulate anti-tumor response, decrease suppressor mechanisms and/or increase tumor immunogenicity [80].
Adoptive T-cell therapy is an example of passive cellular immunotherapy. Adoptive T-cell therapy
involves the infusion of T cells derived from autologous or allogeneic sources with the goal of eliminating a tumor and preventing its recurrence [81]. T cells
can be stimulated and expanded ex vivo prior to infusion in order to enhance their ability to react to a
tumor. T cells can also be genetically engineered to
express chemokine receptors to promote trafficking to
the tumor [82].
Passive humoral immunotherapy includes administration of exogenous monoclonal antibodies. These
antibodies can block function, enhance function or be
conjugated with toxins or radioisotopes. For example,
antibodies to immune inhibitory signals such as PDL1 or CTLA4 prevent the repression of the immune
response and have been shown to enhance tumor
regression in mice [83].
Active specific immunotherapy involves infusion
of autologous or allogeneic tumor cells with the goal
to enhance anti-tumor response. This mechanism also
includes ex vivo loading of dendritic cells with tumor
antigen followed by infusion into a cancer patient
[84]. Further studies have demonstrated that dendritic
cells which have been engineered to express transgenic
tumor antigens or chemokines are more potent inducers of anti-tumor immunity when compared to loaded
dendritic cells alone [85].
Non-specific immunotherapy includes administration of cytokines such as interferon- (IFN ), IL-2
or IL-12 in order to activate macrophages and NK cells
and enhance anti-tumor activity [86]. Synthetic CpG
oligodeoxynucleotides (CpG ODNs) are agents that
have been demonstrated to stimulate both innate and
adaptive immunity by enhancing antibody dependent
cellular cytotoxicity (ADCC) or serving as an adjuvant
to elicit an anti-tumor immune response [87].
A major frustration in effective immunotherapy regimens are Tregs. However, there are a number of mechanisms that have been demonstrated to
decrease Treg activity, clearing the way for an effective anti-tumor immune response. For example, low
dose cyclophosphamide treatment selectively depletes
Tregs, denileukin diftitox (an IL-2-diptheria toxin
fusion protein) reduces the percentage of Tregs in the
peripheral blood of cancer patients and daclizumab
(a CD25 monoclonal antibody) inhibits Tregs in
metastatic breast cancer patients who also received a
multipeptide cancer vaccine [88].
Harnessing the immune system to generate an
effective, long-lasting anti-tumor response is a major
challenge. Continued efforts will make attempts to tip
the balance towards the elimination or equilibrium
phases of immune surveillance (Figure 4.5).
References
Concluding remarks
Human malignancies are the result of acquired genetic
changes that deregulate normal cellular proliferation,
differentiation and death. Though we have now characterized hundreds of genes and molecular processes
that underlie the development and progression of cancer, a comprehensive understanding of tumor biology
remains a distant reality. The new paradigm of cancer
stem cell theory has revealed that many of the main-
45
46
47
48
Section 2
Chapter
of life, particularly if there was little discussion regarding alternatives [1518]. Women under 40 years of age
are likely to be offered genetic counseling and testing,
and this is increasingly performed prior to definitive
surgery. The emergence of neoadjuvant chemotherapy shortly after biopsy and before definitive surgery
for women with stage II or higher tumors increases
the complexity and reduces time for standard fertility preservation procedures such as controlled ovarian
stimulation (COS) and oocyte retrieval. How can decisions regarding fertility preservation be incorporated
into the already crowded and emotionally charged
interval immediately following diagnosis?
In this chapter we will cover those issues most likely
to be raised by young women who have been recently
diagnosed with breast cancer or those at high risk of
the disease contemplating assisted fertility procedures.
Questions likely to be posed by women with a
recent cancer diagnosis are: (1) Are the benefits from
chemotherapy worth the possible loss of fertility? (2)
What are the chances that the planned treatment will
result in menopause or the loss of ability to become
pregnant? (3) If I am still able to conceive will a subsequent pregnancy alter my prognosis? (4) If you alter
my treatment to help preserve fertility will it change
my prognosis? (5) How much do fertility preservation
procedures cost and are they covered by insurance?
(6) How is fertility preservation orchestrated with the
rest of my treatment? Young women at increased risk
of breast cancer want to know if ovarian stimulation
is likely to further increase their already elevated risk
of breast cancer. All members of the treatment team
should be able to provide at least a general response to
these questions.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
49
50
Regimen
AC
FAC
FEC
CEF
ACT
TAC
CMF
Cycles
Dose
Adriamycin
60 mg/m2
Cyclophosphamide
600 mg/m2
4 and 4
6 or 12
Frequency
5 Fluorouracil
500 mg/m2
Adriamycin
50 mg/m2
Cyclophosphamide
500 mg/m2
5 Fluorouracil
500 mg/m2
Epirubicin
100 mg/m2
Cyclophosphamide
500 mg/m2
Cyclophosphamide
75 mg/m2
D1D14
Epirubicin
60 mg/m2
D1 and D8
5 Fluorouracil
500 mg/m2
D1 and D8
Adriamycin
60 mg/m2
Every 3 weeks
Cyclophosphamide
600 mg/m2
Docetaxel (Taxotere)
100 mg/m2
Every 3 weeks
or Paclitaxel
175 mg/m2
Docetaxel (Taxotere)
75 mg/m2
Every 3 weeks
Adriamycin
50 mg/m2
Cyclophosphamide
500 mg/m2
Every 3 weeks
Cyclophosphamide
600 mg
D1 and D8
Methotrexate
40 mg/m2
D1 and D8
5 Flunoorouracil
600 mg/m2
D, day.
hormone (GnRH) agonist to chemotherapy tamoxifen improves disease-free survival by a relative factor
of 25% and may also improve overall survival [19,
2224]. Use of a GnRH agonist or removal of the
ovaries is probably most effective in women who are
still menstruating or who have premenopausal levels
of estradiol following chemotherapy [25].
51
52
ER+ BrCa
Good ovarian
reserve
Less favorable
prognosis
Unfavorable risk
Ovary harvest
with cryopreservation
chemotherapy followed by
510 years anti-hormone
therapy + GnRha
Intermediate risk
Consider COS with
letrozole + GnRHa +
oocyte retrieval 5 years
tamoxifen + GnRHa
Good prognosis
Figure 5.1 Fertility preservation in women with estrogen receptor positive (ER+) tumors. BrCa, breast cancer; COS, controlled ovarian
stimulation; GnRHa, gonadotropin-releasing hormone agonist.
53
ERBrCa
Good ovarian
reserve
Less favorable
prognosis
Immediate chemotherapy
with cyclophosphamide
containing regimen
pretreatment ovarian
harvest with
cryopreservation
Good prognosis
* Neoadjuvant chemotherapy
Two cycles of non-cyclophosphamide
regimen
oocyte retrieval, followed by
adjuvant chemotherapy and
surgery
Definitive surgery
COS with letrozole +
GnRHa and oocyte retrieval
adjuvant chemotherapy
54
Cyclophosphamide
Worst
Carboplatin
Cisplantin
Anthracyclines
Intermediate
Taxanes
Vincas
Antimetabolites
Least
over 1216 weeks can count on adding an approximate 10 years to her ovarian reproductive age, such
that if she is aged 30 at the time her chemotherapy
is initiated, her ovarian reproductive age will be the
equivalent of a 40-year-old woman when her treatment
is completed. While she is likely to resume menses,
her capacity for a birth without assisted reproductive techniques will be marginal. The mechanism of
ovarian toxicity resulting from chemotherapy is not
completely understood, but it is likely to result from
apoptotic changes in pre-granulosa cells that subsequently develop into follicles [56, 57]. Recent investigations suggest that women with greater levels of
neutropenia (often associated with higher doses and
more cycles) are more likely to have permanent amenorrhea as are women with certain single nucleotide
polymorphisms in enzymes responsible for cyclophosphamide metabolism [58, 59]. Drugs more and less
likely to be associated with amenorrhea are given in
Table 5.2.
Rates of amenorrhea that have been observed with
regimens given to good-prognosis node negative premenopausal women are 33% for 6 cycles of CMF and
4 cycles of AC either dose dense or non-dose dense
(there are no published rates for the 4 cycle taxotere
and cyclophosphamide combination) [6065].
Rates of amenorrhea for chemotherapy regimens
often given to high-risk node negative or node positive women such as 6 cycles of FEC or FAC, 6 cycles
of AC or 4 cycles of AC followed by 4 cycles of taxotere are generally double that of the good prognosis
regimens ranging from 5065% [6670]. However, it
appears that giving only three cycles of FEC followed
by thre cycles of taxotere may have less ovarian toxicity
than six cycles of FEC [20]. Fifteen to fifty percent of
women younger than age 40 at diagnosis will eventually resume menses. Recovery rates are higher for regimens with less total cyclophosphamide. Amenorrhea
is likely to be permanent in 90% of women aged over 40
55
cost not included) and ovarian harvest and cryopreservation is $6000. Other procedures are available but are
rarely utilized at present [77]. Costs for fertility preservation are not covered by most insurance carriers and
so it may be difficult for young women to come up with
funds on short notice. In addition, in order for embryo
cryopreservation to occur, there must be both egg and
sperm available and for many single women this adds
an additional short-term challenge.
56
References
1. Anders CK, Johnson R, Litton J et al. Breast cancer
before age 40 years. Semin Oncol 2009; 36: 23749.
2. SEER Cancer Statistics Review, 2008 update.
http://www.seer.cancer.gov.
3. American Cancer Society. Cancer Facts and Figures.
Atlanta GA: American Cancer Society, 2009.
4. National Center for Health Statistics. Vital Statistics
of the United States, 1993, vol. 2, Mortality, Part A.
Washington DC: Public Health Service, 1993.
5. Partridge AH, Gelber S, Peppercorn J et al.
Web-based survey of fertility issues in young
women with breast cancer. J Clin Oncol 2004; 22:
417483.
6. Mueller BA, Simon MS, Deapen D et al. Childbearing
and survival after breast carcinoma in young women.
Cancer 2003; 98: 113140.
7. Cvancarova M, Samuelsen S, Magelssen H et al.
Reproduction rates after cancer treatment: experience
from the Norwegian Radium Hospital. J Clin Oncol
2009; 27: 33443.
8. Rosenberg R and Schwartz RL. Breast cancer in
women younger than 40 years. Int J Fertil 2003; 48:
2005.
9. Anders CK, Hsu DS, Broadwater G et al. Young age at
diagnosis correlates with worse prognosis and defines
a subset of breast cancers with shared patterns of gene
expression. J Clin Oncol 2008; 26: 332430.
10. Fredholm H, Eaker S, Frisell J et al. Breast cancer in
young women: poor survival despite intensive
treatment. PLoS One 2009; 4: e7695.
57
58
59
60
61
Section 2
Chapter
Age at
diagnosis (years)
1519
0.2
White
0.2
Black
2024
1.4
1.3
1.9
2529
8.1
7.8
11.2
3034
25.6
25.3
30.4
3539
58.5
58.0
64.4
4044
118.4
119.3
119.9
4549
185.3
188.1
178.7
5054
229.0
234.7
224.1
5559
288.5
296.1
282.7
6064
351.9
366.8
321.4
6569
394.4
416.5
351.2
7074
415.4
435.9
382.3
7579
441.9
465.7
387.6
8084
428.7
447.9
376.7
85+
342.1
350.5
335.8
From Horner et al. [2], based on November 2008 SEER data submission, posted to the SEER website, 2009.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
62
All races
63
64
Table 6.2 Reported rates of amenorrhea with common breast cancer regimens
Regimen
No. of cycles
CMF (cyclophosphamide,
methotrexate, fluorouracil)
312
1865
7497
Variable
3246
73100
4 AC/EC 4 taxane
646
3586
65
66
tion in order to receive several years of hormonal therapy may also affect a womans chance of successful conception. To date, no randomized studies have examined the effect of hormonal therapy on pregnancy outcome [45]. Given the lack of data, many patients with
hormone receptor-positive breast cancer who desire
pregnancy, and their treating physicians, struggle with
whether to shorten the duration of endocrine therapy
or to temporarily interrupt endocrine therapy in order
to achieve pregnancy.
Embryo cryopreservation
To date, embryo cryopreservation is considered the
more effective approach to fertility preservation. It has
been routinely used for storage of surplus embryos
after in vitro fertilization treatment for infertility. The
post-thaw survival rate of embryos is in the range of
3590%. Successful implantation rates range from 35
to 90%, and if multiple embryos are stored, cumulative pregnancy rates can be 60% [48]. This approach
requires either a male partner or sperm donor. Typically, the patient undergoes 2 weeks of ovarian stimulation with daily injections of follicle stimulating hormone (FSH) beginning at the onset of menses. Follicle
development is monitored by serial ultrasound imaging and blood tests. At the appropriate time, human
chorionic gonadotropin is administered to stimulate
ovulation and oocytes are collected using transvaginal aspiration under ultrasound guidance. Oocytes are
Oocyte preservation
Cryopreservation of unfertilized oocytes is an option
for women for whom a partner is unavailable. The
ovarian stimulation and harvesting process is identical to that used for embryo cryopreservation, but
the oocytes are stored unfertilized and then subsequently thawed and fertilized in vitro. Thus, this technique is associated with the same issues regarding timing, potential delay of chemotherapy and exposure to
hormones. Unfortunately, oocyte freezing is technically more complex than embryo cryopreservation and
unfertilized oocytes are more prone to damage during cryopreservation; thus, the overall successful pregnancy rate is likely lower with this procedure [51].
2000 [52], and since then there have been several case
reports of successful pregnancies resulting from these
procedures [5355]. Ovarian tissue is removed laparoscopically (this procedure requires general anesthesia)
and frozen, and can be thawed and re-implanted at a
later date, either orthotopically (in the pelvis) or heterotopically (in subcutaneous tissue in the forearm or
abdomen); studies have reported restoration of ovarian function with both methods. Although the cryopreservation technique is highly effective, a large number of follicles may be lost due to ischemia at the
time of re-implantation, so typically the cortex from
an entire ovary is used.
One hypothethical concern with this method is the
potential for re-introduction of metastatic cancer cells.
However, in breast cancer patients without evidence
of systemic disease, the likelihood of occult ovarian
metastases appears to be extremely low, and in the limited reported literature, no cases of cancer recurrence
after ovarian transplantation have been noted [46].
67
function preservation in patients receiving chemotherapy (n = 62) compared with retrospective controls
(n = 55), the percentage of patients resuming ovulation and menses was significantly higher in the GnRH
group [58]. In the first prospective randomized trial
of this strategy, 17 women were randomized to either
buserelin or control prior to and during chemotherapy; at 3 years, 4 of 8 women receiving buserelin
and 6 of 9 controls were amenorrheic. In a more
recent randomized study that included 80 women
receiving chemotherapy for breast cancer, the addition of GnRH agonist prior to and during chemotherapy significantly improved the rate of resumption
of menses (89.6% versus 33.3%) and spontaneous
ovulation (69.2% versus 29.6%) [59]. Though compelling, these data require confirmation; the Southwest Oncology Group (SWOG) is currently conducting SWOG S0230 a randomized trial with an accrual
goal of 458 patients evaluating GnRH agonists for
ovarian function preservation in women with hormone receptor-negative breast cancer who receive
chemotherapy [46].
68
significantly [70]. Annual risk of recurrence for hormone receptor-positive tumors is more stable, and
is relatively constant from years 15 and then drops
slightly but stays constant from years 512. As younger
women are more likely to develop hormone receptornegative disease, these risks must be considered when
making decisions regarding pregnancy. With regard
to hormone-receptor positive disease, it is important to note that the currently recommended duration of adjuvant endocrine therapy for premenopausal
women is 5 years, and the decision to attempt conception necessitates discontinuation of endocrine therapy.
Thus, the decision regarding when to attempt conception should be individualized.
mine which modalities can successfully aid in protecting and aiding reproductive potential in women
undergoing breast cancer therapy who desire future
fertility.
References
1. Jemal A, Siegel R, Xu J and Ward E. Cancer statistics.
CA Cancer J Clin 2010; 60(5): 277300.
2. Horner MJ, Ries LAG, Krapcho M et al. (eds.). SEER
Cancer Statistics Review 19752006. Bethesda MD:
National Cancer Institute. http://seer.cancer.
gov/csr/1975 2006/.
3. Brinton LA, Sherman ME, Carreon JD and Anderson
WF. Recent trends in breast cancer among younger
women in the United States. J Natl Cancer Inst 2008;
100(22): 16438.
4. Fredholm H, Eaker S, Frisell J et al. Breast cancer in
young women: poor survival despite intensive
treatment. PLoS One 2009; 4(11): e7695.
5. Collaborative Group on Hormonal Factors in Breast
Cancer. Familial breast cancer: collaborative reanalysis
of individual data from 52 epidemiological studies
including 58,209 women with breast cancer and
101,986 women without the disease. Lancet 2001;
358(9291): 138999.
6. Lindor NM, McMaster ML, Lindor CJ and Greene
MH. Concise Handbook of Familial Cancer
Susceptibility Syndromes, 2nd edn. New York: Oxford
University Press, 2008.
7. Antoniou A, Pharoah PD, Narod S et al. Average risks
of breast and ovarian cancer associated with BRCA1 or
BRCA2 mutations detected in case series unselected
for family history: a combined analysis of 22 studies.
Am J Hum Genet 2003; 72(5): 111730.
8. Liu Q, Wuu J, Lambe M et al. Transient increase in
breast cancer risk after giving birth: postpartum
period with the highest risk (Sweden). Cancer Causes
Control 2002; 13(4): 299305.
9. Lambe M, Hsieh C, Trichopoulos D et al. Transient
increase in the risk of breast cancer after giving birth.
N Engl J Med 1994; 331(1): 59.
10. Albrektsen G, Heuch I, Thoresen S and Kvale G.
Family history of breast cancer and short-term effects
of childbirths on breast cancer risk. Int J Cancer 2006;
119(6): 146874.
11. Lyons TR, Schedin PJ and Borges VF. Pregnancy and
breast cancer: when they collide. J Mammary Gland
Biol Neoplasia 2009; 14(2): 8798.
12. Ives AD, Saunders CM and Semmens JB. The
Western Australian gestational breast cancer project: a
population-based study of the incidence, management
and outcomes. Breast 2005; 14(4): 27682.
69
70
71
72
Section 2
Chapter
Background
There are currently an estimated 270 000 survivors
of childhood cancer in the USA [1]. The 5-year survival rate for all childhood cancers is approximately
80% [2], but survival is often associated with a cost.
Treatment with past and contemporary regimens of
chemotherapy and radiation can affect future fertility. Sterility, infertility or subfertility can result from
gonadal removal or damage to germ cells from adjuvant therapy. Damage to the gonads by irradiation
or chemotherapy depends upon the cancer survivors
gender, age at time of treatment, dose of radiation and
fractionation schedule, and total dose and nature of
chemotherapy given [3, 4]. This chapter reviews pediatric cancer therapy and its consequences on fertility.
Effects of chemotherapy
Chemotherapy can cause infertility, premature ovarian failure, menstrual irregularity and delayed puberty.
The effects of chemotherapy on ovarian function are
both agent and dose-dependent, and this effect may be
additive to that resulting from abdominopelvic radiotherapy. Alkylating agents such as cyclophosphamide
affect the resting oocyte in a dose-dependent, cell
cycle-independent manner [10] by affecting undeveloped oocytes and possibly pre-granulosa cells of primordial follicles [11]. The results of alkylating agents
are more pronounced in post-pubertal as compared
to pre-pubertal females, due to the fact that postpubertal females have fewer remaining oocytes. Risks
of menstrual irregularity, ovarian failure and infertility increase with age at treatment, as the normal
aging process is accompanied by an ongoing depletion of oocytes. Amenorrhea and premature ovarian
failure occur more commonly in adult women treated
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
73
74
Effects of radiation
Abdominal, pelvic and total body irradiation (TBI)
may result in ovarian and uterine damage. Damage
induced by radiotherapy results in progressive and
irreversible damage in the ovary, causing amenorrhea
and infertility. The oocyte is very sensitive to radiation
and undergoes irreversible damage at 2 Gy in 50% of
patients [26]. Radiation causes a decrease in the number of ovarian follicles, impaired follicular maturation,
cortical fibrosis and atrophy, generalized hypoplasia
and hyalinization of the capsule.
The frequency of ovarian failure following abdominal radiotherapy is related to the age at time of irradiation and the radiation therapy dose received by the
ovaries. Females treated prior to the onset of puberty
have a greater number of ova than older females;
thus ovarian function is more likely to be preserved
after radiotherapy in pre-pubertal females compared
to post-pubertal females [27, 28]. Whole abdominal radiotherapy produces severe ovarian damage.
Wallace et al. demonstrated that 25% of female
childhood cancer survivors who received abdominal
irradiation therapy doses of 20003000 cGy had premature menopause before the age of 16 years [29].
Minimum ovarian
dose (Gy)
Effect
0.6
None
1.5
2.55.0
58
Effects of chemotherapy
Testicular dysfunction is among the most common
long-term side effect of chemotherapy in men. The
germinal epithelium is very susceptible to injury by
cytotoxic drugs secondary to its high mitotic rate.
Compared with the germinal epithelium, Leydig cells
are relatively resistant to the effects of chemotherapy
[3739]. However, with more intensive gonadotoxic
regimens, reductions in testosterone concentrations
have been noted that may be clinically relevant.
Similar to females, regimens that include high
doses of alkylating agents are the most toxic to gonadal
tissue. Total dose of cyclophosphamide is one factor that influences the severity of testicular damage. In survivors of Ewings and soft tissue sarcoma,
who received a cumulative dose of cyclophosphamide
7.5 g/m2 , there was an increased risk of persistent oligo- or azoospermia [40]. In studies of male
childhood survivors of acute lymphoblastic leukemia
(ALL), Blatt et al. reported normal testicular function in boys treated for ALL with therapy that did
not include cyclophosphamide or intravenous cytosine arabinoside [41]. Drugs such as procarbazine,
cyclophosphamide and chlorambucil have been shown
to produce prolonged azoospermia in 90% of men
[4244]. Combination chemotherapy that includes an
75
alkylating agent and procarbazine causes severe damage to the testicular germinal epithelium [42, 4446].
Studies of adolescent and young adult male survivors of Hodgkins lymphoma have shown that both
the chemotherapeutic regimen and dose intensity are
important variables affecting reproductive potential
[4, 44, 47]. Those treated with 6 cycles of chemotherapy that included nitrogen mustard, vincristine, prednisone and procarbazine had 90% of infertility due
to azoospermia [44, 47]. However, the adolescent and
young adult males who received 3 or fewer cycles of
identical therapy had only a 50% rate of infertility
[47]. In a study examining more contemporary therapy
in Hodgkins lymphoma, authors found that patients
treated with a regimen of adriamycin, bleomycin, vinblastine and dacarbazine had a 33% risk of infertility [4]. In a recent cohort study conducted by the
European Organization for Research and Treatment
of Cancer, exposure to alkylating chemotherapy was
associated with a significantly higher risk of gonadal
dysfunction among male patients and longer recovery
time of gonadal function [48].
Chemotherapy that includes platinum compounds
can cause prolonged azoospermia in up to 50% of men
[49]. Conditioning regimens that include high doses of
alkylating agents and TBI used for stem cell transplantation cause prolonged azoospermia in more than 50%
of survivors [50, 51].
Chemotherapy appears to lower healthy sperm
counts in cancer survivors but, after an adequate time
off of therapy, some studies suggest that DNA integrity
of sperm is re-established similar to age-matched controls; the amount of time off therapy required for this
recovery has not been adequately quantified [52].
Effects of radiation
When testes are exposed to radiation, sperm count
begins to decrease. Dependent upon dosage, temporary or permanent sterility may result [53]. The degree
and permenancy of radiotherapy-induced testicular
damage also depends upon the treatment field and
fractionation schedule. Table 7.3 shows the effects of
fractionated radiation on spermatogenesis and Leydig
cell function [54]. Males who receive radiation to the
abdominal or pelvic region may still regain partial or
full sperm production depending upon the amount of
injury to the testes. Unlike the germinal epithelium,
Leydig cell function may be more prone to damage
from irradiation in pre-pubertal life than adulthood
76
[55]. Testicular radiation with doses 20 Gy is associated with Leydig cell dysfunction in pre-pubertal boys,
while Leydig cell function is usually preserved with
doses of as much as 30 Gy in sexually mature males.
Exposing the testes to ionizing radiation at a dose
6 Gy causes disturbances of spermatogenesis and
altered spermatocytes with recovery periods dependent on dose [54]. Doses 6 Gy cause permanent
infertility by destroying all stem cells [56].
The testes are directly irradiated in situations such
as testicular relapse in ALL, and the high doses of
radiotherapy required (often 2400 cGy) results in both
sterilization and Leydig cell dysfunction [57]. Total
body irradiation used for stem cell transplantation
conditioning can cause permanent gonadal failure in
approximately 80% of males [21]. Craniospinal irradiation produced primary germ cell damage in almost
20% of children with ALL in a study that utilized
the Childhood Cancer Survivor Study [58]. This study
demonstrated the testes sometimes receive radiation
via body scatter. Scatter occurs when X-rays interact
with tissues near the target of interest, resulting in secondary X-rays that then hit the target [59]. The amount
of scattered radiation is a function of the proximity of
the radiation field to the target, the field size and shape,
the X-ray energy and the depth of the target. Of these,
distance from the field edge is the most important factor. Scatter dose to the testes may be an issue when
treating a field that extends into the pelvis.
Table 7.3 The effects of fractionated radiation on spermatogenesis and Leydig cell function
Effect on spermatogenesis
<10
No effect
No effect
1030
Temporary oligospermia
No effect
3050
No effect
50100
No effect
100200
No change in testosterone
200300
No change in testosterone
1200
Permanent azoospermia
2400
Permanent azoospermia
for oncologists to help address the possibility of infertility with patients treated during their reproductive
years [66]. The guidelines state that oncologists should
be prepared to discuss fertility preservation options or
to refer the patient to reproductive specialists for further information prior to initiation of therapy. However, there is no consensus on when the appropriate age
of reproductive potential actually occurs or at what age
patients should be referred to a reproductive specialist.
Preservation of fertility
The options for fertility preservation before treatment
differ between females and males. Males have more
available options that are less invasive, less expensive
and more effective.
technique [9]; this is the only non-experimental pretreatment fertility preservation option for adolescent
and young adult women. This intervention is technically complex and can delay initiation of cancer therapy by 24 weeks. This intervention is also limited to
females who are either involved in a stable relationship
or willing to identify a known or anonymous sperm
donor. These issues often preclude this intervention
as an option for adolescent females. Also, this technique requires ovarian stimulation, which precludes
it being an option for those females with estrogensensitive tumors.
Ovarian tissue cryopreservation is a process in
which normal, functioning ovarian tissue is excised
from the ovary and stored cryogenically and is the
only option that can be offered to pre-pubertal girls
[67]. Currently this technique is available only in certain parts of the USA as an experimental protocol. This
technique involves obtaining primordial follicles from
a laprascopic biopsy. The immature follicles are smaller
than mature oocytes and more tolerant to freezing and
thawing [6]. Hundreds of immature oocytes are cyropreserved without the necessity of ovarian stimulation
and subsequent delay of cancer treatment. Ideally, the
stored ovarian tissue is thawed and autotransplanted
into the donor once treatment has been completed.
Studies in humans are still in their infancy, though
there are scattered published case reports that demonstrate its efficacy; a recent paper details 8 years of
77
experience in adult female cancer patients with positive results [68, 69]. Kim et al. reported on four young
cancer survivors who had their ovaries removed prior
to the initiation of cancer therapy and then had a
heterotopic autotransplantation of their ovarian tissue
over the course of a few years after completing therapy. Their report concluded that ovarian function was
re-established in all three patients [69].
78
Role of physician
References
1. Hollen PJ and Hobbie WL. Establishing
comprehensive specialty follow-up clinics for
long-term survivors of cancer. Providing systematic
physiological and psychosocial support. Support Care
Cancer 1995; 3(1): 404.
2. Jemal A, Siegel R, Ward E et al. Cancer statistics, 2009.
CA Cancer J Clin 2009; 59(4): 22549.
3. Sklar C. Reproductive physiology and
treatment-related loss of sex hormone production.
Med Pediatr Oncol 1999; 33(1): 28.
4. Waring AB and Wallace WH. Subfertility following
treatment for childhood cancer. Hosp Med 2000; 61(8):
5507.
5. Jahnukainen K, Ehmcke J, Soder O et al. Clinical
potential and putative risks of fertility preservation in
children utilizing gonadal tissue or germline stem
cells. Pediatr Res 2006; 59(4): R407.
6. Pfeifer SM and Coutifaris C. Reproductive
technologies 1998: options available for the cancer
patient. Med Pediatr Oncol 1999; 33(1): 3440.
7. Kim JG, Suh CS, Kim SH et al. Insulin-like growth
factors (IGFs), IGF-binding proteins (IGFBPs), and
IGFBP-3 protease activity in the peritoneal fluid of
patients with and without endometriosis. Fertil Steril
2000; 73(5): 9961000.
8. Bines J, Oleske DM and Cobleigh MA. Ovarian
function in premenopausal women treated with
adjuvant chemotherapy for breast cancer. J Clin Oncol
1996; 14(5): 171829.
79
80
81
82
Section 2
Chapter
Introduction
Cancer is a rare disease in childhood. In developed
countries, only about 0.5% of all cases occur in children under 15 years of age. The incidence rates range
between 96 and 138/million children per year for
males and between 70 and 116/million children for
females [1]. Contrary to what is observed in adults,
epithelial cancers (carcinomas) are rare among children, and in this age group solid tumors are predominantly embryonal. Therefore, it is more appropriate to classify childhood cancers according to their
histology. According to the International Classification of Childhood Cancer [2], childhood tumors are
classified into 12 major diagnostic groups: leukemias,
lymphomas, central nervous system (CNS) tumors,
sympathetic nervous system tumors, retinoblastomas,
renal tumors, liver tumors, bone tumors, soft tissue sarcomas, germ cell tumors, epithelial tumors
and other and unspecified malignant cancers (Table
8.1 [2]). Leukemias, brain tumors and non-Hodgkins
lymphomas (NHLs) are the most frequent pediatric
cancers in developed countries, representing almost
60% of all cases, while in developing countries NHLs
are more common than brain tumors (Figure 8.1)
[3]. Each year, an estimated 160 000 cancers are
diagnosed in children worldwide, and an additional
240 000 cancers in adolescents and young adults (aged
1524 years) [1].
Descriptive epidemiology
Childhood cancer
According to the last edition of Cancer Incidence in
Five Continents, the highest cancer incidence rates,
for all tumor sites except skin, are observed in
Kuwait (non-Kuwaitis, 418/million), Italy (Brescia and
Naples, 250 and 234 new cases/million, respectively),
Brazil (Sao Paulo and Braslia, 224 and 219/million,
respectively), Switzerland (Vaud, 214/million), Croatia (203/million) and the USA (non-Hispanic white
population, 179/million) for males, while for females
high incidence is noted in Kuwait (non-Kuwaitis, 500
new cases/million), Brazil (Brasilia and Sao Paulo,
195 and 190/million, respectively), Cyprus (190/million), Italy (Salerno, 172/million), Germany (Munster, 168/million), Portugal (southern region, 160/million) and the USA (non-Hispanic white population,
155/million) (Figure 8.2a, b) [1]. On the other hand,
mortality rates are usually higher in medium-income
countries rather than in high-income countries, with
the highest rates (4 deaths/100 000 habitants per
year) observed in Ukraine, Republic of Moldova,
Romania, Cuba, Latvia, Russia, Estonia and Mexico for
both males and females (Figure 8.3a,b) [4].
In Europe, data from the Automated Childhood Cancer Information System (ACCIS) Project
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
83
Group
Subgroup
Description
Ia
Ib
Ic
Id
Ie
II
IIa
IIb
IIc
IId
IIe
III
IIIa
IIIb
IIIc
IIId
IIIe
IIIf
IV
IVa
IVb
Retinoblastoma
VI
VIa
VIb
Vic
Renal tumors
Nephroblastoma and other nonepithelial renal tumors
Renal carcinomas
Unspecified malignant renal tumors
VII
VIIa
VIIb
VIIc
Hepatic tumors
Hepatoblastoma
Hepatic carcinomas
Unspecified malignant hepatic tumors
IX
IXa
IXb
IXc
IXd
IXe
Xa
Xb
Xc
Xd
Xe
XI
XIa
XIb
XIb
XIc
XId
XIe
XII
XIIa
XIIb
84
35
30
More-developed
countries
Percentage
25
Less-developed
countries
20
15
10
5
0
Leukemias
Hodgkins
lymphomas
(a)
New Zealand
Cyprus
Brazil, Cuiab
USA, SEERNon-Hispanic White
Latvia
Italy, Veneto
Portugal, South
Colombia, Cali
Serbia
China, Guangzhou
Italy, Milan
Croatia
Brazil, Braslia
Switzerland, Vaud
Italy, Florence and Prato
Brazil, So Paulo
Italy, Umbria
Italy, Naples
Italy, Brescia
Kuwait: non-Kuwaitis
50
100
150
200
250
300
350
400
450
100
200
300
400
500
600
85
(a)
Germany
Kyrgyzstan
Norway
Canada
USA
Austria
Singapore
South Africa
Japan
Tajikistan
Venezuela
Mexico
Russian
Albania
Latvia
Cuba
Moldova
Ukraine
Romania
Azerbaijan
(b)
Germany
Finland
The Netherlands
France
USA
Canada
Tajikistan
Austria
Japan
South Africa
Albania
Panama
Mexico
Cuba
Lithuania
Russian Federation
Moldova
Ukraine
Azerbaijan
Romania
86
(a)
New Zealand
Cyprus
Brazil, Cuiab
USA, SEERnon-Hispanic white
Latvia
Italy, Veneto
Portugal, South
Colombia, Cali
Serbia
China, Guangzhou
Italy, Milan
Croatia
Brazil, Braslia
Switzerland, Vaud
Italy, Florence and Prato
Brazil, So Paulo
Italy, Umbria
Italy, Naples
Italy, Brescia
Kuwait: nonKuwaitis
50
100
150
200
250
300
350
400
450
(b)
Australia, Western
Germany, Munster
Canada, Ontario
Australia, New South Wales
France, Haut-Rhin
New Zealand
Israel
Belarus
Spain, Navarra
Australia, South
USA, SEERnon-Hispanic white
Canada, Manitoba
Italy, Romagna
France, Vendee
Italy, Salerno
Brazil, So Paulo
Australia, Queensland
Italy, Naples
Italy, Florence and Prato
Italy, Brescia
10
20
30
40
50
60
87
Table 8.2 International Classification for Cancers in Adolescents and Young Adults
Group
Subgroup
Description
1.1
1.2
1.3
Leukemias
Acute lymphoid leukemia
Acute myeloid leukemia
Other and unspecified leukemia
2.1
2.2
Lymphomas
Non-Hodgkins lymphoma
Hodgkins disease
3.1
3.2
3.3
3.4
3.5
3.6
4.1
4.2
4.3
4.4
Osseous and chondromatous neoplasms, Ewing tumors and other neoplasms of bone
Osteosarcoma
Chondrosarcoma
Ewings tumor
Other specified and unspecified bone tumors
5.1
5.2
5.3
6.1
6.2
7.1
7.2
8.1
8.2
8.3
8.4
8.5
8.6
8.7
9.1
9.2
10
Survival
Survival for children with cancer has continued to
improve over the past 20 years. In high-income coun-
88
tries, since the late 1990s, 5-year survival rates are close
to 80% [9].
In Europe, according to data from 83 populationbased cancer registries in 23 countries participating in the EUROCARE-4 project, 5-year survival
rates for all cancers combined was 81% for children and 87% for adolescents and young adults.
Among children under 15 years of age, very high survival rates were observed for retinoblastoma (97.5%),
(a)
Greece
Sweden
The Netherlands
France
USA
Canada
Austria
Germany
China, Hong Kong
Japan
Venezuela
Lithuania
Northern Ireland
Russian
Panama
Romania
Azerbaijan
Costa Rica
Mexico
Ukraine
Guatemala
10
12
(b)
Greece
Sweden
The Netherlands
France
USA
Canada
Austria
Germany
China, Hong Kong
Japan
Mexico
Panama
Venezuela
Ecuador
Cuba
Romania
Azerbaijan
Costa Rica
Russian Federation
Ukraine
Guatemala
10
tumors) to 97.0% (retinoblastoma) [11]. Contrasting with the finding for adults, in which cancer survival for European adults is lower than that observed
in USA, no systematic differences were observed
between these two regions regarding childhood cancer
survival [12].
Unfortunately, in spite of advances in diagnosis and treatment, survival rates are still poor
in low and middle-income countries, such as
India (Chennai, 40%) [13]. In several low-income
89
90
Preconception exposure
Linet et al. have called attention to the difficulties
in assessing results from studies evaluating the association between maternal preconception exposure to
diagnostic medical radiation and childhood cancer
[21]. According to these authors, the data are scarce
and difficult to interpret due to lack of standardization
of the time window of exposure, lack of emphasis on
key anatomic sites of exposure, absence of assessment
of the medical indication for the radiological examination and failure to adjust the results for therapeutic and
occupational radiation exposure [21]. The early results
of the OSCC indicated an association between abdominal X-ray exposures prior to marriage and childhood
cancer [24], but a subsequent analysis of data collected during the course of OSCC did not support
this finding (OR of all childhood cancer for preconception X-ray 1.1, 95% CI 0.91.2) [25]. Most of the
further studies also did not detect an association [19,
2630].
91
Infections
It has been long proposed that infections might cause
childhood leukemia and NHLs, based on two nonmutually exclusive mechanisms. Greaves hypothesized
that a delayed exposure to common infections in
infancy and early childhood leads to an increased
risk of ALL [52], while Kinlen suggested that a large
influx of people into relatively isolated areas (population mixing) might facilitate the spread of a viral infection that occurs when infected and susceptible individuals come in contact with each other, leading to an
increased risk of childhood leukemia [53]. Although
isolated studies have suggested varicella [54], influenza
[55] and EpsteinBarr virus (EBV) [56] as possible etiological factors, no single agent has been compellingly
implied as a cause.
EpsteinBarr virus is linked to Burkitts lymphoma (BL), and this association is based on seroepidemiological studies and the identification of EBV
genomes in tumor specimens from individuals living
in endemic areas [57]. Endemic BL is predominantly
found among children living in equatorial Africa and
in Papua New Guinea, where it is strongly associated
with endemic malaria. In these areas, pediatric lymphomas account for up to 80% of all cancers in children. On the other hand, areas with low incidence of
BL have a much lower (30%) proportion of BL associated with EBV [58], while areas with an intermediate
pattern, like South America, show percentages ranging
between 25 and 70% [5962].
92
Lifestyle factors
The effects of maternal lifestyle during pregnancy on
embryonic and fetal development are well known and,
therefore, effects on the subsequent risk of cancer in
the offspring might be expected [57]. Several features
of maternal lifestyle during pregnancy have been studied regarding their association with childhood cancer, including diet, breastfeeding, smoking and alcohol consumption and the use of cosmetics. In addition,
paternal exposures during the preconception period
are also hypothesized as having effects on the risk
of germ cell mutations [57], and habits like tobacco
smoking and alcohol consumption have also been
assessed regarding the risk of childhood cancer.
context of the UK Childhood Cancer Study, including 3500 cases (1637 with leukemia) and 6964 controls,
have demonstrated a small protective effect for breastfed children, both for leukemia (OR = 0.89; 95% CI
0.801.00) and for all cancers combined (OR = 0.92;
95% CI 0.841.00) [68]. A first meta-analysis comprising 14 case-control studies (including 6835 ALL
cases and 1216 acute myeloid leukemia [AML] cases)
have shown a negative association between long-term
breastfeeding (6 months) and both ALL (OR = 0.76;
95% CI 0.680.84) and AML risk (OR = 0.85; 95%
CI 0.730.98) [69]. In 2005, Martin et al. published
another meta-analysis, including 26 publications (92%
were case-control studies), suggesting that breastfeeding was associated with a 9% risk reduction for ALL
(OR = 0.91; 95% CI 0.840.98), 24% for Hodgkins disease (OR = 0.76; 95% CI 0.600.97) and 41% for neuroblastoma (OR = 0.59; 95% CI 0.440.78) [70].
Results from the Northern California Childhood
Leukemia Study, including 282 case-control sets of
children, in which maternal diet (12 months before
pregnancy) was evaluated through a food-frequency
questionnaire, have shown that the consumption of
vegetables (OR = 0.65; 95% CI 0.500.84), fruits
(OR = 0.81; 95% CI 0.651.00), proteins (OR = 0.55;
95% CI 0.320.96) and legumes (OR = 0.75; 95% CI
0.590.95) were associated with a reduced risk of ALL
[71]. Another study from the same group have also
investigated the role of childs diet on leukemia risk
and authors have reported that the regular consumption of oranges/bananas (OR = 0.49; 95% CI 0.26
0.94) and orange juice (OR = 0.54; 95% CI 0.310.94)
during the first 2 years of life were both associated
with a reduction in risk of childhood leukemia [72].
In addition, a recent Taiwanese report has pointed out
the role of the childs consumption of cured/smoked
meat and fish on increasing the risk of acute childhood leukemia (OR = 1.74; 95% CI 1.152.64), while
it also has shown a protective effect linked to a frequent consumption of vegetables (OR = 0.55; 95% CI
0.370.83) and bean-curd food (OR = 0.55; 95% CI
0.340.89) [73].
Several studies have investigated the role of maternal diet during pregnancy as a risk or protective
factor in relation to pediatric CNS tumors, and the
most compelling and studied hypothesis concerns the
high risk of disease in the offspring of those mothers with a high intake of N-nitroso compounds and
precursors. Results from an international collaborative
case-control study have shown specific associations
between cured meat consumption and pilocytic astrocytomas (OR = 2.5; 95% CI 1.15.8) or ependymomas
(OR = 2.0, 95% CI 1.42.9), as well as negative associations between cruciferous vegetables and anaplastic astrocytomas (OR = 0.4; 95% CI 0.30.7) and also
fresh fish consumption and malignant gliomas (OR =
0.5; 95% CI 0.30.6). Oil products intake was associated with an increased risk of medulloblastoma (OR =
1.5; 95% CI 1.02.2) [74].
93
94
Recreational drugs
Parental illicit drugs use has been associated with several types of childhood cancer, including leukemia,
neuroblastoma, brain tumor and rhabdomyosarcoma.
A case-control study conducted by Robison et al.
reported that the maternal use of mind-altering drugs
(mainly marijuana) prior to or during the index pregnancy was associated to an increased risk of acute nonlymphoblastic leukemia [93]. Kuijten et al., using data
from a case-control study conducted in the USA in
the late 1980s, reported that maternal use of any recreational drug, and particularly marijuana, was associated with an increased risk of astrocytoma in the offspring (OR = 2.8, 95% CI 0.99.9) [94].
A case-control study carried out in the USA,
including 322 cases and 322 matched controls, has
shown that maternal use of marijuana during the year
before the child birth conferred a threefold higher risk
of rhabdomyosarcoma (RMS) (95% CI 1.46.5) [95].
Maternal use of cocaine was also associated with RMS
(OR = 5.1, 95% CI 1.025.0). Both paternal marijuana
smoking (OR = 2.0, 95% CI 1.33.3) and cocaine use
(OR = 2.1, 95% CI 0.94.9) were also associated with
an increased risk of this type of tumor [95].
More recently, findings form a study conducted in
the USA showed that maternal use of marijuana in
the first trimester may also increase the risk of their
child developing neuroblastoma (OR = 4.8, 95% CI
1.616.5) [96].
Zahm and Devesa have reiterated the difficulties needed to make an accurate assessment of these
Melanoma
The etiology of melanomas is not fully understood,
although epidemiological studies have pointed out
several risk factors for developing the disease, including high solar exposure in early childhood, sunburns,
light hair and eye color, pale white skin and sunbed use
[98]. However, the major environmental risk factor for
melanoma is undoubtedly the exposure to UV radiation [98]. A meta-analysis published in 2005, including 57 studies from Europe, North America, Australia,
New Zealand, Argentina, Brazil and Israel, has found
a significant increase in the risk of melanoma associated with total sun exposure (RR = 1.34, 95% CI
1.021.77), with studies published after 1990 showing
a stronger effect (RR = 1.75, 95% CI 1.312.35). Intermittent sun exposure (RR = 1.61, 95% CI 1.311.99)
and sunburn history (RR = 2.03, 95% CI 1.732.37)
were also confirmed as significant risk factors for this
cancer [99].
Testicular cancer
Cryptorchidism is the best characterized risk factor
for testicular cancer. This association was first doc-
Cervical cancer
Infection with HPV is the main risk factor for cervical
cancer, and viral DNA is found in almost 100% of the
tumors. Human papilloma virus oncogenic types 16,
18, 31, 33 and 35 are the most associated with invasive
cervical carcinomas, with the types 16 and 18 causing
approximately 70% of the cancers worldwide. Human
papilloma virus is now recognized as the most common sexually transmitted infection in most populations. Infected women usually clear the infection
within 2 years, but those who become chronically
infected with high-risk HPV types are at greatest risk
for developing cervical cancer [101]. Female adolescents are one of the population groups with highest risk of becoming infected by HPV. It is estimated
that 75% of all new HPV infections are diagnosed
among those women aged 1524 years, with oncogenic HPV types accounting for nearly 50% of the
infections among adolescents. Recently, two vaccines
against HPV were developed: a quadrivalent (protecting against HPVs 6, 11, 16 and 18) and a bivalent vaccine (HPVs 16 and 18). Clinical trials have
shown almost 100% of efficacy of the vaccine in preventing high-grade, pre-cancerous cervical lesions,
caused by the HPV types prevented by the vaccination, for women nave to HPV types included in
the vaccine. Both vaccines are now licensed for use
in women more than 90 countries. Since 2006, the
quadrivalent vaccine is approved for use in the USA,
in girls aged 926 years, and the Advisory Committee
on Immunization Practices (ACIP) currently recommends that all 1112 year-old girls receive this vaccine
95
Thyroid cancer
The only established risk factor for thyroid cancer, particular for papillary carcinomas, is ionizing radiation.
Numerous reports have described the epidemiological aspects of thyroid cancer in areas associated with
nuclear plants, accidents and atomic bombs. Studies
that have assessed thyroid cancer incidence after the
explosion of atomic bombs in Hiroshima and Nagasaki
have demonstrated that the risk was highest among
individual aged 10 years at the time of exposure.
Moreover, a striking increase in thyroid cancer incidence among children was registered in areas surrounding Chernobyl, after the power plant accident
in 1986. In Belarus, the incidence rate previous to
the accident was 1 new case/million per year and it
has increased to more than 100 new cases/million per
year in the post-accident period. Diagnostic and therapeutic uses of radiation are also associated with an
increased risk of thyroid cancer, even if low doses are
applied [103].
Conclusions
Cancer is the second commonest cause of death, after
accidents, among children in high-income countries,
while in low and medium-income countries, improvements in the control of infectious diseases and perinatal mortality will lead to the emergence of cancer in
children as an important public health problem [57].
Incidence rates of childhood cancer have been increasing in the last decades in most countries. It is unclear
whether this reflects a real phenomenon or an artifact
due to better diagnosis and reporting, with improvement of the quality of cancer registries [5]. Current
knowledge on etiological factors remains limited, particularly for tumors other than leukemia and CNS neoplasms. Known causes of childhood cancer include
ionizing radiation, several infectious agents and possibly some behavioral factors; however, they explain
only a small proportion of the cases. Future collaborative studies should be conducted, aiming to have
enough power to test specific hypothesis as well as
to integrate knowledge from different areas, including
pediatric oncologists, epidemiologists and investigators working on basic research. Etiological hypotheses would be better tested in prospective studies, but
very large populations of newborn and children need
96
References
1. Curado MP, Edwards B, Shin HR et al. Cancer
Incidence in Five Continents, vol. 9. Lyon: IARC,
2007.
2. Steliarova-Foucher E, Stiller C, Lacour B and
Kaatsch P. International Classification of Childhood
Cancer, third edition. Cancer 2005; 103: 145767.
3. Ferlay J, Bray F, Pisani P and Parkin DM.
GLOBOCAN 2002: Cancer Incidence, Mortality and
Prevalence Worldwide. Lyon France: IARC Press, 2004.
http://www-dep.iarc.fr. (Accessed Nov. 10,
2009)
4. World Health Organization. WHO Mortality
Database, Geneva: WHO, 2008. http://www-dep.iarc.
fr. (Accessed Dec. 1, 2009)
5. Steliarova-Foucher E, Stiller C, Kaatsch P et al.
Geographical patterns and time trends of cancer
incidence and survival among children and
adolescents in Europe since the 1970s (the ACCIS
project): an epidemiological study. Lancet 2004; 364:
2097105.
6. Linabery AM and Ross JA. Trends in childhood
cancer incidence in the US (19922004). Cancer 2008;
112: 41632.
7. Birch JM, Alston RD, Kelsey AM et al. Classification
and incidence of cancers in adolescents and young
adults in England 19791997. Br J Cancer 2002; 87:
126774.
8. Bleyer A, OLeary M, Barr R and Ries LAG. Cancer
Epidemiology in Older Adolescents and Young Adults 15
to 29 Years of Age, Including SEER Incidence and
Survival: 19752000. Bethesda MD: National Cancer
Institute, 2006.
9. Smith M and Hare ML. An overview of progress in
childhood cancer survival. J Pediatr Oncol Nurs 2004;
21: 1604.
10. Gatta G, Zigon G, Capocaccia R et al. Survival of
European children and young adults with cancer
diagnosed 19952002. Eur J Cancer 2009; 45:
9921005.
97
98
99
100
Section 3
Chapter
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
101
102
103
25.0
P < 0.001
22.5
20.0
NH2
Oxidation
O
H
N
TUNEL (%)
NH
17.5
15.0
12.5
10.0
NH
7.5
HO
N
NH2
8-hydroxy,2deoxyguanosine
(8OHdG)
5.0
2.5
0.0
10
15
20
25
30
35
8OHdG (%)
104
Significance of
spermiogenesis in the
etiology of DNA damage
Spermiogenesis, the process by which round spermatids differentiate into spermatozoa, is a key event
in the etiology of DNA damage in the male germ line.
As indicated above, it is possible that some of the
DNA breaks seen in spermatozoa are the result of an
abortive apoptotic process initiated during spermiogenesis in response to some disruptive event. Equally,
DNA fragmentation in spermatozoa may also be
the result of unresolved strand breaks created dur-
105
(a)
(b)
30
P < 0.001
50
P < 0.001
CMA3 (%)
25
8-OH-dG (%)
40
20
15
30
10
20
0
0
10
10
15
20
25
30
35
TUNEL (%)
(c)
0
TUNE L
0
10
15
20
CMA3 (%)
correlations between DNA damage in human spermatozoa and elements of the conventional semen profile (specifically sperm count and morphology) that,
in turn, reflect the efficiency of the spermatogenic
process.
Not only is the disruption of spermiogenesis correlated with DNA damage; it is specifically correlated
with oxidative DNA damage as reflected by 8OHdG
formation (Figure 9.2). We postulate that this relationship exists because the poorly remodeled chromatin detected by CMA3 is particularly vulnerable to
oxidative attack by ROS originating from a number
of potential sources including infiltrating leukocytes,
depleted antioxidant systems and excessive free radical
generation by the spermatozoas own mitochondria.
We further propose that of all these potential sources,
the sperm mitochondria are the most important [5].
Experimental conditions associated with the induction of high levels of oxidative DNA damage, such as
exposure to radio frequency electromagnetic radiation
(RFEMR) [9] or the triggering of apoptosis through
the suppression of PI3 kinase with wortmannin [R. J.
Aitken, unpublished observations] invariably involve
the release of considerable amounts of ROS from the
sperm mitochondria.
106
107
RFEMR
PUFA
Infection
Loss of antioxidants
Heat
Oxidative stress
in testes
Testicular torsion
Toxicants
Disrupted
spermiogenesis
Defective spermatozoa
become senescent in male
tract and undergo
apoptosis
Apoptosis
Mitochondrial ROS
Figure 9.3 Hypothesis for the creation of DNA damage in human spermatozoa. This hypothesis posits that a variety of different clinical,
genetic and environmental factors can induce oxidative stress in the testis. One of the processes affected by oxidative stress is the
differentiation of spermatozoa during spermiogenesis. As a result of disrupted spermiogenesis DNA damage will occur in the spermatozoa via
at least three potential routes: (1) Physiological, topoisomerase-mediated strand breaks that occur during spermiogenesis to relieve the
torsional stresses associated with DNA packaging are not resolved and persist in the mature gamete. (2) Defective spermiogenesis generates
spermatozoa possessing poorly compacted, inadequately protaminated DNA that is vulnerable to free radical attack originating from a variety
of sources including depleted antioxidant protection, leukocytic infiltration, redox-cycling xenobiotics and ROS generation by the
spermatozoa themselves. (3) Defective human spermatozoa respond to their imperfect state by prematurely engaging in a process of
programmed senescence equivalent to apoptosis. During apoptosis caspases are activated, the spermatozoa lose their motility,
phosphatidylserine (PS) externalization occurs and the mitochondria start to generate ROS. As a consequence of routes (2) and (3) the DNA is
oxidatively attacked, generating the base adduct, 8-hydroxy-2-deoxyguanosine (8OHdG), which ultimately leads to the creation of abasic
sites and DNA strand breakage. In addition, we propose that as cells enter the terminal stages of senescence, topoisomerase, possibly acting
in concert with endogenous endonucleases [7880], completes the final destruction of the DNA.
(endonuclease G, apoptosis inducing factor) or activated in the cytosol (caspase-activated DNase) during the apoptotic cascade, the physical architecture of
these cells prevents these nucleases from translocating
to the sperm nucleus for two reasons:
108
109
Conclusions
DNA damage in human spermatozoa has been correlated with a range of adverse clinical outcomes including subfertility, miscarriage and morbidity in the offspring. A majority of this DNA damage is oxidative
and is correlated with the disruption of spermiogenesis and the generation of spermatozoa with poorly
compacted chromatin. We hypothesize that the disruption of spermiogenesis is the result of oxidative
stress, which can be induced by a wide range of factors including age, exposure to electromagnetic radiation in the form of heat or RFEMR, smoking, alcohol, insecticides, herbicides and heavy metals (Figure
9.3). The poorly protaminated spermatozoa generated
as a consequence of defective spermiogenesis may not
only carry unresolved physiological strand breaks
but are also vulnerable to oxidative attack. We further
hypothesize that this attack originates largely from the
generation of ROS by defective spermatozoa as they
prematurely enter a default pathway of programmed
senescence characterized by a truncated apoptotic cascade featuring ROS generation, PS externalization and
caspase activation. However, the endonucleases that
translocate to the nucleus during somatic cell apoptosis are prevented from doing so in spermatozoa by
virtue of the unique architecture of these cells. As
a result, apoptosis in spermatozoa initially generates
oxidative DNA damage followed by non-enzymatic
DNA fragmentation. Enzymatic DNA cleavage may
subsequently be induced by topoisomerase and other
uncharacterized nucleases that are bound up in the
structure of sperm chromatin. We propose that the
110
References
1. Weng SL, Taylor SL, Morshedi M et al. Caspase
activity and apoptotic markers in ejaculated human
sperm. Mol Hum Reprod 2002; 8: 98491.
2. Paasch U, Grunewald S, Agarwal A and Glandera HJ.
Activation pattern of caspases in human spermatozoa.
Fertil Steril 2004; 81 (Suppl. 1): 8029.
3. Barroso G, Morshedi M and Oehninger S. Analysis of
DNA fragmentation, plasma membrane translocation
of phosphatidylserine and oxidative stress in human
spermatozoa. Hum Reprod 2000; 15: 133844.
4. Almeida C, Sousa M and Barros A.
Phosphatidylserine translocation in human
spermatozoa from impaired spermatogenesis. Reprod
Biomed Online 2009; 19: 7707.
5. Koppers AJ, De Iuliis GN, Finnie JM, McLaughlin
EA and Aitken, RJ. Significance of mitochondrial
reactive oxygen species in the generation of oxidative
stress in spermatozoa. J Clin Endocrinol Metab 2008;
93: 3199207.
6. Aitken RJ and De Iuliis GN. On the possible origins of
DNA damage in human spermatozoa. Mol Hum
Reprod 2010; 16: 313.
7. Aitken, RJ, Koopman P, Lewis SE. Seeds of concern.
Nature 2004; 432: 4852.
8. Sakkas D, Moffatt O, Manicardi GC et al. Nature of
DNA damage in ejaculated human spermatozoa and
the possible involvement of apoptosis. Biol Reprod
2002; 66: 10617.
9. Aitken RJ, De Iuliis GN and McLachlan RI. Biological
and clinical significance of DNA damage in the male
germ line. Int J Androl 2009; 32: 4656.
10. Muriel L, Garrido N, Fernandez JL et al. Value of the
sperm deoxyribonucleic acid fragmentation level, as
measured by the sperm chromatin dispersion test, in
the outcome of in vitro fertilization and
intracytoplasmic sperm injection. Fertil Steril 2006; 85:
37183.
11. Zini A, Boman JM, Belzile E and Ciampi A. Sperm
DNA damage is associated with an increased risk of
pregnancy loss after IVF and ICSI: systematic review
and meta-analysis. Hum Reprod 2008; 23: 26638.
12. Aitken RJ and De Iuliis GN. Origins and
consequences of DNA damage in male germ cells.
Reprod Biomed Online 2007; 14: 72733.
111
112
113
Section 3
Chapter
10
Introduction
The follicle has a fundamental reproductive role in the
ovary. It consists of the oocyte (in various stages of
growth and development) and its surrounding layers
of supportive cells, the granulosa cells (GCs), cumulus
cells (CCs) (during the antral phase of follicular development) and thecal cells (TCs). These somatic cells
perform essential roles in ensuring optimal development and maturation of the oocyte. They receive signals from the external environment and pass them on
to the oocyte to orchestrate its growth and development until ovulation.
It is now widely recognized that there is bidirectional communication between the oocyte and
surrounding somatic cells and that this is essential
for the creation of a favorable follicular microenvironment in which the gamete can develop. Many complex and intricate molecular pathways are likely to be
involved in the production of an optimal follicle and a
viable gamete. This chapter aims to provide an insight
into the molecular mechanisms likely to be involved in
the various stages of follicular development, as determined from animal and human studies.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
114
As the follicle undergoes pre-antral to antral transition, the GCs continue to proliferate and as they
do so, follicular fluid accumulates and coalesces to
form a single follicular antrum. The follicular antrum
enlarges during the antral phase but the oocyte does
not increase in size during this period and remains suspended in fluid surrounded by the CCs (which differentiates from GCs during antrum formation). The CCs
remain connected to the rim of GCs by a thin stalk
of cells [7].
Follicle growth takes 85 days in humans and most
follicles become atretic at some stage [8] with the
oocyte continuing to play a critical role in follicular
control and the regulation of oogenesis, ovulation rate
and fecundity [911]. Only a minority of the developing follicles reach ovulation and, in most cases, only
one will complete growth each month, reaching about
100 m in diameter just before ovulation [12]. This
oocyte will be ovulated in response to the mid-cycle
LH surge, which effects the meiotic and cytoplasmic
maturation of the oocyte. The oocyte will complete
meiosis I and arrest at metaphase of meiosis II (MII)
[13, 14]. Meiosis II is only resumed when fertilization
occurs. The rest of the follicle collapses after ovulation
and now becomes the corpus luteum. Both GCs and
TCs contribute to the formation of the corpus luteum
with a switch of morphology and endocrine functions.
The CCs continue to be associated with the oocyte
after ovulation, assisting in oocyte transport along
the Fallopian tube. The cumulus matrix and CCs also
participate in fertilization by influencing spermatozoa
binding and penetration of the cumulus oocyte complex (COC) [15, 16]. Subsequently, the intimate contact between the CCs and the oocyte is broken by the
withdrawal of the cytoplasmic processes.
115
116
activation of Akt and hence elevated phosphorylation (activation) of ribosomal protein S6 (rpS6), which
is dependent on mTOR complex 1 [45]. Therefore,
PTEN, which inhibits the actions of PI3K, governs follicular activation through control in the initiation of
oocyte growth via PI3K, Akt and Foxo3a [46].
FOXL2, a single exon gene of 2.7 kb, belongs to the
family of winged-helix/fork-head transcription factors
[47] and is expressed in somatic cells of the developing
human ovary before and during follicle formation [48].
This gene product is essential in pre-granulosa cells to
mediate the squamous-to-cuboidal transition and proliferation. In the absence of FOXL2, the whole pool of
primordial follicle gets prematurely activated without
synchronizing with the differentiation and division of
surrounding pre-granulosa cells. This results in follicles that lack multiple layers of functional GCs, ultimately leading to atresia of these follicles [49, 50]. It is
likely that FOXL2 in pre-granulosa cells provides an
inhibitory mechanism via gap junction or paracrine
secretion and maintains the primordial follicles in a
dormant state [42].
Upon activation of primordial follicles, progression from the primary (single GC layer) to secondary
(pre-antral) stage of follicular development requires
further oocyte expansion, GC proliferation and investment of an LH-responsive theca cell layer. These vital
steps are not FSH dependent but remain critically
dependent on paracrine signaling by members of the
TGF superfamily [51]. In primary to pre-antral follicles, activin subunit (INHBA and INHBB) and AMH
[41] gene expressions predominate [52].
Activins (homodimers or heterodimers of INHBA
and INHBB) enhance GCs proliferation and promote
their responsiveness to FSH. At the same instance,
LH responsive thecal androgen synthesis will be suppressed. Appropriate stimulation by FSH will then
divert the formation of activins to that of inhibins
which promote androgen synthesis. Androgen synthesis is in turn linked to FSH augmentation of inhibin
synthesis with the primary aim to ensure estrogen synthesis from the aromatizing of androgens [51].
Anti-Mullerian hormone expression peaks in GCs
of secondary, pre-antral and small antral follicles
4 mm in diameter. In larger antral follicles (4
8 mm), AMH expression gradually disappears [53].
Anti-Mullerian hormone has been shown to suppress
early stages of follicular growth and onset of responsiveness to FSH in vitro, exerting a controlling influence on the rate at which follicles become available
This communication is so crucial that genetic deletion of the oocyte specific gap junctional subunit,
connexin-37, leads to female sterility in mice, resulting from a lack of mature follicles, failure to ovulate
and development of numerous inappropriate corpora
lutea. In addition, oocyte development in connexin-37
deficient mice is arrested before meiotic competence is
achieved [62].
In addition to oocyte growth and GCs proliferation, the pre-antral follicle further increases its size
and complexity through formation of a basal lamina
between the GC and TC layers in addition to multiplication and differentiation of TCs into two theca layers:
the inner vascular theca interna and the fibrous capsule, theca externa (Figure 10.1).
117
Figure 10.1 Primordial follicle to secondary follicle formation (gonadotrophin independent phase): The oocyte, under the stimulatory
effects of retinoic acid, recruits squamous type granulosa cells (GCs) to form the primordial follicle. Steroid hormones inhibit the formation of
primordial follicles. Under the activating effects of stem cell factor (SCF)/kit ligand, basic fibroblast growth factor (bFGF), bone morphogenetic
proteins 4 and 7 (BMP-4 and -7), platelet derived growth factor (PDGF), leukemia inhibitory factor (LIF) and keratinocyte growth factor (KGF),
the primordial follicle further develops into the primary follicle with accompanying morphological changes of the supportive cells, i.e. from a
monolayer of squamous type GCs to cuboidal type GCs. Thecal cells then surround the now multiple layers of GCs which encloses the
developing oocyte to form the secondary follicle. Repressive signals like phosphatase and tensin homolog (PTEN), Foxo3 and anti-Mullerian
hormone (AMH), tuberous sclerosis complexes (TSC) 1 and 2 and FOXL2 inhibits the development of primordial follicles to secondary follicles.
See plate section for color version.
118
119
AMH
Secondary follicle
FSH, activins,
inhibins
androgens,
estrogens,
GDF9, BMP15
Primary
oocyte
with zona
pellucida
Basement
membrane
Pre-antral follicle
Thecal layer
Antrum formation
Cumulus cell
layer
enclosing the
oocyte
Antral follicle
Figure 10.2 Pre-antral phase to antral phase transition. As the secondary follicle develops, it becomes more gonadotrophin sensitive with
the expression of gonadotrophin receptors. Under the activation of follicle stimulating hormone (FSH) and the effects of activins and inhibins,
androgen synthesis with estrogen production take place, resulting in the development of the secondary follicle into the pre-antral follicle.
Anti-Mullerian
hormone (AMH) has been shown to suppress early stages of follicular growth and onset of responsiveness to FSH in vitro,
exerting a controlling influence on the rate at which follicles become available for pre-ovulatory development. Enhancing effects from
growth differentiation factor (GDF-9), bone morphogenetic protein 15 (BMP-15) and insulin-like growth factors enables the pre-antral follicle
to develop further into the antral follicle with the formation of the antrum. At the same time, the GCs proliferate and differentiate into the
cumulus cell layer which encloses the developing oocyte. With the antral cavity filled with follicular fluid, the pre-antral follicle now becomes
the antral follicle. See plate section for color version.
stimulation with gonadotrophins, triggers the resumption of meiosis as well as cumulus expansion [94].
The MAPK appears to mediate LH-induced oocyte
maturation by interrupting cell-to-cell communication through phosphorylation of connexin-43 within
the ovarian follicle [95]. The breakdown of communication arrests the supply of cAMP from somatic
cells to oocyte, thereby reducing the levels of cAMP
within the oocyte [96]. This is probably mediated
by termination of gap junctions between CCs and
the oocyte [97]. The rapid increase of cAMP in
CCs may activate phosphodiesterase 3A (PDE3A)
and decrease cAMP level in oocytes, possibly by
cAMP-dependent guanine nucleotide exchange factor (GEF)/PI3K/phosphoinositide-independent protein kinase 1 (PDK1)/protein kinase B (PKB) pathway
[98, 99].
Following the LH surge, a cascade of events is initiated that leads to CC proliferation. The competence to
undergo expansion is a unique characteristic of CC differentiation [100], which has been shown to be critical
120
121
Gonadotrophin-independent phase
Ovarian follicular
development
Gonadotrophin-dependent phase
Figure 10.4 In the ovary, follicular development constitutes of a gonadotrophin-independent and a gonadotrophin-dependent phase.
Coordination of multiple growth factors, hormones and biochemical molecules signaling at timely intervals with the activation of several
pathways, e.g. PI3K, JAK/STAT, WNT/-catenin and MAPK between the growing oocyte, surrounding somatic cells and wider endocrine
system ensures optimum follicular development with the release of a meiotically and developmentally competent oocyte. See plate section
for color version.
122
Concluding remarks
Follicular development consists of gonadotrophinindependent and gonadotrophin-dependent phases
(Figure 10.4). These two phases require the fine coordination of multiple growth factors, hormones and
biochemical molecules signaling at timely intervals
between the growing oocyte, surrounding somatic
cells and the wider endocrine system. The precise control and balance of these systems is essential if the ovulation of a meiotically and developmentally competent
oocyte is to be achieved.
References
1. Byskov AG. Differentiation of mammalian embryonic
gonad. Physiol Rev 1986; 66: 71117.
2. Faddy MJ, Gosden RG, Gougeon A, Richardson SJ
and Nelson JF. Accelerated disappearance of ovarian
follicles in mid-life: implications for forecasting
menopause. Hum Reprod 1992; 7: 13426.
3. Gondos B, Westergaard L and Byskov AG. Initiation
of oogenesis in the human fetal ovary: ultrastructural
and squash preparation study. Am J of Obstet Gynecol
1986; 155(1): 18995.
123
124
125
126
127
128
Section 3
Chapter
11
Introduction
Cryobiology as a science deals with the effects of
reduced temperatures on living organisms, their constituent parts and their products. Understanding the
basics of cryobiology to develop improved cryopreservation procedures has been a major challenge to scientists all over the world. Since the complexity of cryobiology is deeply rooted in the complexity of living systems, historically this has entailed coordinated
research efforts among biological and physical sciences
and involved the participation of biologists, chemists,
physicists, engineers, mathematicians and others using
carefully designed empirical evaluations, as well as
investigation and utilization of specific cellular characteristics in theoretical models, all in efforts to apply
cryopreservation to a broad range of cells and tissues
[1].
For the most part, reproductive cryobiology has
been developed to gain time, either for flexibility (e.g.
extension of fertility), logistics (e.g. gamete donation),
back-up (for repeat embryo transfer) or some combination of these [2].
Many misconceptions related to the cryopreservation or vitrification methods stem from the use of
potentially confusing terminology that has evolved in
cryobiology literature. At one level, this occurs in the
labels we place on various categories of general methods for cryopreservation. For example it is common
to refer to methods on the basis of cooling rate such
as slow cooling methods or rapid cooling methods
[35]. In other cases cryobiologists refer to methods as
equilibrium methods, non-equilibrium methods
or even quasi-equilibrium methods [6]. Still, in other
cases the methods are described by the type of con-
tainer or device in which the cells or tissues are cryopreserved such as an Open Pulled Straw method [7],
a needle-immersed vitrification method [8], a Cryoloop method [3] and so on.
The key to sorting-through what all this means in
terms of outcomes is to understand how these labels
(methods) relate to the underlying physiochemical
conditions produced that fundamentally determine
whether cells and tissues survive (or fail to survive)
the series of extreme conditions we subject them to
during the cryopreservation process. For the purposes
of organizing this chapter and to potentially clarify
misconceptions and/or misuses of terminology with
respect to cryobiological processes, the subject has
been divided into three parts consisting of: (1) classical cryopreservation; (2) vitrification; and (3) preservation of reproductive cells and tissues using classical
cryopreservation and vitrification procedures.
Classical cryopreservation
Cryopreservation has been developed to store viable
biological systems at ultra-low temperature (196 C)
in a cryogenic medium such as liquid nitrogen for
extended periods of time. At such ultra-low temperatures, all cellular divisions and metabolic activities
are arrested, such that the systems can be revived and
restored to the same living state as before they were
stored. Most currently used methods for cryopreservation of mammalian cells are indeed derived from centuries of low temperature work on reproductive cells
and tissues [9].
Cells and tissues lose viability (are killed) during
classical cryopreservation due to two major causes:
(1) the formation of ice inside the cells (intracellular
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
129
130
essentially contain little or no freezable water and consequently IIF cannot occur during a subsequent plunge
at 196 C [17]. The first successful cryopreservation
of mouse embryos using equilibrium slow freezing
was reported by Whittingham et al. and Wilmut and
Rowson [18, 19]. These authors reported a high percentage of embryo survival when frozen at 1 C/min
to 70 C under near-equilibrium conditions. However, extreme dehydration, as would be encountered
during equilibrium slow freezing, can cause volume
excursions (shrinkage) beyond what some cells can
tolerate and produce irreversible damage to cellular components. Additionally, as described above,
unavoidable excessive salt concentrations can also play
a significant role in damaging the cells.
Alternatively, if the cooling rate is not slow enough
to maintain equilibrium, IIF becomes possible due to
supercooling of intracellular solution in relation to the
frozen extracellular solution at a given temperature.
However, there is an alternate slow freezing approach
often referred as non-equilibrium slow freezing or
interrupted slow freezing [20] during which the cells
are initially cooled slowly (slow enough to avoid IIF) to
an intermediate temperature to render some degree of
dehydration. At such an intermediate temperature, it is
expected that the concentration of unfrozen fraction of
the suspending solution and cytosol are significantly
high enough to enable vitrification and form amorphous glass upon rapid cooling by immersing abruptly
in liquid nitrogen. However, high survivals with this
procedure usually require that the cells be warmed
rapidly during thawing. The reason for this is that
after the initial slow cool to the intermediate temperature, the cells still probably contain a small amount of
freezable water and that water undergoes damaging recrystallization unless the subsequent warming is rapid
[17]. Further, if the concentration of unfrozen fraction is not enough an unstable glass may form during
rapid immersion in liquid nitrogen and devitrification
and/or re-crystallization can occur upon re-warming
[9].
131
Vitrification
As described in many recent research papers and
reviews, vitrification has become an increasingly
accepted method for preserving embryos, oocytes and,
recently, even sperm [2]. However, despite the growing
popularity of this type of preservation, misconceptions
in the literature abound. Vitrification may be simply
defined as the process of converting a supercooled liquid into a glass-like amorphous solid which is free of
any crystalline structure, either by the quick removal
or addition of heat and/or by mixing with an additive. Vitrification occurs at the glass transition temperature (Tg ) which is lower than the melting point
(Tm ) [32].
In the context of cryobiology, the supercooled liquid is almost always water and the additive is a combination of solutes, typically including salts and one or
more permeating or non-permeating CPAs. The relationship between the quick removal of heat (i.e. cooling rate) and total additive (solute) concentration can
be expressed conceptually as:
PV (S P )B ;
132
q = hA(Ts Ta );
where q is the heat flow, h is the heat transfer coefficient, A is the surface area, Ts is the surface temperature, and Ta is the reference temperature [42]. Using
this approach we can show the relationship between
the heat transfer coefficient (h) of a sample, and how
this is related to cooling rate, the size of the sample
and the required CPA concentration in the sample to
achieve vitrification at various cooling rates. Plunging samples into LN2 is an example of a pool boiling
approach. Flowing LN2 over a sample is an example of
a forced flow boiling approach. Oscillating heat pipe
(OHP) technology [41] is an example of new technologies that are being developed to increase our ability
to apply vitrification approaches to biological samples
(Figure 11.1 [41]).
11
misperception, it is helpful to consider the relationship between the heat transfer coefficient (h) of a sample, and how this is related to cooling rate, the size of
the sample and the required CPA concentration in the
sample to achieve vitrification.
The heat transfer coefficient comes from Newtons
law of cooling: the dissipation of heat from a solid to
a fluid is proportional to the temperature difference
between the solid and the fluid. This can be expressed
as:
10
Pool boiling
Forced flow
Boiling
New
technology
h = 103 W/m2 K
h = 104 W/m2 K
h = 105 W/m2 K
6
5
h = 106 W/m2 K
4
0.0
0.2
0.4
0.6
0.8
Sample dimension (cm)
1.0
133
NEV
Tm
20
Temperature (C)
40
EV-1
Supercooled
(Metastable)
Th
60
EV-2
80
Unstable
100
Td
120
140
Tg
160
10
20
30
40
50
60
70
80
100
Concentration (w/w%)
rewarmed tissue was severely impaired both functionally and structurally after exposure for 20 h in
unfrozen CPA medium at 79 C [46]. Subsequent
experiments by Elford and Walter determined that the
toxic effects of DMSO are not solely responsible for the
loss of tissue function, and in fact its function was only
improved after adequate steps were taken to optimize
the solute concentration (the ionic composition) of the
CPA medium during freezing [47]. Nevertheless, Farrants eminent procedure has been the basis for most
of the equilibrium vitrification methods that followed
[4851]. Despite some success achieved, this equilibrium approach has not been actively pursued, presumably because the technique requires lengthy periods
of exposure to toxic solutes at high temperatures and,
more importantly, the equilibration at sub-zero temperatures may not be achievable either in a practical
time scale or without exceeding the tolerance limits of
solute toxicity of the tissue [47, 52].
Vitrication is also possible using less concentrated
cryoprotective solutions if sufficiently rapid cooling
rates are employed. This approach requires the best
possible cryoprotective concentration (critical CPA
concentration) that ideally balances the glass formation ability and toxicity at the temperature of addition
[48]. These optimally adjusted cryoprotectants could
be vitrified by supercooling at realistically feasible
cooling rates through the metastable non-equilibrium
zone between the melting temperature and glass transition temperature on the phase change diagram (NEV
on Figure 11.2) [53]. The success of non-equilibrium
134
effective preservation for a number of cells, including monocytes [56], organized tissues [44, 4851, 57],
mouse ovaries [58] and pancreatic islets [59].
135
136
Cryobiology of oocytes
Cryopreservation of mammalian oocytes is one of the
most intensively studied topics in the field of cryobiology. For example, due to their regular spherical
shape and relatively large size, mouse oocytes were
used as model cells for numerous theoretical investigations and the tests of newly developed cryopreservation methods. Human oocyte cryopreservation plays
not only an important clinical role in assisted reproduction as an adjunct to sperm and embryo cryopreservation, but also a representative of the application of both the freezing and vitrification approaches.
Therefore, in this section, the discussions are concentrated on the current status of human oocyte cryopreservation.
sulfoxide (Me2 SO) survived and only half of those fertilized [76]. Oocytes frozen in PG tended to survive
better (32% and 75% of those fertilized). The second
report of a live birth came in 1987 using an equilibrium
method with DMSO as the permeating CPA [78].
Since 2001, at least 10 reports have appeared
describing the results of freezing human oocytes using
a standard equilibrium method with 1.5 M PG and
sucrose. Some of these reports show very high survival
and early development rates, and this has lead some
individuals to emphatically defend human oocyte
cryopreservation and suggest that the clinical results
are nearly equivalent to the use of fresh embryos.
Other investigators have been more cautious in their
interpretation [79], and in two recent reports with a
very large number of cycles, the results were still rather
poor [80, 81], especially when compared to the use of
fresh embryos [81].
Vitrification has also been utilized as a means to
cryopreserve human oocytes in recent years, gaining
more popularity than traditional equilibrium methods in recent years. Kuleshova et al. described a birth
resulting from an oocyte vitrified with a solution containing ethylene glycol and sucrose [82]. For this work,
the investigators utilized an OPS for the procedure.
Since this time, other reports on human oocyte vitrification have been published [83, 84], with all of these
reports utilizing open container systems to achieve socalled ultra-rapid cooling. However, the use of open
containers represents a potential problem for the possibility of disease transmission in the storage vessel
[85]. On average, the results from the vitrification trials
have been better than the trials using slow cooling (as
measured by the number of oocytes to achieve a pregnancy). However, the results are still limited in number and general conclusions are difficult to reach at this
time. Overall, progress on human oocyte cryopreservation has been significant during the past few years,
as evident by the number of reports appearing in the
literature. However, the procedure is still regarded as
suboptimal and experimental in nature.
based solution, with similar rates of maturation, fertilization, and cleavage compared to controls [87]. In
a different study, the maturation rate and fertilization
rate of cryopreserved and control oocytes was similar, but blastocyst development was lower [88]. Vitrification has also proved successful with GV human
oocytes [89], with rates of maturation, fertilization and
early development similar between frozen and nonfrozen oocytes. Despite these successes, to date, only
one report describing a live birth after cryopreserving a
GV stage human oocyte appeared in the literature [90].
Clearly, more basic research needs to be undertaken to
improve this technology.
Ovarian tissue
Successful ovarian tissue cryopreservation is one of the
effective clinical options for preserving female fertility.
For example, orthotopic re-implantation of cryopreserved ovarian cortical strips is a promising technique
for restoring ovarian function in women treated with
sterilizing chemotherapy for cancer. Both equilibrium
freezing and vitrification methods have been applied
for human ovarian tissue cryopreservation. Hovatta et
al., using histological assessment, showed that a high
proportion of follicles could survive an equilibriumfreezing method with human ovarian tissue [91, 92].
In the first report, they showed very good morphological survival of ovarian follicles after cryopreservation using either DMSO or PG, with no obvious
differences between frozenthawed and non-frozen
tissue. In the second report, using an in-vitro culture system, they present data suggesting that follicle
development occurs in the previously cryopreserved
tissue, with development rates similar between the
frozenthawed and unfrozen tissue samples. Shortly
after these reports, a study undertaken to determine
the diffusion rates of cryoprotectants into human ovarian tissue was published [93]. The results from this
study suggest that ethylene glycol (EG) and DMSO
diffuse into the tissue more rapidly than PG and
glycerol. These results supported the previous findings of these authors which showed EG and DMSO
to be superior cryoprotectants compared to PG and
glycerol [94], suggesting that the rate of cryoprotectant permeation is a critical factor in successful cryopreservation of human ovarian tissue. A similar conclusion was reached in a different report, where the
time of equilibration prior to cooling had a significant
effect on the proportion of intact follicles post-thaw
[95]. Therefore, it is of both practical and theoretical
137
22.0
R 2 = 0.99
22.5
Log (D )
23.0
EG
DMSO
PG
23.5
24.0
24.5
25.0
25.5
0.34
0.36
100/T
0.38
0.40
importance to address the issue of optimizing cryoprotectant permeation into human ovarian tissue by
measuring permeability parameters of the relevant
cells and using this information to model the mass
transport of water and cryoprotectant. Various physical models regarding the permeation of cryoprotectants into tissues have been established. A differential scanning calorimetry method to measure the
in ovarcryoprotectantwater mutual diffusivity ( D)
ian tissues at both super and sub-zero temperatures
was also established [96]. Figure 11.3 shows that the
Arrhenius relationship was strictly followed for the
values of D with different cryoprotectants. Due to the
relatively low values of D (106 cm2 /s), it has been
suggested that the optimal cooling rate for equilibrium
freezing procedures for ovarian cortical strip should
be at the order of 0.5 K/min. For the cryoprotectant
perfusion procedure before cooling, a stepwise perfusion is preferred and the perfusion time should be controlled to approximately 1 h.
Follicle loss after transplantation is likely due in
part to ischemic damage [9799]. However, using fresh
or frozen ovarian tissue from cynomolgus monkeys,
Schnorr et al. showed that only two of four recipients had functional frozen and thawed transplants, yet
five of six of the ovarian transplant recipients receiving fresh tissue had functional transplants [100]. In a
more recent study using ovarian tissue from cynomolgus or rhesus macaques, immediate post-thaw viabil-
138
Cryobiology of embryos
Because of its central importance, the cryopreservation of human embryos has become an integral part
of almost every ART program. According to International Committee Monitoring ART (ICMART), the
majority of the children (up to 40% worldwide) born
after ART are now born from cryopreserved embryos
[111], and this number is likely to increase in future.
Embryo cryopreservation is also a widely used method
of fertility preservation for cancer patients [112].
Cryopreservation allows the storage of excess viable
embryos for future use in an in vitro fertilization (IVF)
treatment cycle. In addition, cryopreservation makes
feasible the postponement of embryo transfer in the
event of a mother becoming ill or with patients at high
risk of ovarian hyperstimulation syndrome [113].
Over the years, major empirical advances have
been made to develop successful protocols for the
cryopreservation of embryos. Literature shows that
majority of the studies either used glycerol, EG or
DMSO as CPAs. Recently, however, these cryoprotectants have been increasingly replaced by 1,2 propanediol, which is generally used in combination with
other permeable and non-permeable cryoprotectants.
Propanediol is considered to have higher penetration permeability and is less toxic than DMSO. Both
slow cooling and vitrification have been studied for
embryo cryopreservation. The slow cooling technique
was developed independently in 1972 by Wilmut and
Rowson for bovine embryos and Whittingham et al.
for mouse embryos using DMSO as CPA [18, 19].
The same freezing protocol was applied to human
early-cleavage stage embryos and resulted in the first
report of pregnancy in 1983 [114] and live births in
1984 [115]. In 1985, PG along with sucrose was first
introduced in human embryo freezing and proved
to be quite efficient for 1, 2 or 3 day-old embryos
[116]. Therefore, this method was rapidly applied and
has become the widespread application for embryo
preservation in virtually every full scale IVF program
worldwide [117]. In general, slow freezing of embryos
employ progressive pre-equilibration in a CPA solution and then cooling to a seeding temperature at
which deliberate ice nucleation is initiated to prevent
supercooling. After equilibration for about 1015 min,
the cells are cooled slowly (1 C/min) to about
30 C till they are sufficiently dehydrated and then
finally plunged and stored in liquid nitrogen. Normally, frozen embryos are thawed slowly to prevent
excessive osmotic stresses. Finally, the CPA is removed
either by successive dilution or by using a sucrose dilution technique [118].
Slow cooling, however, is a long process and
requires extensive resources like controlled rate freezers and large quantities of liquid nitrogen. Further, the
studies using slow freezing have reported significant
differences in survival and developmental rates after
warming between laboratories, developmental stages
and quality [119121]. Vitrification, on the other hand,
has increased greatly in use in recent years particularly
for freezing reproductive cells, and avoids the multihour long process of slow cooling to 196 C. For vitrification, embryos are usually loaded with high concentrations of CPA and then plunged directly in LN2
using some specialized carrier or supporting device.
For thawing, usually high warming rates are used to
prevent ice crystallization during devitrification. There
are, however, several factors that need to be taken
into consideration while undertaking the vitrification
of embryos. The type of CPA used, its concentration
139
Conclusion
Within the realm of reproductive cryobiology, a better understanding of the basic features of what we have
described here as classical cryopreservation and vitrification is critical to accelerate advancement and to
allow individual clinical laboratories to make processing choices appropriate to reaching their desired outcomes. This includes clarification of terms, theoretical and technical details, and a balanced, pragmatic
evaluation of possible risks and potential (or definite)
gains [2]. As the survey of preservation methods and
results for gamete, tissue and embryos presented here
indicates, the ability to preserve these cells and tissues
has had a profound effect on humankind with millions
of births around the globe facilitated by these efforts
while truly optimum protocols are yet to be uncovered. Because of the complex nature of the biological
systems as well as the physical phenomenon in question, a scientific approach balanced between theoretical and empirical methods is absolutely necessary to
ensure procedures continue to be further optimized.
References
1. Woods EJ, Benson JD, Agca Y et al. Fundamental
cryobiology of reproductive cells and tissues.
Cryobiology 2004; 48: 14656.
2. Vajta G, Nagy ZP, Cobo A, Conceicao J and Yovich J.
Vitrification in assisted reproduction: myths, mistakes,
disbeliefs, and confusion. Reprod Biomed Online 2009;
19 (Suppl. 3): 17.
3. Oberstein N, ODonovan MK, Bruemmer JE et al.
Cryopreservation of equine embryos by Open Pulled
Straw, Cryoloop, or conventional slow cooling
methods. Theriogen 2001; 55: 60713.
4. Noyes N, Knopman J, Labella P et al. Oocyte
cryopreservation outcomes including
pre-cryopreservation and post-thaw meiotic spindle
evaluation following slow cooling and vitrification of
human oocytes. Fertil Steril 2010, Feb. 24. [Epub ahead
of print]
5. Smith GD, Serafini PC, Fioravanti J et al. Prospective
randomized comparison of human oocyte
cryopreservation with slow-rate freezing or
vitrification. Fertil Steril 2010, Feb. 18. [Epub ahead of
print]
6. Mazur P. Equilibrium, quasi-equilibrium, and
nonequilibrium freezing of mammalian embryos. Cell
Biophysics 1990; 17: 5392.
7. Vajta G, Holm P, Kuwayama M et al. Open Pulled
Straw (OPS) vitrification: a new way to reduce
140
141
142
143
144
Section 3
Chapter
12
Introduction
In the field of assisted reproductive technology, vitrification is becoming an increasingly popular method of
cryopreserving cells, tissues and even entire organs [1
4]. The National Library of Medicines public database
(PUBMED) lists an exponentially increasing number
of citations on vitrification over the past decade, many
of them pertaining to vitrification in reproductive biology.
Vitrification in the present context is the process
whereby an aqueous solution is transformed into an
amorphous solidified system as a result of a significant
decrease in temperature [5]. The three-dimensional
arrangement of the molecules in such a system resembles that of a liquid (i.e. remains disordered), but
the viscosity and associated shear relaxation time are
more solid-like. This is in contrast to freezing methods, where molecular rearrangements in the form of
extensive ice formation occur in the solution during
the cooling process.
Vitrification as a means of cryopreservation is an
attractive alternative to freezing for several reasons.
The two most obvious are the complete elimination
of ice formation and its consequent damage, and the
ability to avoid chilling injury by rapidly cooling a
sample through the temperature zone known to be
damaging [68]. Other advantages include reduced
labor and equipment costs [9]; the lack of need to
discover optimum cooling and warming rates; and
scalability from cells to tissues and even to whole
organs. Some recent reports suggest that vitrifying
oocytes and embryos is a more effective means of
cryopreservation than is freezing [1013], provid-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
145
to use the minimum possible concentrations of cryoprotectants), some understanding of the physics of ice
formation is necessary to understand the risks associated with different vitrification methods and, indeed,
to be able to determine whether vitrification is complete or partial. Interested readers are encouraged to
consult a number of authoritative reviews for further
details [35, 9, 2229].
146
% Glucose
% Sucrose
% Glycerol
% Me2 SO
% EG
% MeOH
Resultb
40
30
40
30
30
44
30
40
F, NF
30
40
IS
30
40
NF
30
35
10
IS
30
30
10
30
30
10
NF
20
30
10
IS
20
30
20
IS, NF
20
25
25
IS
20
20
20
20
20
20
20
20
20
10
IS
%w/v glucose and sucrose, % v/v glycerol, Me2 SO, EG and MeOH. EG, ethylene glycol; MeOH, methanol. Experiments carried out from
12/21/1979 to 3/17/1980; previously unpublished data.
Results determined after 1119 days of storage in triplicate 15ml samples in test tubes. F, frozen; IS, ice spheres; NF, not frozen.
to present the idea in public for the first time [48] and
published confirmation of the utility of high pressures
[50]. More thorough expositions of the idea were provided in 19821984 [22, 49, 51, 52].
By 1984, Fahy had clearly defined the physicochemical requirements for true vitrification; had
demonstrated the possibility of highly concentrated
yet minimally toxic solutions for vitrification; had
explained the idea of combining permeating and nonpermeating cryoprotectants to induce vitrification in
a manner consistent with the vitrification tendency
of the intracellular compartment; had provided efficient methods for adding and removing vitrifiable
concentrations; had described taking advantage of
osmotic shrinkage for facilitating vitrification; had
demonstrated methods for avoiding fracturing even
in objects larger than rabbit kidneys; and had presented his new preservation method as one that should
be applicable not just to organs but to all cells and
tissues [22]. However, few cryobiologists concerned
themselves with organ cryopreservation, and therefore few read about these developments or realized
their broader applicability. In addition, there still
remained no actual demonstration that living cells vitrified according to Fahys methods could actually survive after re-warming.
147
148
G
Ice
Pure water
Aqueous solution
Tm Solution
Tm Pure water
0
Temperature (C)
Figure 12.1 An example of the relative values of G for pure water,
water containing dissolved solutes and ice. A physical state is more
thermodynamically stable than an alternative state if its value for G
is lower than that alternate states value. As can be seen, Gice is
lower than Gpure water at low temperatures; hence ice is the more
favorable state ( G is negative). The temperature where these lines
cross is the melting point of water (Tm pure water; 0 C), and at that
point, both states are equally stable. Adding solutes to water causes
the curve to shift to the left, decreasing the value of G relative to
pure water. This decreases the temperature at which ice becomes
the more stable state (Tm solution). The curve for solutions would
continually be shifted to the left if additional solutes were added,
further depressing the melting point. The relative difference
between the curves for water and ice at a specific temperature (as
indicated by arrows) is an indication of the degree of supercooling
for a solution remaining liquid below Tm , and can be thought of as a
driving force for a phase transition. Hence, as a liquid is lowered
below its melting temperature, the strength of the driving force
continually increases. Note the difference in the length of the
arrows for G for water-ice (single arrowheads) and solution-ice
(double arrowheads) at specific temperatures. Modified from Angell
and Senapati [59].
Ice nucleation
Ice growth
Relative rates
Tg
150
125
Tm
100
75
50
Temperature (C)
Figure 12.2 The maximum rates of ice nucleation and ice growth
occur at very different temperatures in effective vitrification
solutions. The curves represent the temperature dependence of
these two rates for a specific vitrification solution (M22) [58].
Whereas the maximum rate for nucleation occurs near Tg , the
maximum rate for crystal growth occurs near Tm . With permission
from Wowk [61].
149
150
Th
104
Relative time
1
thom
104
thet
108
tmr
1012
Tg
Temperature
Tm
water molecules to arrange themselves into a crystalline structure becomes very long longer than
observable time scales, thus confining ice development
to higher temperatures. Hence, at lower temperatures
crystallization is thermodynamically favorable, but it
does not occur on a normal time scale simply due to
kinetic barriers that preclude the molecules from joining a crystal.
Boutron used a semi-empirical approach to
describe the dependence of the total quantity of ice
crystallized on the cooling rate for given solutions.
Under the assumptions described in his work, and
according to the most accurate model, the quantity
of ice crystallized during cooling is given by equation
12.1:
1
1
1
+ 3 arctg(( 3x 3 )/(2 + x 3 )) = k4/|V|
[12.1]
151
CPAs in water
The warming rates necessary to avoid devitrification, i.e. the critical warming rates (vcwr ), are generally higher than those needed to vitrify (critical cooling
rates, vccr ) by several orders of magnitude. Baudot and
Odagescu [68] reported, for example, the values of vcwr
for solutions with 50, 45 and 40% (w/w) EG in water as
853, 1.04 106 and 1.08 1010 C/min, respectively.
Such rates reflect the increasingly short times needed
PG in water
(a)
(b)
Figure 12.6 Effect of the carrier (physiological support) solution on vcwr . (a) When vcwr is plotted as a function of cryoprotective agent (CPA)
concentration only, it can be seen that the presence of the sugar-rich EuroCollins solution carrier (EC) can lower vcwr for 30% w/w
2,3-butanediol (2,3-BD) by 4 orders of magnitude, whereas salt-rich carriers (PBS [circles] and St Thomas solution [stars]) have a smaller
effect. (b) If the same data are plotted as a function of total solute concentration (CPA plus carrier solutes), it can be seen that the effect of the
carrier is approximately the same, gram for gram, as that of the cryoprotectant, at least for 2,3-BD solutions. For solutions of propylene glycol
(PG), the presence of salt carrier solutions may actually slightly increase vcwr . These results allow the contribution of the carrier to vitrification
solution stability to be estimated fairly easily as a rough rule of thumb. (Data from [66, 67, 74].) Modified from Fahy and Rall [3].
152
Thermo-mechanical instability in
vitreous materials and the problem of
glass fracturing
of mouse oocytes when using a more complex vitrification solution (EAFS 10/10, consisting of 10% [v/v]
EG, 10.7% [v/v] acetamide, 24% [w/v] Ficoll-70 and
0.4 mol/l sucrose [in a stock solution of PB1 medium])
[82]. They found that maximum survival was attained
with a cooling rate as low as 500 C/min provided the
warming rate was approximately 3000 C/min (lower
warming rates reduced survival). They report that the
cooling rate needed to avoid ice formation in this solution is 500 C. Although we have not been able to find
any reference to a critical warming rate for EAFS 10/10,
it is likely to be 3000 C/min based on Figure 12.6.
Although Figure 12.7 supports the ability of cells
to survive after devitrification, it and Seki and Mazurs
observations, emphasize that attaining a high warming rate is more critical than attaining a high cooling
rate, a point that is often ignored due to preoccupation only with ensuring vitrification during cooling.
The same observations also make it apparent that survival alone is not unequivocal proof of vitrification
[35]. This point is often overlooked in the reproductive cryobiology literature, and is particularly relevant
to some of the current methodologies utilized for vitrification. Many of these methodologies might be better characterized as nucleated vitrification procedures
or as one-way vitrification methods in which appre-
153
temperature, elastic strain is insignificant and the solution acts like a liquid. Below the set temperature, the
elastic strain dominates and the fluid acts like a solid
(see Steif et al. [85] for more details).
To avoid stress buildup to the point of fracture, it
is important to consider the effect of thermal gradients through the sample. During cooling at a constant
rate, significant thermal gradients inevitably become
established within all but very tiny systems. Above the
set temperature, minimum stress exists within the system as cooling proceeds. As the system approaches the
storage temperature (usually below the set temperature), the thermal gradients established during cooling can result in significant stress build-up within the
system as the system approaches thermal equilibrium
(e.g. as the still-pliant center of the system cools and
pulls against the rigid outer layers). If the stress developed from this process exceeds the yield stress, fracturing results.
Using a continuum mechanics approach to modeling the stresses built up as a result of temperature
gradients during cooling, Steif and colleagues developed an analytical expression to approximate the maximum tensile stress ( max , which occurs at the center
of a sample) associated with various ideal geometries,
given by the following equation:
max = gE T/(1 v)
[12.2]
[12.3]
154
1e + 14
1e + 13
1e + 12
1e + 11
1e + 10
1e + 9
1e + 8
1e + 7
1e + 6
1e + 5
1e + 4
1e + 3
1e + 2
1e + 1
1e + 0
1e 1
1e 2
20
(a)
Time in years
to equal 10 s at 0C
(b)
1e + 8
1e + 7
1e + 6
1e + 5
1e + 4
1e + 3
1e + 2
1e + 1
1e + 0
1e 1
1e 2
1e 3
1e 4
1e 5
1e 6
1e 7
20
40 60 80 100 120
Temperature (C)
10000 years
100 years
10 years
1 year
1 month
1 week
1 day
1 hour
1 minute
20
20
40 60 80 100 120
Temperature (C)
cells [99101]. A recent meta-analysis of the published literature of full reports over the past 10 years
identified fewer than 10 vitrification solutions having
been tested on in vivo matured human oocytes to date
[102]. This is undoubtedly due, at least in part, to the
155
156
threshold concentration needed for the solution to vitrify under standard conditions (qv , pronounced cue
vee star). According to analyses of many VSs tested on
rabbit renal cortical slices, viability is high when qv is
low, and vice versa, indicating that weak glass forming agents are less toxic than strong ones. A similar
trend is apparent from the results of Ali and Shelton
[98] when the toxicity of 13 vitrification solutions on
mouse morulae is assessed [S. F. Mullen, unpublished
results].
This is interpreted to mean that it is preferable to
choose cryoprotectants that compete less strongly with
cellular constituents for access to water, and that water
is actually more available to hydrate biomolecules in
VSs that have lower absolute water concentrations at
qv [65]. However, it is often preferable to use mixtures
of cryoprotectants rather than attempting to select
only the weakest available glass former because the
concentrations needed for any one agent to vitrify tend
to be high enough to introduce specific toxic effects for
that agent that defeat its theoretical advantages [65].
Nevertheless, Ralls choice of glycerol + albumin (VS3
[106]) is consistent with qv theory because glycerol is
a particularly ineffective glass former, polar group for
polar group, compared to other agents. Unfortunately,
glycerol tends to be limited by its low permeability, and
in some systems is able to dramatically lower adenosine triphosphate (ATP) levels by being phosphorylated at the expense of ATP [107].
MacFarlane and colleagues have examined the
physicochemical basis of glass formation in aqueous cryoprotectant solutions by nuclear magnetic resonance (NMR) spectroscopy techniques, and were
able to establish that glass-forming efficacy is directly
related to the water-cryoprotectant hydrogen bond
strength (cryoprotectant basicity) [26, 108]. These fundamental observations might in principle be used to
quantitatively relate qv to hydrogen bond strength
and therefore to relate viability directly to hydrogen
bond strength, but this depth of analysis has not to date
been pursued.
of having toxicity that can be blocked by the simultaneous presence of Me2 SO, such that, for example, a
50% w/v total concentration resulting from the combination of 20% w/v formamide with 30% w/v Me2 SO
can have no toxicity even though 20% w/v formamide
by itself can lower viability by 60% [110]. The mechanisms involved are unknown, but sufficient clues are
available to enable mechanisms to be elucidated [109].
In the meantime, CTN enables the exceptionally poor
vitrification tendency of amides to be traded off against
the strong glass-forming tendency of Me2 SO and supplemented with the use of other intrinsically weak
glass formers to enable solutions of exceptionally low
toxicity, high concentration and high overall stability
against ice formation [58, 65, 111].
The use of amides in VSs can be controversial when
considered outside the context of CTN. However, the
toxicological effects of amides in vivo are not meaningful in the context of low temperature addition and
removal in the presence of Me2 SO in vitro, and can
be avoided if necessary by ensuring that all amides are
removed prior to re-warming to 37 C or by very rapid
washout at 37 C.
The choice of acetamide in the original VS1
[55] solution may have been suboptimal [65, 109].
Formamide currently appears to be the amide of
choice, combining high permeability [112], exceptionally poor glass-forming ability [24], lack of denaturing character [109], full toxicity neutralization potential [109] and lack of any documented mutagenicity
or carcinogenicity. Thus far, CTN has been demonstrated only in kidney and liver tissue, and seems not to
apply to brain tissue. Nevertheless, solutions based on
CTN have been effective for brain slices [113], mouse
oocytes [65] and many other systems.
157
Summary
Vitrification is becoming an increasingly common
means to cryopreserve reproductive cells and tissues.
Efforts to understand the physics of vitrification have
been underway for decades, and the basic principles
of vitrification are now reasonably well understood,
although many important practical details remain to
be investigated. Achieving vitrification in a laboratory
setting is generally believed to require the use of multimolar concentrations of cryoprotectants, which have
the potential to be toxic to living cells. Fortunately,
some general principles underlying the relationship
between molecular structure, vitrifiability, and toxicity have been elucidated in recent years, increasing
our ability to develop vitrification solutions on a rational basis. While the avoidance of the damaging effects
of ice formation is the basis for choosing vitrifica-
158
tion as a means of cryopreservation, mechanical damage due to fracturing of the vitreous material can also
cause serious injury, in some cases negating the benefits of vitrification. However, utilizing interdisciplinary
knowledge of the fundamental principles of vitrification, the fundamental cryobiology of the cells being
preserved and the derived parameters associated with
current technology, continued improvements in vitrification methods for reproductive cells, tissues and
organs should be possible, enabling the achievement
of superior fertility preservation results.
Acknowledgements
We would like to thank Brian Wowk for stimulating
discussions on thermodynamic aspects of vitrification,
and his assistance in the clarification of those concepts.
We also thank Mehmet Toner for his consent to publish
his estimates in Figure 12.5. This work was supported
by 21st Century Medicine, Inc.
References
1. Baudot A, Courbiere B, Odagescu V et al. Towards
whole sheep ovary cryopreservation. Cryobiology
2007; 55(3): 23648.
2. Cobo A, Kuwayama M, Perez S et al. Comparison of
concomitant outcome achieved with fresh and
cryopreserved donor oocytes vitrified by the Cryotop
method. Fertil Steril 2008; 89(6): 165764.
3. Fahy GM and Rall WF. Vitrification: an overview. In:
Tucker MJ, Liebermann J (eds.), Vitrification in
Assisted Reproduction. London: Informa Healthcare,
2007: pp. 120.
4. Pegg DE. The role of vitrification techniques of
cryopreservation in reproductive medicine. Hum Fertil
(Camb) 2005; 8(4): 2319.
5. MacFarlane DR. Physical aspects of vitrification in
aqueous solutions. Cryobiology 1987; 24: 18195.
6. Martino A, Songsasen N and Leibo SP. Development
into blastocysts of bovine oocytes cryopreserved by
ultra-rapid cooling. Biol Reprod 1996; 54(5): 105969.
7. Mazur P, Cole KW, Hall JW, Schreuders PD and
Mahowald AP. Cryobiological preservation of
Drosophila embryos. Science 1992; 258(5090): 19325.
8. Steponkus PL, Myers SP, Lynch DV et al.
Cryopreservation of Drosophila melanogaster
embryos. Nature 1990; 345(6271): 1702.
9. Vajta G and Nagy ZP. Are programmable freezers still
needed in the embryo laboratory? Review on
vitrification. Reprod Biomed Online 2006; 12(6):
77996.
159
160
161
162
163
Section 4
Chapter
13
Introduction
Radiation and chemotherapeutic regimens required
for life-threatening diseases, such as cancer or exposure to environmental toxicants, may jeopardize the
fertility of men of reproductive age causing permanent or temporary azoospermia. Young pre-pubertal
patients who cannot supply a semen sample for cryopreservation of sperm have particularly poor fertility prognosis. In the USA alone about 17 000 men
aged 1545 years old are diagnosed each year with
Hodgkins disease, lymphoma, bone and soft tissue sarcomas, testicular cancer or leukemia. Of these, over
3000 are treated with doses of alkylating agents, platinum drugs or radiation that are sufficient to induce
prolonged azoospermia. In addition, over 6000 boys
under the age of 15 are diagnosed each year with cancer, including leukemia, nervous system tumors, lymphomas and other solid tumors. About 80% of them
receive chemotherapy or gonadal radiation, and about
550 of the long-term survivors are azoospermic when
they reach adulthood.
Environmental and occupational toxicants can
also produce prolonged azoospermia. This was most
dramatically shown with dibromochloropropane
(DBCP), as highly exposed manufacturing and
agricultural workers had an increased incidence of
azoospermia [1, 2]. In addition, sterility can be developed during development such as due to cryptorchidism or aging.
Methods to prevent these effects on male fertility
and to restore normal testicular function are of great
importance. A variety of biochemical and biological
approaches (thiol radioprotectors, prostaglandin analogues, growth factors, blockers of apoptotic pathways,
and reduction in blood flow) have been tested to protect the testes in experimental animal model systems
against radiation and chemotherapy (reviewed in [3]).
However, utmost research interest in this field, including all clinical trials, has involved hormonal modulation in attempts to prevent or reverse damage to
the germ line from radiotherapy and chemotherapy.
In this chapter, I will discuss the current knowledge
of hormonal suppression as a means to preserve or
restore fertility in males.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
164
165
X
X
X
(a)
(b)
*
*Sp
Sp
*Sp
Sp
*
(c)
(d)
Figure 13.1 Photomicrographs of Lewis brownNorway F1 hybrid (LBNF1) rat testes sections showing normal spermatogenesis in control
testis (a), radiation-induced block in the differentiation of spermatogonia (b,c) and the reversal of this block by gonadotropin-releasing
hormone (GnRH) antagonist treatment. The testes were harvested 13 weeks after 5 Gy irradiation with or without GnRH antagonist treatment
during weeks 37 after irradiation. Note that with no GnRH antagonist treatment all tubules are atrophic (X) and contain only Sertoli cells and
type A spermatogonia (b), with normal (half-open arrow) and dividing (filled arrow) spermatogonia shown at higher magnification (c). With
GnRH antagonist treatment all tubules are repopulating ( ) with mature spermatids in many of them (Sp) (d). Bars = 50 m.
166
Normal
rat
Irradiated
rat
Spermatogonium
T independent
+T
Spermatocyte
T independent
T independent
R. spermatid
+T
+T
Sperm
[41]. The endogenous hormone primarily responsible for the inhibition of spermatogonial differentiation in toxicant-treated rats was testosterone, although
FSH also had a minor inhibitory effect [35], and
other exogenously administered androgens were also
inhibitory [34]. It was also observed that hormonal
suppression after irradiation of mouse testes modestly
but significantly increased the percentage of tubules
in which differentiation of surviving spermatogonia
occurred [G. Wang and M. L. Meistrich, personal communication].
Hormonal suppression with GnRH analogues or
hypophysectomy has also been shown to promote the
survival and differentiation of spermatogonia that are
transplanted into testes of animals that were depleted
of endogenous stem cells. Although the effects were
most dramatic when rat testes depleted by cytotoxic
treatments were used as recipients [12, 42], enhanced
proliferation and differentiation of transplanted spermatogonia were also observed when mouse testes
were used as recipients [4345; G. Wang and M. L.
Meistrich, personal communication].
The inhibition of spermatogenesis by testosterone
after cytotoxic exposure such as radiation appears to
be contradictory to its well-studied role in stimulating and maintaining spermatogenesis (Figure 13.2). In
normal unirradiated and irradiated rats, testosterone
has a similar role in spermiogenesis, as shown by the
failure to find cells that have developed past the round
spermatid stage in both these cases when testosterone
167
Table 13.1 Results of hormone suppression treatments given before and during cytotoxic therapy on spermatogenic recovery in men
Recoveryb
Cytotoxic
therapya
Hormone
treatment
Hormonetreated
Controls
References
Hodgkins
MOPP
36 cycles
GnRH agonist
1/5
No controls
Johnson et al.
[48]
Hodgkins
MVPP, ChlVPP
GnRH agonist +
testosterone
0/20
0/10
Waxman et al.
[49]
Hodgkins
MOPP 4 cycles
Testosterone
70%/23c
70%/22c
Redman and
Bajorunas [50]
Testis Ca
PVB + ADR/CY +
radiation
Medroxy-progesterone
0/4 (2/12)d
2/3 (7/13)d
Fossa et al.
[51]
Testis Ca
PVB
GnRH agonist
6/6
8/8
Kreuser et al.
[52]
Testis Ca
(Seminoma)
Radiation
GnRH agonist +
antiandrogen
12/12
8/8
Brennemann et al.
[53]
Nephritis
Cyclophosphamide
Testosterone
5/5
1/5
Masala et al.
[54]
Disease
is suppressed. But, unlike normal rats in which spermatogonial survival and differentiation are qualitatively
independent of testosterone, in irradiated rats the survival and differentiation of type A spermatogonia are
inhibited by this hormone.
The mechanism by which somatic cells in rat testis
are protected from damage if testosterone suppression
treatment is given at the time of cytotoxic exposure is
not known. Neither is the mechanism by which testosterone suppression after cytotoxic exposure enhances
the ability of the somatic elements of the testis to
maintain the differentiation of spermatogonia. Previous studies ruled out the possibility that the protective
effect of hormonal suppression given before cytotoxic
drug exposure was due to reduced delivery to the tissue or altered metabolism of the drug [46]. We have
recently shown that the block in spermatogonial differentiation in rats treated with cytotoxic agents is associated with the increased levels of interstitial edema
in the testes [47]. Based on data indicating that more
rapid stimulation of recovery of spermatogonial differentiation in irradiated rats was achieved by elimination of Leydig cells with ethane dimethane sul-
168
Clinical trials
Seven clinical trials have been performed in attempts
to demonstrate protection of spermatogenesis in
humans by hormone suppression treatment before
and during cytotoxic therapy, but six indicated no
protection (Table 13.1 [4854]). Three of the trials
involved patients treated for Hodgkins lymphoma
and three involved testicular cancer patients. Treatment with GnRH agonist resulted in only 20% of
169
Table 13.2 Summary of effects of hormone suppression on protection and/or stimulation of recovery of spermatogenesis after cytotoxic
treatment in different species
Treatment
Before
Mouse
Procarbazine,
doxorubicin,
cyclophosphamide
Cisplatin
Radiation
Radiationa
Rat
After a delay
References
n.d.
n.d.
n.d.
n.d.
++
++
Procarbazinea
++
++
n.d.
Busulfan
n.d.
DBCP
Immediately after
++
Aging
n.d.
Monkey
Radiation
Human
Chemotherapeutic drug
combinations
( radiation)
/?
Radiation
n.d.
Cyclophosphamide
++
n.d.
DBCP, dibromochloropropane.
a Instances in which the type A spermatogonia showed a block in differentiation.
+, ++, relative effectiveness at protection or stimulation.
, no protection/stimulation.
?, no protection observed but could not be demonstrated or ruled out with experimental design.
n.d., the effect not tested.
170
(procarbazine, chlorambucil, busulfan) and radiation as measured by stem cell death and prolonged azoospermia, respectively [60]. However, there
are also differences as spermatogonial stem cells in
mice are sensitive to, and killed, by doxorubicin
(Adriamycin) but not cyclophosphamide, whereas
cyclophosphamide, but not doxorubicin, strongly
induces long-term azoospermia in humans. It has to
be noted that although both primate and mouse spermatogenesis are sensitive to radiation, the human [61
63] and monkey [64] appear be more sensitive to radiation than mice [6567]. Thus, one of the directions of
future research should be on elucidating the reasons
for greater sensitivity of primate stem spermatogonia
to radiation and certain chemotherapeutic agents in
order to develop methods to protect them.
One contribution to the difference in the stimulation of recovery by hormone suppression after cytotoxic treatment may be the interspecies differences in
the block in differentiation of spermatogonia. In rats
exposed to moderate doses of cytotoxic agents, the
induction of a block in spermatogonial differentiation
is a much more likely cause of prolonged azoospermia
than is spermatogonial stem cell death. The reversal
of this block in spermatogonial differentiation appears
to be the mechanism by which hormone suppression protects or restores spermatogenesis in toxicantexposed rats [27], and the occurrence of such a block
should indicate whether hormonal suppression might
stimulate spermatogenic recovery in another species.
Although, in many cases, the seminiferous tubules in
testicular biopsies taken from men with chemotherapy
or radiotherapy-induced azoospermia contain only
Sertoli cells and no spermatogonia [68], occasionally the presence of isolated spermatogonia have been
observed at relatively long times after chemotherapy
treatment [69]. In addition, spontaneous recovery of
spermatogenesis in some men more than 1 year after
radiation [70] or chemotherapy [71] also implies a
block in the differentiation of spermatogonia that survive these cytotoxic exposures. These results suggest
that after some cytotoxic therapy regimens, there is a
potentially reversible block to spermatogonial differentiation in men. It should be noted that there is no
evidence of a similar spermatogonial block in monkeys [13]. The human data emphasize that the cytotoxic therapy regimens need to be carefully selected in
these clinical trials from a range of regimens. To have a
chance of success, doses of cytotoxic therapies should
be chosen at which there is an appreciable block to
171
the initial action of the hormones. It may be possible to suitably modulate those targets in primates to
release the blockade in spermatogenic differentiation,
while maintaining hormone levels, which should allow
spermatogonial differentiation. In addition, elucidation of mechanisms for greater sensitivity of primate
germ cells to cancer therapeutic agents and development of ideal conditions for germ cell transplantation
in primates may open strategies to recover spermatogenesis and sperm count in toxicant-exposed human
males.
Acknowledgements
The authors research in this review was supported by
Research Grant ES-08075 from the US National Institute of Environmental Health Sciences (National Institutes of Health). I am thankful to Dr. Marvin Meistrich
for his valuable help in preparing this review.
References
1. Whorton D, Milby TH, Krauss RM and Stubbs HA.
Testicular function in DBCP exposed pesticide
workers. J Occup Med 1979; 21(3): 1616.
2. Slutsky M, Levin JL and Levy BS. Azoospermia and
oligospermia among a large cohort of DBCP
applicators in 12 countries. Int J Occup Environ Health
1999; 5(2): 11622.
3. Meistrich ML, Zhang Z, Porter KL, Bolden-Tiller OU
and Shetty G. Prevention of adverse effects of cancer
treatment on the germline. In: Anderson D and
Brinkworth MH (eds.), Male-Mediated Developmental
Toxicity. Cambridge: Royal Society of Chemistry,
2007: pp. 11423.
4. Glode LM, Robinson J and Gould SF. Protection from
cyclophosphamide-induced testicular damage with an
analogue of gonadotropin-releasing hormone. Lancet
1981; 1: 11324.
5. da Cunha MF, Meistrich ML and Nader S. Absence of
testicular protection by a gonadotropin releasing
hormone analog against cyclophosphamide-induced
testicular cytotoxicity in the mouse. Cancer Res 1987;
47: 10937.
6. Meistrich ML, Wilson G, Zhang Y, Kurdoglu B and
Terry NHA. Protection from procarbazine-induced
testicular damage by hormonal pretreatment does not
involve arrest of spermatogonial proliferation. Cancer
Res 1997; 57: 10917.
7. Meistrich ML. Quantitative correlation between
testicular stem cell survival, sperm production, and
fertility in the mouse after treatment with different
cytotoxic agents. J Androl 1982; 3: 5868.
172
173
174
175
Section 4
Chapter
14
Cryopreservation of spermatozoa
Old routine and new perspectives
E. Isachenko, V. Isachenko, R. Sanchez, I. I. Katkov and R. Kreienberg
Introduction
Cryopreservation of male and female gametes has
been long established, and nowadays low-temperature
storage of human spermatozoa is a routine technique in assisted reproduction. This technique offers
the following advantages over the use of fresh ejaculated spermatozoa: (1) storage of both homologous
or donor sperm for subsequent intrauterine insemination, in vitro fertilization (IVF) and intracytoplasmic
sperm injection; (2) long-term storage of known quality donor semen; (3) the ability to quarantine donor
semen until appropriate testing can be completed;
(4) preservation of epididymal or testicular spermatozoa/tissue for subsequent intracytoplasmic sperm
injection or necessary diagnosis [1, 2]. Recently, fertility preservation has been a hot topic and attracted
much attention because chemotherapy and radiotherapy result in a significant reduction of spermatozoa
quality and, as a consequence, a following indefinite
period of infertility [35]. Human genome banking
is one specific approach that can be used to preserve
male genetic material before sterilization, chemotherapy or radiotherapy. It can also be used for males
with autoimmune diseases [2, 4, 68] or before some
treatments and special kinds of surgical procedures
which may lead to testicular failure or ejaculatory
dysfunction [9].
The empirical methods of cryopreservation developed in the 1950s are still used today [10, 11] and based
on use of a relatively slow cooling rate (1170 C/min)
in a region of critical temperatures (10 to 60 C). At
present the major steps used for cryopreservation of
different kind of cells can be summarized as follows:
(1) Adding cryoprotective agents (CPAs) before
freezing. This substance enhances post-thaw survival
by limiting the crystallization of water. (2) The seeding
Vitrification: a promising
new direction
The success of these current procedures brings up the
reasonable question: Why do we need to develop
other technique and what advantages will it give
us compared to the current system? This new
technique is vitrification and it could be beneficial
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
176
60]. The question of diminished spermatozoa motility after cryopreservation is crucial since this variable
is known to be the first affected by cryopreservation
[61], although the mechanism of sperm impairment
and its mechanical and/or physicalchemical etiology
remains unclear. The reason for cell damage could be
due to mechanical cell injury, as a consequence of
intracellular or extracellular ice crystal formation, and
osmotic damage due to extensive cell shrinkage during
conventional freezing. The warming process also has a
negative influence on cells, possibly through excessive
osmotic swelling [10, 62]. Both these factors accompany slow freezing due to the chemical and physical damage of the sperm cell membranes, which is
the result of changes in lipid phase transition and/or
increased lipid peroxidation and active production of
reactive oxygen species [63] and the subsequent loss
of sperm motility [24, 64]. The permeable and nonpermeable cryoprotectants, used during conventional
freezing to prevent intracellular ice formation, can be
damaging due to the so-called CPAs toxicity [30]. This
toxicity includes damage to the cells membrane due to
the osmotic and chemical influence of CPAs on cells
during freezing and/or thawing [62, 65], which activates an apoptosis-like mechanism [66] that can also
lead to chromatin damage [67]. All of these findings
suggest that, quite apart from ice crystal formation,
slow cooling, especially the thawing of spermatozoa,
is intrinsically deleterious.
Compared to conventional slow freezing, the vitrification technique, even though it has its own peculiarities, is a real alternative.
177
178
In other words, the faster the cooling and warming is undertaken, the lower the critical solute concentration necessary to obtain ice-free vitrification
[34, 41, 77]. Given the biological and physiochemical effects of cryoprotectants and the high concentrations used in vitrification, it is not surprising that
cryoprotectant toxicity has been described as a key
limiting factor in the cryobiology of vitrification [34,
41]. In an attempt to avoid this toxicity, we achieve
ultra-rapid cooling and warming rates, in the range of
0.51.0 106 K/min, using a relatively dilute vitrification medium (around 12% total solute content) of
similar solute concentration to semen or blood; thus,
resurrecting the original vitrification approach of
Luyet.
179
Table 14.1. The relationship between the size (approximate) of mammalian sperm heads and stability of gametes to cold shock
Boar
Bull
Ram
Rabbit
Cat
Dog
Stallion
Human
Length (m)
9.0
9.1
8.1
7.7
7.7
7.0
6.5
4.6
5.0
4.7
4.0
4.5
3.2
5.0
3.4
3.2
37.5
34.2
28.0
++++
+++
+++
++
15.2
10.8
that cryoprotectants are not always needed for successful vitrification. The first investigations in this direction had very contradictory results. Luyet, the pioneer in vitrification investigation, showed that a small
specimen cooled very rapidly could be vitrified without substantial loss of viability [34]. There followed
subsequent investigations by Jahnel [103] and then
Parkes [104] in which they performed cryoprotectantfree cryopreservation of human spermatozoa cooled
in LN2 and liquid helium (269.5 C) using glass or
metal tubes as packaging with a large volume of sperm
suspension (a few milliliters). They did not get such
promising results, seemingly due to the difficulty of
achieving a quick enough mode of warming. All subsequent attempts to vitrify mammalian spermatozoa
using this approach resulted in low or null survival [43,
44], mostly because of the critical speed of freezing and
warming, which is very high for low concentrations of
cryoprotectants. Such situations were why it was considered that the vitrification could only be achieved
using high concentrations of permeable cryoprotectants alone or in combination with non-permeable
cryoprotectants [105]. However, classical vitrification
requires a high percentage of permeable cryoprotectants in medium (3050% compared to 57% with
slow freezing) and is unsuitable for the vitrification of
spermatozoa due to the lethal osmotic effect [65, 70].
It was reported that, by optimizing regimens for the
addition and removal of the CPAs, it may be possible
to significantly reduce the damaged effect of these substances on male gametes during the equilibration time
before freezing [13, 62, 65, 106]. But, based on numerous publications dedicated to this theme, it must be
emphasized that generally, in the majority of species,
sperm cannot tolerate the high concentrations of cryoprotectants conventionally used for vitrification. One
peculiarity of male gametes could be a significant factor: the shape and size of the sperm head, which defines
the cryosensitivity of the cell. Comparative studies on
180
Scandinavian IVF Science, Sweden). The swim-up prepared spermatozoa, conventionally frozen with cryoprotectants immediately after thawing, showed 38%
motility compared to 49% after vitrification without cryoprotectants. The number of morphologically
normal spermatozoa after applying both cryopreservation methods was not significantly different (27%
for slow frozen with permeable cryoprotectants versus 26% for vitrified without permeable cryoprotectants, P 0.01). Programmable freezing in the absence
of cryoprotectants dramatically reduced all sperm
variables.
The negative influence of freezing on cells can lead
to chromatin damage and is strongly correlated with
mutagenic effects [23]. It has been shown that there is a
correlation between fertilization rates in IVF [110] and
intracytoplasmic sperm injection (ICSI) [111] with
fragmented DNA. We performed the comet assay to
investigate the occurrence of apoptosis in spermatozoa DNA after vitrification (Figure 14.1). Our albumin estimates [26] showed that devitrification (especially intracellular) did not occur [112] during cooling
or, especially, during re-warming/resuscitation, due to
the small amount of specimen and cells, the high vis-
cosity of the solution and the high speed of cooling and warming [30]. Our investigation showed that
no significant difference in DNA integrity was found,
independent from the mode of cooling and the presence of cryoprotectants (P 0.05) (Figure 14.2) [26].
We found that the DNA integrity of vitrified spermatozoa was comparable with standard slow frozen
and thawed spermatozoa. The DNA integrity (P
0.05) was found to be unaff1ected by the vitrification method. Our results supported the results of the
Schrader group [113] and Duty [114]. They found no
difference in sperm chromatin structure assay results
for cryopreserved or non-cryopreserved sperm, or for
slowly or flash-frozen specimens [113]. They suggested
that the unique packaging of sperm DNA protects
it from intracellular fluid shifts and the formation
of the nuclei of crystallization during the cooling
warming cycle. The use of spermatozoa vitrified using
this method for IVF of human oocytes resulted in successful fertilization and development to the blastocyst
stage [33].
The next modification of the vitrification method
was the direct dropping of spermatozoa suspension
into LN2 (Figure 14.3 [115]) [32, 49]. Immediately
following vitrification, 30 l of sample was dropped
directly into LN2 . The temperature of the foil was
determined by an electrical thermometer. After 5 min
of cooling, the solution-precursor-solid (SPS) droplets
were placed into pre-cooled LN2 Cryovials and stored
there until the time of use. The warming of the solid
sphere was performed by directly plunging it into
a centrifuge tube with 10 ml of sperm-preparation
medium (SPM) at 37 C under intense agitation.
Based on our previous data and the peculiarities
of carbohydrates, we decided to investigate the contribution of disaccharide sucrose to sperm cryoprotection during cooling/warming using the dropping
method. The investigation focused especially on the
ability of sucrose to support the motility of spermatozoa, prevent the artificial induction of capacitation or
acrosome reaction and support mitochondrial stability during vitrification/warming. The viable, acrosome
reacted and capacitated spermatozoa (Figure 14.4)
were detected using the double-fluorescence Chlortetracycline (CTC)-Hoechst 33258 staining technique
[116].
The results have shown that the medium supplemented with 1% HSA + 0.25 M sucrose allowed the
motility rate to increase after warming to 57%, compared to HSA alone (45%) or pure medium without
181
105
100
95
90
85
80
75
70
65
Swim up
Swim up
Slowly frozen Slowly frozen
Vitrified
Vitrified
without
with
without
with
without
with
cryoprotectant cryoprotectant cryoprotectant cryoprotectant cryoprotectant cryoprotectant
Method of treatments
Figure 14.2 DNA integrity of spermatozoa according to different treatments and cryopreservation methods. Each bar represents the
medium, 25 and 75 percentile, minimum and maximum values. Bars with different letters inside of each treatment group represent significant
differences (P 0.05). With permission from Isachenko et al. [26].
182
(b)
(a)
(c)
(d)
Figure 14.4 Example of non-capacitated (a), capacitated (b), acrosome-reacted (c) and non-viable (d) spermatozoa. At least 200
spermatozoa were observed in each plate and 3 patterns were identified (see Figure 14.2). (a) A uniform fluorescence on the head of the
spermatozoa (non-capacitated spermatozoa). (b) A band of fluorescence diminished in the post-acrosomal region and a relatively shining
fluorescence in the acrosomal region (capacitated spermatozoa). (c) A fluorescence in the complete head of the spermatozoa, except a
tenuous band of fluorescence in the equatorial segment (acrosome-reacted spermatozoa). The slides were viewed using a Zeiss Axiolab
Epifluorescence microscope that was equipped with an excitation/emission filter of 485 nm/520 nm under 400 magnification. The
non-viable spermatozoa were observed with the filter set 09 (450490 nm). The dead spermatozoa displayed a pattern of blue fluorescence in
the whole head (Figure 14.2d). With permission from Isachenko et al. [115]. See plate section for color version.
document post-thaw sperm survival by determination of mitochondrial activity. It has previously been
demonstrated that mitochondrial activity and viability
are equally susceptible to cryopreservation-induced
damage [25]. It has been suggested, however, that
current conventional freezing techniques for human
sperm cryopreservation are good enough to preserve
all these structures [24, 25]. The non-permeable
cryoprotective substances also showed stable cryoprotective effect on the mitochondrial membrane (Figure
14.5c). It was observed that the medium alone has no
cryoprotective effect (8%). The combination of HSA
and sucrose is more effective then addition of HSA
only (65 and 33%, respectively; P 0.05).
Unfortunately, these methods have a large technological disadvantage: their techniques do not prevent
direct contact with LN2 . At present, numerous publications exist about microbial contamination of the
different kinds of biological material cryopreserved
in LN2 [120123]. Because of this, a special kind of
183
Motility of
spermatozoa
immediately after
warming
100
90
80
70
Motility of
spermatozoa
after 24 h
60
50
Viability
immediately after
warming
40
30
20
10
0
HTF
HTF + HSA + S
HTF + HSA
Vitrification media
(a)
Capacitated/acrosome
reacted spermatozoa (%)
100
Capacitation
80
Acrosome
reaction
60
40
20
0
HTF
HTF + HSA
HTF + HSA + S
Vitrification media
(b)
100
80
60
40
20
0
HTF
(c)
HTF + HSA
Vitrification media
HTF + HSA + S
Figure 14.5 (a) Cryoprotective effect of human tubal fluid (HTF) alone or in combination with human serum albumin (HSA) or with HSA +
sucrose on motility and viability of vitrified spermatozoa. (b) Cryoprotective effect of HTF alone or in combination with HSA or HSA + sucrose
on activation of capacitation and induction of acrosome reaction of vitrified spermatozoa. (c) Cryoprotective effect of HTF alone or in
combination with HSA or HSA + sucrose on the mitochondrial membrane integrity of vitrified spermatozoa. Asterisks denote statistical
differences between respective values of compared groups (P 0.05). With permission from Isachenko et al. [115].
184
it was necessary to try and develop an aseptic vitrification technique that prevented direct contact with
LN2 . To do this, it was necessary to discover if the
cooling or warming rate of vitrification most affected
the spermatozoa parameters. According to the famous
90
80
Viability
70
Capacitation
60
Acrosome reaction
Percent (%)
100
50
Mitochondrial membrane
potential
40
30
20
10
0
HTF
Figure 14.6 Influence of human tubal fluid (HTF) medium alone or in combination with human serum albumin (HSA) or HSA + sucrose on
motility, viability, activation of capacitation, induction of acrosome reaction and mitochondrial membrane integrity before cryopreservation.
No statistical differences were found between the respective values of compared groups (P 0.1) except for motility 24 h after in vitro culture
(P 0.05). With permission from Isachenko et al. [115].
(a)
(b)
185
100
90
80
70
Rate (%)
60
b
Motility
50
DNA integrity
40
30
20
10
0
Fresh (control)
Quick
Mode of cooling
Slow
186
10
12
12
13
15
16 h, 2 PN
and 3 PN
(n)
3 transfer
3 transfer
3 transfer
3 transfer
3 transfer
3 transfer
2 transfer
2 transfer
48 h, 46
blastomeres
(n)
56 h, EB and
BL (n)
Oocytes
(n)
16 h, 2PN
and 3PN
(n)
48 h, 46
blastomeres
(n)
56 h, EB
and BL
(n)
(2PN) oocytes with two pronuclei; (3PN) oocytes with three pronuclei; (EB) early blastocysts; (BL) blastocysts.
No differences between spermatozoa vitrified using quick and slow cooling (P 0.01).
With permission from Isachenko et al. [27].
Oocytes
(n)
Total
oocytes
(n)
Oocytes
(n)
16 h, 2PN
and 3PN
(n)
48 h, 46
blastomeres
(n)
56 h, EB
and BL (n)
Table 14.2. Fertilization properties of spermatozoa after cryoprotectant-free cryopreservation with rapid and slow cooling: results of in vitro fertilization and culture
(a)
(b)
Figure 14.9 Two modifications of aseptic vitrification technique for spermatozoa. (a) Photograph of container and method for Open Pulled
Straw vitrification and warming of spermatozoa. (1) Open pulled straw. (2) Suspension of spermatozoa (510 l). (3) Meniscus of suspension.
(4) 90 mm straw. (5) Heat-sealed end of 90 mm straw. (6) Marked end of Open Pulled Straw. (7) Tube for warming. (8) Warming medium.
White arrow indicates the direction of thawing and swim-up of sperm suspension. (b) Photograph of container and method for open straw
vitrification and warming of spermatozoa. (1) Tip of pipette. (2) Open straw. (3) Drop of spermatozoa (0.52 l). (4) A 90 mm straw. (5)
Heat-sealed end of a 90 mm straw. (6) Marked end of open straw. (7) Tube for warming. (8) Warming medium. White arrow indicates the
direction of thawing and swim-up of sperm suspension. With permission from Isachenko et al. [49].
sealed from both sides with a hand-held sealer (Medical Technology GmbH, Bruckberg, Germany) and
plunged into LN2 . The warming of the spermatozoa
sample was performed a day earlier than the planned
intracytoplasmic sperm injection procedure as fol-
188
3
1
4
189
*
*
*
(a)
(d)
(b)
(e)
(g)
(h)
(c)
(f)
Figure 14.12 The vitrification procedure for big volume of spermatozoa suspension. (a) The 0.25 ml plastic straws are halved and
dark-marked from one side (dark asterisks) and a 0.5 ml packaging straw (white asterisks). (b) The non-marked side of the half-straw is filled
with spermatozoa suspension. (c) A 0.25 ml half-straw, hermetically closed from both sides, is filled with spermatozoa suspension. (d) Holding
it in a horizontal position, the 0.25 ml half-straw filled with spermatozoa suspension is marked from one side (arrowed) and placed into a
0.5 ml packaging straw, closed from both sides. (e) Using tweezers and keeping it in a horizontal position, this closed packaging system is
directly immersed into liquid nitrogen (LN2 ) and submerged for over 5 s to prevent the flow of spermatozoa suspension spreading into
packaging straw. (f) The vitrified sample is stored in LN2 . Using tweezers, the dark-marked part of the Sealed Pulled Straw (SPS), approximately
1.01.5 cm, is removed from the LN2 and the end of packaging straw is cut. (g) With the help of a 200 l pipette tip (Eppendorf AG, Hamburg,
Germany), the suspension-filled fixed straw is quickly removed from the packaged straw and (h) immersed into a 15 ml plastic tube
containing 6 ml of human tubal fluid (HTF) and human serum albumin (HSA) prewarmed to 37 C with gentle agitation to accelerate the
melting and removing of content. See plate section for color version.
190
100
90
After warming
80
70
Motility (%)
achieve the final concentration of 5 106 spermatozoa/ml. For vitrification, one half of the 0.25 ml plastic straw was used. For this, the straw was cut into
two parts and a dark mark made from one side. All
subsequent manipulations were done strictly in a horizontal position to prevent the loss of spermatozoa
suspension. The half-straw was filled from the nonmarked side with 100 l of spermatozoa suspension.
Then sticky tape in a horizontal position was put into a
0.5 ml packaging straw, previously hermetically closed
from the other side, and hermetically closed. Then
the straw packaging system (SPS), held by tweezers to
strictly keep it in a horizontal position, was directly
immersed into LN2 and submerged there for over 5 s
to prevent the flow of spermatozoa suspension into the
packaging straw. It was then stored in LN2 for at least
24 h before use.
The warming of spermatozoa suspension was done
as follows (Figure 14.12g,h): Before removing the SPS
from the LN2 it was necessary to find the side of
the dark-marked spermatozoa suspension-filled straw.
Using tweezers, this part of the SPS, approximately
1.01.5 cm, was removed from the LN2 . The end of
the packaging straw was cut and then, with the help
of pipette tip for 200 l, the suspension-filled straw
was fixed, quickly removed from the packaged straw
and immersed into a 15 ml plastic tube containing
10 ml of HTFHSA prewarmed to 37 C with gentle agitation to accelerate the melting and removing of content. After warming, the spermatozoa were
concentrated by centrifugation at 340 g for 5 min
and the sediment was resuspended with HTFHSA.
As a control, before cryopreservation we tested the
influence of basic medium (HTFHSA), vitrification
medium (HTFHSAsucrose) and the TESTyolk
buffer (TYB) medium used for conventional freezing,
on the physiologic parameters of spermatazoa (Figures
14.13, 14.14). The motility of spermatozoa 1 h after
warming, with subsequent incubation at 37 C in a CO2
atmosphere, and after 24 h in vitro culture is shown in
Figure 14.13. The data shows that our newly developed
method of vitrification for relatively large volumes of
spermatozoa suspension (100 l, compared to small
volume 130 l) achieves high spermatozoa motility (62%) after warming and satisfactory motility after
24 h in vitro culture (22%). This is not statistically different from slow frozen spermatozoa at 24 h (24%).
Indeed, all of the data are similar to the slow-freezing
technique.
60
After 24 h
*
*
50
40
30
20
10
0
Native
Vitrification
Slow
Type of treatment
The influence of the type of treatment on the stability of mitochondrial membrane potential M ,
the plasmatic and acrosomal membrane, induction of
cryo-capacitation (phosphatidylserine translocation)
and level of DNA fragmentation is shown in Figure
14.14. The data shows that aseptic vitrification had
a significantly stronger protective effect on the mitochondrial membranes (Figure 14.14a) potential stability (72%) compared to conventionally frozen spermatozoa (30% P 0.05).
The integrity of the cytoplasmic membrane (Figure 14.14b) was significantly affected by both cryopreservation methods, but protection of the cytoplasmic membrane by conventional slow freezing was significantly lower (30%, P 0.05) than by vitrification
(54%).
Both aseptic vitrification and conventional freezing negatively influence the acrosomal membrane
integrity (28.0 6.9% versus 41.4 2.5%, respectively; P 0.05; Figure 14.14c). Although the acrosomal membrane integrity was lower after applying aseptic vitrification, the index of spermatozoa integrity was
non-statistically different from the number of conventionally frozen spermatozoa with non-damaged acrosomal membrane.
It was noted that this vitrification technique
strongly prevented the translocation of phosphatidylserine (2%; Figure 14.14d) compared to
phosphatidylserine quantitative indexes of conventionally frozen spermatozoa (20%; P 0.01). Both
191
100
Mitochondrial membrane
potential (JC-1)
80
103
60
40
101
PSAFITC 2 VII 280607.001
10
10
1
*
Native
Vitrification
Slow
Type of treatment
100
101
102
103
104
10
20
Translocation of
phosphatidylserine (anexin V)
103
15
102
10
Native
Vitrification
Slow
100
101
Type of treatment
8
7
6
5
4
3
2
1
0
104
10
25
(d)
(e)
10
10
10
10
10
Fragmentation of DNA
(TUNEL)
104
Damage of acrosomal
membrane (PSA-FITC)
10
I VII 290607.031
103
104
102
SYBR-14
103
10
102
20
10
101
10
101
10
10
10
30
10
100
40
Slow
10
Vitrification
Type of treatment
Native
Native
Vitrification
Type of treatment
Slow
100
10
4
1
20
50
Acrosomal membrane
damage ( %)
40
(b)
(c)
60
10
Phosphatidylserin
(anexin V) translocation (%)
Integrity of cytoplasmatic
membrane (SYBR-14)
Slow
Type of treatment
10
Cytoplasmic membrane
integrity (%)
80
Vitrification
100
Native
(a)
100
102
20
0
PM VIT 2 250607.076
104
Mitochondrial membrane
potential (%)
100
101
102
103
104
Figure 14.14 Influence of type of sperm treatment on spermatozoa mitochondrial membrane potential integrity, plasmatic and acrosomal
membrane integrity, stability of phosphatidylserine and level of DNA fragmentation. (a) Mitochondrial membrane potential integrity. (b)
Plasmatic membrane integrity. (c) Acrosomal membrane integrity. (d) Phosphatidylserine (anexin-V) translocation ability. (e) Level of DNA
fragmentation. Different superscripts indicate significant differences (P 0.05).
cheap, quick and successful for different types of reproductive (different stages of maturity), somatic, stem
cells and, seemingly, tissues. Due to the lowering of
a solutions glass transition temperature, permeable
cryoprotectants:
r prevents actual freezing and
r maintains some flexibility in a glassy phase.
192
The other future perspective in the long-term storage of male gametes is lyophilization. This method
provides a low cost for storage and transport of preserved materials because the biological material can
be stored at 4 C and shipped at ambient temperatures. This technique is very suitable for preservation
of the genome (nucleus), because nuclear viability is
not equivalent to cell viability and is not destroyed by
freezing/drying [131135]. In 2003, Ward, from the
Yanagimachi group, wrote wrote that: (1) the freezedrying procedure itself causes some abnormalities in
spermatozoa but freezing without cryoprotection does
not; and (2) long-term storage of both frozen and
freeze-dried spermatozoa is not deleterious to their
genetic integrity [136]. Freezing without cryoprotection is highly successful, simple and efficient but, like
all routine sperm storage methods, requires LN2 . Liquid nitrogen is also required for freeze-drying but
sperm can then be stored at 4 C and shipped at ambient temperatures. Both preservation methods are successful, but rapid freezing without cryoprotection is
the preferred method for preservation of spermatozoa
from mouse strains carrying unique genes and mutations. The genomic integrity of cells can be maintained
after freeze-drying and it is possible to produce offspring from the cells using nuclear transfer techniques
[131, 137, 138]. In this case, the long-term preservation of mouse sperm by desiccation is economically
and logistically attractive [139].
Conclusions
References
1. Keel BA and Webster BW. Semen analysis data from
fresh and cryopreserved donorejaculates: comparison
of cryoprotectants and pregnancy rates. Fertil Steril
1989; 52: 1005.
2. Nijs M and Ombelet W. Cryopreservation of human
sperm. Hum Fertil 2001; 4: 15863.
3. Meistrich ML, Vassilopoulou-Sellin R and Lipshultz
LI. Adverse effect of treatment: gonadal dysfunction.
In: DeVita VT, Hellman S and Rosenberg SA (eds.),
Cancer. Principles and Practice of Oncology, 7th edn.
Philadelphia: Lippincott Williams and Wilkins, 2005:
pp. 256074.
4. Trottmann M, Becker AJ, Stadler T et al. Semen
quality in men with malignant diseases before and
after therapy and the role of cryopreservation. Eur Urol
2007; 52: 35567.
5. Quinn B and Kelly D. Sperm banking and fertility
concerns: enhancing practice and the support available
to men with cancer. Eur J Oncol Nurs 2000; 4: 558.
6. Sanger WG, Oslon JH and Sherman JK. Semen
cryobanking for men with cancer criteria change.
Fertil Steril 1992; 58: 10247.
7. Brannigan RE and Sandlow JI. Cryopreservation of
sperm after chemotherapy. J Androl 2008; 29: e12.
8. Ginsberg JP, Olge SK, Tuchman LK et al. Sperm
banking for adolescent and young cancer patients:
sperm quality, patient, and parent perspectives. Pediatr
Blood and Cancer 2008, 50: 59498.
9. Donnelly ET, Steele EK, McClure N et al. Assessment
of DNA integrity and morphology of ejaculated
spermatozoa from fertile and infertile men before and
after cryopreservation. Hum Reprod 2001; 16: 11919.
10. Gao D, Mazur P and Critser J. Fundamental
cryobiology of mammalian spermatozoa. In: Karow
AM and Critser JK (eds.), Reproductive Tissue Banking.
London: Academic Press, 1997: pp. 263328.
193
194
195
196
197
198
Section 4
Chapter
15
Transplantation of cryopreserved
spermatogonia
Jill P. Ginsberg and Ralph L. Brinster
Background
Over the last several decades, survival rates for childhood cancer have steadily increased. In fact, with
the overall cure rate for pediatric malignancies now
approaching 80%, current estimates indicate that 1
in every 640 young adults in the USA will be a survivor of childhood cancer [1]. Unfortunately, many
survivors struggle with medical late effects of their
treatment including disorders of the endocrine system, cardiac and pulmonary dysfunction, secondary
neoplasms and infertility. Gonadal damage is a relatively common consequence of the treatments used
to cure pediatric cancer. The extent of cytotoxic germ
cell damage depends on the specific chemotherapeutic
agents used and the cumulative doses received. Alkylating agents (particularly cyclophosphamide, ifosfamide, nitrosureas, chlorambucil, melphalan, busulfan and procarbazine) are the most common class
of drugs known to effect gonadal function and their
impact has been studied extensively [2]. Additionally,
the testes have a very low threshold for radiation exposure, and even small doses are known to be gonadotoxic. As treatment regimens for pediatric oncological
malignancies have improved, more and more survivors are entering their reproductive years [3]. Maintenance of fertility is extremely important with regard
to long-term quality of life for these survivors [4, 5].
Consideration must be given to whether a childs fertility is likely to be impacted by his treatment. Ideally, this should occur before the start of therapy, when
a window of opportunity may exist to preserve the
patients future reproductive potential [48]. Pubertal
males can produce a semen sample prior to starting
gonadotoxic therapy and cryopreserve the sperm for
future use. Because current methods for oocyte fertilization can utilize as few as one motile sperm (e.g.
intracytoplasmic sperm injection [ICSI]), this method
has proven to be successful even when the number of
cryopreserved sperm is small [5, 9, 10].
Unfortunately, pre-pubertal males pose a particular challenge for fertility preservation because
these boys cannot produce mature spermatozoa for
cryopreservation. During embryonic development,
primordial germ cells (PGCs) migrate to the genital
ridge and differentiate into gonocytes [11, 12]. In the
mammalian postnatal testis, gonocytes are the first cell
population committed to male germline development.
Before puberty, these cells then give rise directly to
spermatogonial stem cells (SSCs). In the mouse, which
has a short pre-pubertal period (3 weeks), some
gonocytes transition to SSCs and others undergo an
early differentiation directly to type A1 spermatogonia
by day 6 of life. During the first 23 months after birth
in humans, which have a long pre-pubertal period
(12 years), the gonocytes are replaced by adult
dark (Ad) and adult pale (Ap) spermatogonia that
are thought to represent the reserve SSC and active
SSC pool [12, 13]. These Ad and Ap spermatogonia
undergo activation beginning at approximately age
5 years, particularly to type B spermatogonia. By
age 10 years, these SSC represent about 10% of total
spermatogonia. During puberty, the SSCs in all species
provide the foundation for spermatogenesis, through
self-renewal and differentiation to daughter cells.
Although the germ cells of the pre-pubertal testis contain a small number of the self-renewing SSCs they do
not yet have mature spermatozoa. For these at risk prepubertal boys, current practice does not provide any
options for fertility preservation at cancer diagnosis.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
199
Culture
Transplantation
Cryopreservation
Sperm
production
Testis
biopsy
Chemotherapy
or irradiation
Figure 15.1 Male germline stem cell preservation. Before treatment for cancer by chemotherapy or irradiation, a boy could undergo a
testicular biopsy to recover stem cells. The stem cells could be cryopreserved or, after development of the necessary techniques, could be
cultured. After treatment, the stem cells would be transplanted to the patients testes for the production of spermatozoa. From Brinster [14]
with permission. See plate section for color version.
200
Development of spermatogonial
transplantation
Spermatogonial stem cells are responsible for the continual production of spermatozoa throughout adult
life. Both SSCs and the surrounding cells in the seminiferous tubules regulate the biological activity of
these cells. Considerable research has been dedicated
to understanding the interactions between SSCs and
the surrounding somatic cells for proper sperm production. For example, Sertoli cells are thought to be
extremely important for SSC growth and development
by secreting growth factors that regulate these germ
cells [19, 20]. Importantly, a critical breakthrough in
the characterization of SSCs has been the development
of the germ cell transplantation technique [11, 15, 21].
In 1994, Brinster and Avarbock developed the first animal model of SSC transplantation [21]. Injection of
spermatogenic cells into the seminiferous tubules gave
rise to donor cell-derived foci of spermatogenesis in
the recipient testes.
Figure 15.2 presents a schematic overview of spermatogonial transplantation in mice. First, transgenic
mice carrying a LacZ or green fluorescent protein
(GFP) transgene are used as donor mice. The marked
Germ
cells
Enzyme
In situ
injection
digestion
Testis with
transgene
35 months
Donor genes
transmitted
X Mate
Progeny
Figure 15.2 Testis cell transplantation method. A single-cell suspension is produced from a fertile donor testis. The cells can be cultured or
microinjected into the lumen of seminiferous tubules of an infertile mouse. Only a spermatogonial stem cell can generate a colony of
spermatogenesis in the recipient testis. When testis cells carry a reporter transgene that allows the cells to be stained blue, colonies of donor
cell-derived spermatogenesis are identified easily in the recipient testes as blue stretches of tubule. Mating the recipient male to a wild-typed
female produces progeny, which carry donor genes. From Brinster [11] with permission. See plate section for color version.
Morphological identification of
spermatogonial stem cells
The number of SSCs is very low in the testis of
an adult mouse. It is estimated that SSCs consti-
201
202
Tight
junction
Sertoli cell
Sertoli
cell
-1
GFR
Human
spermatogonial
stem cell
SFK
PI3K AKT
PI3K/AKT
Etv5, Bcl6b
Self-renewal
Differentiation
Figure 15.3 A proposed model of human spermatogonial stem cell (SSC) self-renewal regulation by glial cell line-derived neurotrophic
factor (GDNF), which has been demonstrated to have an essential role in regulating rodent SSC self-renewal. The model is similar to those
suggested for mouse SSC self-renewal. In this model, GDNF binds to RET and the GFR1 coreceptor with possible intracellular protein kinase
signaling through SFK and PI3K/AKT downstream pathways to regulate the expression of specific genes, such as Etv5 and Bcl6b, which are
involved in SSC self-renewal. However, other genes not regulated by GDNF (e.g. Zbtb16, Taf4b and Lin28), are likely controlled by different
signals and may block differentiation but not be involved directly in self-renewal. Genes for these regulatory molecules have been shown to
be highly expressed in pre-pubertal human spermatogonia, mouse gonocytes and mouse SSCs. The basement membrane (green), on which
the SSC rests, is generated by the peritubular myoid cells (dark brown) and Sertoli cells (tan). From Wu et al. [27] with permission. See plate
section for color version.
203
204
Cryopreservation of spermatogonial
stem cells
Once spermatogonial transplantation systems were
established in vivo, it became critical to determine
whether SSCs could be cryopreserved for potential fertility preservation in humans that would otherwise
be infertile due to their gonadotoxic pediatric cancer treatments. To date, these procedures are primarily experimental, and the current success lies within
animal models, not actual human patients. Notably,
the process of SSC transplantation in animal models includes a number of steps in the handling of
SSCs, and preparing the receiving testis for transplantation. Stem cells must be appropriately isolated and
enriched. For practical purposes in human testis banking, SSCs and related germline cells must be cryopreserved. Cryopreservation enables future manipulation and evaluation of SSCs in culture over a period
of time to select cells with the desired properties (i.e.
non-malignant). Investigators have routinely collected
testis cells from many species after enzyme digestion,
frozen these cells and stored them in liquid nitrogen [14, 16, 38, 39]. When ready to be utilized, cells
are thawed and transplanted into sterile recipients.
This procedure has been determined experimentally to
be quite effective, as infertile mice that receive cryopreserved SSC transplants develop donor-cell derived
spermatogenesis [16]. To date, several groups have
demonstrated that SSCs can be cryopreserved and not
lose their ability to restore spermatogenesis in animal models [11, 16, 38]. Transplantation of cryopreserved and fresh testis cells from non-rodent species,
including human, into testes of immunodeficient mice
has been successfully completed and the data demonstrate that cryopreserved donor human spermatogonia colonize mouse testes similar to fresh spermatogonia (Figure 15.4) [17]. Recently, primate testicular
cell suspensions were frozen [38] and, after thawing,
viability was found in 58% of the cryopreserved cells.
Importantly, Keros et al. have documented successful freezing protocols using human testicular tissues
[39]. Specifically, slow programmed freezing with 5%
dimethyl sulfoxide (DMSO) as a cryoprotective agent
is efficient in maintaining spermatogonia, Sertoli cells
and the stromal compartment from testicular biopsies during the freezing, thawing and subsequent tissue
culture procedures required for successful accession
and utilization of human clinical samples. It has been
estimated that 5066% of spermatogonial cells can
(a)
(b)
Figure 15.4 Detection of human germ cells transplanted into seminiferous tubules of recipient mouse testes using a baboon testis specific
antibody that identifies human spermatogonia. (a) Donor human spermatogonia in mouse tubule 4 months after transplantation of
cryopreserved cells. This panel shows that cryopreserved cells also colonize mouse testes as observed with freshly transplanted cells. (b)
Donor human spermatogonia in mouse testis 5 months after transplantation. These donor cells were transplanted without cryopreservation.
C 2002 American Society for
Bar = 100 m (a) and 40 m (b). From Nagano et al. [17] with permission from Elsevier Science, Inc.
Reproductive Medicine. See plate section for color version.
205
Ethical concerns
As testicular cryopreservation is an experimental procedure in humans, efficacy and safety research is governed under the auspices of federal regulations for
clinical trials involving children. These federal regulations mandate that federally funded clinical research
protocols including children that involve greater than
minimal risk, and that present the potential of direct
benefit to these subjects, must be reviewed by an Institutional Review Board (IRB) [48]. The IRB approval
will only be granted when direct benefits of the
experimental treatment outweigh its risks, and that
research is likely to provide important information
that leads to better understanding of the conditions
therein [48]. Strict adherence to these guidelines is
crucial regardless of the funding source for clinical
trials involving children, as they provide a solid ethical cornerstone for clinical and translational research
[48].
Tissue banking centers that accrue gonadal tissue for pediatric cancer patients must inform the parents of the options for disposition of these materials at a future time [49]. These issues are not trivial,
as the legal system upholds the prior wishes from an
individual regarding the disposition of reproductive
material, which is controlling after death. Therefore,
tacit instructions that biological materials must be
destroyed, or alternatively released to research, have to
be honored. Clarity in the handling of biological samples is particularly important in the situation where
pre-pubertal patients have cryopreserved gonadal tissue. Specifically, parents or their legal guardians must
give directions, in writing in advance, for future tissue disposition, and they must be urged to specify
what should be done with banked tissue if their child
dies.
As a young child cannot give his consent for the testicular biopsy cryopreservation, parental consent must
be granted. Testicular biopsy is an invasive procedure
and purely experimental [50]. In practice, a testicular biopsy should be performed under the same general anesthesia that is used to insert a central line for
chemotherapy. Although harvesting gonadal tissue
may be of high risk if taken in isolation, within the
context of the childs illness it may pose minimal additional risk [49]. Therefore, it should be combined with
a procedure that requires anesthesia. Lastly, it is important to remember that testicular tissue cryopreservation is still at a highly experimental stage and careful
206
counseling and consent is essential. To be valid, consent must be informed, voluntarily obtained and given
by a competent person. The consent process must not
raise unrealistic expectations [49, 51].
Conclusions
Little is known about the biology and regulation of
human germline cells, particularly regarding maintenance and regulation of SSCs, which are the foundation of spermatogenesis throughout adult life.
Spermatogonial stem cells reside on the basement
membrane of the testis seminiferous tubule, and their
decision regarding self-renewal versus differentiation
determines the efficiency of spermatozoa production. In mice, approximately 0.03% of testes cells are
stem cells, and they resemble other early differentiating spermatogonia, a morphological characteristic
observed phylogentically across mammalian species
[14, 52]. Spermatogonial stem cells arise directly from
gonocytes within days in mice and over a few months
in humans. The limited knowledge about these critical cells arises from their rarity and relative absence of
distinguishing morphological characteristics. To date,
cryopreservation is feasible in that all species in which
it has been tried; cryopreservation of testicular tissue
has been successful with significant numbers of SSCs
being recovered. Unfortunately, cryopreservation procedures results in some loss of SSCs and the SSCs
from human testicular biopsies are insufficient to reestablish fertility after transplantation. An important
goal of future basic and translational research is to generate methods to expand the SSCs in number. In addition to the basic science issues regarding the accession
and maturation of SSCs for transplantation, a plethora
of practical and ethical issues also need to be addressed
for appropriate application of human SSC usage for
fertility preservation in boys following gonadotoxic
pediatric cancer treatments.
Acknowledgements
Financial support was provided by an Ethel Foerderer
Award (JPG), National Institute of Child Health
and Human Development (NICHD) grant HD061217
(JPG), NICHD grant HD044445 (RLB), NICHD grant
HD 052728 (RLB) and the Robert J. Kleberg, Jr. and
Helen C. Kleberg Foundation (RLB).
References
1. Hewitt M, Weiner SL and Simone JV (eds.). Childhood
Cancer Survivorship: Improving Care and Quality of
Life. Washington DC: National Academies Press,
2003.
2. Lee SJ, Schover LR, Partridge AH et al. American
Society of Clinical Oncology recommendations on
fertility preservation in cancer patients. J Clin Oncol
2006; 24(18): 291731.
207
208
Section 4
Chapter
16
Introduction
Due to remarkable advances in the treatment of childhood cancer, we have seen great improvements in life
expectancy with up to 80% of children surviving their
disease, resulting in a growing population of adult
long-term survivors of childhood malignancies [1].
Although oncological treatments are highly effective, a major concern is their adverse impact on fertility [2, 3].
Since rapidly dividing cells are the target of chemoand radiotherapy, these treatments act not only on cancer cells, but also on germ cells. Differentiating spermatogonia proliferate the most actively and are thus
extremely susceptible to cytotoxic agents, although the
less active stem cell pool may also be depleted [4].
Consequently, although the pre-pubertal testis
does not complete spermatogenesis, there is evidence
that cytotoxic treatment given to pre-pubertal boys
affects fertility [5]. In addition, the presence of a steady
turnover of early germ cells that undergo spontaneous
degeneration before the haploid stage is reached [6, 7]
may possibly explain why the pre-pubertal state does
not offer any protection against gonadotoxic treatments.
Recovery of sperm production after a cytotoxic
insult depends on the survival and ability of mitotically quiescent stem spermatogonia (type A dark) to
transform into actively dividing stem and differentiating spermatogonia (type A pale) [8].
The somatic compartment of the testis may be
more resistant to chemotherapeutic treatment, since
these cells have a low or absent mitotic rate. Neverthe-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
209
Malignant
r
r
r
r
r
r
Leukemia
Hodgkins disease
Non-Hodgkins
lymphoma
Myelodysplastic
syndromes
Solid tumors
Soft tissue sarcoma
Non-malignant
1
therefore remains the only option for fertility preservation in pre-pubertal males.
Cell suspensions
Fertility preservation options before
gonadotoxic therapies
In order to reduce the deleterious effects of gonadotoxic therapies, different strategies have been tested.
Improving therapeutic regimens using less
gonadotoxic protocols [14] could enable spontaneous recovery of spermatogenesis, but their use is
not always possible without compromising patient
survival.
Limiting radiation exposure by shielding or removing the testes from the radiation field should be implemented whenever possible [3].
Minimizing testicular damage from cancer treatment or protecting SSCs in vivo is another approach.
Gonadal protection through hormonal manipulation
was reviewed in Chapter 13.
Anti-apoptotic agents such as sphingosine-1phosphate [15] and AS101 [16] and various other
cytoprotective substances [17, 18] have also been used
with partial success in rodents.
Currently available drugs to prevent testicular
damage from cytotoxic therapy have not proved helpful in humans so far. Testicular tissue cryopreservation
210
Tissue pieces
Cryopreservation of testicular tissue pieces may be
considered as an alternative method capable of maintaining cell-to-cell contacts between Sertoli and germinal stem cells, and therefore preserving the stem cell
niche necessary for their survival and subsequent maturation [26]. Other advantages of this method may be
preservation of the Sertoli cells, since there is evidence
of their reversion to a dedifferentiated state as a consequence of chemotherapy [9], and Leydig cells, whose
preservation may be useful to alleviate the hormonal
imbalance caused by cytotoxic therapy [2].
Whole testis
Due to the small number of SSCs contained in a testicular biopsy and the small size of a childs testis, it is
possible that cryopreservation of a whole testis may be
more appropriate, with a view to later organ autografting. Cryopreservation methods for whole testes need
to be developed, however, as has been done for whole
ovaries [32].
211
(Non)
controlled
Outcome
(germ cells)
Outcome
(endocrine
compartment)
Reference
Cryoprotectant
Kvist et al.
[20]
EG 1.5 M Sucrose
0.1 M
Slow controlled
Start: 1 C,
2 C/min to
9 C, hold 5 min
+ seeding,
0.3 C/min to
40 C,
10 C/min to
140 C, LN2
Culture 2
weeks
Well-preserved
interstitial cells
Testosterone and
inhibin levels
similar to fresh
tissue
Keros et al.
[21]
DMSO 0.7 M
Slow controlled
Program 1:
Start: 4 C, hold 30
min, 1 C/min to
0 C, hold 5 min,
0.5 C/min to
8 C, seeding,
hold 10 min,
0.5 C/min to
40 C, hold 10
min, 7 C/min to
70 C, LN2
Culture 24 h
70 7% ISTs in
frozen-cultured
tissue (versus
71 7% in fresh
tissue and 77 4%
in fresh-cultured
tissue) 94 1%
intact SG in
frozen-cultured
tissue (versus
93 2% in fresh
tissue and 83 1%
in fresh-cultured
tissue)
Undamaged
stromal structure:
80 29% of
frozen-cultured
samples (versus
99.49 0.88% of
fresh samples and
97 2% of fresh
cultured samples)
Rapid
controlled
Program 2:
Start: 4 C, hold 30
min, 1 C/min to
8 C, seeding,
hold 10 min,
10 C/min to
80 C, LN2
20 14% ISTs in
frozen-cultured
tissue 50 43%
intact SG in
frozen-cultured
tissue
Undamaged
stromal structure:
29 28% of
frozen-cultured
samples
Slow controlled
Start: 0 C, hold 9
min, 0.5 C/min
to 8 C, hold 5
min + seeding,
hold 15 min,
0.5 C/min to
40 C, hold 10
min, 7 C/min to
80 C, LN2
Immediate
post-thaw
evaluation
Not assessed
Xenografting
3 weeks
Xenografting
6 months
55 42% ISTs in
frozen-grafted
tissue 3.7 5.5% SG
recovery 21%
proliferating SG
Differentiation up to
pachytene stage of
prophase
Wyns et al.
[22]
Wyns et al.
[31]
DMSO 0.7 M
Sucrose 0.1 M
DMSO 0.7 M
Sucrose 0.1 M
Slow controlled
Freezing rate
Type of
evaluation
Start: 0 C, hold 9
min, 0.5 C/min
to 8 C, hold 5
min + seeding,
hold 15 min,
0.5 C/min to
40 C, hold 10
min, 7 C/min to
80 C, LN2
Signs of
steroidogenic
activity by 3-HSD
IHC and TEM
DMSO, dimethyl sulfoxide; EG, ethylene glycol; HSD, hydroxysteroid dehydrogenase; IHC, immunohistochemistry; LN2 , liquid nitrogen; SG,
spermatogonia; (I)ST, (intact) seminiferous tubule; TEM, transmission electron microscopy.
212
213
differentiate through transplantation of cell suspensions. Experiments on human germ cell transplantation were not able to achieve this goal since, after
6 months xenotransplantation to immunodeficient
mice, only proliferative activity was observed [49].
Hence, studies in animals will help us elucidate some
important considerations for clinical application.
The potential of frozen murine testicular cells
to resume spermatogenesis after transplantation was
demonstrated for the first time by Avarbock et al. in
1996 [58]. Live birth of offspring achieved after transplantation of frozen testicular cell suspensions provided final proof of successful cryopreservation [59].
Although it appears that the functional capacity of
mouse SSCs may be compromised by cryopreservation
[60], this was not observed by Kanatsu-Shinohara et al.
[59]. Moreover, rhesus SSCs retained normal colonization capacity after freezing and transplantation to mice
[48], suggesting that possible functional impairment
due to cryopreservation involves germ cell differentiation rather than their ability to recolonize stem cell
niches.
214
215
216
(a)
(c)
Control
3 HSD
N
Nm
50 m
TEM
Fresh
L
M
Cm
Fresh
2.8 m
50 m
SER
F/T/G
L
Nm
Cm
SER
F/T/G
2.8 m
L
50 m
217
Donor
species
Recipient
species
Graft
localization
Tubule
reconstitution
Sperm
differentiation
Rat (after
culture)
Nude
mouse
Back skin
Yes
Few putative
spermatogonia
No further
differentiation
IHC identification of
Leydig cells
Production of
bioactive
testosteronea
Mouse,
rat
Nude/SCID
mouse
Back skin
Yes
Round
spermatids
Offspring from
mouse-testis-cell
transplants
Not assessed
Yes
Not assessed
Honaramooz
et al. [84]
Pig
Nude/SCID
mouse
Back skin
Yes
Complete
spermatogenesis
IHC identification of
Leydig cells
Production of
bioactive
testosteronea
Arregui et al.
[70]
Sheep
Nude
mouse
Back skin
Yes
Complete
spermatogenesis
Production of
bioactive
testosteronea
Zhang et al.
[87]
Bovine
(after
culture
37 days)
Nude
mouse
Back skin
Testis
Yes
Yes
No germ cells
No germ cells
Not assessed
Watanabe et al.
[88]
Neonatal
pig
Nude/SCID/NOGBack skin
mouse
Yes
Complete
spermatogenesis
Not assessed
Reference
Pig
Steroidogenesis
Safety issues
Cancer cell contamination
The most important, life-threatening concern of
spermatogonial transplantation is the risk of reintroducing malignant cells. Indeed, the majority of
pediatric malignancies metastasize through the blood,
thus carrying a high risk of malignant contamination
of the testes. The risk is greater with hematological cancers, as the testes can act as sanctuary sites for leukemic
cells. Indeed, it has already been shown that as few as
20 leukemic cells injected into a testis can induce disease relapse [91].
Germ cell isolation and cell-sorting techniques
enabling complete purification of SSCs therefore need
218
to be validated before safe transplantation can be contemplated. While cell-sorting methods have shown
promising results in animal studies, the same cannot
be said of humans [92].
One of the reasons for suboptimal cell sorting
may be that the surface antigens are shared by other
stem cells, namely hematopoietic stem cells involved
in hematological cancers. Immunophenotyping malignant cells from each patient, followed by inclusion of
patient-specific cancer antigens for cell sorting, should
therefore improve the success of the technique. For
this purpose, we strongly advise storing patient blood
and/or tumor samples before therapy.
As no specific marker exclusive to SSCs has yet
been identified, allowing positive selection of these
cells through cell sorting techniques, further research
on surface markers should focus on the complete elimination of cancer cells from cell suspensions before
sorted preparations can be safely transplanted.
Cancer cell contamination is also a major
concern in tissue autografting. Since it has been
reported that leukemic cells can survive cryopreservation/xenotransplantation and increase the incidence
of generalized leukemia in the nude mouse host
[93], testicular tissue autografting after cure can
only be considered for patients in whom there is no
risk of testicular metastases or who have undergone
gonadotoxic therapies for non-malignant disease.
Infectious transmission
Due to the risk of infectious transmission from animals to humans, testicular xenografting should not be
considered for reproductive purposes at present. This
approach is nevertheless useful for the evaluation of
the functional capacity of germ cells and should therefore form part of the assessment of germ-cell cryopreservation protocols, for the understanding of testicular physiology and pathophysiology and for testing
malignant contamination of tissue before autografting.
Ethical concerns
Learning that a child has cancer is devastating for all
concerned and treatment needs to begin quickly, leaving very little time for the impact of possible future
sterility to sink in. However, the inability to father
ones own genetic children might have a huge impact
on the psychological well-being of patients in adulthood [10], so it is crucial to inform them of the potential consequences of their therapy on future fertility. Ethical concerns have been expressed about ITT
cryopreservation, highlighting the importance of the
risk/benefit balance [95]. Because of the small size of
testes from pre-pubertal children, immature gonadal
tissue sampling may be considered too invasive a procedure, which must therefore be done for good reason. However, in the two available studies on testicular tissue harvesting in young cancer patients [21, 22],
no major surgical complications occurred during testicular biopsy. Mean biopsy volume was about 5% of
testicular volume which, according to morphological
studies [6], should provide enough germ cells for fertility preservation. Furthermore, in a follow-up study of
cryptorchid boys who had undergone testicular biopsy
during orchidopexy, no adverse long-term effects were
reported [96]. Regarding general anesthesia, since this
biopsy is generally performed under the same anesthesia as that used for placement of the central line for
chemotherapy, there is no additional risk involved.
When considering the benefits of tissue harvesting,
the safety and effectiveness of fertility preservation and
restoration procedures are essential issues. Children
and their parents should be informed of the experimental nature of this approach and the fact that there
is no guarantee of fertility restoration. Parental consent
and the childs ascent, meaning he was given the opportunity to discuss the procedure, should be sought. As
obtaining fully informed consent from children is difficult, substituted consent from parents should for now
be limited to the safekeeping of tissue [95, 97].
Conclusion
Providing young people undergoing gonadotoxic
treatment with adequate fertility preservation strategies is a challenging area of reproductive medicine, but
every patient should be given the chance to consider
fertility-sparing options because the detrimental effect
of such therapy on gonadal function remains unpredictable. Hormonal or cytoprotective drug manipulation aimed at enhancing spontaneous recovery of
spermatogenesis remains a possibility for the future.
Preservation of SSCs offers the prospect of several realistic applications, although none is feasible in humans
at this point in time. Future advances in fertility preservation technology rely on improved understanding of
the cryobiology of gonadal tissue and cells.
Before considering fertility restoration options,
patient selection is essential, since risks vary according to disease. No single (or simple) algorithm can
so far summarize all the possible strategies for fertility preservation and restoration in case of gonadotoxic therapy in pre-pubertal boys, but the most appropriate course of action may be selected according to
the scheme shown in Figure 16.4. Over the next few
years, research should focus on how to extend successful experiments in animals to young boys and on the
identification of the ideal microenvironment for SSC
development. Resolving numerous important technical issues discussed in this chapter should lead to safe
and efficient methodologies for fertility restoration
219
Spermatids/
sperm
High risk of
cancer cell
contamination
ITT
cryobanking
ICSI
IVM
Diploid cells
Low risk of
cancer cell
contamination
Spontaneous
conception
IUI
IVF
ICSI
ICSI
Figure 16.4 Fertility restoration strategy after gonadotoxic therapy in pre-pubertal boys. ICSI, intracytoplasmic sperm injection; ITT,
immature testicular tissue; IVF, in vitro fertilization; IUI, intrauterine insemination; IVM, in vitro maturation; MRD, minimal residual disease;
RT-PCR, real-time polymerase chain reaction.
after storage of preserved gametes, and the development of ethically accepted pilot protocols, which will
then need to be submitted for further ethical approval
before definitive and universal clinical implementation. Until then, samples should at least be banked after
providing careful counseling and obtaining informed
consent, making sure the patient understands there is
no guarantee of success. Preservation of testicular tissue from todays pre-pubertal patients will allow them
to consider various fertility restoration options that
will emerge in the next 2030 years, giving them hope
of fathering children with their own genetic heritage.
References
1. Magnani C, Pastore G, Coebergh JW et al. Trends in
survival after childhood cancer in Europe, 19781997:
report from the Automated Childhood Cancer
Information System project (AGCIS). Eur J Cancer
2006; 42: 19812005.
2. Howell SJ and Shalet SM. Testicular function
following chemotherapy. Hum Reprod Update 2001; 7,
3639.
3. Wallace WH, Anderson RA and Irvine DS. Fertility
preservation for young patients with cancer: who is at
risk and what can be offered? Lancet Oncol 2005; 6:
20918.
4. Bucci LR and Meistrich ML. Effects of busulfan on
murine spermatogenesis: cytotoxicity, sterility, sperm
abnormalities, and dominant lethal mutations. Mutat
Res 1987; 176: 25968.
5. Kenney L, Laufer MR, Grant FD, Frier H and Diller
L. High risk of infertility and long term gonadal
damage in males treated with high dose
220
221
222
223
224
Section 4
Chapter
17
Introduction
Recent advances in the treatment of cancer have lead
to greater longevity and an increased recognition
that quality of life including paternity is significant
issues for cancer survivors. We will focus primarily
on patients with testicular cancer and lymphoma that
generally affects younger patients in the reproductive
window with an excellent overall survival. However,
one must realize in our modern society the age of desiring paternity has increased due to postponement of
marriage as well as for other social reasons. Therefore, this chapter will focus not only on those who
completed chemotherapy as children but adult cancer
patients as well.
Chemotherapeutic effects
The effect of chemotherapeutics on spermatogenesis
varies by both the drug administered as well as the
cumulative dosage and will be discussed in other
chapters. However, it is obvious that due to their high
proliferative index, chemotherapeutics will be toxic to
germ cells while Leydig cells seem more resistant to
the effect of systemic chemotherapy. Therefore, serum
levels of testosterone may be within normal limits
and yet impairment in sperm production may lead to
infertility.
There is no diagnostic test to tell whether spermatogenesis will return after chemotherapy. Men with
return of sperm to the ejaculate may even conceive naturally and all reproductive options are open to them.
However, a focus on men who are rendered azoospermic after therapy remains a clinically important subgroup of patients who are candidates for treatment. Up
to 13.8% of men are azoospermic prior to chemother-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
225
226
from the contralateral testicle, which was also evaluated for concomitant carcinoma in situ (CIS). The
lymphoma patients underwent bilateral TESE. The
sperm retrieval rate was 43% (6/14) in testicular cancer and 47% (8/17) in patients with Hodgkins or nonHodgkins lymphoma [22].
There also is the possibility of vasal or epididymal sperm extraction after orchiectomy from the
orchiectomy specimen. Baniel and Sella reported three
azoospermic patients with testicular cancer who at the
time of radical orchiectomy had vasal and epididymal sperm preserved [23]. After the specimen had
been resected, sterile extraction of sperm with cryopreservation of sperm from the vas and epididymis
was done. They reported two pregnancies from three
couples [23].
Preserving tissue or sperm from the tumor containing testis can be done on a separate sterile field
(a backtable) after orchiectomy or, in cases of partial orchiectomy, the surrounding normal testicular
parenchyma can be examined and sent for cryopreservation. A number of groups have reported on these
techniques in a limited number of patients with good
success at sperm retrieval [2426].
227
affect antegrade ejaculation. Partial ejaculatory function may be preserved depending on the degree of
nerve sparing. The preservation of ejaculatory function obviously has a tremendous impact on fertility
rates. In Norwegian testicular cancer survivors who
had chemotherapy, those with intact antegrade ejaculation have an 83% paternity rate, while rates for paternity were only 10% in the anejaculatory group [27].
An option for anejaculatory patients is to undergo
electroejaculation. This is a procedure generally performed under general anesthesia in the sensate patient.
We do not routinely catheterize the patient prior to
electroejaculation. The patient is placed in the lateral
decubitus position. Anoscopy is performed to confirm that the rectum is empty and no rectal mucosal
abnormalities are present. The rectal probe is inserted
completely into the rectum with the electrodes oriented anteriorly, over the prostate and seminal vesicles.
Stimulation is carried out with a standard electrical
stimulation system. The pattern of electrical stimulation has been empirically evaluated but appears to
work best with a gradually increasing voltage peaked
sine wave stimulation that is abruptly ceased, with at
least 57 s delays between stimulations. The procedure
is also monitored by observation of penile tumescence
and rectal temperature. Typically, penile tumescence
is noted first, followed by seminal emission. When
seminal emission ceases, rectal temperature of 38 C is
observed, or a maximum of 30 volts is attained, then
electrostimulation is stopped. Anoscopy is performed
again to insure that there is no rectal mucosal injury,
which is a potential complication of this procedure.
The patient is turned supine and urethral catheterization is carried out. An initial retrograde specimen
is diluted in human tubal fluid (HTF) buffered with
HEPES and plasmanate, pH 7.4, and sent for immediate processing, as is the antegrade ejaculate. The bladder is then irrigated with HTF, and this second retrograde specimen is sent for immediate processing as
well.
Ohl et al. performed electroejaculation in 24
testicular cancer patients (23 of which had undergone retroperitoneal lymphadenectomy) and observed
seminal emission in all 24 patients [28]. Greater than
10 million motile and progressive sperm were obtained
in 88% (21/24) of patients. In total, 17 couples underwent IUI and the overall cycle fecundity rate was 9%.
Seven clinical pregnancies were detected and there
were five live births. Electroejaculation has also been
successfully combined with IVF (and obtained a 53%
228
Eversion of
testicular
parenchyma
for microdissection
Centrifugal vessels
and that of others suggest that mTESE yields the highest sperm retrieval rate in this population.
Evaluation prior to mTESE includes a through history, sexual history, chemotherapy history physical
exam and hormonal profile. On physical exam, attention is paid to the fullness of the epididymis as well
as testicular volumes. The technique of mTESE is as
follows. This technique involves placing a wide incision in the tunica albuginea in an avascular region
and eversion of the testicular parenchyma for microdissection (Figure 17.1). With high power magnification, subtunical vessels as well as intratesticular
vessels can be identified and preserved. Microscopic
dissection and direct examination of seminiferous
tubules allow identification of the rare regions that
contain sperm in men with non-obstructive azoospermia (NOA). The tubules with spermatozoa are wider
and more opaque than the fibrotic Sertoli cell-only
tubules (Figure 17.2). Overall, mTESE has been shown
to result in a higher number of sperm harvested,
increased chance of retrieving sperm and decreased
testicular tissue removed [32, 33]. The only predictor of successful treatment is the most advanced stage
seen on biopsy and not the predominant stage [34].
Testicular volume, serum FSH levels and the etiology of NOA appear to have little or no effect on the
chance of sperm retrieval [14, 34, 35]. Postoperative
ultrasound has demonstrated fewer acute and chronic
changes after micro-dissection as compared to conventional TESE [36]. Of course, an increased number of
biopsies is always counterbalanced by a greater risk
of damage to the vascularity of the testis, and so the
surgeon must be constantly aware of this. For selection of the initial side, we prefer to start on the side
with larger testicular volume or the side with the more
advanced spermatogenic pattern seen on histology if a
prior biopsy was done (with the most advanced being
normal spermatogenesis followed by late maturation
arrest, early maturation arrest and Sertoli-cell only pattern, in that order).
At Weill Cornell Medical College we have
performed 81 mTESEs in 70 post-chemotherapy
patients. These patients presented with a variety
of malignancies, with the most common being
Hodgkins lymphoma, leukemia, testicular cancer and
229
Table 17.1 Patients with underlying medical conditions treated with chemotherapy
Medical condition
Percentage
Hodgkins lymphoma
29
35.8
Testicular cancer
13
16.0
13
16.0
Non-Hodgkins lymphoma
12
14.8
Sarcoma
8.6
Neuroblastoma
3.7
Medulloblastoma
1.2
Wilms tumor
1.2
1.2
Nephrotic syndrome
1.2
230
Donor sperm
Third-party reproduction
Additional counseling is recommended for those
patients who choose the assistance of third-party
reproduction. There is universal agreement that the
psychosocial, emotional and ethical complexities of
donor conception require thorough exploration both
for those donating and those receiving gametes [39
41]. In most clinics, a mental health professional
(MHP) meets with prospective donors and recipients to explain the known psychological, social and
legal implications of third party reproduction. It
has been argued that the assistance of a MHP is
essential to promote complete examination of the
many dilemmas faced by those who receive gametes
[4244].
The MHP may need to help a patient address previously unresolved grief regarding the cancer diagnosis and treatment. When donor back-up treatment is
being considered, it should be carefully explored prior
to treatment and should not be a decision by default
at the last minute (e.g. after poor TESE results and
oocyte retrieval during an IVF cycle). Often times, a
patients hopes that viable sperm will be found may
interfere with their ability to fully consider all aspects
of a donor sperm back-up plan. While a patient may
have grieved at the time of the cancer diagnosis, a
diagnosis of infertility may reopen the grieving process by adding another dimension to the illness and
may interfere with the desire to move forward with a
donor.
231
232
to make all decisions related to disclosure, the pregnancy and the upbringing of the potential child.
The infertility counselor must help all of the parties
involved explore their motivations, concerns, expectations, wants, hopes and fears regarding the process.
233
234
so that the child does not feel different and the husband
does not feel embarrassed, and to allow the parents to
feel that they are an ordinary family.
Religion and culture often influence attitudes about
the use of donor sperm. Donor insemination remains a
morally questionable treatment option in many countries. Using donor gametes is forbidden in some religions. In Islam, only a married couples eggs and sperm
may be used in procedures to treat infertility because
religious law dictates the preservation of the genetic
line. The Vatican does not endorse any assisted reproductive techniques, even insemination with the husbands own sperm [72]. Some Catholic bioethicists
have supported assisted reproductive techniques if no
other alternatives exist to allow a couple to have children but still do not find use of donor sperm acceptable
[73].
More often, couples are being encouraged to provide genetic disclosure to offspring. This emerging
trend of more openness may reflect legislative and cultural changes as well as the impact of counseling and
the advent of guidance materials parents can use for
talking to their children [51]. The main reasons that
couples choose to disclose include: the children had a
right to this information; the parents wanted to avoid
the burden of secrecy and the risk of disclosure by
somebody else or accidental discovery; and the parents believed that technical advances in genetics could
result in a genetic mismatch discovered by the offspring, again resulting in inadvertent disclosure [74,
75].
If couples choose to disclose the information to
their children, they should also understand the distinctions between openness and privacy. Couples still
have the right to discern who will know, how they will
find out and when they will share the information.
Given the stigmatizing nature of using donor sperm
and male-factor infertility, the couple needs to be in
agreement about with whom they will share information regarding infertility treatment, diagnosis and
their use of donor sperm. If they do not intend to talk
to their future child about his or her donor sperm origins, they must attempt to ensure that their confidants
will not disclose this to the child accidently nor discuss
the issue of donor sperm with others.
Information sharing will not only be an individual decision based on personal preference but will
also depend on cultural and religious factors, such
as the degree of acceptance of donor sperm as well
as legal practices regarding access to information and
donor identity. Research on how parents of donor offspring make decisions about disclosure reveals that
even when couples are initially opposed to disclosing
to their offspring, most ultimately come to a united
disclosure decision either intuitively, or after discussions influenced by the couples local sociopolitical
environment, professional opinion, counseling, religious and cultural background, family relationships
and personal, psychological and ethical beliefs [76].
Moreover, when gamete donation is used because of a
parents history of cancer, telling the child may provide
reassurance about his or her own lifetime cancer risk.
Age of disclosure appears to be important in determining donor offsprings feelings about their donor
conception. It seems that it is less detrimental for children to be told about their donor conception at an
early age [77]. Those told later in life report more negative feelings regarding their donor conception than
those told earlier. Offspring from heterosexual couple families are more likely to feel angry at being lied
to by their mothers than by their fathers. The most
common feeling toward fathers was sympathetic.
While the research in this area is somewhat limited, the
largest prospective follow-up of donor insemination
children to date documents normal social and psychological adjustment of children and families as the
cohort reached 12 years, despite the decision of almost
all parents to maintain secrecy with their offspring
[78]. Golombok et al. compared natural, adopted in
infancy, donor sperm and IVF children up to the age
of 12 years and found that parents who conceived
without difficulty had higher levels of parenting stress
than the other groups [79]. No significant differences
were found between types of families in the childrens
behavior, adjustment at home or school or feelings
toward their mothers or fathers. In assessing the quality of the parentchild relationship, Golombok also
found that IVF, DI and adoptive mothers had higher
levels of warmth and emotional involvement with their
children [79]. In vitro fertilization or DI fathers were
rated as displaying more warmth toward their children
than natural conception or adoptive fathers.
Overall, cancer survivors tend to view parenthood
in a positive manner and feel that their experience with
cancer would make them better parents [4]. Although
cancer survivors can become parents through options
such as adoption and third party reproduction [80],
most prefer to have genetic offspring [5, 81], even
if they have concerns about abnormalities that could
result if the parent conceived before cancer treatment
Acknowledgements
Doctor Hsiao is supported by a grant from the Frederick J. and Theresa Dow Wallace Fund of the New York
Community Trust.
References
1. Lass A, Akagbosu F, Abusheikha N et al. A
programme of semen cryopreservation for patients
with malignant disease in a tertiary infertility centre:
lessons from 8 years experience. Hum Reprod 1998;
13: 325661.
2. Ragni G, Somigliana E, Restelli L et al. Sperm
banking and rate of assisted reproduction treatment:
insights from a 15-year cryopreservation program for
male cancer patients. Cancer 2003; 97: 16249.
3. Zebrack BJ, Casillas J, Nohr L, Adams H and Zeltzer
LK. Fertility issues for young adult survivors of
childhood cancer. Psychooncology 2004; 13: 68999.
4. Schover LR, Brey K, Lichtin A, Lipshultz LI and Jeha
S. Knowledge and experience regarding cancer,
infertility, and sperm banking in younger male
survivors. J Clin Oncol 2002; 20: 18809.
5. Schover LR, Rybicki LA, Martin BA and Bringelsen
KA. Having children after cancer. A pilot survey of
survivors attitudes and experiences. Cancer 1999; 86:
697709.
6. Schover LR, Brey K, Lichtin A, Lipshultz LI and Jeha
S. Oncologists attitudes and practices regarding
banking sperm before cancer treatment. J Clin Oncol
2002; 20: 18907.
7. Glaser AW, Phelan L, Crawshaw M, Jagdev S and
Hale J. Fertility preservation in adolescent males with
cancer in the United Kingdom: a survey of practice.
Arch Dis Child 2004; 89: 7367.
8. Heath JA and Stern CJ. Fertility preservation in
children newly diagnosed with cancer: existing
standards of practice in Australia and New Zealand.
Med J Aust 2006; 185: 53841.
235
236
237
238
Section 5
Chapter
18
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
239
Hypothalamus
Progesterone
GnRH
+
Pituitary
Estradiol
Inhibin
FSH
+
LH
+
Ovary
E2/P4
+
Uterus
240
Dose
Gonadotropin-releasing hormone analogues can be
administered in many formulations with different
durations of action. In human studies, the dose of
GnRHa has been evaluated by the ability to suppress
ovarian and testicular steroid hormone production.
Whether this dose is sufficient for ovarian protection depends on the potential mechanism of protective effect. Some propose that GnRHa may protect
the ovary by inducing a pre-pubertal state. Whether
dose and formulation of GnRHa influences the level
of pituitary suppression at the end of the labeled depot
period has been examined in children using GnRHa
to treat central precocious puberty. With each repeat
dose there is the possibility of an increase in FSH
release if the pituitary suppression from downregulation has decreased. The rise in FSH and LH after repeat
dosing was higher with the lower dose, longer depot
duration formulations (Depotleuprolide 3.75 mg
1 month, 11.25 mg 3 months) than the higher dose,
shorter depot duration formulation tested (Depot
leuprolide 7.5 mg 1 month). While a greater magnitude of pituitary responsiveness was seen with the
lower dose, longer duration formulations, gonadal
steroid production and pubertal stage did not change
[14]. Mohamed et al. also documented a statistically
significant increase in pituitary response to GnRH
administration after 2 months of downregulation with
goserelin 3.6 mg subcutaneous pellet in 21 patients
with endometriosis [15]. Changes in ovarian steroids
were not reported; therefore, it is not known whether
the pituitary responsiveness at the end of the depot
period is clinically significant. Pending additional
studies with biochemical and clinical outcomes, the
relevance of these fluctuations of FSH at the time of
repeat dosing is unknown.
The influence of obesity upon GnRHa effectiveness has been questioned. Obese men using GnRHa
for treatment of prostate cancer exhibit higher testosterone levels than men with normal body mass indexes
[16]. The GnRHa active peptide is hydrophilic and
is therefore most influenced by the constant volume
of the extracellular fluid compartment. The synthetic
polymer linked to the GnRHa, however, is lipophilic.
Agarwal therefore postulated that adiposity may influence the rate of enzymatic breakdown of the lipophilic
Side effects
The most common side effects of GnRH analogues are
related to the subsequent estrogen deprivation. Vasomotor symptoms, hot flushes, night sweats, vaginal
dryness and headaches can occur. With longer duration of administration and the subsequent estrogen
deprivation, bone loss can occur. Side effects can be
mitigated by hormone replacement. A combination
estrogenprogestin product such as a birth control pill
will protect bone and prevent vasomotor symptoms. If
estrogen exposure is contraindicated, a progestin-only
formulation can be of benefit.
241
242
of 5 women, the only woman without a resumption of menses received a more aggressive course of
chemotherapy over 34 weeks for stage IVA Hodgkins
disease, compared to 1624 weeks for stage IIA or B
Hodgkins disease in the other women [25].
The marrow-ablative regimen required for stem
cell transplant resulted in ovarian failure in all
30 women despite prior treatment with leuprolide
3.75 mg [23]. Most of the women received busulfan
4 mg/kg for 4 days and cyclophosphamide 60 mg/kg
for 2 days; other women received variants that
included anti-thymocyte globulin (ATG) or melphalan. Only one of the women had menstrual bleeding during the time of thrombocytopenia following
chemotherapy. Hormonal status was followed over the
following year with FSH consistently elevated (median
35 mIU/ml, range 3080 mIU/ml) and estradiol low
(median 15 pg/ml, range 1025 pg/ml).
Bone density declined during GnRHa therapy in
several studies. In one study of 18 women, the hip
and spine T scores declined by an estimated 0.4 points
during the 6 months of cyclophosphamide, high dose
prednisone and leuprolide. In the following 6 months,
the BMD increased, though not back to baseline, in
the women who had a resumption of menses. It did
not increase in the women with premature ovarian
failure [21]. Another study showed both decreases
and increases in bone density during therapy with
some women with normal density developing osteopenia, several improving from osteoporosis or osteopenia, and many remaining unchanged [27]. Another
reported a significant decline within just 3 months of
goserelin therapy, with median T scores falling from
1.50 to 2.40. Bone density measurements following
the resumption of menses were not reported in this
study [20].
Some studies included add-back estrogen therapy to diminish the side effects of GnRHa therapy. In
the only study that reported side effects, women did
not receive add-back therapy and 97% had hot flashes,
90% headaches, sweating or mood changes, and 59%
reported vaginal dryness [20]. It is not clear how effectively estrogen therapy might relieve these symptoms.
Some women, particularly those with breast cancer
and lupus, are not candidates for estrogen therapy.
18
45
30
5
25
40
92
80
89
89
83
72
96
21
24/25
15
97
Percentage with
ovarian function
27
No. with
ovarian function
Menses
Menses
Menses
Menses
Menses
Either menses or
FSH40
Menses
FSH 40
Definition of
ovarian function
Disease, chemotherapy
ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; chemo, chemotherapy; CML, chronic myeloid leukemia; CYC, cyclophosphamide; FSH, follicle stimulating hormone;
MM, multiple myeloma; OCP, oral contraceptive pill; XRT, radiation therapy.
29
Author (Location of
study, year)
GnRHa
Table 18.1 Clinical data from studies of gonadotropin-releasing hormone agonist (GnRHa) co-therapy without a control group
244
100%
80%
60%
40%
20%
0%
With GnRHa
No GnRHa
Table 18.2 Clinical data from controlled studies of gonadotropin-releasing hormone agonist (GnRHa) co-therapy
GnRHa
Author (Location of
study, year)
75
70
No GnRHa
Percentage
with ovarian
function
Percentage with
ovarian function
93
82
38
46
100
44
30
27
90
26
23
100
83
14
14
100
15
47
100
17
11
65
20
19
20
14
70
50
67
12
12
100
22
36
72
10
39
35
89
39
18
33
downregulation for IVF found no difference in threedimensional power Doppler vascularity color indices
after 2 weeks of daily subcutaneous buserelin [42].
Yu Ng et al.s report of 85 women undergoing luteal
pituitary downregulation for IVF after a median of
10 days of intranasal buserelin also found no difference in three-dimensional power Doppler flow indices
[43]. The goal of GnRHa administration in IVF is to
prevent ovulation without overly suppressing ovarian
response. In the context of these IVF studies using
lower daily GnRHa dosing, GnRHa have not been
reproducibly proven to decrease ovarian blood flow;
the potential effect of higher doses of GnRHa from
depot formulations on ovarian stromal blood flow is
as yet unknown.
95%
245
246
Hypothetical mechanisms of
ovarian protection
Blumenfeld hypothesized additional potential mechanisms for GnRHa to protect ovarian function: via
decrease in secretion of growth factors by suppression of FSH-dependent follicular turnover; potential
upregulation of sphingosine-1-phosphate, an antagonist of the proapoptotic second messenger ceramide;
or by protection of undifferentiated germ-line stem
cells should they be proven to exist in humans
[2]. Whether these putative mechanisms require further study will be predicated upon whether GnRHa
are proven to protect not only the resumption
of menses, but also the retention of true ovarian
function with both sustained cyclical steroidogenesis and live birth in those women desiring future
parenthood.
Given the many and varied influences on different
organ systems attributed to GnRH both centrally and
peripherally, robust evaluation of a role for GnRHa in
fertility preservation during chemotherapy awaits adequately powered randomized controlled trials powered for the true outcomes of interest: live birth, time
References
1. Oktay K, Sonmezer M, Oktem O et al. Absence of
conclusive evidence for the safety and efficacy of
gonadotropin-releasing hormone analogue treatment
in protecting against chemotherapy-induced gonadal
injury. Oncologist 2007; 12(9): 105566.
2. Blumenfeld Z. How to preserve fertility in young
women exposed to chemotherapy? The role of GnRH
agonist cotreatment in addition to cryopreservation of
embrya, oocytes, or ovaries. Oncologist 2007; 12(9):
104454.
3. Meistrich ML and Shetty G. Hormonal suppression
for fertility preservation in males and females.
Reproduction 2008; 136(6): 691701.
4. Ataya K, Rao LV, Lawrence E and Kimmel R.
Luteinizing hormone-releasing hormone agonist
inhibits cyclophosphamide-induced ovarian follicular
depletion in rhesus monkeys. Biol Reprod 1995; 52(2):
36572.
5. Ozcelik B, Turkyilmaz C, Ozgun MT et al. Prevention
of paclitaxel and cisplatin induced ovarian damage in
rats by a gonadotropin-releasing hormone agonist.
Fertil Steril 2010; 93(5): 160914.
6. Lee SJ, Schover LR, Partridge AH et al. American
Society of Clinical Oncology recommendations on
fertility preservation in cancer patients. J Clin Oncol
2006; 24(18): 291731.
7. Periti P, Mazzei T and Mini E. Clinical
pharmacokinetics of depot leuprorelin. Clin
Pharmacokinet 2002; 41(7): 485504.
8. Fulghesu AM, Angioni S, Belosi C et al.
Pituitary-ovarian response to the
gonadotrophin-releasing hormone-agonist test in
anovulatory patients with polycystic ovary syndrome:
predictive role of ovarian stroma. Clin Endocrinol
(Oxf) 2006; 65(3): 396401.
247
248
249
Section 5
Chapter
19
Ovarian transposition
Carrie A. Smith, Erin Rohde and Giuseppe Del Priore
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
250
2% of cancer patients would be potential ovarian transposition candidates. A few years later, Mariani et al.
used age 35 which increased the potential number
of affected patients to 10% [8]. Finally, a New York city
group reported in 2004 that more than 40% of their
patients would be interested in fertility preservation
[9]! This group used age 40 as the cut off, reflecting a
significant change in definition and perspective from
the earliest study. Obviously none of these studies had
an objective basis for their methods. Instead the arbitrary cut offs were based on contemporary and fluid
definitions of reproductive age. Because of the uncertain estimates of the size and definition of the affected
population, all patients should be counseled regarding fertility preservation, including the use of ovarian
transposition.
Ovarian function is compromised when damaged during surgery, exposed to radiation, and/or
chemotherapy. Studies have shown that the amount
of radiation to eliminate 50% of a young womans
primordial follicles is 24 Gy, with 20 Gy of radiation exposure resulting in complete and permanent
ovarian failure [1014]. Pelvic radiation to treat gynecological cancers can be as much as 70 Gy planned
prescribed dose to the ovarian fossa. Given the lack of
precision inherent in radiation planning versus delivery, the actual received dose that the ovaries absorb can
be significantly higher.
In addition, after decades of a relatively stable
delivery system using linear accelerators, radiation
delivery has developed many more options designed to
improve cancer care, increase cures and decrease toxicity [1012, 14]. However there is little to no information on the affect of these modalities on in situ or
transposed ovaries. The general impression and hope
is that there will be less toxicity; however, caution must
be used in counseling patients treated with these latest
modalities that transposition is or is not indicated until
data become available.
Chemotherapy has been found to have a highly
variable chance of acute ovarian failure [14, 15].
There is also a highly variable increased long-term
chance of premature ovarian failure [16]. As discussed
below, the variation in reported chemotherapy affects
is due largely to study design and outcomes. Ovarian transposition done to preserve ovarian function
may paradoxically increase the susceptibility to subsequent toxic exposure. This is only a theoretical concern
based on the reports of ovarian failure following other
pelvic surgeries, especially hysterectomies. Disruption
251
Figure 19.1 Radiation field. See plate section for color version.
252
Figure 19.2 Surgical anatomy. See plate section for color version.
trary definition of success is used. However, the consensus estimate is that a substantial minority of transposed ovaries will cease to function after transposition.
Techniques have been devised to minimize this complication.
For instance, a report indicated that nearly all ovarian function can be preserved, at least in the short
term, using a retro-peritoneal approach. Briefly in this
technique, the ovary and its distal blood supply are
brought retroperitoneal superior and lateral to the distal incised utero-ovarian ligament. A window is made
in the paracolic gutter peritoneum and the ovary gently pulled through this opening (Figure 19.2).
This technique theoretically improves outcomes by
preventing ovarian peritoneal inclusion cyst as the
ovary retains its normal relationship with the peritoneal surface [21, 28]. In addition, this technique
avoids the creation of a potential internal hernia by
a taunt gonadal pedicle. When the ovary is directly
attached to the abdominal wall, it should be done so
with as little tension as possible on the gonadal vascular bundle. Ideally it should lie flat against the side
wall without tension. There have been no direct comparisons of the two techniques.
How the ovary is attached to the lateral abdominal wall varies by operator preference [22, 24, 25].
Suture has an advantage of being able to precisely place
253
254
References
1. McCall ML, Keaty EC and Thompson JD.
Conservation of ovarian tissue in the treatment of
carcinoma of the cervix with radical surgery. Am J
Obstet Gynecol 1958; 75: 590600.
2. Thomas PR, Winstanly D, Peckham MJ et al.
Reproductive and endocrine function in patients with
Hodgkins disease: effects of oophoropexy and
irradiation. Br J Cancer 1976; 33(2): 22631.
3. Hamilton BE, Martin JA and Sutton PD. Births:
preliminary data for 2003. Natl Vital Stat Rep 2004; 53:
117.
4. Del Priore G, Smith JR, Boyle DC et al. Uterine
transplantation, abdominal trachelectomy, and other
reproductive options for cancer patients. Ann N Y
Acad Sci 2001; 943: 28795.
5. Shover LR, Rybicki LA, Martin BA et al. Having
children after cancer: a pilot study of survivors
attitudes and experiences. Cancer 1999; 89: 697709.
6. Chalian R, Licciardi F, Rebarber A and Del Priore G.
Successful infertility treatment in a cancer patient with
a significant personal and family history of cancer. J
Womens Health (Larchmt) 2004; 13(2): 2357.
7. Maddux HR, Varia MA, Spann CO, Fowler WC and
Rosenman JG. Invasive carcinoma of the uterine
cervix in women age 25 or less. Int J Radiat Oncol Biol
Phys 1990; 19(3): 7016.
24. Huang KG, Lee CL, Tsai CS, Han CM and Hwang LL.
A new approach for laparoscopic ovarian transposition
before pelvic irradiation. Gynecol Oncol 2007; 105(1):
2347.
255
256
Section 5
Chapter
20
Introduction
Patients with early cervical cancer (FIGO stage IB1
or less) are conventionally considered treated with a
surgical approach while those with more advanced
disease are treated by radiotherapy with concurrent
chemotherapy. Surgical treatment usually involves a
radical hysterectomy with pelvic lymphadenectomy
except for those with early microscopic disease (FIGO
stage IA1) where conisation or extrafascial hysterectomy lymph-node dissection would be sufficient.
With the good implementation of cervical screening
programs in most developed countries, more women
are presenting with early cervical invasive disease. For
these women, the long-term prognosis is good, with
survival of over 90%. With good long-term survival,
preservation of fertility potential and quality of life
become more important issues and have been the focus
of recent surgical advances in the treatment of cervical
cancer.
For FIGO stage IA1 without lymph-vascular space
involvement, cold-knife conisation with a complete
lesion resection is recommended as a fertilitypreserving treatment, since the rate of parametrium
and lymph-node involvement is negligible. For larger
tumors, or in case of lymph-vascular space involvement, due to risk of lymphatic extension, resection of
the pelvic lymph nodes and of the proximal part of
the parametrium is recommended. For these patients,
preserving the fertility was a challenging option until
recently.
The recent development in surgical oncological
technologies aims at improving their effectiveness
while decreasing their morbidity. Therefore, the endoscopic and in particular laparoscopic techniques were
introduced in surgical oncology. With these concepts
of optimization of the treatments and reduction of
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
257
Operative technique
In our team, the operation always begins with a laparoscopic pelvic lymph-node dissection, which includes
sentinel node identification. This technique of the
sentinel node identification discovers, in a significant proportion of cases (1520%), lymphatic drainage
channels with unusual locations of sentinel nodes.
Identification of the sentinel node allows the realization of frozen sections, their result being of paramount
importance for the next surgical step. Indeed, if the
frozen-sections evaluation is positive, the planned radical trachelectomy is cancelled and the patient receives
a full laparoscopic pelvic, common iliac and paraaortic lymph-node dissection followed by radiotherapy and chemotherapy. Detection of the sentinel node
is performed with a dual technique of isotopic colloid
particle (Nanocys ) and color (Patent Blue ) injection in the cervix (in the four quadrants) (Figure 20.1),
and then the sentinel nodes are identified through
laparoscopy (Figure 20.2). This identification is followed by a full pelvic lymph-node dissection performed from the obturator membrane until the iliac
bifurcation, with the removal of all external iliac lymph
nodes (below the vein and at the contact of the external iliac vein and artery). In the case of a tumor larger
than 2 cm in maximal diameter, the cellulo-adipose
tissue of the distal parametrium is dissected through
laparoscopy (corresponding to a lymph-node dissection of the parametrium).
The next step is that of the radical trachelectomy performed through the vagina [8]. The steps
of this operation reproduce those of the intervention
of SchautaStoeckel. Dargents operation includes the
following steps. The first step is the realization of a
vaginal cuff. It is rarely necessary to remove more than
R
258
12 cm of vaginal mucosa. Thus, a rim of vaginal tissue is delineated circumferentially using 68 Kocher
forceps (Figure 20.3). In order to reduce bleeding and
to help for the following dissections, 20 cc of xylocaine
plus epinephrine is injected at regular intervals in the
vaginal fold. A round incision of the vaginal wall is
made just above the Kocher forceps. Once the incision is fully made, the Kocher forceps are removed and
the anterior and posterior edges of the vaginal cuff are
grasped together with 46 Chroback clamps in order
to isolate the cervix and to prevent tumor spillage.
These clamps allow good traction onto the operative
specimen in order to help the following steps. Then
dissection of the vesico-vaginal septum is performed
Figure 20.3 Performance of the vaginal cuff. See plate section for
color version.
on the midline with scissors. The space is usually avascular and, when fully dissected, the bladder base is
mobilized with a vaginal retractor. The following step
is the opening of the para-vesical space. On the left
side, the opening is obtained while dissecting along
the vaginal wall at 3 oclock level. It should be noticed
that the upper part of this space has been dissected
through laparoscopy helping the vaginal approach. A
retractor is put into the para-vesical space, and palpation of the bladder pillar and the ureter is performed
between the surgeon finger and the retractor. The precise location of the ureter can easily be defined. And
so, the bladder pillars are excised after coagulation.
The goal of this dissection is to evidence the ureter
(Figure 20.4), and then to fully cut the bladder pillars
in order to free the bladder base and to move away
the lower part of the ureter from the operative field.
The same procedure is performed on the patients right
side. The following steps are then performed on the
posterior aspect of the operative specimen. It first consists of opening the Douglas pouch. This opening is
performed with scissors on the midline, as for a simple vaginal hysterectomy. Then a vaginal retractor is
put in place in order to evidence the utero-sacral ligaments. These ligaments are coagulated and divided
leading to opening of the para-rectal space and identification of the posterior aspect of the parametrium.
Then the next steps are modifications of the ones performed during the Schauta operation. Indeed, the division of the parametrium is carried out while preserving the uterine artery. With retractors in place in the
para-vesical and para-rectal spaces, the anterior and
posterior aspects of the parametrium and the ureter
259
260
Figure 20.9 Set up of the isthmic cerclage. See plate section for
color version.
Our experience
In our department, from December 1986 to December 2008, Dargents operation was proposed to 171
patients. For 11 of these patients, the final intervention was changed either because of a lymph-node invasion (6 cases) or because of isthmic involvement by
the tumor (5 cases). Thus, 160 radical trachelectomies
were carried out. The average age of the patients was
31.5 years. The proportion of nuliparous patients was
56%.
The initial stages of the tumors were: stage IA1
or (+ lymph vascular space invasion) IA2 in 38 cases
(24%); and stage IB1 in 122 patients (76%). The histological types were: squamous in 123 cases (77%), glandular in 35 cases (22%) and other types (of which 1
was neuroendocrine) in 2 cases (1%). The maximum
tumoral size was 2 cm in 130 cases (81%) and 2 cm
in 30 cases (19%). The presence of lymph vascular
space invasion (LVSI) was found in 51 cases (32%).
The mean operating time was 192 min (including
the associated laparoscopic pelvic lymph-node dissection). The only intraoperative complications observed
(2 cases, 1%) were a ureteral laceration and a bladder breach, which were sutured without further damage. The average hospital stay was 4.6 days. During
the postoperative course a notable complication was
observed in 14 patients (19%). Nine (6%) patients presented a postoperative bleeding requiring a surgical
revision. Two pelvic lymphocysts required a surgical
marsupialization. Two abscesses (pelvic and parietal)
were drained. A case of uretero-vaginal fistula required
a ureteral anastomosis without sequellas. It should be
noted that the rate of complication was significant,
especially in the first-operated cases because of the
experimental character of the surgical technique. Concerning the last 70 patients who went through surgery,
no operative complication was observed and the postoperative course was marked by 3 (2%) of the 14 complications previously described.
In nine patients, adjuvant radiotherapy was recommended either because of a lymph-nodal or parametrial tumor involvement at the final histological examination (seven cases) or because of a massive LVSI
(two cases). The average follow-up of the patients is 92
months. To this date, 8 (5%) relapses were observed,
leading to death in 6 cases. A neuroendocrine case of
cervical cancer presented 2.5 years after surgical treatment with distant metastases leading to the death of
the patient. Four nodal relapses were observed (located
in the latero-pelvic, common iliac and para-aortic
areas). Despite the treatment of these recurrences
(surgery and radiotherapy), three of these patients
died. A patient with a stage IB1 adenocarcinoma presented a central-pelvic relapse on the uterine isthmus. She was operated on and irradiated and is currently doing well. Two patients presented relapses in
the parametrium and unfortunately could not be saved
despite loco-regional treatments.
The prognostic factors of the relapses were evaluated. After exclusion of the case of neuroendocrine
cancer, which represents an inadequate indication of
radical trachelectomy, the only statistically significant
factor for relapse is tumoral size. Thus, in the event of
maximum tumoral size 2 cm, 6 relapses (20%) were
observed against 1 relapse (1%) when the size was
2 cm (P 0.05). The presence of LVSI is associated
with an increased risk of recurrence, but the difference
is not statistically significant: 5 relapses (10%) in the
event of the presence of LVSI, against 2 relapses (3%)
in the absence of LVSI (P = NS). The histological type
has no effect on the risk of recurrence.
The last update of the pregnancies obtained after
radical trachelectomy was carried out in 2005. At that
time, we had observed the birth of 49 live healthy children following radical trachelectomy performed in our
department. All the childbirths were carried out by
cesarian section. The evaluation of the fertility after
Dargents operation made it possible to find infertility in relation to the operation (primarily by isthmic
stenosis) in 16% of the cases. The rate of late miscarriage is relatively important in our practice since it is
15% of all the pregnancies. However, this rate of late
miscarriage was lowered by the introduction of various procedures: the use of isthmic cerclage, which
made it possible to halve the rate of miscarriage, and
then the closing of the uterine cervix according to the
Saling procedure, which made it possible to reduce
the rate of late miscarriage in our experience from 21
to 12%.
261
Discussion
Dargents operation or radical trachelectomy was
invented to enable preservation of fertility among
women with early cervical cancer. The current followup of the first cases over 20 years confirms the hopes of
Professor Dargent: women benefiting from this operation have an excellent opportunity to have a healthy living child after this treatment without increased oncological risk [8]. The benefits of Dargents operation are
linked to the laparoscopic approach, which reduces the
262
Conclusion
Dargents operation was developed during a period
of reduction in aggressive operative techniques in the
263
References
1. Dargent D, Brun JL, Roy M, Mathevet P and Remy I.
La trachelectomie elargie: une alternative a`
lhysterectomie radicale dans le traitement des cancers
infiltrants developpes sur la face externe du col uterin.
JOBGYN 1994; 2: 28592.
2. Plante M, Renaud MC, Hoskins IA et al. Vaginal
radical trachelectomy: a valuable fertility-preserving
option in the management of early-stage cervical
cancer. A series of 50 pregnancies and review of the
literature. Gynecol Oncol 2005; 98: 310.
264
17. Lee CL, Huang KG, Yen CF and Lai CH. Laparoscopic
radical trachelectomy for stage IB1 cervical cancer. J
Am Assoc Gyn Laparosc 2003; 10: 11115.
18. Cibula D, Ungar L, Palfalvi L, Bitio B and Kuzcl D.
Laparoscopic abdominal radical trachelectomy.
Gynecol Oncol 2005; 97: 7079.
265
Section 5
Chapter
21
Introduction
Conservative and functional surgery is increasingly
used in surgical oncology. Its aim is to preserve organ
functionality and to limit radical resections. The development of new surgical procedures in oncologic gynecological surgery is a perfect example of this evolution.
Although radical surgery remains the gold standard
for the treatment of ovarian and cervical cancer, a conservative approach can be considered in patients with
early stage disease, in order to preserve their ovarian
function and fertility. These procedures are proposed
to selected patients, depending on the histological subtypes and prognostic factors. Ovarian cancers are classified as epithelial (including borderline and malignant
tumors) and non-epithelial cancers.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
266
the presence of an MP pattern was an adverse prognostic factor [9]. However, Eichhorn et al. reported that
the evolution of patients with non-invasive implants
associated with an MP pattern was similar to that of
patients with non-invasive implants without an MP
pattern [10]. The real prognostic impact of this entity
continues to fuel debate.
logic analysis of the margins to rule out any microscopic invasion [17]. Nevertheless, in the recent series
reported by Papadimetriou, among 18 patients who
underwent a cystectomy with histologically free margins, 7 relapsed [18].
In order to reduce the rate of relapse on the remaining ovary, some authors propose initial complete staging surgery with routine biopsies of the spared ovary.
In our series, we performed this procedure in 14
patients with a macroscopically normal ovary. We
found no tumor implants. Like Bostwick and Tazelaar, we observed only one relapse on the macroscopically normal ovary, that was routinely biopsied [19,
20]. It is important to note that this is not a harmless procedure, as it may induce infertility because of
postoperative ovarian adhesions. Macroscopic inspection appears to be sufficient. Biopsies should be performed only in case of macroscopically suspicious
lesions.
The high recurrence rate implies that the optimal
treatment in patients with an intraoperative diagnosis
of BOT, is a unilateral adnexectomy, which reduces the
risk of relapse. A cystectomy should be performed only
in cases of bilateral tumors and/or in patients with only
one ovary (a previous history of adnexectomy). In case
of a recurrent BOT on the remaining ovary, further
conservative management (cystectomy) may be proposed to these patients, in order to preserve fertility.
In our series, eight patients with recurrent BOT were
reoperated on conservatively for their recurrent disease [16]. Six pregnancies were obtained in five of these
patients [16, 21]. In the series by Gotlieb et al., three
cases of conservative management of recurrent BOT
were reported [15]. Those 3 patients were alive after 6
months, 2 years and 7 years of follow-up [15].
267
Disease stage
Data in the literature concerning conservative management of BOT with peritoneal implants are rare
(Table 21.1) [22, 2932]. To date, only two large series
268
Table 21.1 Literature review of recurrence rates in advanced stage serous ovarian borderline tumor (in particular with invasive implants)
after conservative treatment
No. of invasive
implants
Death in patients
with invasive
implants
No.
Zanetta et al. [22]
25
10
10
21
21a
41
22
Total
97
40 (41.2%)
2 (2%)
11
No. of
deaths
Recurrence in
patients with
invasive implants
No. of
recurrences
269
Table 21.2 Literature review of cases of ovarian induction or in vitro fertilization (IVF) procedures in patients with a previous history of a
borderline ovarian tumor
Series
Nijman et al. [37]
No. of
patients
Ovarian
induction (no.)
No. of
IVFs
No. of
pregnancies
No. of recurrences
(after stimulation)
2 (1 spontaneous
after IVF)
1 (III C
micropapillary)
Rapidly progressive
peritoneal disease
11
30
27
13
1 (IIIC nodal)
270
No. of
stages II/III
Table 21.3 Literature review of results conservative management in epithelial ovarian cancer (5 series reported including 10 cases)
Stage IA
grade 1
Stage IA
grade 2
Stage IA
grade 3
Stage IC
grade 1
Stage IC
grade 2
Stage IC
grade 3
1 recurrence out
of 24 patients
3 recurrences
out of 8 patients
1 recurrence out
of 4 patients
No recurrence
out of 10
patients
1 recurrence out
of 6 patients
No recurrence
out of 3 patients
2 recurrences
out of 33
patients
2 recurrences
out of 6 patients
No recurrence
out of 3 patients
No recurrence
out of 5 patients
1 recurrence out
of 3 patients
No recurrence
out of 2 patients
1 recurrence out
of 13 patients
4 recurrences
out of 14
patients
1 recurrence out
of 3 patients
2 recurrences
out of 2 patients
No patient
1 recurrence out
of 1 patient
1 recurrence out
of 29 patients
No recurrence
out of 3 patients
4 recurrences
out of 4 patients
1 recurrence out
of 15 patients
1 recurrence out
of 2 patients
2 recurrences
out of 2 patients
Anchezar et al.
[57]
1 recurrence out
of 10 patients
No patient
1 recurrence out
of 1 patienta
No recurrence
out of 3 patients
No recurrence
out of 1 patient
No recurrence
out of 1 patient
Total
6 recurrences
out of 109
patients
9 recurrences
out of 31
patients
7 recurrences
out of 15
patients
3 recurrences
out of 35
patients
8 recurrences
out of 12
patients
3 recurrences
out of 9
patients
Italian series
Zanetta et al.
/Colombo et al.
[52, 53]
American series
Schilder et al. [54]
French series
Morice et al. [55]
Patient considered as having a stage IA grade 3 tumor after pathological review of the initial tumor at the time of the recurrence.
271
272
?
?
37
No. conservative
management
Menstration
maintained
No. pregnancies
No. conservative
management,
stages II/III/IV
11
24/26
50
14
16
19/20
19
1e
43/45
74
74
Low et al.
2000 [70]
46
72
Tewari et al.
2000 [69]
46
55 in 32
patients
128/130
138
169
Zanetta
et al. 2001
[71]
108
No. patients
26
129
Series
69
Brewer
Peccatori Mitchell
et al. 1995 et al.a 1999 et al.b
[67]
[66]
1999 [68]
11 (9 pregnancies)
11
11
26
32/40c
38 in 29
patients
39
55
Zanagnolo
et al. 2004
[73]
64
106
Tangir
et al. 2003
[72]
Table 21.4 Literature review of fertility results following conservative management in germ cell tumors (series published after 1995)
10d
15
29
8 (4
?
pregnancies)
6 in 5
patients
19/23c
23
23
Ayhana
Boran
b et al. 2005 et al. 2005
[74]
[75]
15
15
20
Kangb
et al. 2008
[76]
4 pregnancies
19 in
12 patients
39/40
41
52
De La Motte
Rougea
et al. 2008 [77]
274
few papers are devoted to the conservative management of such tumors (mostly case reports or short
series). In the series by Zhang et al., among the 376
women treated for SCST, 71 young patients underwent
uterine preservation for stage I disease [78]. The survival of patients treated conservatively and radically
was similar [78].
Two important characteristics observed in granulosa tumors exert an impact on conservative surgery:
bilaterality is uncommon (between 2 and 8% of cases
[79] and these tumors are frequently associated with
endometrial disorders (hyperplasia or cancers). Consequently, random biopsies of the contralateral ovary
are not required (if macroscopically normal) but uterine curettage should be systematically performed. The
overall prognosis of granulosa cell tumors is good
in early stage disease (stage IA) and conservative
management could be considered in young patients
with a similar stage. However, conservative management should not be proposed for higher stages
(or in the case of ovarian capsule rupture during
initial surgery) because the prognosis is less clear
cut.
The use of completion surgery after childbearing
continues to be debated in SCST [79].
Conclusions
Conservative treatment yields good fertility results
and does not affect the survival of patients with borderline ovarian tumors. It should be considered for young
women desiring fertility, even if peritoneal implants
are discovered at the time of initial surgery. In case of
infertility, medically-assisted procreation techniques
may be proposed to patients with stage I BOT with a
limited number of stimulation cycles.
In patients with epithelial ovarian cancer, conservative surgery of an ovary and the uterus can only be
considered in adequately staged patients, with a stage
IA, grade 1 (and probably 2) serous, mucinous or an
endometrioid tumor and careful follow-up. Such management could probably also be safely proposed for
stage IC, grade 1 disease.
In patients with non-epithelial ovarian cancer, conservative surgery is highly applicable, particularly in
patients with malignant germ cell tumors.
Acknowledgement
The authors thank Lorna Saint Ange for her editing.
References
1. Taylor HC. Malignant and semi-malignant tumors
of the ovary. Surg Gynecol Obstet 1929; 48:
20430.
2. Hart WR and Norris HJ. Borderline and malignant
mucinous tumors of the ovary. Cancer 1973; 31:
103144.
3. Bell DA, Weinstock MA and Scully RE. Peritoneal
implants of ovarian serous borderline tumors.
Histologic features and prognosis. Cancer 1988; 62:
221222.
4. Duvillard P. Tumeurs ovariennes a` la limite de la
malignite. Ann Pathol 1996; 16: 396405.
5. Gershenson D, Silva E, Levy L et al. Ovarian serous
borderline tumors with invasive peritoneal implants.
Cancer 1998; 82: 1096103.
6. Gershenson D, Silva EL, Tortolero-Luna G et al.
Serous borderline tumors of the ovary with
non-invasive peritoneal implants. Cancer 1998; 83:
215763.
7. Seidman JD and Kurman RJ. Subclassification of
serous borderline tumors of the ovary into benign and
malignant types. A clinicopathologic study of 65
advanced stage cases. Am J Surg Pathol 1996; 20:
133145.
8. Kane A, Uzan C, Rey A et al. Prognostic factors in
patients with ovarian serous low malignant potential
(borderline) tumors with peritoneal implants.
Oncologist 2009; 14: 591600.
9. Seidman JD and Kurman RJ. Treatment of
micropapillary serous ovarian carcinoma: the agressive
variant of serous borderline tumors. Cancer 2002; 95:
6756.
10. Eichhorn JH, Bell DA, Young RH et al. Ovarian
serous borderline tumors with micropapillary and
cribriform patterns: a study of 40 cases and
comparison with 44 cases without these patterns. Am J
Surg Pathol 1999; 23: 397409.
11. Trimble CL, Kosary C and Trimble EL. Long-term
and patterns of care in women with ovarian tumors of
low malignant potential. Gynecol Oncol 2002; 86:
347.
12. Tinelli R, Tinelli A, Tinelli FG et al. Conservative
surgery for borderline ovarian tumors: a review.
Gynecol Oncol 2006; 100: 18591.
13. Morice P. Borderline tumours of the ovary and
fertility. Eur J Cancer 2006; 42: 14958.
14. Swanton A, Bankhead CR and Kehoe S. Pregnancy
rates after conservative treatment for borderline
ovarian tumours: a systematic review. Eur J Obstet
Gynecol Reprod Biol 2007; 135: 37.
275
276
277
278
Section 6
Chapter
22
Introduction
Survival rates after cancer have increased significantly
in recent decades; however, these treatments also have
drawbacks and patients (or parents in the case of children) must be informed of the long-term side effects
of oncological treatments and the possible options for
preserving the fertility of these patients. It is important to set out clearly the possible risks of developing
ovarian failure or azoospermia with oncological treatments. These will depend on the age of the patients and
on the type, dose and duration of chemotherapy, and
on the field, dose and duration of radiotherapy.
The strategy must be individualized in each case
depending on:
r Patients age.
r Type and stage of the cancer.
r Therapeutic plan to be followed.
r Foreseeable long-term effects.
r Possibility or impossibility of delaying the start of
treatment.
r Whether or not the patient has a partner/spouse.
r Biology of the tumor and potential for metastasis
in ovary.
If ovarian stimulation is possible, embryo cryopreservation is the method with the greatest chance of success so far. However, it is only possible if there is a
partner or if the patient accepts donor sperm. It is
very important to clarify which are the safest ovarian stimulation protocols to be used in these patients,
although current data suggest that in certain cancers a
cycle of ovarian stimulation does not increase the risk
of recurrence.
Since the first pregnancy obtained from cryopreserved human embryos [1], the yield and safety
of embryo cryopreservation programs have improved
notably with successful freezing of zygotes and cleaving of embryos and blastocysts, and pregnancies
are regularly obtained after thawing and transfer
of embryos in any of these stages. Our first pregnancy after transferring a previously frozen embryo
was obtained even though only one of the embryos
original four cells survived thawing and was transferred [2].
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
279
Cycles
Transfers
Thawed embryos
Survival
Pregnancy/transfer
Babies born
6715
5354
20.318
66%
22.5%
807
Frozen time
Pregnancy rate/transfer
57 years
16/44
36.4%
79 years
5/16
31.3%
9 years
5/8
62.5%
5 years
853/2479
33.68%
5 yearsa
26/68
38.2%
Totals
One baby born after 10 years and 1 baby born after 14 years of
cryopreservation.
280
patients have opted for this strategy and the first births
achieved in this way have been published [21].
In applying embryo-freezing techniques to preserve the fertility of oncology patients, it is very important to know the couples preference for the disposition of any unused embryos. Couples are normally
offered the possibility of choosing, in the event of death
or unforeseen circumstances, between the destruction
of the embryos, donation for research, anonymous
donation to other infertile couples or leaving the decision to the surviving partner. The oncology patients
make similar choices to the infertile patients who
undergo IVF with regard to donation of the embryos
for research or to other infertile couples, but most
oncology patients reject destruction of the embryos
and accept this option in a much lower percentage than
normal IVF patients [14].
Conclusions
Up to now, embryo cryopreservation has been the only
clinically accepted method for preserving the fertility
of oncology patients before they undergo chemotherapy and/or radiotherapy. The post-thawing pregnancy
rates are acceptable and are around 30% per cryoreplacement depending on the number of embryos available and their quality.
However, this option does have some drawbacks
such as:
(a) It is not feasible for pubertal girls.
(b) Ovarian stimulation may be contraindicated and
oocyte retrieval and IVF may cause a delay in the
initiation of oncological treatment that may not
be acceptable in some cases.
(c) Spermatozoa from a male partner or from a donor
is required.
(d) Possible religious or moral objections.
References
1. Trounson A and Mohr L. Human pregnancy following
cryopreservation, thawing and transfer of an eight-cell
embryo. Nature 1983; 305(5936): 7079.
2. Veiga A, Calderon G, Barri PN and Coroleu B.
Pregnancy after the replacement of a frozenthawed
embryo with less than 50% intact blastomeres. Hum
Reprod 1987; 2(4): 3213.
3. Wenherholm UB, Sodertrom Anttila V et al.
Children born after cryopreservation of embryos or
oocytes: a systematic review of outcome data. Hum
Reprod 18(6): 81520.
281
21. Yang D, Brown SE, Nguyen K et al. Live birth after the
transfer of human embryos developed from
cryopreserved oocytes harvested before cancer
treatment. Fertil Steril 2007; 87(6): 1469 e14.
282
Section 6
Chapter
23
Oocyte cryopreservation
Slow freezing
Andrea Borini and Veronica Bianchi
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
283
intracellular water from leaving the cell and, consequently, result in ice crystal formation and cell death.
To partially overcome to this issue, CPAs have been
introduced; in fact one role of CPAs is to protect biological materials against cooling injuries from ice crystals and high concentrations of solute during cryopreservation [7, 8].
Since the introduction of glycerol as a permeable cryoprotective agent in sperm cryopreservation
by Polge et al., and the subsequent discovery and use
of dimethyl sulfoxide (DMSO), many cells and tissues have been frozen but the cooling procedures were
mostly theoretical [9, 10].
All freezing methods developed to date rely on the
presence of one or more CPAs in molar concentrations.
In general cryoprotectants may be divided into two
groups:
(a) Permeating agents such as glycerol, DMSO,
ethylene glycol and 1,2-propanediol (PROH). All
of which are chemically characterized as having a
relatively low molecular weight and can penetrate
the lipid bilayer of the cell with a slower
permeability than water.
(b) Non-permeating agents that remain in the
extracellular solution as a result of their size or
polarity. These CPAs include sugars and
macromolecules such as sucrose, Ficoll and
raffinose, as well as proteins and lipoproteins.
Cryoprotective agents can reduce the toxic effects
of high concentrations of other compounds in the
solution [11], and their protective effects are related to
their hydrophilic ability to create hydrogen bond and
consequently to interact with water and to achieve high
aqueous solubility.
With glycerol and PROH, the hydrogen bonding is
between the hydrogen of the OH groups and the water.
The oxygen on the DMSO molecules bind to the water
protons with the release of heat. During slow cooling,
when the cells are very dehydrated and are surrounded
by concentrated salts, the cryoprotectants appear to
reduce damage caused by the high levels of salt.
These CPAs, however, can have dramatic osmotic
effects upon cells during the freezing/thawing procedures; when exposed to permeating solutes the oocyte
undergoes extensive initial dehydration and shrinkage
followed by a return to near the original volume as the
cryoprotectant enters the cell and replaces the intracellular water. This causes a double flux across the membrane (the water exits the cell while the CPA enters)
284
that influences both the intracellular solute concentration and the cell volume. The extent of shrinkage and
swelling can cause damage or even cell death due to
the osmotic stress acting on the oocyte membrane. To
reduce stress from excessive swelling, non-permeating
molecules are added to the CPA mixture. Sucrose is the
most commonly used, but trehalose and choline have
also been utilized in cooling protocols [1214].
Usually, in the most widely adopted slow cooling
protocols, cryoprotectant concentrations are around
1.5 M for the penetrating agent (mainly PROH or
DMSO) and 0.10.3 M for the non-penetrating agent
(usually sucrose).
The analysis of cell-volume change dynamics is
important to evaluate approaches to improve freezing protocols. Addition of cryoprotectants can cause
cell damage due to volume changes and chemical toxicity; optimal exposure should aim to minimize
osmotic stress while avoiding chemical toxicity and
allow sufficient permeation and dehydration to achieve
protection from freezing injury.
Several authors approached those issues with different ideas: Yang et al. tried to increase cryoprotectant exposure temperature to achieve faster dehydration rates [15].
Quintans et al. adopted a stepwise addition of
the permeating cryoprotectant (PROH) to reduce volume excursion [16], while Boldt et al. tried to use a
sodium-depleted freezing medium combined with a
lower seeding temperature to improve post-thawing
recovery [17].
However, no measurement of the actual response
of the cells to these changes was performed in these
studies. To address this, Paynter et al. tried to improve
the cryopreservation technique by evaluating several
factors involved during the equilibration of oocytes
to cryoprotectant interaction and decreased temperatures [18]. The authors measured the osmotic response
by monitoring the oocytes for 10 min during a twostep addition of the permeating cryoprotectant PROH
(0.75 and 1.5 M PROH). Following this, the oocyte
osmotic response to 1.5 M PROH and 0.2 or 0.3 M
sucrose was measured. Those authors found that each
oocyte shrank during the first exposure to the cryoprotectant (0.75 M PROH) as water left the cell and
then gradually re-expanded as water and cryoprotectant entered. The entire volume equilibration process
took around 10 min to complete.
During the second exposure to an increased cryoprotectant concentration (1.5 M PROH) the oocytes
underwent the same phases of shrinkage and reexpansion, while in the last set of exposure experiments (1.5 M PROH plus sucrose) the shrinkage rate
was faster with an obvious reduction of cell volume
before freezing to avoid intracellular ice crystal formation.
This paper targeted the important concept of a
shrinkage and re-expansion time frame in which
oocytes can recover after PROH exposure.
An equally important consideration for oocyte survival involves the removal of permeating cryoprotectants. When a cell containing cryoprotectant is placed
in medium with a lower concentration of cryoprotectant, water enters the cells to dilute the cryoprotectant at a faster rate than the cryoprotectant can leave
the cells, which causes swell and potential burst. This
problem can be overcome by controlled removal of the
cryoprotectants in a series of decreasing concentration
steps.
This does not prevent the cells from swelling, but
reduces the corresponding magnitude and achieves the
desired outcome of gradual CPA removal and rehydration in a controlled manner. Cells are moved from one
solution to the next after they re-acquired their normal
level of hydration.
An alternative method for removing cryoprotectants from cells is to use a high concentration of a
non-penetrating molecule such as sucrose; this extracellular high concentration serves to balance the
high intracellular cryoprotectant concentration as it
reduces the difference in osmolarity between the intra
and extracellular environments. The oocytes shrink
proving that both the cryoprotectant and the water are
leaving the cell reducing the intracellular volume. The
use of high sucrose concentrations allows a one step
thawing protocol that is simpler and faster than multistep dilution procedures.
Morphological variables
In addition to surviving the cryopreservation/warming process, the oocyte needs to maintain competence
to fertilize and develop in vitro to the appropriate
embryonic stage without any structural alterations.
It has been widely demonstrated that low temperatures and cryopreservation procedures in general may induce critical damage to the cell substructures, especially due to the peculiar characteristics of
the metaphase II (MII) oocyte. At this maturational
stage the chromosomes are aligned on the metaphase
285
286
oocytes. They demonstrated that the use of PROHfreezing protocol for human oocytes resulted in extensive cortical granule exocitosis. Those results were
later confirmed by Nottola et al. who showed that the
amount and density of cortical granules appeared to
be abnormally reduced in some frozenthawed samples, despite the slow-freezing protocol [51]. These
abnormal features were frequently associated with an
increased density of the filamentous texture related to
the occurrence of zona hardening. The same result
was obtained by these authors using EG and sucrose
as cryoprotectants [44]. As previously mentioned, the
MII oocyte has a complex subcellular structure that
includes the meiotic spindle, cortical granules and
other features like mithocondria or smooth endoplasmatic reticulum (SER). These ultrastructural components were not taken into account in the initial studies,
since oocyte survival after thawing was primarily correlated to morphological appearance of the egg. Lately,
several authors focused their attention on the possible consequences related to subcellular injuries. Nottola et al. analyzed fresh and frozenthawed oocytes
using a slow cooling method with PROH and sucrose
(0.1 or 0.3 mol/l) as cryoprotectants [51]. The oocytes
were then processed for electron microscopy observations. All the oocytes showed a homogeneous cytoplasm and an intact zona pellucida (ZP) with abundant and uniformly dispersed organelles (mainly
mitochondriasmooth endoplasmic reticulum aggregates and mitochondriavesicle complexes). Nevertheless, a degree of microvacuolization was detected in
the ooplasm of some frozenthawed oocytes, particularly in those treated with higher sucrose concentration. Another interesting study comparing fresh and
frozen thawed eggs (PROH plus 0.3 mol/l sucrose)
was published by Gualtieri et al. [53]. The authors
showed evidence that in fresh samples mitochondria
had a regular shape with few short cristae, whereas in
the frozenthawed group a high percentage of oocytes
(72%) showed a variable and, in some cases, a very high
fraction of mitochondria with decreased electron density of the matrix or ruptures of the outer and inner
membranes. Moreover in those oocytes, the mitochondrial damage was associated with SER swelling.
287
288
ences between the two protocols applied in the literature at that time (using 0.1 and 0.3 M sucrose) [63]. It
was surprising that, while the survival and fertilization
rates were significantly improved using higher sucrose
concentration, the pregnancy and implantation were
much improved using low sugar-based solutions. This
may be due to the subtle equilibrium between dehydration enhanced by sucrose and possible subcellular
damages caused by the same cryoprotectant. At this
point it was clear that, besides the biological good
outcome, the high sucrose-based protocol was not producing good results in term of pregnancy and implantation.
Starting from this important observation, Bianchi
et al. established a modified cryopreservation protocol
in which the freezing solution contains 1.5 M PROH
and 0.2 M sucrose in order to reduce the impact of
shrinkage during cooling procedures [64]. The higher
sucrose concentration (0.3 M) was used during thawing, after Fabbri et al. [40], in keeping with the original
idea of Lassalle et al. [54], who used a thawing solution in which sucrose concentration was higher than
that employed in the freezing solution. The survival
rate reported was high and comparable to the previous studies (76.0%) as is the rate of fertilization (76.2%)
and embryo cleavage (93.7%) [64]. The significant difference was evidenced in the pregnancy rate, which
was 21.2, 18.9 and 21.8% per embryo transfer, thaw
cycle and patient, respectively, and the implantation
rate was 13.4%. Pregnancy rate per cryopreserved
thawed oocyte was 4.9% and the implantation rate per
oocyte was 6.9%. These results represent an important achievement in the everyday application of oocyte
freezing.
In a multicenter study, Borini et al. showed that,
out of 2046 patients involved in oocyte cryopreservation, the overall survival rate of thawed oocytes was
55.8% [65]. In 940 thaw cycles, the mean numbers
of inseminated oocytes and fertilization rates were
significantly decreased versus fresh cycles outcomes
(2.6 0.7 versus 2.9 0.2 and 72.5 versus 78.3%,
respectively), as were the rates of implantation (10.1
versus 15.4%), pregnancy rates per transfer (17.0 versus 27.9%) and pregnancy rates per cycle (13.7 versus
26.2%). Even though differences in clinical outcome
were found among centers, a pregnancy rate per thawing cycle above 14% was achieved by most clinics.
It is evident that oocyte cryopreservation is able to
add a valuable option to routine IVF procedures, especially in countries with severe law restrictions.
289
References
1. Chen C. Pregnancies after human oocyte
cryopreservation. Lancet 1986;1(8486): 8846.
2. Jackowski S, Leibo SP and Maxur P. Glycerol
permeabilities of fertilized and infertilized mouse ova.
J Exp Zool 1980; 212: 32941.
3. Leibo SP. Water permeability and its activation energy
of fertilized and unfertilized mouse ova. J Membr Biol
1980; 53: 17988.
4. Arrhenius S. Quantitative Laws in Biochemistry.
London: J. Bell, 1915.
5. Mazur P. Freezing of living cells: mechanisms and
implications. Am J Physiol 1984; 247: 12542.
6. Mazur P. Equilibrium, quasi-equilibrium, and
non-equilibrium freezing of mammalian embryos. Cell
Biophysiol 1990; 17: 53.
7. Elliot K and Whelan J. (eds.). CIBA Foundation
Symposium 52. The Freezing of Mammalian Embryos.
Amsterdam: Elsevier, 1977.
8. AshwoodSmith M J and Farrant J. (eds.). Low
Temperature Preservation in Medicine and Biology.
Bath UK: Pitman Press, 1980.
9. Polge C, Smith A and Parkes A. Revival of
spermatozoa after vitrification and dehydration at low
temperatures. Nature 1949; 164: 6667.
10. Lovelock J and Bishop MW. Prevention on freezing
damage to living cells by dimethyl sulphoxide. Nature
1959; 183: 13945.
11. Fahy GM, Vitrification, in low temperature
biotechnology. In: McGrath JJ and Diller KR (eds.),
Low Temperature Biotechnology: Emerging
Applications and Engineering Contributions. New York:
American Society of Mechanical Engineers, 1989: pp.
11346.
12. Eroglu A, Toner M and Toth TL. Beneficial effect of
microinjected trehalose on the cryosurvival of human
oocytes. Fertil Steril 2002; 77: 1528.
290
291
292
Section 6
Chapter
24
Introduction
In principle, the aims we expect from a cryopreservation technique that allows us to store a biological
material at the low temperature of liquid nitrogen
(196 C) with the tissue free of any crystalline structure, and with arrest of all molecular diffusion and
chemical processes which would otherwise precipitate
degradation and aging, are as follows:
1.
2.
3.
4.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
293
250
200
200
161
157
132
122
150
102
100
64
50
12 15 14
1 4 1 6 1 5 6 5 5
21
52
31
22 23 30 29
19
66
19
77
19
79
19
84
19
86
19
88
19
90
19
92
19
94
19
96
19
98
20
00
20
02
20
04
20
06
20
08
48 48
96
73 74
Years
Figure 24.2 The explosion of interest in the use of vitrification is beginning to affect clinical embryo and oocyte storage. (Data derived from
the Scopus Library the worlds largest abstract and citation database of research literature and web sources; it offers access to 245 million
references and 27 million abstracts from over 15 000 peer-reviewed journals.)
294
Donor
oocytes
Autologous
oocytes
Warming cycles
12
21
53/64 (83%)
140/187 (75%)
Fertilization (%)
48/53 (91%)
101/140 (72%)
Transfers
12
17
Pregnancies/embryo
transfer (%)
9 (75%)
5 (29%)
Implantations (%)
9/24 (37.5%)
7/48 (14.5%)
from these cryostored oocyte pregnancies [38], regardless of the historical concerns that cryopreservation
of mature oocytes might disrupt the meiotic spindle and thus increase the potential for aneuploidy in
the embryos arising from such eggs. These concerns
have mostly been allayed by publications that show
no abnormal or stray chromosomes from previously
frozen oocytes [39], and fluorescence in situ hybridization (FISH) comparison of embryos from fresh and
thawed oocytes show no increase in anomalies [40].
There also appears to be adequate recovery of the
meiotic spindle post-cryopreservation whether using
conventional or vitrification technology [4143]. With
respect to cryostorage of donated oocytes, there have
been a number of reports that have described success with this approach [23, 30, 32, 44, 45]. Indeed
this approach has now been formally commercialized.
Cryostorage of womens own oocytes was originally reported with three pregnancies established in
the late 1980s by two centers [46, 47]. Following a hiatus of several years, these early reports were reproduced by others [17, 30, 48]. These pregnancies mostly
arose from the freezing of oocytes that had been
collected for purposes of infertility therapy where
couples may have had religious or ethical concerns
with embryo cryopreservation; or when couples consented to research studies; or even when spermatozoa
were unexpectedly unavailable after oocytes have been
retrieved during an IVF cycle. The latter circumstances
would have occurred following an unsuccessful testicular biopsy or when the partner was unable to produce
a semen sample for a variety of unanticipated reasons.
295
Figure 24.3 (a) Mature metaphase-II (MII) stage oocyte; (b) immature MI stage oocyte; and (c) germinal vesicle (GV) stage oocyte.
296
Cryoloop
Hemi-straw system
Number of oocytes
448
480
Oocyte survival
361 (80.6%)a
410 (85.4%)b
Overall survival
a
b
738/890 (82.9%)
297
7.5
0.0
7.5
Vitrification solution
15
0.5
15
Warming solution
1.0
Dilution solution
0.5
Holding solution
0.0
for single women concerned with their future reproductive choices (insurance freezing/self donation)
[64]. In accepting that human oocyte cryopreservation is here to stay, it is of great importance that we
research the consequences of this technology carefully,
to ensure that we do no harm, through establishment
of an effective registry of outcomes and births from
oocyte cryopreservation.
Conventional cryopreservation has seen significant improvement in consistency of outcomes the past
few years in the form of the introduction of ICSI
as the routine insemination approach after cryostorage, choline-based (sodium-depleted) cryo-media and
use of higher concentration sugar solutions. Within
the confines of conventional freezing technology there
may be subtle variations in approach that will only be
revealed through ongoing clinical studies. Major leaps
of technology, such as injection of trehalose into the
oocytes before freezing [65], while theoretically attractive, nevertheless impose an increased level of invasiveness that seems to run contrary to the wish to simplify oocyte freezing. However, we should remain open
minded given that little has been done to date to assess
oocyte quality at the cellular level, other than to determine the status of the meiotic spindle [41].
With regard to vitrification as a means to cryopreserve oocytes, increased speed of cooling through use
of better designed carriers and protocols that lessen the
concentration of cryoprotectant used, while hastening
exposure and procedure times, have put this technology on the map [66], and excellent embryo quality can
be obtained from vitrified oocytes [67]. Furthermore,
the most widely used protocol for vitrification of biological material is a two-step partial equilibration in
the cryoprotectant, and a three-step warming procedure in sucrose only (Table 24.3).
298
(a)
(b)
(c)
(d)
(e)
(f)
(g)
(h)
(i)
(a)
(b)
(c)
(d)
(e)
(f)
(g)
(h)
(i)
Open system
(Direct contact
with LN2
Cooling rates
>20 000C)
Closed system
(No direct
contact with LN2
Cooling rates
~1750C)
vation: the oocytes themselves must be able to fertilize normally at an acceptable rate; develop morphologically as would fertilized fresh oocytes; and ultimately to implant at a clinically appropriate rate. Poor
embryonic cleavage is especially indicative of cryptic
cyto-skeletal damage that may have incurred during
the cryopreservation process. It has to be remembered,
however, that oocytes are only as good as they were
299
Vitrification/warm (% survival)
120
100
80
60
40
20
0
% Survival
5
Trials
300
at their time of collection, and fundamental compromise of oocyte quality may occur during ovarian stimulation, retrieval and, more problematically, innate
quality issues related to a womans physiology may
make assessing these developmental issues less clear
cut. Indeed, there exists a natural variability between
women, and often between stimulation cycles from the
same woman; such fluctuations in oocyte quality may
have a significant negative impact on innate competency of such eggs. However, it is our mission during
oocyte cryopreservation to minimize any deleterious
conditions that reduce oocyte quality further on a permanent basis.
metal surfaces [70] remains to be seen. Such developments do detract from the simplicity and low-cost benefits of vitrification; however, even higher cooling rates
may reduce even further the need for the relatively
high concentrations of cryoprotectant and so allow vitrification to be an even more low-impact cryopreservation approach for oocytes.
References
1. Whittingham DG, Leibo SP and Mazur P. Survival of
mouse embryos frozen to 196 and 269 C. Science
1972; 178: 41114.
2. Rall WF and Fahy GM. Ice-free cryopreservation of
mouse embryos at 196 C by vitrification. Nature
1985; 313: 5735.
3. Liebermann J, Dietl J, Vanderzwalmen P and Tucker
MJ. Recent developments in human oocyte, embryo
and blastocyst vitrification: where are we now? Reprod
Biomed Online 2003; 7: 62333.
4. El-Danasouri I and Selman H. Successful pregnancies
and deliveries after a simple vitrification protocol for
day 3 human embryos. Fertil Steril 2001; 76:
4002.
5. Jelinkova L, Selman HA, Arav A et al. Twin
pregnancy after vitrification of 2-pronuclei human
embryos. Fertil Steril 2002 77: 41214.
6. Liebermann J, Tucker MJ, Graham JR et al. Blastocyst
development after vitrification of multipronuclear
zygotes using the Flexipet denuding pipette. Reprod
Biomed Online 2002; 4: 14650.
7. Kuwayama M, Vajta G, Ieda S and Kato O.
Comparison of open and closed methods for
vitrification of human embryos and the elimination of
potential contamination. Reprod Biomed Online 2005;
11: 60814.
8. Al-Hasani S, Ozmen B, Koutlaki N et al. Three years
of routine vitrification of human zygotes: is it still fair
to advocate slow-rate freezing? Reprod Biomed Online
2007; 14: 28893.
9. Rama Raju GA, Haranath GB, Krishna KM, Prakash
GJ and Madan K. Vitrification of human eight-cell
embryos, a modified protocol for better pregnancy
rates. Reprod Biomed Online 2005; 11: 4347.
10. Rama Raju GA, Jaya Prakash G, Murali Krishna K
and Madan K. Neonatal outcome after vitrified day 3
embryo transfers: a preliminary study. Fertil Steril
2009; 92: 1438.
11. Desai N, Blackmon H, Szeptycki J and Goldfarb J.
Cryoloop vitrification of human day 3 cleavage-stage
embryos: post-vitrification development, pregnancy
outcomes and live births. Reprod Biomed Online 2007;
14: 20813.
301
302
303
304
Section 6
Chapter
25
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
305
3.8
3.2 2.7 2
0.6
Leukemia
11.3
Hematological
malignancies
44.3
9.6
Hodgkin's
lymphoma
22
22.6
Breast cancer
Ovarian cancer
Rhabdomyosarcoma
Cervical cancer
Liposarcoma
Leukemia
Non-Hodgkin's
lymphoma
11
306
from the stromal cells. After investigating for follicles in the different interfaces, we found 63% in the
phosphate-buffered saline (PBS)1.06 Ficoll interface
and 36.9% at the 1.061.09 Ficoll interface, which represents 99.9% of total recovered follicles. Analysis by
vital fluorescent staining showed that 95.8% of follicles treated with Ficoll were totally viable. The Ficoll
density gradient method thus allows us to maximize
recovery of isolated human ovarian follicles and minimize manipulation time, while maintaining high follicular viability [32].
Figure 25.2 Enzymatically isolated human follicles seen under a
stereomicroscope.
307
308
References
1. Donnez J, Dolmans MM, Demylle D et al. Livebirth
after orthotopic transplantation of cryopreserved
ovarian tissue. Lancet 2004; 364: 140510.
2. Meirow D, Levron J, Eldar-Geva T et al. Pregnancy
after transplantation of cryopreserved ovarian tissue in
a patient with ovarian failure after chemotherapy. N
Engl J Med 2005; 353: 31821.
3. Demeestere I, Simon P, Emiliani S, Delbaere A and
Englert Y. Fertility preservation: successful
transplantation of cryopreserved ovarian tissue in a
young patient previously treated for Hodgkins disease.
Oncologist 2007; 12: 143742.
4. Andersen CY, Rosendahl M, Byskov AG et al. Two
successful pregnancies following autotransplantation
of frozen/thawed ovarian tissue. Hum Reprod 2008; 23:
226672.
5. Silber SJ, DeRosa M, Pineda J et al. A series of
monozygotic twins discordant for ovarian failure:
ovary transplantation (cortical versus microvascular)
and cryopreservation. Hum Reprod 2008; 23:
15317.
6. Piver P, Amiot C, Agnani G et al. Two pregnancies
obtained after a new technique of autotransplantation
of cryopreserved ovarian tissue. Hum Reprod 2009; 24
(Suppl. 1): i15 0035.
7. Sanchez-Serrano M, Crespo J, Mirabet V et al. Twins
born after transplantation of ovarian cortical tissue
and oocyte vitrification. Fertil Steril 2010; 93(1): 286
e1113.
8. Donnez J, Martinez-Madrid B, Jadoul P et al.
Ovarian tissue cryopreservation and transplantation: a
review. Hum Reprod Update 2006; 12: 51935.
9. Meirow D, Hardan I, Dor J et al. Searching for
evidence of disease and malignant cell contamination
in ovarian tissue stored from hematologic cancer
patients. Hum Reprod 2008; 23(5): 100713.
10. Shaw JM, Bowles J, Koopman P, Wood EC and
Trounson AO. Fresh and cryopreserved ovarian tissue
samples from donors with lymphoma transmit the
cancer to grafts recipients. Hum Reprod 1996; 11:
166873.
11. Meirow D, Ben Yehuda D, Prus D et al. Ovarian tissue
banking in patients with Hodgkins disease: is it safe?
Fertil Steril 1998; 69: 9968.
12. Kim SS, Radford J, Harris M et al. Ovarian tissue
harvested from lymphoma patients to preserve fertility
may be safe for autotransplantation. Hum Reprod
2001; 16: 205660.
13. Seshadri T, Gook D, Lade S et al. Lack of evidence of
disease contamination in ovarian tissue harvested for
cryopreservation from patients with Hodgkin
26. Osborn SM, Gook DA, Stern C and Speirs AL. The
isolation and culture of human primordial follicles
from fresh ovarian tissue. Hum Reprod 1997: 12: 192
[Abstr.].
27. Roy SK and Treacy BJ. Isolation and long-term culture
of human preantral follicles. Fertil Steril 1993; 59:
78390.
28. Schroder CP, Timmer-Bosscha H, Wijchman JG
et al. An in vitro model for purging of tumour cells
from ovarian tissue. Hum Reprod 2004; 19: 106975.
29. McShane P, Sutton R, Gray DW and Morris PJ
Protease activity in pancreatic islet isolation by
enzymatic digestion. Diabetes 1989; 1:1268.
30. Linetsky E, Bottino R, Lehmann R et al. Improved
human islet isolation using a new enzyme blend,
Liberase. Diabetes 1997; 46(7): 11203.
31. Dolmans MM, Michaux N, Camboni A et al.
Evaluation of Liberase, a purified enzyme blend, for
the isolation of human primordial and primary
ovarian follicles. Hum Reprod 2006; 21: 41320.
32. Martinez-Madrid B, Dolmans MM, Van Eyck AS
et al. Ficoll density gradient method for recovery of
isolated human ovarian primordial follicles. Fertil
Steril 2004; 82: 164853.
33. Carroll J and Gosden RG. Transplantation of
frozenthawed mouse primordial follicles. Hum
Reprod 1993; 8: 11637.
34. Rodgers RJ, Irving-Rodgers HF and Russell DL.
Extracellular matrix of the developing ovarian follicle.
Reproduction 2003; 126, 41524.
35. Dolmans MM, Martinez-Madrid B, Gadisseux E
et al. Short-term transplantation of isolated human
ovarian follicles and cortical tissue into nude mice.
Reproduction 2007; 134: 25362.
36. Skinner MK. Regulation of primordial follicle
assembly and development. Hum Reprod Update 2005;
11: 46171.
37. Qu J, Godin PA, Nisolle M and Donnez J.
Distribution and epidermal growth factor receptor
expression of primordial follicles in human ovarian
tissue before and after cryopreservation. Hum Reprod
2000; 15: 30210.
38. Dolmans MM, Yuan Yuan W, Camboni A et al.
Development of antral follicles after xenografting of
isolated small human preantral follicles. Reprod
Biomed Online 2008; 16: 70511.
39. Amorim CA, Rondina D, Lucci CM et al.
Cryopreservation of sheep primordial follicles. Reprod
Domest Anim 2007; 42(1): 537.
309
Section 6
Chapter
26
Introduction
This book is primarily about prevention; its emphasis is on interventions that can be done at the time
of cancer diagnosis modifications of treatment and
techniques for storing gametes, tissues or embryos for
future use. By contrast, this final chapter of Section 6
explores the options open to the cancer survivor some
time after the diagnosis has been made and treatment
completed. If preventive treatment was successful, in
some cases, normal fertility has been preserved. Other
survivors may conceive using the gametes, embryos
or tissue that were obtained and cryopreserved before
their gonadotoxic treatment.
However, in some cases, prevention may not have
been successful. This chapter is written to provide
insight into the fertility management of cancer survivors with compromised or absent ovarian function
and without cryopreserved material.
Before embarking on a pregnancy, no matter how
it is to be achieved, a cancer survivor and her physicians must look at her general health, at her uterine function, at her chance of recurrence and, finally,
at her prognosis for long-term survival. Since these
issues are common to a cancer survivors pregnancies
whether achieved in the natural cycle, through assisted
reproductive technology (ART) or through donor egg
(DE) they will be discussed together in the first part
of this chapter.
The second part of this chapter will focus on situations where the cancer survivor does not conceive
spontaneously but her fertility is amenable to treatment. In situations where ovarian function persists but
is attenuated, ART may be able to overcome treatmentrelated damage. For some cancer survivors, specific
modifications of ART may be indicated.
Prognosis
Before other evaluation is done, the treating oncologist
should be consulted about the prospective mothers
prognosis. Has the survivor been disease free for a
sufficient period of time? Is her prognosis for cure
generally good? Is there a risk that pregnancy would
increase cancer recurrence? Kavic and Sauer have suggested that in some cases, but particularly in cases
where cancer is considered to be in remission but not
cured, a second opinion from an independent oncologist may be helpful [1]. Patients and their physicians
may elect to proceed with pregnancy in the presence
of a poor prognosis, or even in situations where pregnancy may increase the risk of recurrence. However,
this should not take place without full discussion and
fully informed consent from all parties involved.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
310
No
Psychological evaluation
and ethical considerations
Yes
No
No
Routine preconceptual
counseling
MFM consult
(Is pregnancy contraindicated?)
Yes
Test for ovarian function
Consider gestational
carrier
Yes
Attempt pregnancy
with ART
No
Donor egg
311
Uterine evaluation
If a cancer survivor does not achieve a pregnancy in the
first few months of attempting conception, she should
generally have a uterine cavity evaluation a hysterogram or sono-hysterogram to rule out adhesions,
intraluminal pathology or a significant septum. The
uterine evaluation is especially critical for a woman
who has had pelvic or total body irradiation.
Pelvic irradiation often results in damage to the
uterine musculature and vasculature. Childhood radiation, especially, may result in poor uterine growth
during puberty. The smaller uterine volume may
diminish implantation, cause second trimester loss,
preterm labor or placenta accretia [7]. A tripling of
spontaneous miscarriage and a 10-fold increase in lowbirth weight infants have been reported in patients
who had received pelvic radiation therapy (RT) compared to the general population [8].
These risks are affected by the dose of RT delivered to the uterus and by the temporal association
with puberty. There is, for example, a linear correlation between the size of the uterus, the response to hormone treatment and the age at which RT was administered [8]. Some investigators have emphasized that
many reported pregnancies post-pelvic radiation are
delivered prematurely and do not result in the delivery of a healthy child [9].
Although the adult uterus is less sensitive to
the effects of radiation, adult cancer survivors who
received RT to the pelvis should delay pregnancy for
at least a year following RT; it has been suggested that
pregnancies that occur 1 year after RT have a higher
rate of low birth weights and miscarriages [10]. (For a
more detailed discussion of the effects of pelvic radiation on uterine function, see Chapter 2.)
312
tissue as long as she is advised of the presumed prognosis in doing so. By contrast, a patient who exhibits
extremely high FSH levels, very low inhibin or AMH
levels or has a negligible antral follicle count, may
wish to discuss oocyte donation or other reproductive
alternatives. If she has previously cryopreserved eggs
or embryos, she should be encouraged to use the
cryopreserved material.
Ovarian stimulation
The physician treating a cancer survivor who wishes
to have children will want to keep one central principle in mind: if the patient was treated with gonadotoxic
agents or pelvic RT, no matter what her present age or
ovarian function, she is likely to have reduced ovarian function earlier than her age would suggest. Therefore, as soon as she is ready, she should be encouraged to attempt conception. If she is not successful within 612 months, a thorough evaluation and
intervention should be undertaken promptly. Finally,
strategies to achieve pregnancy in the shortest time
frame possible are reasonable, if not imperative, in this
population.
For these reasons, we should recommend IVF for
the infertile cancer survivor who fails to conceive a
pregnancy after a limited trial of ovulation induction
and intrauterine insemination (IUI). In vitro fertilization (IVF) offers the greatest chance for pregnancy
in the shortest time interval. This is additionally
important in women with estrogen sensitive tumors in
order to minimize their exposure to multiple courses
of ovarian stimulation.
For women who have stored gametes or embryos
prior to gonadotoxic treatments, the following caveats
are equally important: when ovarian function still
exists and they are prepared to become pregnant, they
should be encouraged to conceive either on their own
or with ART. It should be emphasized that having
frozen oocytes and embryos does not guarantee pregnancy success; rather they should be considered as
the last option should the woman become sterile. One
should not allow women who can still conceive to delay
childbearing because of a false sense of security that
their future reproductive potential is guaranteed by
cryopreservation of either eggs or embryos. And, in
general, and especially if the patient is considering having more than one child, consideration should be given
to maintaining the cryopreserved eggs or embryos in
reserve. If the patient has reasonable ovarian function
313
314
antag
hCG
FSH 150 IU
Tamoxifen
with FSH
Tamoxifen 60 mg/days
antag
Letrozole
with FSH
hCG
FSH 150 IU
10
Letrozole 5 mg/days
Letrozole 5 mg/day
12
Cycle days
of these women will achieve a pregnancy spontaneously. However, for others, fertility potential will be
reduced. In general, women who have been treated
with gonadotoxic therapy will experience diminished
ovarian reserve and menopause at earlier ages than
their peers.
Women with ovarian function, but diminished
ovarian reserve, can probably safely undergo ART and
can be reassured that, so far, recurrence rates are no
different when compared to women who elected not
to attempt pregnancy. Stimulation with standard protocols for IVF or novel protocols in patients with hormonally sensitive tumors can be undertaken. However,
the success rates in cancer survivors who received systemic chemotherapy or pelvic RT are not equivalent
to their age-matched peers without a history of cancer treatment. Despite advances, some will not achieve
genetic pregnancies. Some will choose to pursue adoption or child free living. Others may choose to pursue
pregnancy through oocyte donation.
315
Donor
Recipient
Screening
Medical/infectious disease
Medical
Psychological
Psychological/psycho-education
Genetic
Informed consent
Informed consent
Matching
Matching
Ovarian stimulation
Preparation/synchronization of endometrium
Retrieval of oocytes
Fertilization
Transfer
Recruitment of donors
Third-party reproduction is one of the most ethically complex aspects of reproductive health care. Even
societies with generally similar values, legislate donor
recruitment very differently [33]. Thus, some countries
have mandated anonymity (Spain, France, Belgium,
316
Genetic screening
By contrast, genetic testing of US donors is not legally
mandated. The appropriate professional society sets
the standard of care: ASRM recommends that all
donors be tested for cystic fibrosis (CF) mutations
and that additional testing be based on risk factors
identified by history. According to ASRM guidelines,
donors and their first-degree relatives should be free
of mendelian disorders, major malformations, significant familial diseases with a known genetic component and mental retardation of undocumented etiology [35].
Genetics is progressing rapidly and recommended
screening tests change very quickly. The American
College of Medical Genetics has published excellent
guidelines regarding ethnicity and population-based
genetic screening (http://www.acmg.net). Of course,
the absence of legal mandates does not decrease US
physicians responsibility to act in the best interests
of the donor, the recipient parents and the potential
child. Of note is that screening gamete donors is not
the same as preconceptual testing in women attempting pregnancy. The testing of young, often unmarried oocyte donors and the dissemination of information so acquired carry different ethical, medical and
317
318
Psychological screening
Psychological evaluation of the donor and the recipient and, when applicable, their families can be helpful
to all involved. Third-party reproduction is a complex
action with potential long-term repercussions. Participants have both the right and the responsibility to
explore the consequences and emotions that accompany these decisions. We can listen, explore feelings,
support and educate donors and recipients in a way
that helps all parties to make choices that are right for
them.
Donor
The donor should be free of significant psychiatric
pathology. The psychologist should assess current life
stressors, traumatic past events (including any unresolved history of abuse or neglect) and coping skills.
Psychosocial history should include such items as family history, interpersonal relationships, educational
background, employment history, sexual history and
any history of past or present substance abuse. Current and past prescribed psychoactive medication use
should also be assessed.
Informed consent will probably be formally
obtained at another point in the evaluation. However, the psychological interview provides a valuable
moment to ascertain that the prospective donor is
aware of the procedures involved and of their potential
risks.
The donor should be participating without coercion. If her donation is anonymous, we can give her
information about program precautions that are taken
to protect her anonymity. In addition, we want to
apprise her of the limits of anonymity in a complex
technological society and in a world in which values
and laws change rapidly [35].
When the donor is known, it is, perhaps, even more
important that situations of coercion be identified and
that the prospective known donor be supported by the
medical team if she is uncomfortable with egg donation. In family situations, in particular, and even more
so when the prospective recipient is a cancer survivor,
coercion can have many faces. It can be intergenerational, it can threaten family membership and it can
stir up long dormant feelings of anger and resentment
[44].
Even when the prospective donor is happy that she
can be of help, there are details to be discussed. Both
families should have discussed and come to agreement about disclosure (and timing of disclosure) to the
donor-conceived person(s) and to other family members, including genetic half siblings. The cancer prognosis should be openly and truthfully discussed among
family members. In cases where the prognosis is poor,
painful as it is to do, a discussion should be held about
how relationships might change were the recipient to
die.
Recipient
Psychological consultation can be very helpful for all
recipients of donated oocytes and is specifically recommended for recipients considering known donation
[44, 49]. These sessions are, at least partly, psychoeducation.
The psychologist and recipient family can discuss
feelings about pregnancy, positive and negative aspects
of disclosure and the family can be provided with a bibliography of childrens and adult books related to ovum
donation. Hopefully, any differences between the partners in attitudes towards disclosure can be resolved
well in advance of the birth of a child.
Additionally, the session offers the recipient and
her partner the opportunity to discuss their feelings about the loss of the hypothetical genetic child
that they might have made together. Couples often
carry many fears and fantasies about gamete donation,
including concerns that the donor will try to reclaim
the offspring or that the child will wish to seek out
his/her real mother. Most recipient mothers wonder
how they will feel carrying another womens genetic
child and whether they will bond to that child in utero,
after birth and throughout his or her lifetime. In situations where either blame or guilt has colored the process, these feelings, too, can be discussed. To the extent
that resentment of the process might later surface as
resentment towards the child, these conversations are
helpful steps towards building a healthy family.
In the case of recipients who are cancer survivors,
other issues and feelings may be layered onto the decision for DE. Anger and resentment about the cancer
and its treatment, seemingly resolved, may resurface.
And, while a pregnancy may restore confidence in the
319
320
hCG
Finding threshold
FSH
1 mg leuprolide acetate daily
OCP 25 days
Day 1
21
25
14
Retrieval
Figure 26.3 A dual suppression protocol used by us in 1986, still valid today. FSH, follicle stimulating hormone; hCG, human chorionic
C 1987 American Society for
gonadotropin; OCP, oral contraceptive pill. Reprinted with permission from Rosenwacks [59], courtesy of Elsevier,
Reproductive Medicine.
GnRH analogue
trigger
GnRH antag
FSH
Confirm LH
Surge
FSH + LH
Menses
Oocyte retrieval
Donor oocyte retrieval does not differ from oocyte
retrieval for IVF. Under conscious sedation, and using
a transvaginal ultrasound-guided needle, the follicles
are aspirated and the oocytes identified in the follicular fluid. Complications of the procedure include
infection and intraperitoneal bleeding, as well as anesthetic complications. All complications are rare and yet
potentially serious.
The incidence of pelvic infection following
transvaginal ultrasound-guided oocyte retrieval
ranges from 0 to 1.3% [67, 68]. In a review of 2670
oocyte retrieval procedures, Bennett et al. reported 18
cases resulting in infection (0.6%), 9 of which were
severe with pelvic abscess formation (0.3%) [56]. The
severity of infection can range from a minor infection
with pyrexia, leukocytosis and abdominal pain, to a
12 13 14
Retrieval
321
in exaggerated form, to the intraperitoneal bleeding that can occur with natural ovulation. Clinical
observations suggest that intraperitoneal bleeding, like
OHSS, occurs more commonly when more oocytes are
retrieved. This is physiologically plausible: the larger,
more highly vascularized ovaries and the increased
number of follicular punctures would likely predispose
to bleeding.
322
Estrace oral
Progesterone vaginal
Intramuscular progesterone
150
125
100
75
50
25
1
9 10 11 12 13 14
Progesterone (mg)
Estrace (mg)
15 16 17 18 19 20 21 22 23 24 25 26 27 28
Figure 26.5 A replacement protocol used by us in 1986, still valid today. Reproduced with permission from the American Society for
Reproductive Medicine.
0.2 mg/day as the donor begins her gonadotropin stimulation, and increase to 0.4 mg/day for approximately
4 days prior to the donors ovulation. During the luteal
phase, patches are decreased back to 0.2 mg/day [71).
Most patients tolerate the patches well although
redness or rash at the patch site may be seen in up to
1% of patients. As in the natural cycle (where follicular
phase length may vary considerably without affecting
fertility), there is considerable latitude in the length
of the estrogen-only phase of the replacement cycle
[72, 73]. This is fortunate; without this flexibility,
synchronization would be difficult.
Progesterone replacement, however, is a different matter. Its administration must be synchronized
closely to the donors cycle. The presence of adequate
endometrial levels of progesterone at the correct time
in the cycle is critical both for the preparation of the
endometrium for implantation and for the support of
the early embryo [73].
Progesterone can be administered either by the
vaginal route or as intramuscular injections of progesterone in oil [74]. Intramuscular progesterone has
the disadvantage of being irritating to muscle; patients
often experience pain at the injection site and occasionally develop fever and leukocytosis. On the other
hand, serum levels can be reliably used to monitor and
titrate the dose, which is not true of the better-tolerated
vaginal suppositories. (Oral micronized progesterone
is variably absorbed and is rarely used for recipient
replacement cycles.)
In our practice, we begin progesterone on the day
after the donor receives her ovulatory trigger at a dose
of 2 mg/day (im) or, more rarely, 300 mg/day (suppository). On the following day, we double the dose and
continue it either until the pregnancy test is negative
or, if positive, until the placenta takes over steroidogenesis, usually by 10 or 11 weeks of pregnancy [75].
If intramuscular progesterone is used, we monitor
serum levels and adjust the dose to keep the serum
concentration above 20 ng/ml. The estradiol dose is
titrated to yield a serum concentration between 100
and 200 pg/ml (Figure 26.6).
Conclusion
A young cancer patient may find significant psychological benefit in preserving oocytes or embryos prior
to embarking on a grueling course of chemotherapy.
This is true even if she ultimately does not need to
use the cryopreserved tissue. Many times, especially
if she is young, ovarian function will return following
treatment. However, even those survivors with regular
menses, may find that ovarian reserve and egg quality have been compromised. The cancer survivor with
regular menses needs to understand this and, after a
safe interval, be encouraged to attempt conception as
soon as she is ready. Physicians should discuss the
advantages of an abbreviated evaluation for a couple
unable to conceive within a few months. Often, effective intervention will shorten the time to pregnancy.
Sometimes, ART is the best initial approach.
When conception does not occur and when prevention has not been done, or has not been successful, a cancer survivor may consider pregnancy through
323
0.6
50
50 mg/day P4
0.4
0.4 mg/day E2
Estradiol
Progesterone
25
25 mg/day P4
0.2
0.2 mg/day E2
Progesterone im (mg)
Transdermal micronized E2
0.8
0.2 mg/day E2
0.1 mg/day E2
.1
0 2
28
10
12
14 15 16 18 22 23 24 26
hCG
Embryo transfer
BhCG
Aspiration
Progesterone day 15
324
References
1. Kavic SM and Sauer MV. Oocyte donation treats
infertility in survivors of malignancies: 10-year
experience. J Assist Reprod Genet 2001; 18(3): 1813.
2. Edgar A, Morris EMM, Kelnar CJH and Wallace WH.
Long-term follow-up of survivors of childhood cancer.
Endoc Dev 2009; 15: 15980.
3. Del Mastro L, Catzeddu T and Venturini M.
Infertility and pregnancy after breast cancer: current
knowledge and future perspectives. Cancer Treat Rev
2006; 32(6): 41722.
4. Meirow D, Epstein M, Lewis H, Nugent D and
Gosden RG. Administration of cyclophosphamide at
different stages of follicular maturation in mice: effects
on reproductive performance and fetal malformations.
Hum Reprod 2001; 16(4): 6327.
5. Hawkins MM. Pregnancy outcome and offspring after
childhood cancer. BMJ 1994; 309(6961): 1034.
6. Sanders JE, Hawley J, Levy W et al. Pregnancies
following high-dose cyclophosphamide with or
without high-dose busulfan or total-body irradiation
and bone marrow transplantation. Blood 1996; 87(7):
304552.
7. Larsen EC, Loft A, Holm K et al. Oocyte donation in
women cured of cancer with bone marrow
transplantation including total body irradiation in
adolescence. Hum Reprod 2000; 15(7): 15058.
8. Bath LE, Critchley HO, Chambers SE et al. Ovarian
and uterine characteristics after total body irradiation
in childhood and adolescence: response to sex steroid
325
326
327
Section 7
Chapter
27
Introduction
Fertility preservation is now recognized as the most
essential quality of life issue in young cancer survivors. Although several strategies to preserve fertility in women have been developed, most of them are
still experimental. Ovarian cryobanking, either freezing ovarian tissue or the whole ovary, is currently perceived as a promising technology for fertility preservation which draws enormous attention not only from
scientific communities but also from the general public.
Although still in its developmental stage, ovarian tissue cryopreservation followed by transplantation has proven to be successful in many animals.
Furthermore, we have been witnessing the successful
restoration of fertility after ovarian transplantation in
humans since 2004. To date, 14 healthy babies have
been born worldwide after transplantation of frozen
thawed ovarian tissue [17].
It is exhilarating to see the steady progress and
increasing enthusiasm for clinical applications of this
technology. However, ovarian cryopreservation and
transplantation should remain experimental until the
efficacy of this technology is proven. Indeed, there are
numerous technical and ethical issues that should be
resolved with this technology. In this chapter, three
urgent and critical problems involved with ovarian
tissue cryopreservation and transplantation (cryoinjury, ischemic tissue damage, cancer cell transmission) are discussed. In addition, the current status of
human ovarian tissue transplantation and whole ovary
transplantation by vascular anastomosis are briefly
addressed.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
328
Prize, reported the first ovarian transplantation by vascular anastomosis in cats. Since then, successful transplantation of the whole ovary with microanastomosis
of vascular pedicles has been reported in many animals including dogs, cats, rodents, rabbits, sheep and
primates [1113]. In 1987, Michel Leporrier in France
reported the successful heterotopic transplantation of
the whole ovary with vascular anastomosis before
pelvic irradiation to treat Hodgkins disease [14], the
first successful whole ovary transplantation (heterotopic) in humans. In 2009, the first baby was born
as a result of orthotopic transplantation of the intact
ovary by vascular anastomosis between monozygotic
twins [15]. Indeed, the surgical complexity of vascular anastomosis is no longer a barrier of human ovary
transplantation. The real challenge of vascular transplantation of the whole ovary is perfecting cryotechnology for organ cryopreservation. The first success in
restoring fertility after vascular transplantation of the
cryopreserved whole ovary was achieved in 2002 in
rats [16].
329
Ischemic-reperfusion injury
Cryoinjury
Cryopreservation of ovarian tissue by slow freezing
and rapid thawing is successful (5080% follicle survival rates), but current methods are still not perfect
and require further optimization to minimize the loss
of follicles and ovarian function. Two main mechanisms of cryoinjury are intracellular ice formation and
salt deposits. The most damaging phase (increased ice
formation and growth) of slow freezing is during cooling between 10 and 40 C, especially when the liquid phase is supercooled. However, significant cryoinjury can occur during the thawing (re-expansion)
phase because of changes in the composition of the
surrounding milieu, possibly mediated by temporary
leakage of the plasma membrane [18]. Indeed, the
thawing rate is important in maintaining cell viability.
Newton and Illingworth noticed the higher follicle survival and in-vitro maturation rates when samples were
thawed at 27 C rather than at 37 C in a mouse model
[19].
330
5C
Rapid cooling
Optimal
cooling
Slow cooling
<10C
Lethal
intracellular
ice formation
External ice
formation, no
intracellular
freezing
Excessive
dehydration,
high internal
and external
salt concentrations
(a)
(b)
sitate a short equilibration time to minimize the toxicity. Unlike individual cells, tissue requires a longer
exposure to high concentration of CPAs to reach
optimal CPA penetration. This is a dilemma for
vitrification of ovarian tissue. The antifreeze proteins
331
(a)
(b)
(c)
Figure 27.3 Protein expression patterns assessed by two-dimensional gel electrophoresis in: (a) fresh ovarian tissue; (b) slow frozenrapidly
thawed ovarian tissue; (c) vitrifiedwarmed ovarian tissue.
332
Ischemic injury
It has been proven that autotransplantation of frozen
thawed human ovarian tissue can restore endocrine
function as well as fertility. However, the follicular
loss in the grafted ovarian tissue is unacceptably high,
and it is mainly caused by tissue hypoxia after grafting while waiting for angiogenesis. In rodents, ovarian tissue slices become revascularized within 23 days
after grafting [24]. If the ischemic period is longer than
24 h, irreversible hypoxic tissue damage is unavoidable in the ovarian graft [25]. Primordial follicles are
more resistant to ischemia than growing follicle or cortical stromal cells. Nevertheless, most primordial follicles die of ischemia rather than of cryoinjury, and
only between 5 and 50% of follicles survive after grafting [26, 27]. The future of ovarian tissue transplantation depends on the development of new strategies to
To investigate the effect of exogenous administration of gonadotropin on angiogeneisis after ovarian grafting, we evaluated gene expression of angiogenic growth factors including HIF-1, VEGF, TGF
and angiopoietin-1, 2 and 3 in rat ovarian grafts 2, 7
and 30 days after transplantation. The mRNA expression of HIF-1 and angiopoietin-1 were low on day 2
but increased on days 7 and 30. Angiopoietin-2 and
3, VEGF and TGF were increased from day 2 and
peaked on day 30 after transplantation. When we compared the VEGF expression and apoptosis in the ovarian graft between the gonadotropin-treated and control groups, we did not find any added benefits of
exogenous gonadotropin for angiogensis or follicular
development in ovariectomized animals [30]. Nevertheless, a significant increase in the VEGF188 isoform
in the gonadotropin-treated group suggested the positive role of gonadotropin in the early stage of angiogensis in ovarian grafts. Of note, the mRNA expression
of VEGF188 increased on day 2 and returned to normal on days 7 and 30. On the other hand, VEGF164
and VEGF120 showed no changes on days 2 and 7 but
increased on day 30.
The optimal graft sites should be further determined to minimize ischemic damage and to improve
follicular survival after avascular ovarian grafting. It is
reasonable to expect better graft survival when ovarian cortical tissue is grafted to vascular sites, such as
muscle tissue or kidney capsule, rather than subcutaneous tissue. Furthermore, the importance of vascular smooth muscle cells and pericytes in sustaining vascular and tissue integrity after transplantation
has been demonstrated [31]. Perhaps, the secure strategy to prevent ischemic damage is using whole ovary
transplantation with vascular anastomosis. However,
cryopreservation of the whole human ovary along with
its vascular pedicles is a huge technical challenge.
333
334
Orthotopic autotransplantation
Although its efficacy should be further probed,
restoration of fertility by orthotopic autotransplantation has been demonstrated in humans as well as in
many animals. For orthotopic transplantation, ovarian tissue can be either transplanted onto the remaining ovary or into the peritoneal pocket of the fossa
ovarica. It appears that grafting ovarian tissue in or
onto the remaining ovary has advantages and more
likely results in natural conception, unless the size of
the ovary is too small as a result of atrophy [35]. In
the 5 years since the first report of a live birth in
2004, the total number of babies born after orthotopic transplantation of frozenthawed ovarian tissue
has increased to 11 (including unpublished data). It is
still too early to determine the efficacy of this technology, but the current data is encouraging: 6 live births
out of 12 pregnancies following 30 transplantations
and several ongoing pregnancies as of the end of 2008
[17]. Six pregnancies occurred spontaneously and five
by in vitro fertilization (IVF) and embryo transfer. In
the IVF group, 15 embryos (including 2 blastocysts)
were transferred, which resulted in 6 pregnancies
(2 miscarriages, 1 ectopic pregnancy and 3 live births).
The details of orthotopic autotransplantation of ovarian tissue is discussed in Chapter 29.
Heterotopic autotransplantation
Heterotopic autotransplantation is an attractive alternative to orthotopic autotransplantation as it can avoid
invasive procedures and make the recovery of oocytes
easy. In particular, it is a practical and cost-effective
technology when repeated transplantation is required,
because of the shortened life span of the ovarian grafts;
or a hostile pelvic environment due to previous radiation; or severe pelvic adhesions precluding orthotopic
transplantation. The duration of ovarian function
Ovarian transplantation
Oophorectomy
Tissue processing
Whole ovary
prepared
with vessels
FREEZE
(Slow freeze/vitrifit)
FREEZE
(Slow freeze/
vitrifit)
THAW
THAW
Autotransplantation
Or thotopic
Autotransplantation
Heterotopic
Xenotransplantation
In vitro culture
Heterotopic
with vascular
anastomosis
Orthotopic
with vascular
anastomosis
Figure 27.4 Theoretical strategies for oocyte maturation in cryopreserved ovarian tissue and intact ovary. To date, the only strategy that
produced live births is orthotopic autotransplantation of ovarian tissue. See plate section for color version.
after heterotopic transplantation of human ovarian tissue varies widely (between 3 and 60 months).
In theory, the optimal site should be rich in blood
supply, convenient to implant, easy for oocyte recovery
and mimic the surrounding environment of the ovary.
Various heterotopic sites have been tested, which
include subcutaneous tissue of the abdomen [36], forearm [37] or hip [23], rectus muscle [38], breast tissue [38], uterus [D. Nugent, unpublished data], as
well as subperitoneal tissue beneath the abdominal fascia [39]. Nevertheless, the optimal site for heterotopic
transplantation of human ovarian tissue is still elusive.
335
Advantages
Disadvantages
Non-invasive procedure
336
Figure 27.5 The process of heterotopic transplantation of frozenthawed human ovarian tissue to the space between the rectus muscle
and the rectus sheath. Reproduced with permission from Elsevier. See plate section for color version.
120
FSH (IU/l)
100
80
60
40
20
0
0
(a)
4
5
6
7
Months after transplant
10
11
12
Estradiol (pg/ml)
250
200
A
B
C
150
100
50
0
0
(b)
4
5
6
7
Months after transplant
environment of heterotopic sites may not be as favorable for normal follicle development.
Xenotransplantation
Numerous animal and human studies support the
value of xenotransplantation of ovarian tissue as a
strategy to preserve fertility and to conserve rare and
endangered species. It has already been demonstrated
that xenotranplantation of ovarian tissue from cat,
wombat, sheep, African elephant, monkey and human
to immunodeficient mice can support follicular development up to the antral stage [23]. Furthermore, generation of live young from xenografted rodent ovaries
proved that it is a valid technology for animal conservation as well as fertility preservation [42].
10
11
12
Historically, xenotransplantation of human ovarian tissue was explored as a strategy to restore fertility in cancer patients with a high risk of cancer cell
re-introduction with autotransplantation. Successful
follicular development, ovulation and corpus luteum
formation in the human ovarian tissue xenografted
after cryopreservation have been demonstrated [43,
44]. Furthermore, Kim et al. succeeded in retrieving human oocytes that had developed in the frozen
thawed ovarian tissue xenografted to SCID mice [45].
All host animals were stimulated with gonadotropin
from 20 weeks after xenografting. Grafts were recovered 36 h after human chorionic gonadotropin (hCG)
administration. Fifty-seven grafts from 30 animals
(total 60 grafts) were recovered, and 12 oocytes were
collected from 26 antral follicles (size larger than
337
Angiogenesis
Oocyte quality
Steroidogenesis
338
Advantages
Disadvantages
Immediate blood
supply to the graft
Minimizing tissue
ischemia
Potential long-term
ovarian function
Ischemia-reperfusion injury
Thromboembolism
Increased morbidity(and
mortality)
Potential risks of cancer
recurrence (especially with
minimal residual disease in
ovarian medulla)
Conclusions
Ovarian tissue cryobanking has rapidly become a
promising strategy for fertility preservation in cancer patients, and its use will extend beyond women
with cancer. Although healthy babies have been born
with orthotopic autotransplantation of frozenthawed
ovarian tissue worldwide, its clinical efficacy and practicability for fertility preservation is still unclear. There
are many unresolved issues with ovarian tissue cryopreservation and transplantation, such as cryoinjury,
ischemic injury, risks of cancer cell transmission and
its efficacy. Heterotopic autotransplantation may be
an alternative method to restore fertility with use of
References
1. Donnez J, Dolmans MM, Demylle D et al. Livebirth
after orthotopic transplantation of cryopreserved
ovarian tissue. Lancet 2004; 364: 140510.
2. Meirow D, Levron J, Eldar-Geva T et al. Pregnancy
after transplantation of cryopreserved ovarian tissue in
a patient with ovarian failure after chemotherapy. N
Engl J Med 2005; 353: 31821.
3. Demeestere I, Simon P, Emiliani S, Delbaere A and
Englert Y. Fertility preservation: successful
transplantation of cryopreserved ovarian tissue in a
young patient previously treated for Hodgkins disease.
Oncologist 2007; 12: 143742.
4. Andersen CY, Rosendahl M, Byskov AG et al. Two
successful pregnancies following autotransplantation
of frozen/thawed ovarian tissue. Hum Reprod 2008; 23:
226672.
5. Roux C, Amiot C, Agnani G et al. Live birth after
ovarian tissue autograft in a patient with sickle cell
disease treated by allogenic bone marrow
transplantation. Fertil Steril 2010; 93: 2413.e1519.
6. Sanchez-Serrano M, Crespo J, Mirabet V et al. Twins
born after transplantation of ovarian cortical tissue
and oocyte vitrification IVF outcome in patients with
orthotopically transplanted ovarian tissue. Fertil Steril
2009; 93: 268.e1113.
7. Silber SJ, DeRosa M, Pineda J et al. A series of
monozygotic twins discordant for ovarian failure:
ovary transplantation (cortical versus microvascular)
and cryopreservation. Hum Reprod 2008; 23:
15317.
339
340
45. Kim SS, Kang HG, Kim NH, Lee HC and Lee HH.
Assessment of the integrity of human oocytes retrieved
from cryopreserved ovarian tissue after
xenotransplantation. Hum Reprod 2005; 20: 25028.
341
Section 7
Chapter
28
Introduction
Early detection and aggressive chemotherapy/
radiotherapy treatments have improved the long-term
survival rates for many young women with various
types of cancer. As a consequence of these cytotoxic
treatments, their reproductive future can be either
short lived or eradicated. For young single women
with cancer, oocyte cryopreservation offers the best
potential option for achieving a future pregnancy
using their own gametes. Unfortunately, the urgent
need to commence cytotoxic treatment often does not
permit adequate time for cryopreservation of mature
oocytes. Conversely, cryopreservation of ovarian
tissue eliminates the delay necessary to obtain mature
oocytes, but the subsequent potential for establishing
pregnancy is currently unknown. Although ovarian
tissue cryopreservation is an attractive alternative and
frequently used for patients with these conditions,
little has been published on the efficacy of various
protocols. Cryopreservation of ovarian tissue is more
complex than that of gametes or embryos, requiring preservation of multiple cell types, which may
vary in volume and water permeability. Essentially,
ovarian tissue cryopreservation is more similar to
organ cryopreservation than to that of gametes or
embryos.
Early attempts to cryopreserve mouse ovarian tissue had shown the potential of this technology with
evidence of intact follicles in cryopreserved tissue
after grafting [1] and subsequent litters from grafts
of cryopreserved tissue [2]. A resurgence of interest in ovarian tissue cryopreservation stemmed from
the birth of a lamb following cryopreservation and
grafting of ovarian tissue reported by Gosden et al.
in 1994 [3]. This success has now been translated
into the human with the demonstration of resump-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
342
1.5 M EG
+0.1 M sucrose
1.5 M DMSO
+0.1 M sucrose
1.5 M DMSO
+0.1 M sucrose
1.5 M DMSO
+0.2 M sucrose
1.5 M DMSO
+0.1 M sucrose
1.5 M PROH
+0.1 M sucrose
551
551
and
10 10 1
552
10 10 1
NP
321
4 C
4 C
4 C
22 C
4 C
30 min
4 C
30 min
22 C
30 or 90 min
NP
NP
0 C
4 C
30 min
4 C
30 min
4 C
30 min
4 C
30 min
Start temp.
7 C
7 C
9 C
7 C
7 C
9 C
7 C
Seed temp.
First-thaw
solution
1.5 M DMSO
1.0 MDMSO
+ 0.1 M sucrose
0.75 M EG
+ 0.25 M sucrose
37 C
2 min
Air 30 s then 37 C
2 min
35 C
23 min
37 C
Thaw temp.
14 oocytes
10 MII
2 2 cell ET
2 6 cell ET
10 oocytes
9 embryos
4 blastocysts
2 blastocysts ET
20 oocytes
6 MII
1 7 cell (no ET)
2 oocytes
1 3 cell ET
6 oocytes
4 2 PN
1 6 cell
1 3 cell
1 2 cell (no ET)
3 oocytes
2 MII
1 4 cell ET
1 5 cell ET
20 oocytes
8 MII
2 2 PN
1 3 cell
1 4 cell ET
Outcome
All cryopreserved using controlled rate slow freezing at 2 C/min to seed temperature followed by 0.3 C/min to 40 C or 50 C.
DMSO, dimethyl sulfoxide; EG, ethylene glycol; ET, embryo transfer; MII, metaphase-II oocyte; NP, not provided; PN, pronuclei; PROH, propanediol.
1.5 M DMSO
+0.1 M sucrose
Cryoprotectants Dehydration
551
and
15 5 2
Size of tissue
slices (mm)
Table 28.1 Cryopreservation methodology used in clinical cases where oocytes and embryos have been derived from heterotopic grafts
C. J. Stern,
personal
communication
Demeestere et al.
[7]
Rosendahl et al.
[6]
Schmidt et al. [10]
Ref.
Typea
Nameb
Characteristic
Follicle diameter
(m)
Population
(%)
Primordial
3540
4156
B/C
Transitory/ intermediary
primordial
3744
4122
Primary
4654
1521
C/D
Proliferating
6077
Secondary
100
Antral
a
b
13
<1
(a)
(b)
(c)
(d)
(e)
(f)
344
(Figure 28.2a) embedded in a dense cortex of stromal cells and collagen bundles. At birth, it is estimated
that between 250 000 and 500 000 primordial follicles
[12] are present within the human ovary, decreasing
(a)
(b)
to approximately 25 000 by the age of 37 [18]. Obviously these are the candidate follicles to preserve with
any cryopreservation regimen. The aim of cryopreservation is not only to preserve the structure and function of both the pre-granulosa cells and oocyte within
individual primordial follicles but also to preserve the
majority of the primordial follicles. This is complicated by the enormous variation in the density of
the ovarian cortex and the distribution of follicles
between patients. Histological examination of ovarian tissue destined for cryopreservation from over 150
patients, ranging in age from the early teens to over
40 years of age [D. A. Gook, unpublished observations] shows that the cortex of the adult human ovary
is extremely fibrous with highly variable distribution
of follicles [19]. In the young teenage ovary, abundant follicles appear to be evenly distributed around
the cortex (Figure 28.2a). However, in ovaries from
women of more advanced reproductive age, follicles
are depleted from specific regions, altering the appearance to that of follicle clusters with fewer individual
follicles which are, in turn, becoming more sparsely
distributed (Figure 28.2b). This lack of uniformity
across the cortex will impact not only on the evaluation
of methodology but also the potential clinical success
in older women requesting ovarian tissue cryopreservation.
Methodology
Preparation of ovarian tissue for
cryopreservation
The most significant loss of follicles results not from
cryopreservation but from ischemia; 65% of sheep follicles are lost following exposure to 0 C for 23 h in a
cell culture medium (Leibovitz L-15) and only a further 7% as a result of cryopreservation [20]. Under the
345
(a)
(b)
Tissue dissection
Obviously, incubation at 37 C in an in vitro fertilization (IVF) medium is also beneficial for the collection of oocyte cumulus complexes (OCC) from
antral follicles during tissue preparation. Generally,
two types of OCC are recovered: those with very few
layers of corona cells, frequently with corona cells in
the perivitelline space (an indicator of atresia); and
those with a dense compact mass of corona cells in
which only a vague outline of the oocyte can be seen.
Our experience suggests that only a minor proportion of these oocytes will mature in culture and this
may take as long as 72 h, at which stage they can
be denuded and cryopreserved. In 2004, Isachenko
346
(c)
Cryopreservation
In contrast to cryopreservation of gametes and
embryos, ovarian tissue cryopreservation is more
closely related to organ cryopreservation [45]. The
underlying problem is that the optimal cryopreservation procedure for one cell type may not be optimal
for other cells within the tissue. Therefore, a compromise may be necessary to preserve a high proportion
of all cell types present in the tissue. There is obviously disparity of cell size between the stromal cells,
the oocyte and the pre-granulosa cells within the primordial follicle, which will limit the ability to optimize the cryopreservation procedure for human ovar-
Cryoprotectants
Cellular density and tissue geometry will affect infiltration of cryoprotectants. Diffusion of cryoprotectants
is relatively rapid in murine ovaries, which consist
almost completely of abutting developing follicles with
almost no fibrotic material and are therefore relatively
porous. However, the densely fibrotic human ovarian
cortex is extremely different. Murine ovarian tissue is
therefore an unsuitable model for the human in this
respect.
In the case of permeating cryoprotectants, the aim
is to gradually displace cellular water without inducing
excessive shrinkage. To overcome the difficulties associated with the density of human ovarian tissue, the use
of higher concentrations of cryoprotectants has been
suggested. However, under these circumstances, the
reduced aqueous phase and hyperosmotic conditions
result in excessive shrinkage of cells and loss of cell
cell communication [48]. This, in turn may result in
subsequent impairment of tissue function even though
morphology may appear normal. The reduction in filamentous actin traversing the zona of oocyte cumulus complexes isolated from cryopreserved murine
ovaries indicates that this may occur in ovarian
tissue [49].
Four permeating cryoprotectants: glycerol (GLY),
dimethyl sulfoxide (DMSO), ethylene glycol (EG) and
propanediol (propylene glycol; PROH) have been used
in human and animal ovarian tissue cryopreservation. For all permeating cryoprotectants, a concentration of 1.4 or 1.5 mol/l has generally been used for
slices of human and animal ovarian tissue [44]. However, higher concentrations of PROH (2 and 4 mol/l at
ambient temperature) do not appear to cause follicular toxicity when compared to control tissue exposed
347
348
insufficient dehydration may have resulted from suboptimal conditions for a specific cryoprotectant and
that, with optimization, a variety of protocols may be
appropriate.
Non-permeating
cryoprotectants,
generally
sucrose at a concentration of 0.1 mol/l, have also
been used to facilitate dehydration of ovarian tissue
[11, 13, 5861]. Recent studies have shown that
increasing the sucrose concentration significantly
improves outcomes for embryo cryopreservation [62]
but addition of various concentrations of sucrose in
combination with DMSO did not appear to result in
improved cryopreservation [55] and no other studies
have specifically compared protocols with and without
sucrose.
The use of sucrose with the permeating cryoprotectant PROH [13] has been investigated in an attempt
to optimize dehydration of both pre-granulosa cells
and oocytes within primordial follicles [63]. Equivalent proportions of morphologically intact oocytes
could be achieved with shorter time exposure when
the sucrose concentration was increased from 0.1
to 0.2 mol/l. However, extended exposure to higher
sucrose resulted in decreased oocyte survival [63]
and the observation, within the surviving oocytes,
that the majority of cytoplasm consisted of vacuolation and lysed mitochondria. This phenomena was
not observed with the lower sucrose concentration
(0.1 mol/l) which, in contrast, resulted in an improvement in the proportion of morphologically intact
oocytes and pre-granulosa cells with time. The proportion of surviving oocytes with normal cytoplasm also
increased with time in 0.1 mol/l sucrose [13]. Extended
exposure to the elevated sucrose concentration may
result in inappropriate osmotic gradients which is also
the likely explanation for the low survival of pregranulosa cells (Figure 28.4b) and poor cytoplasmic
morphology in oocytes when applying two-step compared to single-step dehydration with equivalent final
cryoprotectant concentrations [13]. By manipulation
of dehydration using PROH and sucrose at ambient
temperature, a high proportion of intact pre-granulosa
cells (74%), intact oocytes (91%) and oocytes with normal cytoplasmic appearance (95%) could be achieved
(Figure 28.4c). However, morphology of the stromal
tissue was consistently poor irrespective of the regimen (Figure 28.4b,c) compared to non-cryopreserved
(Figure 28.4a), highlighting the relative complexity
associated with dehydration of tissue containing multiple cell types.
Rate of cooling
(a)
(b)
(c)
In the majority of the above studies the rate of cooling used has been similar to the rates used for controlled rate embryo cryopreservation (2 C/min to ice
seeding temperature followed by 0.3 C/min). As with
embryo and oocyte cryopreservation, the rate of cooling for ovarian tissue has never been systematically
optimized. Mazur estimated that the theoretical rate
of cooling for organs or tissues should be 1 C/min
[45], but a comparison of the standard slow rate
(0.3 C/min) and a faster cooling rate (2 C/min) with
large pieces (200 mm3 ) of ovine ovarian tissue demonstrated a significant improvement in follicular survival
at the faster rate [50]. In an attempt to investigate this
for human ovarian tissue, the slow rate (0.3 C/min)
was compared to two faster cooling rates following
the same prefreeze dehydration and the morphology
of each cell type was determined [13]. Poor morphology of the whole tissue was observed with a rapid rate
(Figure 28.5a; to be discussed in more detail later in
the context of vitrification). At an intermediate rate
(36 C/min), a high proportion of the stromal cells
and collagen bundles appeared normal but only half
the oocytes were intact and almost all pre-granulosa
cells were abnormal (swollen and with swollen nuclei;
Figure 28.5b), again emphasizing the importance of
variation in cell size. With the slow rate, approximately
half of both the stromal and pre-granulosa cells and
over 80% of the oocytes were normal. Normal appearance in the pre-granulosa cells and oocytes could be
further enhanced by applying the slow rate after more
extensive dehydration prior to cryopreservation but
this was at the expense of stromal cell survival (Figure
28.4c) [63, 64].
During controlled rate cryopreservation, cryoprotectant crystallization will occur, and the temperature
at which this occurs is specific for the cryoprotectant.
For PROH or DMSO this occurs at 6 to 8 C. Without manual seeding, crystallization will be initiated at
any solid surface i.e. throughout the tissue. Demirci
et al. [50] reported a dramatic deviation from the normal cooling curve resulting in reduced follicular survival in the absence of manual seeding. Manual seeding at a slightly higher temperature (5 compared to
7 C) appeared to improve follicle survival [54].
Similarly, damage can occur during thawing as
a result of crystallization of water or cryoprotectant. Irrespective of the cryopreservation method used
(controlled rate or vitrification), thawing should be as
349
(a)
(b)
rapid as possible. Although critical, the thawing temperature is rarely reported and, again, almost no studies have systematically investigated this aspect of cryopreservation with animal or human ovarian tissue.
Exposure to some cryoprotectants, such as DMSO,
at higher temperatures may increase their toxicity. A
comparison of thawing of murine ovarian tissue, cryopreserved in DMSO, at 27, 37 or 47 C showed a significant reduction in follicle survival at the higher temperatures [54]. In our experience, thawing tissue cryopreserved in PROH in a 1 ml volume in a Cryovial
requires 3 min at 37 C to achieve liquefaction prior to
immediate removal of the tissue. At this temperature,
at least in the case of PROH, there appears to be no
affect on developmental potential (see later).
Vitrification offers the potential benefit of overcoming many of the issues associated with crystallization discussed above. The problem associated with vitrification of ovarian tissue, however, is how to achieve
the high cooling and warming rates required. Vitrification of murine ovaries has been successful with
subsequent births of pups reported [65, 66] following vitrification in cryostraws and plunging in liquid nitrogen. However, when a faster cooling rate was
achieved by direct contact with liquid nitrogen, it
resulted in better preservation as evidenced by significantly more morphologically normal, viable follicles
and pups [65]. This improvement, however, may also
be partly due to more appropriate dehydration prior
to vitrification. Vitrification in cryostraws resulted in
poorer outcomes relative to controlled rate cryopreservation for all parameters measured. Wang et al. [67]
have also shown the importance of the cooling rate
for vitrification with both mouse and human ovarian
tissue.
Ovine hemi-ovaries have also been successfully vitrified resulting in live births although one of the four
350
Storage
Irrespective of the methodology used for cryopreservation, tissue has generally been stored in Cryovials,
which do not constitute a fully sealed system. A
follicle integrity [75], although this has allowed detection of abnormalities such as oocyte shrinkage [76],
vacuolated areas within the oocyte cytoplasm [77], loss
of mitochondrial cristae [78] and lysis of pre-granulosa
cells [79].
There are only two morphometric studies of
human ovarian tissue which assess cryopreservation
[13, 74], one of which is an evaluation of the most
commonly used procedure [3] using DMSO as a cryoprotectant and controlled rate cooling on tissue from
six patients [74]. In this study, almost half of the follicles and the vast majority of oocytes (81%) were damaged. Parallel assessment of apoptosis confirmed these
observations. The other study assessed the proportion of intact pre-granulosa cells and oocytes together
with the relative normality of the oocyte cytoplasm
as estimated by vacoulation and normal mitochondria within every oocyte for a range of cryopreservation procedures using PROH and sucrose as cryoprotectants [13, 63]. Observations were verified by electron microscopic evaluation of a small number of follicles (Figure 28.6a,b). The highest proportion of both
oocytes (85%) and pre-granulosa cells (74%) with normal morphology was observed following dehydration
for 90 min in 1.5 M PROH + 0.1 M sucrose at ambient temperature followed by a slow controlled rate of
freezing. Although this type of morphometric assessment has provided evidence of morphological normality after cryopreservation, it gives no indication of viability or function.
Viability staining has also been used to assess follicles within a piece of tissue [80, 81] following cryopreservation. Generally, this staining will identify live
cells on the basis of an intact membrane and cytoplasm
which is functionally capable of cleaving a chromagen,
and dead cells on the basis of their inability to exclude
a nuclear stain [51, 57]. Again, this form of assessment has limitations [76]. Tissue pieces must be very
small to facilitate diffusion of dye and only follicles
with a live oocyte are detected. Although the nuclear
stain will detect the germinal vesicles (GV) of lysed
oocytes, these are indistinguishable from the nuclei of
stromal or pre-granulosa cells, resulting in the potential for overestimation of follicle viability. An example
of a lysed oocyte which would be unlikely to be distinguished from a granulosa cell is shown in Figure 28.5a.
Finally, it is important to remember that we cannot
infer, on the basis of viability staining, that the developmental potential of these primordial follicles has been
retained following cryopreservation.
351
(b)
Developmental potential
Assessment of expression of developmental potential in vitro is an attractive possibility, but requires
an understanding of the requirements for initiation
of growth in primordial follicles. Additionally, many
months of culture may be required. More advanced follicles isolated at the secondary stage, in which initiation of granulosa cell proliferation has occurred, can
be successfully grown in culture [8284]. However, the
predominant follicle present in the human ovary is the
primordial. There is evidence, that murine primordial
follicles, grown in a two-phase culture system, can produce live pups [85, 86], but this has not been established using cryopreserved ovarian tissue. The culture
of isolated primordial follicles, whether from fresh or
cryopreserved tissue, has proved problematic in animal models [8789] and in human [59, 90, 91]. However, follicular development to the secondary stage
has been established by culturing primordial follicles
within stromal tissue [92, 93]. More recently, development from primordial to early antral stage in vitro
has been demonstrated using a two-stage culture system with fresh tissue [35], and there is a preliminary
report of this approach being used to demonstrate
developmental potential of human primordial follicles
following vitrification [E. E. Telfer, unpublished data].
Results to date on demonstration of developmental
potential using in-vitro systems, while promising, are
limited to relatively few follicles.
As a result of the difficulties associated with culture of primordial follicles in vitro, various in vivo
approaches involving grafting of cryopreserved ovarian tissue, either alone or in combination with final
maturation in vitro, have been applied. Clear evidence
of preservation of developmental potential has been
established by the birth of live offspring from primordial follicles following grafting of ovine and murine
352
Refrences
1. Green SH, Smith AU and Zuckerman S. The numbers
of oocytes in ovarian autografts after freezing and
thawing. J Endocrinol 1956; 13: 3304.
2. Parrott DMV. The fertility of mice with orthotopic
ovarian grafts derived from frozen tissue. J Reprod
Fertil 1960; 1: 23041.
3. Gosden RG, Baird DT, Wade JC et al. Restoration of
fertility to oophorectomized sheep by ovarian
autografts stored at 196 C. Hum Reprod 1994; 9(4):
597603.
4. Oktay K, Buyuk E, Veeck L et al. Embryo development
after heterotopic transplantation of cryopreserved
ovarian tissue. Lancet 2004; 363(9412): 83740.
5. Poirot C, Piver P, Fayet P et al. Human embryo
development after subcutaneous autograft of
cryopreserved ovarian tissue. Hum Reprod 2006; 21
(Suppl. 1): i155 [AbstrP404].
6. Rosendahl M, Loft A, Byskov AG et al. Biochemical
pregnancy after fertilization of an oocyte aspirated
from a heterotopic autotransplant of cryopreserved
ovarian tissue: case report. Hum Reprod 2006; 21(8):
20069.
7. Demeestere I, Simon P, Emiliani S et al. Ongoing
pregnancy after a second cryopreserved ovarian
transplantation procedure. Hum Reprod 2007;
22(Suppl. 1): i434.
8. Kim SS, Lee WS, Chung MK et al. Long-term ovarian
function and fertility after heterotopic
autotransplantation of cryobanked human ovarian
tissue: 8-year experience in cancer patients. Fertil Steril
2009; 91(6): 234954.
9. Piver P, Amiot C, Agnani G et al. Two pregnancies
obtained after a new technique of autotransplantation
of cryopreserved ovarian tissue. Hum Reprod 2009; 24
(Suppl. 1): i15.
10. Schmidt KL, Yding Andersen C, Loft A et al.
Follow-up of ovarian function post-chemotherapy
following ovarian cryopreservation and
transplantation. Hum Reprod 2005; 20(12): 3539
46.
11. Poirot C, Vacher-Lavenu MC, Helardot P et al.
Human ovarian tissue cryopreservation: indications
and feasibility. Hum Reprod 2002; 17(6): 144752.
12. Gougeon A, Echochard R and Thalabard JC.
Age-related changes of the population of human
ovarian follicles: increase in the disappearance rate of
non-growing and early-growing follicles in aging
women. Biol Reprod 1994; 50(3): 65363.
13. Gook DA, Edgar DH and Stern C. Effect of cooling
rate and dehydration regimen on the histological
appearance of human ovarian cortex following
353
354
355
356
Section 7
Chapter
29
Introduction
According to previous reports, around 700 000 new
cancer cases are expected every year among the female
population of the USA [1] and 8% of these women are
likely to be under the age of 40. In fact, it is now estimated that 1 in every 250 people in the adult population is a childhood cancer survivor [2].
Advances in the diagnosis and treatment of
childhood, adolescent and adult cancer have greatly
increased the life expectancy of premenopausal
women with the disease. Indeed, aggressive chemotherapy and radiotherapy, as well as bone marrow
transplantation, can cure more than 90% of girls
affected by childhood malignancies [3], but have
resulted in a growing population of adolescent and
adult long-term survivors of childhood cancer [2] who
may experience infertility problems due to induced
premature ovarian failure (POF).
Autotransplantation of cryopreserved
human ovarian tissue
There have been numerous reported cases of autotransplantation of cryopreserved ovarian tissue, either
to an orthotopic or heterotopic site [6, 924].
Orthotopic autotransplantation of
cryopreserved human ovarian tissue
In theory, natural pregnancy may be achieved via
orthotopic tissue transplantation if the fallopian tubes
remain intact.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
357
A. Malignant
(a) Extrapelvic diseases
Bone cancer (osteosarcoma, Ewings sarcoma)
Breast cancer
Melanoma
Neuroblastoma
Bowel malignancy
B. Non-malignant
(a) Uni/bilateral oophorectomy
Benign ovarian tumors
Severe and recurrent endometriosis
BRCA-1 or BRCA-2 mutation carriers
Technique
Ovarian biopsy and freezing
Ovarian tissue cryopreservation is undertaken before
chemotherapy. Written informed consent is obtained
358
Cancer diagnosis
Pregnancy
Uterus
Cryopreservation
of tissue
samples
Cancer treatment
Fallopian
tubes
Ovary
Vagina
Complete remission
Grafting or placing
of fragments in the
perinatal cavity
Thawing of tissue
samples
LE SOIR - 10.12.09
Figure 29.1 Illustration describing the different steps from biopsy to re-implantation and pregnancy. See plate section for color version.
Thawing [16]
For thawing, the Cryovials are removed from the liquid nitrogen and left at room temperature for 2 min.
Thawing is subsequently completed by immersing the
Cryovials in a warm (37 C) water bath for 2 min. The
tissue samples are then grasped with small forceps and
placed in a Petri dish containing L-15 medium, which
is replaced 3 times (5 min each wash) to remove the
cryoprotectant completely.
359
Fallopian
tube
Creation of
peritoneal window
Atrophic
ovary
Fimbria
Reimplantation of ovarian
graft in peritoneal window
(a)
(b)
Figure 29.2 (a) Creation of a peritoneal window close to the atrophic ovary. (b) Transplantation of ovarian cortex pieces into the peritoneal
window. See plate section for color version.
360
(a)
(b)
oophorectomy for a non-malignant disease [11]. Follicular development was demonstrated only once by
ultrasonography 15 weeks after transplantation. The
reason why this patient had undergone oophorectomy
and re-implantation is not clear and no further data
were available.
Radford et al. reported a patient with a history
of Hodgkins disease treated by chemotherapy [13].
Ovarian tissue had been cryopreserved before receiving high doses of chemotherapy for a third recurrence
of disease. Histological section of the ovarian cortical tissue revealed only a few primordial follicles due
to the previous chemotherapy. Eight months after reimplantation, estradiol was detected and the follicle
stimulating hormone (FSH) and luteinizing hormone
(LH) levels decreased. One month later, LH and FSH
concentrations returned definitively to menopausal
levels.
361
(a)
(b)
362
In 2003, once the patient had been declared completely disease-free, re-implantation was carried out
in orthotopic sites (see Donnez et al. for techniques
[6, 16]). From 5 to 9 months after re-implantation,
concentrations of FSH, 17 -estradiol and progesterone showed the occurrence of ovulatory cycles. At
11 months, the patient became pregnant and subsequently delivered a healthy baby. This birth was
announced in the Lancet in September 2004 [16].
In 2005, Meirow et al. also published details of
a live birth after orthotopic autotransplantation of
cryopreserved ovarian tissue in a patient with POF
after chemotherapy [17]. Eight months after orthotopic transplantation, the patient spontaneously menstruated. Nine months after transplantation, she experienced a second spontaneous menstrual period. After
a modified natural cycle, a single mature oocyte was
retrieved and fertilized. Two days later, a four-cell
embryo was transferred. The patient became pregnant from this embryo transfer and delivered a healthy
infant weighing 3000 g.
Demeestere et al. reported a pregnancy after natural conception in a woman who had undergone
orthotopic and heterotopic transplantation of cryopreserved ovarian tissue [19]. Unfortunately, this pregnancy, obtained by natural conception, ended in miscarriage at 7 weeks due to aneuploidy. The same team
performed a second re-implantation to an orthotopic
site in the same patient after cessation of graft secretion was evidenced [20]. The patient became pregnant
and delivered a healthy baby. She recently delivered a
second healthy child.
Silber et al. reported a pregnancy following reimplantation of cryopreserved ovarian tissue between
monozygotic twins [21]. It should be noted, however,
that the same woman had already delivered a first
healthy baby after re-implantation of fresh tissue [21].
Andersen et al. described a series of six orthotopic re-implantations of cryopreserved ovarian cortex. In this series, two women became pregnant and
delivered healthy infants [22]. Single mature oocytes
were retrieved during optimized cycles, fertilized and
transferred on day 3. One of the two women was
later naturally pregnant and delivered a second healthy
child.
In 2009, Piver et al. reported the birth of a
healthy baby after orthotopic re-implantation of cryopreserved tissue [31]. The patient became pregnant
naturally. Very recently, Sanchez-Serrano reported a
twin pregnancy after an in vitro fertilization (IVF)
180
160
140
UI/ml
120
100
Mean FSH
Mean estradiol
80
60
40
20
0
0
2
3
4
Months post-transplantation
363
364
Conclusion
Approximately one third of young women exposed
to chemotherapy develop ovarian failure. In 2010, we
believe it is our ethical responsibility to propose cryopreservation of ovarian tissue to all adolescents and
young women under Institutional Review Board (IRB)
protocols having to undergo chemotherapy with alkylating agents.
In conclusion, the 11 live births obtained after
transplantation of frozenthawed ovarian tissue in
humans give hope to young cancer patients, but there
is still much work to be done. Research programs
need to determine whether active angiogenesis can be
induced to accelerate the process of neovascularization in grafted tissue, if isolated human follicles can be
grafted or, indeed, if microvascular re-anatomosis of
an entire cryopreserved ovary is a valuable option.
References
1. Jemal A, Siegel R, Ward E et al. Cancer statistics, 2006.
CA Cancer J Clin 2006; 56: 10630.
2. Blatt J. Pregnancy outcome in long-term survivors of
childhood cancer. Med Pediatr Oncon 1999; 33: 2933.
3. Ries LAG, Percy CL and Bunin GR. Introduction In:
Ries LAG, Smith MA, Gurney JG et al (eds.), Cancer
Incidence and Survival Among Children and
Adolescents: United States SEER Program 19751995.
Bethesda MD: National Cancer Institute, 1999: pp.
115.
4. Donnez J and Bassil S. Indications for
cryopreservation of ovarian tissue. Hum Reprod
Update 1998; 4: 24859.
5. Donnez J, Martinez-Madrid B, Jadoul P et al.
Ovarian tissue cryopreservation and transplantation: a
review. Hum Reprod Update 2006; 12: 51935.
6. Donnez J, Dolmans MM, Demylle D et al.
Restoration of ovarian function after orthotopic
(intraovarian and periovarian) transplantation of
cryopreserved ovarian tissue in a woman treated by
bone marrrow transplantation for sickle cell anaemia:
case report. Hum Reprod 2006; 21: 1838.
7. Slavin S, Nagler A, Aker M et al. Non-myeloablative
stem cell transplantation and donor lymphocyte
infusion for the treatment of cancer and
life-threatening non-malignant disorders. Rev Clin
Exp Hematol 2001; 5: 13546.
8. Mattle V, Behringer K, Engert A and Wildt L. Female
fertility after cytotoxic therapy-protection of ovarian
function during chemotherapy of malignant and
non-malignant diseases. Eur J Haematol 2005; 75:
7782.
9. Wolner-Hanssen P, Hagglund L, Ploman F et al.
Autotransplantation of cryopreserved ovarian tissue to
the right forearm 41/2 years after autologous stem cell
transplantation. Acta Obstet Gynecol Scand 2005; 84:
6958.
10. Donnez J, Squifflet J, Van Eyck AS et al. Restoration
of ovarian function in orthotopically transplanted
cryopreserved ovarian tissue: a pilot experience.
Reprod Biomed Online 2008; 16(5): 694704.
11. Oktay K and Karlikaya G. Ovarian function after
transplantation of frozen, banked autologous ovarian
tissue. N Engl J Med 2000; 342: 1919.
365
366
Figure 1.6 Patrick Steptoe and Robert Edwards at the birth of the worlds first in-vitro
fertilization conceived baby Louise Brown on July 25, 1978. Courtesy of Bourn Hall Clinic.
10 000 000
1 000 000
n =325
r 2 =0.81
100 000
Observed values
WallaceKelsey model
Lower 95% Cl for model
Upper 95% Cl for model
Lower 95% prediction limit
Upper 95% prediction limit
10 000
1000
100
10
5
0
5
10
15
20
25
30
35
40
45
50
Age (in months from conception to birth; in years from birth to menopause)
Figure 2.1 The best model for the establishment of the non-growing follicle (NGF)
population after conception and the subsequent decline until age at menopause is described
by an ADC model. The model has a correlation coefficient r2 = 0.81, fit standard error = 0.46
and F-value = 364. The figure shows the dataset (n = 325), the model, the 95% prediction limits
of the model and the 95% confidence interval (CI) for the model. The horizontal axis denotes
age in months up to birth at age 0 and age in years from birth to 51 years. From Wallace and
Kelsey [8].
55
anti-Mullerian
hormone (AMH),
tuberous sclerosis complexes (TSC) 1
and 2 and FOXL2 inhibits the
development of primordial follicles to
secondary follicles.
AMH
Secondary follicle
FSH, activins,
inhibins
androgens,
estrogens,
GDF9, BMP15
Primary
oocyte
with zona
pellucida
Basement
membrane
Pre-antral follicle
Thecal layer
Antrum formation
Cumulus cell
layer
enclosing the
oocyte
Antral follicle
follicle. Anti-Mullerian
hormone (AMH)
has been shown to suppress early stages
of follicular growth and onset of
responsiveness to FSH in vitro, exerting a
controlling influence on the rate at which
follicles become available for
pre-ovulatory development. Enhancing
effects from growth differentiation factor
(GDF-9), bone morphogenetic protein 15
(BMP-15) and insulin-like growth factors
enables the pre-antral follicle to develop
further into the antral follicle with the
formation of the antrum. At the same
time, the GCs proliferate and differentiate
into the cumulus cell layer which
encloses the developing oocyte. With the
antral cavity filled with follicular fluid, the
pre-antral follicle now becomes the
antral follicle.
Gonadotrophin-independent phase
Ovarian follicular
development
Gonadotrophin-dependent phase
11
10
Pool boiling
Forced flow
Boiling
New
technology
h = 103 W/m2 K
h = 104 W/m2 K
h = 105 W/m2 K
6
5
h = 106 W/m2 K
4
0.0
0.2
0.4
0.6
0.8
Sample dimension (cm)
1.0
(b)
(a)
(c)
(d)
(a)
(b)
*
*
*
(a)
(d)
(b)
(e)
(g)
(h)
(c)
(f)
Figure 14.12 The vitrification procedure for big volume of spermatozoa suspension. (a) The 0.25 ml plastic straws are halved and
dark-marked from one side (dark asterisks) and a 0.5 ml packaging straw (white asterisks). (b) The non-marked side of the half-straw is filled
with spermatozoa suspension. (c) A 0.25 ml half-straw, hermetically closed from both sides, is filled with spermatozoa suspension. (d)
Holding it in a horizontal position, the 0.25 ml half-straw filled with spermatozoa suspension is marked from one side (arrowed) and
placed into a 0.5 ml packaging straw, closed from both sides. (e) Using tweezers and keeping it in a horizontal position, this closed
packaging system is directly immersed into liquid nitrogen (LN2 ) and submerged for over 5 s to prevent the flow of spermatozoa
suspension spreading into packaging straw. (f) The vitrified sample is stored in LN2 . Using tweezers, the dark-marked part of the Sealed
Pulled Straw (SPS), approximately 1.01.5 cm, is removed from the LN2 and the end of packaging straw is cut. (g) With the help of a 200 l
pipette tip (Eppendorf AG, Hamburg, Germany), the suspension-filled fixed straw is quickly removed from the packaged straw and (h)
immersed into a 15 ml plastic tube containing 6 ml of human tubal fluid (HTF) and human serum albumin (HSA) prewarmed to 37 C with
gentle agitation to accelerate the melting and removing of content.
Culture
Transplantation
Cryopreservation
Sperm
production
Testis
biopsy
Chemotherapy
or irradiation
Figure 15.1 Male germline stem cell preservation. Before treatment for cancer by chemotherapy or irradiation, a boy
could undergo a testicular biopsy to recover stem cells. The stem cells could be cryopreserved or, after development
of the necessary techniques, could be cultured. After treatment, the stem cells would be transplanted to the patients
testes for the production of spermatozoa. From Brinster [14] with permission.
Germ
cells
Enzyme
In situ
injection
digestion
Testis with
transgene
35 months
Donor genes
transmitted
X Mate
Progeny
Figure 15.2 Testis cell transplantation method. A single-cell suspension is produced from a fertile donor testis. The
cells can be cultured or microinjected into the lumen of seminiferous tubules of an infertile mouse. Only a
spermatogonial stem cell can generate a colony of spermatogenesis in the recipient testis. When testis cells carry a
reporter transgene that allows the cells to be stained blue, colonies of donor cell-derived spermatogenesis are
identified easily in the recipient testes as blue stretches of tubule. Mating the recipient male to a wild-typed female
produces progeny, which carry donor genes. From Brinster [11] with permission.
Tight
junction
Sertoli cell
Sertoli
cell
GFR
-1
Human
spermatogonial
stem cell
SFK
PI3 /AKT
PI3K
AKT
Etv5, Bcl6b
Self-renewal
Differentiation
Figure 15.3 A proposed model of human spermatogonial stem cell (SSC) self-renewal
regulation by glial cell line-derived neurotrophic factor (GDNF), which has been demonstrated
to have an essential role in regulating rodent SSC self-renewal. The model is similar to those
suggested for mouse SSC self-renewal. In this model, GDNF binds to RET and the GFR1
coreceptor with possible intracellular protein kinase signaling through SFK and PI3K/AKT
downstream pathways to regulate the expression of specific genes, such as Etv5 and Bcl6b,
which are involved in SSC self-renewal. However, other genes not regulated by GDNF (e.g.
Zbtb16, Taf4b and Lin28), are likely controlled by different signals and may block differentiation
but not be involved directly in self-renewal. Genes for these regulatory molecules have been
shown to be highly expressed in pre-pubertal human spermatogonia, mouse gonocytes and
mouse SSCs. The basement membrane (green), on which the SSC rests, is generated by the
peritubular myoid cells (dark brown) and Sertoli cells (tan). From Wu et al. [27] with permission.
(a)
(b)
Figure 15.4 Detection of human germ cells transplanted into seminiferous tubules of
recipient mouse testes using a baboon testis specific antibody that identifies human
spermatogonia. (a) Donor human spermatogonia in mouse tubule 4 months after
transplantation of cryopreserved cells. This panel shows that cryopreserved cells also colonize
mouse testes as observed with freshly transplanted cells. (b) Donor human spermatogonia in
mouse testis 5 months after transplantation. These donor cells were transplanted without
cryopreservation. Bar = 100 m (a) and 40 m (b). From Nagano et al. [17] with permission
C 2002 American Society for Reproductive Medicine.
from Elsevier Science, Inc.
(a)
(b)
(c)
Control
3
HSD
N
Nm
50 m
TEM
Fresh
L
M
Cm
Fresh
2.8 m
50 m
SER
F/T/G
L
Nm
Cm
SER
F/T/G
2.8 m
50 m
100%
80%
60%
40%
20%
0%
With GnRHa
No GnRHa
R
Figure 20.1 Visualization of right pelvic lymphatic channels through laparoscopy after Patent Blue injection in the
cervix.
Figure 20.4 Dissection of bladder pillars and identification of the left ureter.
Figure 20.6 Section of the operative specimen at the level of the uterine isthmus.
Figure 20.8 Operative specimen of a radical trachelectomy showing the vaginal cuff and the proximal parametrial
resection.
Figure 20.10 Pre-treatment epidermoid cervical carcinoma stage IB2 (45 mm) in a 25-year-old young woman,
neoadjuvant chemotherapy is planned.
Figure 20.11 Same patient after four courses of neoadjuvant chemotherapy: complete regression of the lesion.
3.8
3.2 2.7 2
0.6
Leukemia
11.3
Hematological
malignancies
44.3
9.6
Hodgkin's
lymphoma
22
22.6
B re a s t c a n c e r
O v a ri a n c a n c e r
R h a b d o m y o s a rc o m a
C e rv i c a l c a n c e r
L i p o s a rc o m a
Le u k e m ia
Non-Hodgkin's
lymphoma
11
O t h e r c a n c e rs (p a n c re a s , s t o m a c h , )
O ste o sa rc o m a
C o l o re c ta l c a n c e r
E w i n g 's s a r c o m a
H o d g k i n 's l y m p h o m a
N o n - H o d g k i n 's l y m p h o m a
Figure 25.1 Indications for ovarian tissue cryopreservation in case of malignant disease at
Saint Lucs University Hospital, Brussels, Belguim in 2008.
Ovarian transplantation
Oophorectomy
Tissue processing
Whole ovary
prepared
with vessels
FREEZE
(Slow freeze/vitrifit)
FREEZE
(Slow freeze/
vitrifit)
THAW
THAW
Autotransplantation
Or thotopic
Autotransplantation
Heterotopic
Xenotransplantation
In vitro culture
Heterotopic
with vascular
anastomosis
Orthotopic
with vascular
anastomosis
Figure 27.4 Theoretical strategies for oocyte maturation in cryopreserved ovarian tissue and intact ovary. To date,
the only strategy that produced live births is orthotopic autotransplantation of ovarian tissue.
120
FSH (IU/l)
100
80
60
40
20
0
0
(a)
4
5
6
7
Months after transplant
10
11
12
Estradiol (pg/ml)
250
200
A
B
C
150
100
50
0
0
(b)
4
5
6
7
Months after transplant
Figure 27.6 Monthly follicle stimulating hormone (FSH) (a) and estradiol levels (b) after the
second transplantation of ovarian tissue in three cancer patients (AC). Estradiol production
from ovarian grafts was noticed 2 months after transplantation, and serum FSH levels
decreased below10 mIU/ml in 2 patients 3 months after transplantation. Reproduced with
permission from Elsevier.
10
11
12
(a)
(b)
(c)
(d)
(e)
(f)
Figure 28.1 Types of follicles observed in human ovarian tissue: (a) primordial; (b) two
intermediary primordial; (c) an intermediary primordial (top) and primary (bottom); (d)
proliferating; (e) secondary and an intermediary primordial; (f) an antral and a primordial (top
right corner).
(a)
(b)
Figure 28.2 Ovarian cortex from 2 patients: (a) an 18-year-old patient with abundant primordial follicles throughout
the cortex; and (b) a 34-year-old patient with a cluster of primordial follicles.
(a)
(c)
(b)
(a)
(b)
(c)
(a)
(b)
Figure 28.5 Ovarian tissue cryopreserved using: (a) a rapid cooling rate showing remnants of
a primordial follicle; and (b) an intermediate rate of cooling showing a primordial follicle
containing lysed pre-granulosa cells and oocyte.
Cancer diagnosis
Pregnancy
Uterus
Cryopreservation
of tissue
samples
Cancer treatment
Fallopian
tubes
Ovary
Vagina
Complete remission
Grafting or placing
of fragments in the
perinatal cavity
Thawing of tissue
samples
LE SOIR - 10.12.09
Figure 29.1 Illustration describing the different steps from biopsy to re-implantation and pregnancy.
Fallopian
tube
Creation of
peritoneal window
Atrophic
ovary
Fimbria
Reimplantation of ovarian
graft in peritoneal window
(a)
(b)
Figure 29.2 (a) Creation of a peritoneal window close to the atrophic ovary.
(b) Transplantation of ovarian cortex pieces into the peritoneal window.
Figure 29.3 An important vascular network is observed 7 days after the creation of the peritoneal window.
(a)
Figure 29.4 (a) Decortication of the ovarian cortex from the remaining ovary.
(b)
(b) Suture of the cryopreserved thawed cortical strips on the ovarian medulla.
(a)
(b)
2.5
15
10
1.5
1
0.5
3
Progesterone
levels (ngr/ml)
2
1
0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29
71
30
20
10
38
1
Progesterone
level
(ngr/ml)
40
Estradiol
level
(pmol/ml)
Progesterone
level
(ngr/ml)
37
73
36
72
300
200
0.5
100
0
Estradiol
level
(pmol/ml)
Estradiol level
(ngr/ml)
Progesterone level
(pmol/ml)
20
34
35
36
(a)
(b)
Figure 30.1 Endocrine function and fertility after cryopreservation and re-transplant of intact sheep ovaries at 2 and 6
years post-transplantation. (a) Hormonal levels 2 years post-transplantation [50]. (b) Follicles by transillumination 6 years
post-transplantation [51].
Figure 30.2 Whole human ovary cryopreservation with the vascular pedicle utilizing the
same Multi-Thermal-Gradient device and slow cooling, rapid thawing protocol as described in
the text.
(a) Stages of follicle development from primordial to ovulatory. All growing follicles (primary onwards)
must be activated from the finite resting pool of primordial follicles.
Primordial
pool
Growth
activation
Primary
Pre-antral
Early antral
Mid antral
Pre-ovulatory
PIP3
PDK1
RPTK
Pk
p85
p110
PIP3
Akt
mTORC2
Thr308
Ser473
P13K
PTEN inhibits activation
of primordial follicles
TSC1/TSC2
mTORC1
mTORC1 promotes activation
of primordial follicles
PDK1
P
Thr309
P
S6K1
Thr225
rpS6
Figure 33.1 (a) Digrammatic representation of follicle growth from the non-proliferating pool
of primordial follicles. Primordial follicles are continuously activated into the growing
population where they become primary follicles consisting of an oocye arrested at the dictyate
stage of prophaseI of meiosis (yellow) surrounded by granulosa cells (green). Primary follicles
undergo oocyte growth and granulosa cell proliferation and differentiation (purple) when they
form an antral cavity. Antral follicles continue to grow and granulosa cells differentiate into two
subpopulations of cells: (1) cumulus surrounding the oocyte (blue); and (2) mural lining the wall
of the follicle (orange). Exact timings for this developmental sequence to occur in humans are
not known but estimations suggest several months. However, it is not known whether the
growth profile is continuous or whether there are resting phases through follicle
development. (b) Simplified version of the PI3K pathway. The factors initiating this process are
largely unknown but a body of evidence is emerging to show that the
phosphatidylinositol-3-kinase (PI3K-AKT) signaling pathway is a major regulator of early
follicle/oocyte development and that components of this pathway are involved in controlling
the rate of activation from the non-growing population of follicles. The phosphatase PTEN
converts PIP3 to PIP2, which negatively regulates PI3K activity. Signaling mediated by PI3Ks
converge at PDK1. PDK1 phosphorylates Akt and activates it. Akt can phosphorylate and
inactivate tuberous sclerosis complex 2 (TSC2 or tuberin), which leads to the activation of
mTOR complex (mTORC1). mTORC1 can phosphorylate (activate) S6K1. S6K1 subsequently
phosphorylates and activates rpS6, which enhances protein translation that is needed for cell
growth. mTORC1 can be inhibited pharmacologically with Rapamycin and stimulated by
leucine. The manipulation of this pathway could have important clinical applications in the field
of fertility preservation.
Micro-cortex
culture
Removal of pre-antral
follicles by
micro-dissection
Individually cultured
and monitored for
oocyte/follicle
health markers
(b)
(a)
(c)
(e)
IVM of isolated
oocytecumulus complexes
and subsequent fertilization
Step 1
Step 2
Step 3
Step 4
Figure 33.2 Proposed multi-step culture system for activation of human primordial follicles and subsequent follicle/oocyte development.
The stages required for a multi-step culture system are as follows. Activation of primordial follicles within cortical strips (a). Removal of all
growing follicles and most of the underlying stromal cells increases the rate of activation [6]. Flattened strips are cultured free floating in
medium containing human serum albumin (HSA), ascorbic acid and basal levels of follicle stimulating hormone (FSH) [6]. Once follicles have
reached multilaminar stages they are isolated mechanically using needles and cultured individually Isolated follicle culture is to support
development from pre-antral to antral stages (b). The addition of activin at this stage results in improved follicle development and increased
antral formation (c) [6]. Follicles of similar stages that have been grown in vivo have been isolated and grown with alginate drops [7] (d), and
oocytes grow to almost full size within a total of 30 days [7]. The final stages of oocyte growth and development could be achieved by
culturing the oocyte and its surrounding somatic cells outwith the constraints of the large follicle (e).
(a)
(b)
(d)
(e)
(c)
(f)
Figure 33.3 (a) A cluster of quiescent follicles in freshly fixed human ovary. (b) After 6 days in vitro, growing follicles () appear on
the surface of a cultured fragment of human ovarian cortex. (c) A growing follicle protruding from the edge of a fragment of cultured
human cortex. (d) Intact secondary human mechanically dissected with presumptive theca layers attached. (e) Histological image of a
secondary human follicle fixed after 6 days in vitro growth within a cortical fragment. (f) Histological image of human antral follicle
fixed after a total of 10 days in vitro growth.
Antral
Secondary
Primordial
Antrum
Primary
Organ culture
(stromal cells)
Oocyte
Granulosa
Theca
(a)
(b)
(a)
(b)
Section 7
Chapter
30
Introduction
Patients with cancer who desire to preserve their future
reproductive potential but require immediate gonadotoxic treatments (chemo and/or radiotherapy), are
left with few options for fertility preservation. These
options include: (a) cryopreservation of ovarian tissues
as cortical strips; (b) dual cryopreservation of both
ovarian cortical tissue and cryopreservation, after in
vitro maturation, of immature oocytes extracted from
the small antral follicles visible within the ovarian cortex at the time of the harvest; (c) cryopreservation of
one whole ovary [19]. Each of these options is still
considered experimental (thus requiring Institutional
Review Board approval and patients informed consent).
Ovarian cryopreservation and transplantation
[1011], either as heterotopic or orthotopic allografts,
has shown some reproductive success [1214]. At
the time of writing, a total of seven live births from
re-transplantation of ovarian cortical tissue to an
orthotopic location have been reported [1519], while
four more have been announced at a meeting, but not
yet published. Typically, it takes about 45 months
for resumption of endocrine function as evidenced by
menses or serological hormonal evaluation. However,
the re-transplanted cortical pieces only retain ovarian
function for a short time and almost all ceased to
function by 3 years [6, 20, 21].
There are several reasons to explain this transient return of ovarian function followed by the rapid
decline. One reason is that the amount of cryopreserved/thawed cortical tissue re-transplanted during a
graft is limited. Another reason for the short longevity
is that the cortical tissue is grafted without a vascular anastomosis and is, therefore, completely dependent for its survival on the development of a new vas-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
367
368
Slow cooling
In 2002, Jeremias et al. attempted orthotopic transplantation of a whole ovary in an adult sheep by anastomosis of the vascular pedicle [36]. After bilateral
laparoscopic oophorectomy, ovaries were autotransplanted into the abdominal wall, and microsurgical
vascular anastomosis of the ovarian to the inferior epigastric vessels was performed. After noting promising resumption of endocrine function post-transplant
and a high follicular count, the authors concluded that,
in conjunction with an improved protocol for cryopreservation, ovarian autotransplantation with vascular anastomosis may be superior to ovarian tissue
banking and grafting techniques [36].
The same team also performed also one of the
first transplants of an intact frozenthawed ovary via
microvascular anastomosis [44]. After laparoscopic
dissection, the whole ovaries were immediately perfused with heparin, followed by perfusion and immersion in a bath containing Leibovitz L-15 medium,
10% fetal bovine serum (FBS) and 1.5 M DMSO.
Ovaries were perfused via the ovarian artery with the
369
2.5
15
10
1.5
1
0.5
3
Progesterone
levels (ngr/ml)
2
1
0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29
71
30
20
10
1
Progesterone
level
(ngr/ml)
40
Estradiol
level
(pmol/ml)
Progesterone
level
(ngr/ml)
37
73
36
72
38
300
200
0.5
100
0
Estradiol
level
(pmol/ml)
Estradiol level
(ngr/ml)
Progesterone level
(pmol/ml)
20
34
35
36
(a)
(b)
Figure 30.1 Endocrine function and fertility after cryopreservation and re-transplant of intact sheep ovaries at 2 and 6 years
post-transplantation. (a) Hormonal levels 2 years post-transplantation [50]. (b) Follicles by transillumination 6 years post-transplantation [51].
See plate section for color version.
370
Vitrification
While many attempts have been made to cryopreserve
whole ovaries by slow freezing techniques, there have
been few studies that have attempted utilizing vitrification. This method is becoming increasingly recognized as alternative to organ and tissue preservation by
slow freezing as it circumvents the mechanical damage
cause by ice crystal formation [54].
Fahy and colleagues have described two vitrification solutions that may be useful for whole ovary cryopreservation. The first, VS1, contains 20.5% wt/vol.
DMSO, 15.5% wt/vol. acetamide, 10% wt/vol. propylene glycol and 6% wt/vol. polyethylene glycol in a
modified Dulbeccos saline (HB1) [55]. The second,
VS4, contains 2.75 M DMSO, 2.76 M formamide and
1.97 M propylene glycol diluted in BM1 medium [56].
The two solutions, VS1 and VS4, were compared by
Courbiere et al. in a sheep model of whole ovary cryopreservation for toxicity to primordial follicles and
vessels by collecting ovaries with intact pedicles from
5 to 6-month-old lambs [57]. Each ovary was perfused via the ovarian artery with heparinized Ringers
solution, followed by perfusion and immersion in a
bath with VS1 or VS4 solution. Perfusion rate was performed at 0.35 ml/min with a stepwise increase in concentration of cryoprotectant. After perfusion, ovaries
were transferred into cryobags containing the cryoprotectant mix and then plunged into liquid nitrogen.
After storage, the vitrified samples were rapidly rewarmed in a 37 C water bath, and the cryoprotectant
was removed by a reversed concentration gradient perfusion, and then washed in BM1 medium for 5 min
[57].
Follicle viability fell from 75.6% 1.1% without
vitrification to 68.2% 1.9% after vitrification with
VS1, and from 68.0% 3.8% to 60.7% 2.4% after
vitrification with VS4. While these differences between
VS1 and VS4 were not statistically significant, follicle density remained significantly higher in vitrification with VS4 (P 0.05). Histologically, the percent-
age of normal primordial follicles fell after vitrification, with 25.2% 7.0% of follicles remaining normal
with VS1 and 53.5% 3.2% remaining normal with
VS4. There were also more post-vitrification cytoplasmic anomalies with VS4 (P 0.05) but more nuclear
and combined anomalies with VS1 (P 0.05). Fractures occurred in vessels during thawing in 3 of 5 cases
with VS1 and in 8 of 10 cases with VS4, although the
authors noted that the catheters used to re-perfuse the
ovarian artery were fitted without difficulty. This study
showed that whole sheep ovaries can survive vitrification with good immediate follicular viability via histological evaluation; however, the blood vessels were
cryodamaged.
Similar to previous to slow-freezing studies, Courbiere et al. performed a follow-up study in 2008,
attempting vascular anastomosis to the contralateral pedicle in sheep ovaries either fresh or after
vitrification [58]. Successful microsurgical transplantation was performed in both groups, but, not surprisingly, the median ischemia time was significantly longer in the cryopreservation group. Only
one out of five ewes undergoing ovarian vitrification recovered endocrine function six months after
transplantation compared to four out of five in the
fresh transplant group. However, histological evaluation showed total follicle loss in the vitrification
group, suggesting that attempts at cryopreservation
with vitrification were unsuccessful, despite technical
feasibility [58].
371
Table 30.1 Summary of whole human ovary cryopreservation experiments listed according to type of freezing method, cryoprotectant
used and outcomes measured to indicate post-thaw viability
Study
N=
Surgical
method
Freezing method
Cryoprotectant
Outcomes measured
Laparoscopy
Slow cooling
DMSO
Laparoscopy
Slow cooling
(ovaries bisected)
DMSO
Laparoscopy
Slow cooling
DMSO
Apoptosis, ultrastructural
assessment
Laparoscopy
Slow cooling
DMSO
Technical feasibility of
oophorectomy and freezing
11
Laparoscopy,
laparotomy
Slow cooling
EG
Apoptosis, histological
morphology
372
In 2007, the same group described the technique of laparoscopic oophorectomy with the intent to
cryopreserve the whole ovary and its vascular pedicle
[46]. The whole ovary was successfully removed and
cryopreserved by arterial catheterization in all nine
patients in the study. The authors had two main conclusions: first, that the ischemic interval before cryopreservation must be as short as possible, as this is the
time when significant damage is done to the organ; second, that surgeons must leave a long infundibulopelvic
ligament (5 cm) in order to allow easier access to the
ovarian vessels during canalization and perfusion of
the cryoprotectant.
More recently, Bedaiwy et al. described the successful cryopreservation of the human ovary [25].
They performed bilateral oophorectomy in two premenopausal women. In each case, one ovary was cryopreserved intact with its vascular pedicle in the same
method as described earlier. Ovaries were thawed 7
days later, and follicular viability and histology were
assessed, as well as apoptosis via TUNEL assay and
Bcl-2 and p53 protein expression profiles. They found
that overall viability of the primordial follicles in the
two ovaries was 75 and 78% in frozen/thawed ovaries,
and that there were similar primordial follicle counts,
absence of features of necrosis and mean values of
apoptosis when compared to control ovaries. After
demonstrating comparable survival rates and limited
molecular alterations, the authors concluded that this
represents further evidence that an intact human ovary
could be cryopreserved using a slow freezing protocol.
However, there has been confusion about whether the
References
1. Georgescu ES, Goldberg JM, du Plessis SS and
Agarwal A. Present and future fertility preservation
strategies for female cancer patients. Obstet Gynecol
Surv 2008; 63(11): 72532.
2. Bromer JG and Patrizio P. Preservation and
postponement of female fertility. Placenta 2008; 29
(Suppl. B): 2005.
Conclusions
The survival of reproductive-age women with cancer has dramatically improved over the last several
decades and, as a result, researchers and patients are
373
374
375
376
Section 7
Chapter
31
Ovarian transplantation
Whole ovary transplantation
Mohamed A. Bedaiwy and Tommaso Falcone
Introduction
Over the past few decades, considerable attention
has been given to the long-term reproductive function of females undergoing cancer treatment. Many
treatments, such as non-specific chemotherapeutic
agents and ionizing radiation, damage the ovaries
and induce premature ovarian failure (POF), making
future pregnancy impossible. Indeed, the number of
cancer patients at risk of POF who are seeking help to
preserve their fertility has increased dramatically.
One method of preserving fertility in female cancer patients is cryopreservation of ovarian cortical
strips. In this procedure, strips of ovarian tissue are
harvested before cancer treatment, cryopreserved and
then thawed for subsequent use. The practice has
become well established within the last decade. However, the subsequent use of the frozenthawed ovarian
strips to restore fertility remains a challenge. Transplantation is currently the only available option to
restore fertility using cryopreserved ovarian tissue
because in-vitro follicular culture technology is far
from fully developed in humans, although it has been
successful in the rodent model [1].
An option for those undergoing pelvic radiation
therapy is fresh whole ovary autotransplantation. Typically, transposition of the ovaries can be performed.
However, in cases where the radiation beam involves
much of the pelvis and abdomen transposition may
not be possible. In this case, the ovary can be surgically
removed and immediately implanted into an alternate
site that is not exposed to pelvicabdominal radiation. Currently, this procedure can be done using fresh
whole ovaries only. Although whole ovaries have been
successfully frozen and later transplanted back into the
donors in animal models, the procedures needed to
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
377
Post-transplantation ischemic
ovarian damage: morphological and
functional consequences
Avascular OTT is limited by one important fact
the graft completely depends on the establishment of
a new blood supply via the process of neovascularization. Avascular grafts experience an initial period
of ischemia after transplantation while waiting for
neovascularization to occur and consequently, a large
proportion of follicles are lost during this time [14
20]. The main feature in all of the OTT reports that
used cortical strips is the limited graft longevity and
high risk of recurrent ovarian failure (Box 31.1) [2].
The key reason behind this phenomenon is the fact
that most of these experimental transplants were performed without vascular re-anastomosis. As a result,
ischemic injury occurred before full revascularization
of the transplanted tissue from the surrounding vessels
could take place.
The exact mechanism by which the neovascularization process occurs is unknown and so is the time
needed for this process to be completed. Theoretically,
the shorter the time needed for neovascularization,
378
hormone (AMH)
Strategies to prevent
post-transplantation ischemic
ovarian damage
Many strategies have been devised to minimize the initial post-transplantation ovarian ischemia (Box 31.2).
Steps to improve neoangiogenesis have been successfully applied by experiments that have shown early
perfusion of ovarian cortical strips upon their transplantation into granulation tissue [31]. Free oxygen
radicals are produced as a result of the ischemia perfusion process. They have the potential to damage cell
membranes, endothelial membranes and mitochondrial function [32]. The use of exogenous antioxidants
to augment ovarian transplant resistance to oxidative stress-associated damage has been evaluated by
many investigators. In one study, for example, ascorbic
acid and mannitol reduced surgically-induced ovarian ischemic injury in rats [33]. Moreover, injection
of vitamin E before ovarian transplantation improved
the follicular survival rate [21]. These preliminary data
have not been confirmed by others, however [34]. Sapmaz et al. locally administered melatonin and oxytetracycline during intraperitoneal rat ovarian grafting
and assessed the effects on graft function. They found
that the injection reduced ovarian tissue necrosis [35].
In an in vitro model, Kim et al. found that incubating
ovarian tissue with ascorbic acid for up to a maximum
of 24 h reduced apoptosis [26].
It is expected that neoangiogenic growth factors
such as fibroblast growth factor (FGF), transforming
379
380
dant for POF [9] or Turners syndrome [49] have benefited from it.
The upper arm was the first recipient transplantation site tested in humans. There are two reports of successful whole fresh ovary transplantation in the upper
arm that was done prior to sterilizing pelvic irradiation [47, 48]. Leporrier et al. created a cavity in the
forearm arm for the ovary using a testicular prosthesis that was inserted 3 months before the transplantation. In that report, the ovary remained function
for the next 16 years [47]. In the second report, the
transplantation process was performed in the context
of radical hysterectomy for early stage cervical carcinoma. The ovary remained functional for at least 1 year
after transplantation [48].The procedure was unfortunately followed by local vault malignant recurrence.
No long-term follow-up has been reported. Mhatre
et al. successfully implemented the technique in two
patients with Turners syndrome (ovarian function was
restored and the patients developed secondary sexual
characteristics) [49]. An abdominal pfannenstiel incision was used in both cases.
More recently, Silber et al. culminated their efforts
in ovarian transplantation in monozygotic twins discordant for POF by reporting the first full-term pregnancy obtained using orthotopic whole fresh ovary
transplantation with microvascular anastomosis [9]. A
fresh ovary from the fertile twin was implanted in her
monozygotic twin with POF.
Indication for
transplantation
of ovarian tissue
Reference
No. of
patients
Ovarian volume
and site
Whole ovary,
heterotopic
Hodgkins disease
Whole ovary
Cervical cancer
Whole ovary,
orthotopic: Case 1,
vascular pedicle;
Case 2, avascular
transplantation
Turners syndrome
Premature ovarian
failure
A pair of monozygotic
twins discordant for
premature ovarian failure
Outcomes
381
cryopreserved and three of four control grafts. Functional corpora lutea were identified in 3 ewes (1 control; 2 cryopreserved) 1825 weeks after grafting. In
addition, inhibin A levels indicated resumption of follicular development in four cryopreserved and one
control ewes; however, castrate gonadotrophin levels
persisted in five cryopreserved and two control ewes.
The main prominent feature of this whole ovary transplantation experiment is the fact that primordial follicle density was significantly reduced following grafting
in both cryopreserved and non-frozen ovaries [57].
Although transplantation of whole cryopreserved
thawed ovary was not performed in humans, cryopreservation of a whole ovary using a slow-freezing
protocol has been successfully attempted [45]. The
results showed both vascular and follicular integrity
upon thawing after freezing and thawing. More
recently, a multi-gradient-freezing device was used
with promising results [58]. In that study a high follicular viability, normal histological architecture and preserved vessel integrity were reported, supporting the
potential for vascular re-anastomosis.
382
383
384
Conclusion
The current evidence suggests that whole frozen
thawed ovary transplantation may be successful in
humans in the future. However, the significant depletion of the primordial follicle reserve observed after
transplantation in animals is a major problem that
needs to be further addressed at the experimental level
prior to attempting that approach in humans. With
the recent success of whole ovary transplantation with
microvascular anastomosis, reproductive organ transplantation researchers may need to learn from other
transplantation settings. Safer cooling techniques and
new operative steps that minimize vascular thrombosis are needed. Multi-team approaches should be
expanded [42]. Future research should operate within
the framework of the patients needs, namely fertility
and sexuality.
References
1. Picton HM, Harris SE, Muruvi W et al. The in vitro
growth and maturation of follicles. Reproduction 2008;
136(6): 70315.
2. Bedaiwy MA, El-Nashar SA, El Saman AM et al.
Reproductive outcome after transplantation of ovarian
tissue: a systematic review. Hum Reprod 2008; 23(12):
270917.
385
386
387
388
Section 8
Chapter
32
Introduction
With the advent of culture models for studying the
process of ovarian folliculogenesis some 40 years ago,
opportunities arose for the more systematic evaluation
of the factors that regulated ovarian function [1]. The
initial focus of studies using cultured follicles emphasized two of the then widely recognized roles of the
follicle in mammals: the production of ovarian steroid
hormones and of viable oocytes during the process
of ovulation. As our understanding of the molecular
and cellular complexity of this tissue compartment has
evolved and deepened, so too has the need to redefine the major functions of the follicle at both local
ovarian and systemic levels in the context of reproduction in mammalian species, especially as it relates to
the origins and treatments for human infertility [2].
Thus, a shift in the motivation to use cultured follicles in humans has taken place owing primarily to the
rapidly evolving field of fertility preservation. Through
an interesting turn of events dictated by the need to
maintain and propagate human oocytes that would be
capable of supporting term gestations, a dire need has
been recognized that would enable optimization of follicle functions under in vitro conditions in order to
realize ovarian capacity for young women who have
had their fecundity seriously compromised as a result
of genetic, environmental or iatrogenic life-sparing
treatments such as those involved with the management of cancer [35].
The introduction of technology that permits the
cryopreservation on ovarian tissues has opened the
prospect of sustaining and storing primordial follicles
from individuals that could at a later time be thawed
and subjected to prolonged culture. Exactly what con-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
389
390
Table 32.1 Summary of major functional properties for ovarian follicles that have been used in the assessment of tissue integritya
Property
Follicle stage
Biomarker
Vital
Quiescence
Primordial
Chromatin/PTEN
Proliferation
Primary, secondary
Yes (Click-IT)
Apoptosis
Secondary, antral
TUNEL, caspase 3
Autophagy
Primordial (? others)
Beclin/ATG
Differentiation
Antral
Aromatase, LHr
No
Note that many of the biomarkers employed to date draw on the use of immunocytochemical and histological assays requiring tissue
destruction. Also, not all markers are pertinent to all stages of folliculogenesis. Some examples of vital biomarkers that are under development for determining cultured follicle integrity are shown in the last column.
adherent two-dimensional conditions or within matrices of various kinds that retain a three-dimensional
architecture [1520]. While standard protocols
deploying assays for cell proliferation (3H-thymidine,
BrdU incorporation or cell cycle markers such as
phosphohistone-3, PCNA, etc.) or apoptosis (TUNEL,
caspase-3) offer postscripts for the relative fraction
of viable cells within a follicle (Table 32.1), in the
end these are crude and retrospective assays that
add little to the immediate needs of the clinician
requiring a more real-time assessment of follicle
integrity. Towards this end, several new probes have
gained usage in the evaluation of tissue culture models
that take advantage of the speed, sensitivity and
spectral properties of microplate readers. This new
generation of reagents permits resolution of metabolic
activity, including reactive oxygen generation, cell
proliferation and even identification of rapid-versusslowly dividing cells within an organ or tissue culture
using multi-well formats, which should avail the
optimization of conditions that support oogenesis.
Moreover, as discussed below, the link between DNA
damage sensing and repair is fast becoming a major
determinant in the assessment of follicle integrity as
it relates to both somatic and germ cell components,
and these assays have introduced a range of sensitivity
and precision that will materially advance the field of
follicle culture.
As with most in vitro systems, culture environments create adverse conditions that are known to
affect DNA integrity, often due to the generation of
free radicals in response to high oxygen tension [21].
Given these deleterious side effects of culture environments, genomic integrity is one area of follicle evaluation that has received little attention and requires
closer inspection. Many new reagents are available for
evaluating the cascade of events associated with the
(a)
(b)
(c)
(d)
(e)
(a')
(b')
(c')
(d')
(e')
391
(a)
(b)
(c)
(d)
(e)
(f)
(g)
(h)
(i)
392
(a)
(b)
(c)
(d)
(e)
(f)
(g)
(h)
(i)
393
conceptualizing folliculogenesis as an oocentric process rather than one that fulfills the endocrine functions so vital to reproductive physiology [8, 9]. In
this sense, future research into the phases of follicular development that are focused on supporting the
growth stage of oogenesis, as well as in anticipation of
the resumption and completion of meiosis, are likely
to uncover complex feedback pathways that are subject to regulation by the patterns of gene and protein
expression resident within the oocyte [39]. Defining
what these factors are, and how their presence signals
transformations in the behaviors of granulosa and thecal cells, should reveal arrays of biomarkers that in the
end reflect the ongoing role of the oocyte in determining its ultimate fate [2, 4044]. This may require application of more sensitive methods for the detection
of metabolites and substrates whose concentration is
maintained in a microenvironment more reminiscent
of what is present within the intact ovary. This goal is
achievable if the potential of microfluidic technologies
is realized for cultured follicles as it is being realized
for other model developmental systems requiring protracted periods of time in vitro.
A second dimension of future considerations for
the ex vivo maintenance of ovarian follicles concerns
that of time [3, 4, 18]. While the exact course of events
and their chronology that govern the development
of oocytes is not known, it is likely to require some
weeks for the entire process to be brought to fruition.
Besides being of an invasive nature, rote biochemical
and molecular studies while providing large quantities of data that might be expected to serve some value
as biomarkers are unlikely to contribute in a major
way to our understanding of the epigenetics of oogenesis. This rapidly evolving area of contemporary biology
has already entered the front-lines of human ARTs [18,
20, 24] given the well-documented effects that various
in vitro procedures have on the software that drives
the cytoplasmic and nuclear genomes of the early
embryo, all of it inherited from the process of oogenesis. Our biological clocks are changing the way in
which epigenetic modifications of the oocyte genome
play out during the natural course of folliculogenesis
[2, 27], warranting use of caution in future applications of follicle culture technology that would be relied
upon to preserve oocytes for individuals whose germ
line may have been compromised as a result of natural aging or the adverse effects of cancer treatment
[4, 18].
394
Acknowledgements
The support of the ESHE fund, the University of
Kansas Cancer Center and The Scientific and Technological Research Council of Turkey (TUBITAK) is graciously acknowledged.
References
1. Smitz J and Cortvrindt R. Oocyte in-vitro maturation
and follicle culture: current clinical achievements and
future directions. Hum Reprod 1999; 14(Suppl. 1):
14561.
2. Rodrigues P, Limback D, McGinnis LK, Plancha CE
and Albertini DF. Oogenesis: prospects and challenges
for the future. J Cell Physiol 2008; 216(2): 35565.
3. Rosendahl M, Andersen CY, Ernst E et al. Ovarian
function after removal of an entire ovary for
cryopreservation of pieces of cortex prior to
gonadotoxic treatment: a follow-up study. Hum
Reprod 2008; 23(11): 247583.
4. Sanchez M, Alama P, Gadea B et al. Fresh human
orthotopic ovarian cortex transplantation: long-term
results. Hum Reprod 2007; 22(3): 78691.
5. Senbon S, Ishii K, Fukumi Y and Miyano T.
Fertilization and development of bovine oocytes
grown in female SCID mice. Zygote 2005; 13(4):
30915.
6. Telfer EE, Binnie JP, McCaffery FH and Campbell
BK. In vitro development of oocytes from porcine and
bovine primary follicles. Mol Cell Endocrinol 2000;
163(12): 11723.
7. Van Den Hurk R, Abir R, Telfer EE and Bevers MM.
Primate and bovine immature oocytes and follicles as
sources of fertilizable oocytes. Hum Reprod Update
2000; 6(5): 45774.
8. Hutt KJ and Albertini DF. An oocentric view of
folliculogenesis and embryogenesis. Reprod Biomed
Online 2007; 14(6): 75864.
9. Jayawardana BC, Shimizu T, Nishimoto H et al.
Hormonal regulation of expression of growth
differentiation factor-9 receptor type I and II genes in
the bovine ovarian follicle. Reproduction 2006; 131(3):
54553.
10. Walters KA, Binnie JP, Campbell BK, Armstrong
DG and Telfer EE. The effects of IGF-I on bovine
follicle development and IGFBP-2 expression are dose
and stage dependent. Reproduction 2006; 131(3): 515
23.
11. Blondin P, Bousquet D, Twagiramungu H, Barnes F
and Sirard MA. Manipulation of follicular
development to produce developmentally competent
bovine oocytes. Biol Reprod 2002; 66(1): 283843.
395
396
Section 8
Chapter
33
Introduction
Follicular development
Tissue banking of ovarian material is being increasingly offered to a variety of patients as a means of fertility preservation [1]. This tissue comprises of thin
cortical surface biopsies that contain predominantly
immature primordial follicles and currently is the only
option to restore fertility using this tissue is by transplantation [2]; however, this may not be a viable option
for all patients [3]. Increased options to maximize the
potential of this tissue to restore fertility could be realized by the development of in vitro systems to support complete growth from the early primordial stages
through to maturity. This technology would have many
therapeutic applications, including the production of
competent oocytes for assisted reproductive technology (ART); a model system to determine toxicological effects on germ cell development; a method for
the assessment of cryopreserved ovarian tissue prior
to transplantation; as well as providing an experimental model to address basic scientific questions concerning human oocyte development [4, 5]. Complete
oocyte development in vitro from the primordial stage
has been achieved in mice, but the larger size and
longer growth period of human follicles has made the
inter-species translation of these techniques difficult.
Recently progress has been made in defining conditions that support different stages of human follicle
development in vitro [6, 7], and these advances bring
the prospect of achieving a complete in vitro system
that supports oocytes from primordial to maturation
closer [8]. This chapter deals with our current understanding of in vitro development of human oocytes
and highlights the gaps that need to be bridged to
achieve a complete in vitro growth (IVG) system.
Female reproductive function requires cyclical development and maturation of ovarian follicles on a background of continuous activation from the pool of primordial follicles (Figure 33.1a). Primordial follicles are
formed prenatally and represent a finite population of
germ cells from which recruitment for growth will take
place throughout the womans reproductive life. Follicular growth and development involves a series of complex and precisely regulated events. It is characterized
by transition stages that begin with: (1) initiation of
primordial follicle growth and development to the preantral follicle stage; (2) the formation of antral follicles
where expansion to the pre-ovulatory or Graafian follicle is associated with granulosa cell proliferation and
antral fluid accumulation within the basement membrane; and (3) rupture of the Graafian follicle releasing
a cumulusoocyte complex at ovulation in response to
the mid-cycle luteinizing hormone (LH) surge (Figure
33.1a).
During its growth within the follicle the oocyte is
held in meiotic arrest, but as it grows it must acquire
the ability to resume meiosis (meiotic competence)
and the ability to support fertilization and embryonic development (developmental competence). Thus,
the oocyte is dependent upon the local environment
within the follicle for subsequent function as a gamete.
The development of follicles is regulated by a complex mixture of inhibitory and stimulatory endocrine,
paracrine and autocrine signaling by the somatic cells
(granulosa and surrounding theca cells) enhanced by
a range of oocyte specific regulatory factors mediated through bi-directional communication within the
follicle [9].
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
397
(a) Stages of follicle development from primordial to ovulatory. All growing follicles (primary onwards)
must be activated from the finite resting pool of primordial follicles.
Primordial
pool
Growth
activation
Primary
Pre-antral
Early antral
Mid antral
Pre-ovulatory
PIP3
PDK1
RPTK
Pk
p85
p110
PIP3
Akt
mTORC2
Thr308
Ser473
P13K
TSC1/TSC2
mTORC1
mTORC1 promotes activation
of primordial follicles
PDK1
P
Thr309
P
S6K1
Thr225
rpS6
Figure 33.1 (a) Digrammatic representation of follicle growth from the non-proliferating pool of primordial follicles. Primordial follicles are
continuously activated into the growing population where they become primary follicles consisting of an oocye arrested at the dictyate stage
of prophaseI of meiosis (yellow) surrounded by granulosa cells (green). Primary follicles undergo oocyte growth and granulosa cell
proliferation and differentiation (purple) when they form an antral cavity. Antral follicles continue to grow and granulosa cells differentiate
into two subpopulations of cells: (1) cumulus surrounding the oocyte (blue); and (2) mural lining the wall of the follicle (orange). Exact timings
for this developmental sequence to occur in humans are not known but estimations suggest several months. However, it is not known
whether the growth profile is continuous or whether there are resting phases through follicle development. (b) Simplified version of the
PI3K pathway. The factors initiating this process are largely unknown but a body of evidence is emerging to show that the
phosphatidylinositol-3-kinase (PI3K-AKT) signaling pathway is a major regulator of early follicle/oocyte development and that components of
this pathway are involved in controlling the rate of activation from the non-growing population of follicles. The phosphatase PTEN converts
PIP3 to PIP2, which negatively regulates PI3K activity. Signaling mediated by PI3Ks converge at PDK1. PDK1 phosphorylates Akt and activates
it. Akt can phosphorylate and inactivate tuberous sclerosis complex 2 (TSC2 or tuberin), which leads to the activation of mTOR complex
(mTORC1). mTORC1 can phosphorylate (activate) S6K1. S6K1 subsequently phosphorylates and activates rpS6, which enhances protein
translation that is needed for cell growth. mTORC1 can be inhibited pharmacologically with Rapamycin and stimulated by leucine. The
manipulation of this pathway could have important clinical applications in the field of fertility preservation. See plate section for color version.
398
support the partial growth of human follicles and several developmental milestones have been achieved,
namely, follicle activation [6, 1822] pre-antral follicle growth [6, 7, 19, 21, 2325], follicle differentiation
[6, 7] and oocyte maturation [26, 27]. These advances
have been made using both fresh and cryopreserved
human tissue. In the clinical setting progress has been
made with the ability to apply in vitro maturation
(IVM) techniques to immature human oocytes with
subsequent in vitro fertilization (IVF) of these oocytes
leading to pregnancy and live births [2830]. Despite
the successes, stumbling blocks still exist in putting
all of these systems together to achieve complete IVG.
A major difficulty associated with prolonged culture
is oocyte degeneration associated with the disruption
of the contact between the oocyte and its companion
somatic cells. Therefore it is important to understand
how culture conditions impact on the maintenance
of appropriate cell interactions during follicle/oocyte
development.
399
Micro-cortex
culture
Removal of pre-antral
follicles by
micro-dissection
Individually cultured
and monitored for
oocyte/follicle
health markers
(b)
(a)
(c)
(e)
IVM of isolated
oocytecumulus complexes
and subsequent fertilization
Step 1
Step 2
Step 3
Step 4
Figure 33.2 Proposed multi-step culture system for activation of human primordial follicles and subsequent follicle/oocyte development.
The stages required for a multi-step culture system are as follows. Activation of primordial follicles within cortical strips (a). Removal of all
growing follicles and most of the underlying stromal cells increases the rate of activation [6]. Flattened strips are cultured free floating in
medium containing human serum albumin (HSA), ascorbic acid and basal levels of follicle stimulating hormone (FSH) [6]. Once follicles have
reached multilaminar stages they are isolated mechanically using needles and cultured individually. Isolated follicle culture is to support
development from pre-antral to antral stages (b). The addition of activin at this stage results in improved follicle development and increased
antral formation (c) [6]. Follicles of similar stages that have been grown in vivo have been isolated and grown with alginate drops [7] (d), and
oocytes grow to almost full size within a total of 30 days [7]. The final stages of oocyte growth and development could be achieved by
culturing the oocyte and its surrounding somatic cells outwith the constraints of the large follicle (e). See plate section for color version.
meiosis is acquired after its ability to resume the process; moreover, oocyte size is a determinant in reaching metaphase II [48, 49]. Growing oocytes accumulate cytoplasmic organelles and these are dispersed
to the periphery of the cell in readiness for fertilization and pre-implantation embryo development; only
oocytes that have completed both cytoplasmic and
nuclear maturation are capable of fertilization and subsequent embryo development [50]. The aim of invitro follicle development or IVG is to achieve developmentally competent oocytes. Therefore culture systems must provide an environment that will sustain
oocyte growth and support cytoplasmic and nuclear
maturation but without the necessity to develop large
follicular structures [13]. Attempts to recapitulate in
vivo development (timings and size) in vitro have not
been successful and therefore strategies to improve
IVG systems should concentrate on optimizing oocyte
growth within the context of surrounding somatic cells
in the shortest time possible.
400
(a)
(b)
(d)
(e)
(c)
(f)
Figure 33.3 (a) A cluster of quiescent follicles in freshly fixed human ovary. (b) After 6 days in vitro, growing follicles () appear on the
surface of a cultured fragment of human ovarian cortex. (c) A growing follicle protruding from the edge of a fragment of cultured human
cortex. (d) Intact secondary human mechanically dissected with presumptive theca layers attached. (e) Histological image of a secondary
human follicle fixed after 6 days in vitro growth within a cortical fragment. (f) Histological image of human antral follicle fixed after a total of 10
days in vitro growth. See plate section for color version.
401
402
v-shaped micro-well plates has allowed the maintenance of three-dimensional follicular architecture in
vitro while promoting growth and differentiation in
bovine [7678] and human follicles [6, 38, 52] with
antral formation occurring within 10 days. Follicle differentiation has also been reported in bovine follicles embedded in collagen gels and cultured for 13
days [79] and, using a combination of media thickened with polyvinylpyrrolidone, a macromolecular
supplement and microporous membranes, one live calf
was produced from immature bovine follicles cultured
for 14 days [80]. In addition to v-shaped microwell culture plates, follicle encapsulation in alginate
hydrogels has been used to support secondary human
follicle growth in vitro [7]. The novelty of alginate
encapsulation is that it is believed to mimic the extracellular matrix in vivo in terms of its ability to facilitate molecular exchange between the follicle and the
culture medium, while its flexibility can accommodate
cell proliferation but its rigidity prevents dissociation
of the follicular unit. It would appear that the rigidity
of the alginate capsule is of vital importance to follicle development as inhibition of growth and reduced
steroidogenesis have been reported in murine follicles embedded in 1.0 and 1.5% alginate gels, respectively [81, 82], whereas fully grown human oocytes
have been produced using 0.5% gels [7]. The ability of
the reported systems to promote human follicle growth
in vitro from the earliest stages at rates that are accelerated in comparison to the in vivo environment is
indeed promising. The next step is to ascertain whether
the oocytes produced in these systems are capable of
IVM and to determine whether the altered growth is
deleterious to oocyte epigenetic health and normality.
403
404
conditions that enable the transition of these isolated follicles from pre-antral to antral stage of follicle development in vitro, since at present oocyte
somatic cell interactions are not fully supported at the
antral transition; and (3) developing in vitro systems
to support oocyte growth and subsequent maturation
of oocytegranulosa cell complexes taken from the
in-vitro grown early antral follicles, hence removing
the requirement to support large pre-ovulatory follicle
growth in vitro.
Summary
The achievement of complete mouse oocyte development in vitro has led to investigation into the potential of large mammal and human oocyte development
within culture systems. The individual success of several keys stages of this process, i.e. initiation of primordial follicle growth, follicle differentiation, completion of oocyte growth and IVM have been encouraging, although a unified system incorporating all of
these developmental milestones has yet to be defined
for human follicles. Although significant progress has
been made, much optimization is still required to routinely complete the in vitro development of the stages
detailed above. Translation of any in-vitro human follicle growth system into a clinical setting will require
rigorous testing to determine the normality and health
of in-vitro grown oocytes prior to the application of
IVF procedures. Basic research using rodent models suggest that oocyte development in vitro does not
result in adverse developmental outcomes or longterm effects [91]. Whilst this is encouraging, it is also
essential that good models, using large animal models,
are rigorously tested before proceeding with translating human culture systems to a clinical setting.
Whilst the therapeutic potential of in-vitro grown
human oocytes may not be imminently realized, the
methodology itself is central to fertility preservation
programs. The culture systems provide a reproducible
and effective technique to assess the viability of cryopreserved cortical strips prior to transplantation.
Furthermore, these systems allow many basic scientific questions regarding human oocyte development
to be addressed and, as a consequence, factors and
mechanisms involved in its regulation identified. The
knowledge gained from these basic studies will facilitate the development of optimized culture systems,
which could have the prospect of clinical application
References
1. Anderson RA, Wallace WH and Baird DT. Ovarian
cryopreservation for fertility preservation: indications
and outcomes. Reproduction 2008; 136: 6819.
2. Donnez J, Jadoul P, Squifflet J et al. Ovarian tissue
cryopreservation and transplantation in cancer
patients. Best Pract Res Clin Obstet Gynaecol 2010; 24:
87100.
3. Meirow D, Hardan I, Dor J et al. Searching for
evidence of disease and malignant cell contamination
in ovarian tissue stored from hematologic cancer
patients. Hum Reprod 2008; 23: 100713.
4. Telfer EE and McLaughlin M. Natural history of the
mammalian oocyte. Reprod Biomed Online 2007; 15:
28895.
5. Picton HM, Harris SE, Muruvi W and Chambers EL.
The in vitro growth and maturation of follicles.
Reproduction 2008; 136: 70315.
6. Telfer EE, McLaughlin M, Ding C and Thong KJ. A
two step serum free culture system supports
development of human oocytes from primordial
follicles in the presence of activin. Hum Reprod 2008;
23: 11518.
7. Xu M, Barrett SL, West-Farrell E et al. In vitro grown
human ovarian follicles from cancer patients support
oocyte growth. Hum Reprod 2009; 24: 253140.
8. Smitz J, Dolmans MM, Donnez J et al. Current
achievements and future research directions in ovarian
tissue culture, in vitro follicle development and
transplantation: implications for fertility preservation.
Hum Reprod Update 2010; 16(4): 395414.
9. Rodrigues P, Limback D, McGinnis LK, Plancha CE
and Albertini DF. Oogenesis: prospects and challenges
for the future. J Cell Physiol 2008; 216: 35565.
10. Zuckerman S. The number of oocytes in the mature
ovary. Recent Prog Horm Res 1951; 6: 63109.
11. Gougeon A. Regulation of ovarian follicular
development in primates: facts and hypotheses. Endocr
Rev 1996; 17: 12155.
12. Gougeon A and Chainy GB. Morphometric studies of
small follicles in ovaries of women at different ages.
J Reprod Fertil 1987; 81: 43342.
13. Thomas FH, Walters KA and Telfer EE. How to make
a good oocyte: an update on in vitro models to study
follicle regulation. Hum Reprod Update 2003; 9:
115.
14. Eppig JJ and Schroeder AC. Capacity of mouse
oocytes from preantral follicles to undergo
embryogenesis and development to live young after
405
406
407
72. Gosden RG, Mullan J, Picton HM, Yin H and Tan SL.
Current perspective on primordial follicle
cryopreservation and culture for reproductive
medicine. Hum Reprod Update 2002; 8: 10510.
73. Muruvi W, Picton HM, Rodway RG and Joyce IM. In
vitro growth of oocytes from primordial follicles
isolated from frozenthawed lamb ovaries.
Theriogenology 2005; 64: 135770.
74. Gosden RG and Telfer E. Numbers of follicles and
oocytes in mammalian ovaries and their allometric
relationships. J Zool London 1987; 211: 16975.
75. Telfer EE, Binnie JP and Jordan LB. Effect of follicle
size on the onset of apoptotic cell death in cultured
bovine ovarian follicles. Theriogenology 1998; 49:
357.
76. Gutierrez CG, Ralph JH, Telfer EE, Wilmut I and
Webb R. Growth and antrum formation of bovine
preantral follicles in long-term culture in vitro. Biol
Reprod 2000; 62: 13228.
77. Thomas FH, Armstrong DG, Campbell BK and
Telfer EE. Effects of IGF-1 bioavailability on bovine
preantral follicular development in vitro. Reproduction
2007; 133: 11218.
78. Walters KA, Campbell BK, Armstrong DG and Telfer
EE The effects of IGF-I on bovine follicle development
and IGFBP-2 expression are dose and stage dependent.
Reproduction 2006; 131: 51523.
79. Itoh T, Kacchi M, Abe H, Sendai Y and Hoshi H.
Growth, antrum formation, and estradiol production
of bovine preantral follicles cultured in a serum-free
medium. Biol Reprod 2002; 67: 1099105.
80. Hirao Y, Itoh T, Shimizu M et al. In vitro growth and
development of bovine oocytegranulosa cell
complexes on the flat substratum: effects of high
polyvinylpyrrolidone concentration in culture
medium. Biol Reprod 2004; 70: 8391.
408
Section 8
Chapter
34
Introduction
In-vitro culture systems for ovarian follicles are
enabling tools for advancing the study of folliculogenesis and the development of fertility preservation techniques. Folliculogenesis is a complex process regulated
by endocrine, paracrine and autocrine factors, and can
be difficult to study in vivo. In-vitro culture systems
provide a controlled environment in which to investigate the mechanisms driving follicle development. The
usage of these systems has produced significant discoveries about the influence of hormones, mechanics and
extracellular matrix (ECM) proteins in folliculogenesis [16]. In addition to these fundamental observations, these culture systems are providing a foundation
for the development of systems for fertility preservation for cancer survivors [7, 8]. The increase in survival rates for young women with cancer has prompted
the need for fertility preservation techniques [9]. Lifesaving cancer treatments, such as chemotherapy and
radiation, threaten fertility by diminishing the immature follicle pool and triggering early menopause. Current treatments include cryopreservation and transplantation of ovarian tissue, which incurs the risk of
re-introducing cancer cells into the patient [10, 11].
The successful development of follicle culture systems
could circumvent this risk, by allowing in vitro follicle
maturation/fertilization to obtain fertilizable oocytes
from immature follicles. Follicle culture systems have
had some success; however, further developments are
necessary to achieve the consistent growth of human
follicles to produce fertilizable oocytes [12, 13].
Stromal tissue is essential for the development of
the earliest stage follicles. The ovarian stroma con-
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
409
Antral
Secondary
Primordial
Antrum
Primary
Organ culture
(stromal cells)
Oocyte
Granulosa
Theca
410
411
and/or theca cells, have been linked to the primordialto-primary follicle transition. Lee et al. demonstrated
that BMP-7 promotes the activation of primordial follicles in vivo [38, 47]. BMP-7 was injected into the
ovarian bursa of rats, which produced a decrease in
the number of primordial follicles and an increase in
the number of primary, pre-antral and antral follicles.
Nilsson et al. achieved similar results using in-vitro
organ culture and BMP-4 [37]. Thus, these studies support the notion that stroma-to-follicle signaling exists.
412
(a)
(b)
Figure 34.2 In-vitro follicle culture systems. (a) In two-dimensional systems, follicles are cultured on flat surfaces such as tissue culture
plastic (polystyrene). The unnatural geometry/mechanics of these systems disrupts cellcell communication and causes the granulosa cells to
break though the basement membrane, migrate away from the oocyte, and attach to the two-dimensional surface. Two-dimensional systems
lack the ability to support large follicles for extended culture times. (b) In three-dimensional systems, follicles are cultured within biomaterial
scaffolds, such as alginate. These systems maintain the natural spherical geometry and cellcell interactions of the follicle. See plate section
for color version.
ture employed collagen [7074], which is an extracellular matrix protein that is prominent throughout the ovary [75]. Torrance et al. demonstrated the
growth and survival of primary mouse follicles in collagen hydrogels, but did not achieve antrum formation [71]. Later, Hirao et al. produced mature oocytes
from pre-antral pig follicles [74], and Sharma et al.
achieved antral follicles from pre-antral buffalo follicles [72]. In addition to collagen, alginate, a common tissue engineering hydrogel, has shown great
promise as a scaffold for follicle culture [1, 36, 12,
7678]. It is a naturally derived polysaccharide isolated from brown algae. Alginate is a block copolymer
composed of blocks of (14)-linked -D-mannuronic
acid (M units) and its C-5 epimer -L-guluronic acid
(G units) [79]. These blocks can be either similar or
alternating. Alginate chains can be crosslinked with
divalent cations, such as Ca2+ . As a result, alginate
avoids the use of harmful chemicals, ultraviolet light
or temperatures to crosslink the polymer network
[50]. Crosslinking occurs via interaction of the carboxylic acid functional groups in the G-blocks. This
crosslinking leads to the formation of a gel network
while retaining cell viability and the cellular interactions within the follicle. Due to its hydrophilic
nature, alginate discourages protein adsorption and
cell attachment. Thus, cells are unable to specifically
interact or bind with alginate. In order to overcome
this limitation, alginate can be covalently linked to
cell-adhesion molecules, such as RGD (Arg-Gly-Asp)
413
(a)
(b)
414
stromal cells have a significant role in the activation of primordial/primary follicles and theca cell
recruitment/differentiation. The paracrine signaling
involved in these processes is hypothesized to be a
complex time-dependent synergy of unidentified factors [20]. Cellcell contact could also have an important role in these processes. Accordingly, the simple
addition of candidate hormones and growth factors
to the culture media has not yet achieved success.
Until the mechanisms of these processes is elucidated,
follicle culture systems could attempt to incorporate
stromal cells in order to potentially culture smaller
follicles and improve growth, survival and oocyte
quality.
While only a few co-culture studies with stromal
cells have been conducted, these experiments demonstrate positive results and motivate further investigation. Osborn et al. showed that the presence of stromal cells around isolated primordial follicles improved
initial culture success [82]. Building upon this observation, Itoh and Hoshi co-cultured small pre-antral
(primary and secondary) bovine follicles with ovarian mesenchymal cells, granulosa cells and skin fibroblasts for 30 days [83]. Compared to the non-coculture controls, follicle viability was significantly
increased in all three co-cultures (18.6, 17.1 and 10.0%,
respectively) and follicle growth was significantly
increased in the mesenchymal (15.4%) and fibroblast
(30.0%) co-cultures. In a similar fashion, Wu et al. cocultured pre-antral pig follicles with different follicular cells [84]. In contrast to Itoh and Hoshis results,
Wu et al. found that small pre-antral follicle growth
and survival was inhibited by co-culture with multiple follicles (with or without oocytes). The growth
and survival of these follicles was only enhanced
when co-cultured with cumulus cells from antral follicles 3 mm in diameter. Moreover, Ramesh et al.
co-cultured buffalo pre-antral follicles with different
somatic cells (cumulus, granulosa, mesenchymal and
epithelial) [85]. Co-culture with cumulus, granulosa
and mesenchymal cells resulted in better development,
growth rate and survival than the control and epithelial cells. Maximum growth and survival was achieved
via co-culture with cumulus cells, which supports Wus
results. Therefore, these studies clearly demonstrate
the utility and effect of co-culture. Nevertheless, these
co-culture experiments have not yet been conducted in
the current state-of-art three-dimensional culture systems with optimized culture media. Hence, the impact
of co-culture could be significantly enhanced by inte-
415
biomaterial scaffolds is an effective strategy to recapitulate natural cellular environments and restore cell
cell communication.
Co-culture has also been successfully applied to
numerous cell types including skin, cartilage, bone,
liver, blood, nerve and stem cells [8791]. These experiments have demonstrated that co-culture can promote or inhibit differentiation of one or both cell
types. No correlation has yet been found between coculture methods, cell types or differentiation. Nevertheless, co-culture has proven to be effective at
improving the survival, proliferation and function of
cells. For example, Houchin-Ray et al. developed a
co-culture model consisting of primary neurons and
accessory cells in order to promote and direct neurite outgrowth [92, 93]. Co-culture promoted neuron survival and neurite extension. With respect to
reproductive technology, co-culture has been applied
to in vitro maturation (IVM) and in vitro fertilization (IVF). Oocytes and developing embryos have
been co-cultured with various feeder cells including oviduct endothelial, endometrial epithelial, fallopian tube ampullary, cumulus, granulosa and nonhuman cells such as the Vero cell line. While controversial, these experiments suggest that co-culture
may improve IVM and IVF results [9497]. Moreover,
the undifferentiated state of embryonic stem cells has
been maintained via co-culture with feeder cells such
as fibroblasts [98, 99]. In sum, these examples have
demonstrated the utility of co-culture, which potentially could have similar impacts in follicle culture.
Conclusion
The co-culture of ovarian stromal cells and follicles is
another step in the evolution of three-dimensional follicle culture systems. Stromal cell and ovary organ culture experiments have established the significant role
of stromal cells in primordial/primary follicle activation and theca cell recruitment/differentiation. Hence,
co-culture has the potential to activate individual primordial/primary follicles, which do not mature in
any in-vitro culture system and improve growth, survival and oocyte quality. Current follicle culture systems that utilize biomaterials, such as alginate, provide
three-dimensional scaffolds that maintain the natural
geometry of the follicle and could enable the translation to follicles of larger animals and humans. These
culture systems allows for the straightforward integration of stromal cells, which will help recapitulate the
416
Acknowledgements
This research was supported by grants from the
National Institutes of Health (NIH) to the Oncofertility Consortium (Grant Numbers: RL1HD058295,
PL1EB008542) and the Eunice Kennedy Shriver
National Institute of Child Health and Human Development (NICHD) through cooperative agreement as
part of the Specialized Cooperative Center Program in
Reproduction and Infertility Research (Grant Number
U54HD41857). The content of this research is solely
the responsibility of the authors and does not necessarily represent the official views of the NIH or NICHD.
References
1. Pangas SA, Saudye H, Shea LD and Woodruff TK.
Novel approach for the three-dimensional culture of
granulosa cell-oocyte complexes. Tissue Eng 2003;
9(5): 101321.
2. Kreeger P, Fernandes N, Woodruff T and Shea L.
Regulation of mouse follicle development by
follicle-stimulating hormone in a three-dimensional in
vitro culture system is dependent on follicle stage and
dose 1. Biol Reprod 2005; 73(5): 94250.
3. Kreeger P, Deck J, Woodruff T and Shea L. The in
vitro regulation of ovarian follicle development using
alginate-extracellular matrix gels. Biomaterials 2006;
27(5): 71423.
4. Xu M, West E, Shea LD and Woodruff TK.
Identification of a stage-specific permissive in vitro
culture environment for follicle growth and oocyte
development. Biol Reprod 2006; 75(6): 91623.
5. West ER, Xu M, Woodruff TK and Shea LD. Physical
properties of alginate hydrogels and their effects on in
vitro follicle development. Biomaterials 2007; 28(30):
443948.
6. Xu M, West-Farrell ER, Stouffer RL et al.
Encapsulated three-dimensional culture supports
development of nonhuman primate secondary
follicles. Biol Reprod 2009; 81(3): 58794.
7. Woodruff TK. The emergence of a new interdiscipline:
oncofertility. Cancer Treat Res 2007; 138: 311.
8. Woodruff TK. Oncofertility: Fertility Preservation for
Cancer Survivors. New York: Springer, 2007.
417
418
91. Luk JM, Wang PP, Lee CK, Wang JH and Fan ST.
Hepatic potential of bone marrow stromal cells:
419
420
Section 8
Chapter
35
Introduction
Success rates of assisted reproductive technology
(ART) have tremendously improved over the past
three decades. Nevertheless, controlled ovarian stimulation protocols are still associated with a substantial
risk of ovarian hyperstimulation syndrome (OHSS)
which, in its severe form, may result in significant
morbidity and even mortality [1]. The overall incidence of severe OHSS is 2% [2], but in women
with polycystic ovary syndrome (PCOS), who represent up to 30% of women eligible for ART [3], the
risk of OHSS is higher. These patients are therefore at
increased risk of having the embryo transfer cancelled.
Strategies to prevent OHSS in patients with a high
response to gonadotropins include the administration
of a gonadotropin-releasing hormone (GnRH) agonist
instead of human chorionic gonadotropin (hCG) to
induce the final oocyte maturation [4] and in vitro
maturation (IVM) of oocytes. In vitro maturation
avoids the risks and side effects of conventional ovarian hormonal stimulation because it involves retrieving immature oocytes from unstimulated or minimally
stimulated ovaries. After immature oocyte collection,
these oocytes are cultured, matured and fertilized in
vitro. Selected embryos are then transferred to an
adequately primed endometrium or, alternatively, are
cryopreserved and subsequently thawed or warmed in
a natural or artificial cycle.
The first successful pregnancy and birth from IVM
in human was described more than 15 years ago. In
spite of technical advances since then to the IVM protocol and improvements of the maturation method
and culture media, implantation rates do not exceed
15%, they rather stagger at 10%. This often necessitates the transfer of more than one embryo, which
in young women may increase the risk of multiple
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
421
422
423
Hormonal priming
The introduction of hCG administration prior to
immature oocyte retrieval significantly improved maturation, fertilization and pregnancy rates [17, 41, 42],
although some authors did not find a beneficial effect
of hCG priming with regard to the number of oocytes
retrieved, maturation, fertilization or cleavage rates
[33]. The exact mechanism by which hCG exerts an
effect on small follicles is still unclear. At least there
may be a technical advantage to its administration,
in that the CC expansion of the oocytes secondary to
hCG may facilitate detachment of the COC from the
follicle during the aspiration. This in turn may improve
the oocyte yield in hCG-primed IVM cycles compared
to non-hCG primed IVM cycles. Priming with hCG
may also increase the likelihood to obtain some in-vivo
matured oocytes at retrieval, which is associated with
improved blastocyst development rates, and explain
the better implantation rates and clinical pregnancy
rates.
FSH priming
Compared to the use of hCG, the value of FSH priming in IVM cycles is more controversial. Follicle stimulating hormone (FSH) priming would lead to a technically easier oocyte retrieval, higher E2 levels and
improved maturational competence of the oocytes, as
well as improved endometrial priming [43].
In normo-ovulatory patients, Wynn et al. showed
that administration of FSH during the early follicular
phase is associated with a higher oocyte maturation
rate [43], whereas others reported that FSH priming
had no apparent effect on oocyte developmental competence [44, 45]. Suikkari et al. investigated the influence of low dose FSH priming in normo-ovulatory
women starting from the late luteal phase and came to
similar conclusions [46].
Conflicting results were reached in women
with anovulatory cycles; Mikkelsen and Lindenberg
424
demonstrated improved implantation and clinical pregnancy rates in FSH-primed cycles without
additional hCG administration [47], whereas Lin
et al., who prospectively compared FSH-priming
with hCG administration versus hCG alone, found
no additional benefit of FSH-priming in those cases
where FSH and hCG were both administered [48]. In
a large randomized study encompassing 400 women,
Fadini et al. demonstrated that FSH-priming with
150 IU/day FSH for 3 days from day 3 of the cycle
plus hCG priming led to significantly improved
maturation rates, implantation rates and clinical
pregnancy rates, whereas FSH priming and hCG
priming alone showed no significant effects on clinical
outcome [49].
Metformin
Metformin improves insulin sensitivity, lowers serum
LH, total and free testosterone concentrations, and
causes an elevation in serum FSH and sex-hormone
binding globulin levels in obese women with PCOS
[54]. Significantly higher implantation and clinical
pregnancy rates were obtained in a series of 56
metformin pre-treated clomiphene-resistant PCOS
patients undergoing IVM compared to the controls
[55], suggesting that metformin may improve IVM
outcome through optimization of the oocyte microenvironment, although the mechanism by which this
occurs needs to be elucidated.
425
by ICSI and then embryo transfer. However, immature oocytes obtained from conventionally stimulated
ovaries have a lower fertilization rate [68], and the
resulting embryos have high incidence of non-cleavage
and chromosomal anomalies [69]. These observations
appear to indicate that oocytes that remain at the
GV stage after conventional ovarian stimulation are
of inferior quality or have a rare intrinsic follicular or
oocyte-specific defect.
IVM outcome
426
References
1. Delvigne A and Rozenberg S. Review of clinical
course and treatment of ovarian hyperstimulation
syndrome (OHSS). Hum Reprod Update 2003; 9:
7796.
2. Delvigne A and Rozenberg S. Epidemiology and
prevention of ovarian hyperstimulation syndrome
(OHSS): a review. Hum Reprod Update 2002; 8:
55977.
3. Child TJ, Abdul-Falil AK, Gulekli B et al. In vitro
maturation and fertilization of oocytes from
unstimulated normal ovaries, polycystic ovaries, and
women with polycystic ovary syndrome. Fertil Steril
2001; 76: 93642.
4. Engmann L, DiLuigi A, Schmidt D et al. The use of
gonadotropin-releasing hormone (GnRH) agonist to
induce oocyte maturation after cotreatment with
GnRH antagonist in high-risk patients undergoing in
vitro fertilization prevents the risk of ovarian
hyperstimulation syndrome: a prospective randomized
controlled study. Fertil Steril 2008; 89: 8491.
5. Smitz J, Picton HM, Platteau P et al. Principal
findings from a multicenter trial investigating the
safety of follicular-fluid meiosis-activating sterol for in
vitro maturation of human cumulus-enclosed oocytes.
Fertil Steril 2007; 87: 94964.
6. Banwell KM and Thompson JG. In vitro maturation
of mammalian oocytes: outcomes and consequences.
Semin Reprod Med 2008; 26: 16274.
7. Filali M, Hesters L, Franchin R et al. Retrospective
comparison of two media for in vitro maturation
of oocytes. Reprod Biomed Online 2008; 16:
2506.
8. Fadini R, Dal Canto MB, Mignini Renzini M et al.
Effect of different gonadotrophin priming on IVM of
oocytes from women with normal ovaries: a
prospective randomized study. Reprod Biomed Online
2009; 19: 34351.
9. Mikkelsen AL, Smith S and Lindenberg S. Possible
factors affecting the development of oocytes in in vitro
maturation. Hum Reprod 2000; 15: 1117.
10. Nogueira D, Ron-El R, Friedler S et al. Meiotic arrest
in vitro by phosphodiesterase 3-inhibitor enhances
maturation capacity of human oocytes and allows
subsequent embryonic development. Biol Reprod 2006;
74: 17784.
11. Albuz FK, Sasseville M, Armstrong DT et al.
Stimulated physiological oocyte maturation
(SPOM): a novel in vitro maturation system that
substantially improves embryo yield and pregnancy
outcomes. Hum Reprod 2010 [Epub ahead of
print].
427
428
429
430
Section 8
Chapter
36
Introduction
Over the last three decades there has been a significant delay in starting a family in modern Western
societies [1]. The most likely reason of this deliberate
action is extended life expectancy combined with everincreasing involvement of women in education and
the workforce. However, while men are able to reproduce until much later in life, female fertility declines
with increasing age [2, 3]. The current reproductive
paradigm suggests women are born with a finite quantity of oocytes and their reproductive potential usually does not last much beyond the end of the fourth
decade of life. This is a worrisome fact for many modern women deferring child bearing until their late thirties or even forties. In the year 2007, decreased ovarian
reserve was the sole indication for 14% of all reported
assisted reproductive technology (ART) cycles in the
USA [4]. Moreover, the female partner was older than
35 years of age in 71% of all ART cycles performed in
the same year [4].
The natural decline in fertility is markedly accelerated following gonadotoxic therapy for cancer and
other non-oncological conditions. Cancer continues
to be a major health problem despite advances in its
diagnosis and treatment. It is estimated that in 2009
approximately 713 220 women in the USA will be
diagnosed with cancer [5]. Survival rates have enjoyed
a stable increase over the last decades leading to an
increase in the number of cancer survivors every year
[5]. Similar advancements occurred in the treatment
of childhood cancers, and it is estimated that one in
every 250 adults will be a childhood cancer survivor
by the year 2010 [6]. Eventually, a growing number of
female cancer survivors are faced with the risk of infer-
tility resulting from gonadotoxic oncological treatment. Patients who are exposed to gonadotoxic agents
for the treatment of non-oncological diseases, such as
systemic lupus erythematosus, those who are undergoing surgery for endometriosis as well as women
with genetic disorders such as Turners syndrome and
fragile-X premutation face similar risks, further contributing to the population of women who need fertility preservation procedures [710].
Relatively recent advances in the field of reproductive medicine, particularly in cryopreservation methods, have rendered fertility preservation a realistic
option for such unfortunate women. With an increased
awareness of the options available for fertility preservation, a greater number of women are being offered
and are utilizing these technologies. Currently available options for preservation of female fertility are
cryopreservation of oocytes or embryos following in
vitro fertilization (IVF) or in vitro maturation (IVM)
or cryopreservation of ovarian tissue. Although successful transplantation of fresh whole ovary has been
reported in a pair of monozygotic twins discordant
for ovarian failure and encouraging results have been
achieved with frozen ovaries in animal models, cryopreservation of entire ovary has not been successfully performed in humans [11, 12]. Administration of gonadotropin-releasing hormone analogues or
inhibitors of apoptosis have been proposed as alternative strategies for patients undergoing chemotherapy;
however, effectiveness of these techniques remains to
be proved [13]. The most appropriate method for any
woman is determined by several factors including the
indication for fertility preservation, availability of a
male partner and patient preference.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
431
432
GnRH antagonist
Two to five weeks from first
consultation to oocyte
retrieval depending on time
of first consult
>14mm
Scan on
day 2
Cycle 1
day
Orgalutron/cetrotide (antagonist)
10 11 12 13
FSH/hMG stimulation
Collection
hCG
433
Advantages of IVM
In vitro maturation avoids these two important concerns which prevent a substantial amount of patients
at risk of infertility using them. Immature oocyte
retrieval in an unstimulated menstrual cycle or from
ovarian tissue biopsies followed by IVM and oocyte
or embryo cryopreservation provides a novel fertility
preservation strategy [38, 39]. Avoiding ovarian stimulation provides several important advantages for cancer patients. Compared to 25 weeks required for a
stimulated IVF cycle, immature oocyte retrieval can
be done within 210 days, depending on the patients
menstrual status [39]. Immature oocytes can be col-
434
435
Table 36.1 The number of patients to date who underwent different fertility preservation procedures at the McGill
Reproductive Centre, Montreal, Canada
Malignancy
IVM/EV
IVF/EV
IVM/OV
IVF/OV
Hematological
10
15
15
Breast
31
36
Gynecological
Brain
Sarcoma
Gastrointestinal tract
Melanoma
Autoimmune diseases
Desmoid tumor
Total
45
20
70
44
EV, embryo vitrification; IVF, in vitro fertilization; IVM, in vitro maturation; OV, oocyte vitrification.
436
Fertility preservation
strategies for women
undergoing
gonadotoxic treatment
Chemotherapy cannot be
delayed and/or hormonal
stimulation contraindicated
Chemotherapy can be
delayed and hormonal stimulation
not contraindicated
Ovarian tissue
cryopreservation
Ovarian stimulation
Mature oocyte retrieval
Male
partner
available
No male
partner
available
Male
partner
available
No male
partner
available
Embryo
cryopreservation
Oocyte
vitrification
Embryo
cryopreservation
Oocyte
vitrification
References
1. Martin J, Hamilton B, Sutton P et al. National vital
statistics reports. National Vital Statistics Reports 2009;
57(7).
2. Heffner LJ. Advanced maternal age how old is too
old? N Engl J Med 2004; 351(19): 19279.
3. Tan SL, Royston P, Campbell S et al. Cumulative
conception and livebirth rates after in-vitro
fertilisation. Lancet 1992; 339(8806): 13904.
4. Society of Assisted Reproductive Technology. Clinic
Summary Report, 2008. https://www.sartcorsonline.
com/rptCSR PublicMultYear.aspx?ClinicPKID=0.
5. American Cancer Society. Cancer Facts and Figures
2009. http://wwwcancerorg/docroot/STT/STT 0asp.
6. Bleyer WA. The impact of childhood cancer on the
United States and the world. CA Cancer J Clin 1990;
40(6): 35567.
437
438
439
Section 9
Chapter
37
Future technologies
Introduction
Stem cells
Stem cells are undifferentiated cells characterized by
their ability both to renew themselves and to give rise
to various types of specialized cells. Adult stem cells are
multipotent, i.e. they have the ability to differentiate
to a limited number of cell fates, and can be obtained
from various tissues, including blood [6], bone marrow [7], fat [8], skin [9] and testis [10].
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
440
Epigenetic regulation
The DNA code is more or less the same in all the cells
of the human body. What differs is the way this code
is utilized or, in other words, which combinations of
genes that are active in a specific cell at a given time.
The secret behind this is the epigenetics, the heritable
changes in gene function that occur without a change
in the sequence of the DNA [29, 30]. This includes a
wide range of mechanisms, such as DNA methylation,
histone modifications, genomic imprinting and higher
order chromatin conformation [29]. The overall epigenetic state of a cell is referred to as the epigenome.
The creation of the diverse cell types from pluripotent
stem cells depends ultimately on the establishment and
maintenance of specific patterns of gene expression,
a process under careful control of epigenetic features.
This information can be reversed and changed, but also
passed on through cell divisions and generations, such
as an epigenetic memory.
There is an extensive cross talk between different
types of epigenetic regulation that carefully adjust the
level of gene expression at any given moment. Some
adjust rapidly upon a stimulus and others remain
steady through cell divisions, distinguishing different
cell types. This fine-tuned control of gene expression is
the key factor behind stemness and differentiation, and
it is an absolute necessity that we learn to understand
these features in order to continue making progress
in the field of stem cell research and differentiation.
What is the difference between various types of cells?
How many different cell types are there in the human
body? This is a key question if we are to create one kind
of cell from another. However, first we need to define
cell type. Depending on who answers, the answer will
probably range from a few hundred to a few million. The epigeneticist would most likely state that
there are as many types of cells in the human body
as there are number of cells in total, because no cell is
epigenetically the same as another. The approximately
2 m of DNA is modified and folded in an unique way
441
442
443
444
restored the somatic diploid chromosome complement and developed into blastocysts.
it is always changing (as responses to various stimulus, from within the cell or the surrounding), and this
dynamic feature is technically challenging to investigate. In particular, there are very few single cell analyses available, where the epigenome of one induvidual
cell could be measured.
The use of stem cells has provided a research tool to
study development and disease in details never before
possible. But even though treatment using stem cells is
giving great hope towards the cure of various human
disorders, such as leukemia, brain damage, cancer and
infertility, there are many fundamental questions that
still need to be answered, not least at an epigenetic
level. How do we ensure that cells differentiated in vitro
have the same phenotype as corresponding cells differentiated in vivo? Which are the fundamental genetic
and epigenetic steps during differentiation and development?
In the future there may be potential for using
gametes differentiating from hESCs or hiPSCs as a
therapy for infertile couples. The advantage of using
iPSCs, compared to hESCs, is the hope to generate
patient specific cells, where somatic cells such as skin
cells from a patient could be utilized. The main benefit
to this would be that men and women lacking egg and
sperm, for any reason, could potentially get the opportunity to conceive children that are biologically their
own. The major obstacle for the moment is the technical difficulties deriving a sufficient amount of iPSCs
from differentiated cells without using a virus and/or
risking epimutations in the host genome.
References
1. Marques-Mari AI, Lacham-Kaplan O, Medrano JV,
Pellicer A and Simon C. Differentiation of germ cells
and gametes from stem cells. Hum Reprod Update
2009; 15(3): 37990.
2. Ziebe S, Loft A, Petersen JH et al. Embryo quality and
developmental potential is compromised by age. Acta
Obstet Gynecol Scand 2001; 80(2): 16974.
3. Howe G, Westhoff C, Vessey M and Yeates D. Effects
of age, cigarette smoking, and other factors on fertility:
findings in a large prospective study. Br Med J (Clin
Res Ed) 1985; 290(6483): 1697700.
4. Lampe H, Horwich A, Norman A, Nicholls J and
Dearnaley DP. Fertility after chemotherapy for
testicular germ cell cancers. J Clin Oncol 1997; 15(1):
23945.
5. Joki-Korpela P, Sahrakorpi N, Halttunen M et al. The
role of Chlamydia trachomatis infection in male
infertility. Fertil Steril 2009; 91(Suppl. 4): 144850.
445
446
447
Section 9
Chapter
38
Future technologies
Artificial ovary
Christiani A. Amorim
Introduction
The reader may feel it is somewhat fanciful to see a
chapter on artificial ovary assembly for further transplantation, when no studies have been published on
this topic to date, and no one has yet been able to
endorse its feasibility. However, we believe it is the
right time to consider the artificial ovary as a promising technology in reproductive medicine, as it could
represent an alternative to ovarian tissue transplantation. In addition, positive results with different types
of artificial organs and tissues, such as bone, cartilage,
skin, heart, liver, kidney, brain, cornea and teeth, indicate that construction of an artificial ovary is wholly
achievable. Since there are no available studies on artificial ovary assembly for grafting purposes, this chapter may provide the starting point for this technology,
where indications, advantages, strategies, techniques
and possibilities will be discussed.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
448
Advantages
Although the main concern is being able to provide an
alternative to restore fertility in patients who cannot
benefit from ovarian tissue transplantation, the assembly and grafting of an artificial ovary would offer additional advantages compared to transplantation of ovarian tissue.
were found to increase vessel density, size and maturity when added to scaffolds grafted to mice [14].
According to Bergmann and West, these factors could
well influence the differentiation of mesenchymal stem
cells from the bloodstream into endothelial cells and
encourage microvascularization [10].
449
450
interactions between GCs and oocytes, which is essential to regulate follicular growth and development. The
3D arrangement of follicles is also influenced by cell
migration and proliferation, induced by interaction of
the scaffold and cells.
In order to modulate cell adhesion, behavior and
function, polymer properties, such as crystallinity,
morphology and surface, can be modified. Some of
these important properties are discussed below.
Crystallinity
Crystallinity refers to the degree to which molecules of
a polymer are oriented toward a repeating model. It is
therefore favored by polymers with a chemically simple structure. Crystallinity may influence the response
of cells to the scaffold, since it affects several surface characteristics, such as polarity and irregularity
[32]. Degirmenbasi et al. reported that growth rates of
fibroblasts varied according to the crystallinity range
of poly(L-lactide) (PLLA); when the polymer was
highly crystalline, fibroblasts showed lower growth
rates [33].
Morphology
Scaffold topography and dimensionality play an
important role in cell behavior. Topography can influence the morphology of cells, which consequently
affects their orientation, proliferation, gene expression
and function. Studies have shown that cells behave in
a completely different manner when cultured on irregular surfaces (e.g. edges, grooves and ridges) rather
than smooth surfaces [32, 34]. For example, ridges can
influence cell orientation and migration of different
types of cells. This cellular locomotory response is
termed contact guidance. Regarding dimensionality,
preservation of 3D structure is not only important for
follicles, but also for cells. A 3D scaffold would provide
normal polarity and spatial regulation of cells, and it
may also influence the cellular response to existing
physical and biochemical signals [32].
Surface
In tissue engineering, adhesion of cells to the scaffold surface is crucial because their activities (proliferation, migration, differentiation, etc.) depend on this.
In order to improve cell attachment and thereby its survival and behavior in the material, modifications can
be made to the scaffold surface. The advantage of treating polymer surfaces is that only the external part of
the material is changed, preserving the polymer bulk.
This is extremely useful, since materials with satisfactory bulk properties usually do not have the surface
characteristics required for clinical application [35]. In
this way, different polymers can be combined for specific purposes (i.e. surfaces with superior biocompatibility and bulk, with suitable mechanical properties).
Methods such as addition of adsorbed proteins
and immobilized functional groups have been proposed to alter the surface chemistry of scaffolds. Polymer surfaces can be pre-treated with matrix proteins
(fibronectin, collagen, laminin, etc) to increase cellular adhesion, which affects scaffold biocompatibility.
Although ECM proteins show high adhesiveness and
adsorb to practically all polymer surfaces, they may
induce an immunoresponse in the host, since they are
usually prepared from animal tissues. An alternative
could be isolation and purification of these proteins,
but the procedure is time-consuming and very expensive [31].
Incorporation of small biologically active functional groups, for example oligopeptides, saccharides
and glycolipids, has also been attempted with the
aim of isolating some features of ECM molecules
associated with cell adhesion [34]. Some amino acid
sequences, such as Arg-Gly-Asp (RGD), which is the
cell-binding domain of fibronectin, play an essential
role in cell binding and mediate cell adhesion. RGD
is not exclusively linked to fibronectin; it can also be
found in several ECM proteins, such as collagen, vitronectin and laminin. In addition, Tyr-Ile-Gly-SerArg (YIGSR) and Ile-Lys-Val-Ala-Val (KVAV) laminin
sequences also show cell binding activity and appear to
mediate adhesion in some cell types [34].
Alternatively, chemical groups can be added
through plasma, which consists of highly excited
atomic, molecular, ionic and radical species [35].
Plasma treatment has been used to attach chemical
groups or atoms to material surfaces. Use of oxygen
plasma, for example, has been shown to increase
the hydrophilicity of materials [35], which in turn
increases material biocompatibility. Addition of
chemical groups usually changes surface wettability
and, consequently, improves cell adhesion [34]. Wettability can be defined as the ability of a liquid to
adhere to a solid and spread over the surface. The
surface wettability of a material is a measure of
its hydrophilicity, hydrophobicity and surface-free
energy [32], and it is associated with the crystallinity
and chemical composition of the material [36].
Studying different types of mammalian cells, in 1978,
451
Porosity
Scaffolds of appropriate size with good pore distribution and interconnectivity are essential for uniform cell seeding and distribution and tissue ingrowth.
Porosity also influences invasion and migration of cells
from the host, which positively affects biodegradation
and neovascularization of the scaffold. In addition, an
interconnected pore network is necessary for transportation of signaling molecules, growth factors, oxygen and nutrients, as well as metabolic waste removal.
Another parameter to bear in mind is follicular
diameter. Human primordial follicles have a diame-
452
Bioactivity
In order to assemble an artificial ovary, the scaffold
should act as a vehicle to graft isolated follicles, ensuring preservation of their 3D structure. However, it
should also have a bioactive function, regulating the
growth of these follicles. It is known that many biologically functional molecules, ECM components and
cells interact on the nanoscale, creating a highly specialized microenvironment that is essential for proper
cell development and continued function. For this reason, in order to induce and coordinate folliculogenesis in a grafted scaffold, it is necessary to program it
to deliver bioactive molecules, such as factors that may
positively influence neovascularization, follicle growth
and development and oocyte maturation. These factors
could be added using polymeric systems, which would
allow regulation of the localization, duration, delivery and availability of different inhibitory and growth
factors [42]. To this end, several strategies have been
applied, such as multiple levels of encapsulation, noncovalently bonding bioactive factors to peptides with a
range of dissociation constants that mimic the immobilization of growth factors in the ECM [43]. Encapsulation, for example, can protect these factors from
denaturation that could occur if they were directly
adsorbed onto the scaffold, which would result in their
complete degradation in a very short space of time.
The released quantity of factors may be modulated
by the encapsulated quantity in the microspheres, by
the number of microspheres incorporated in the scaffold or by the composition of the microspheres. Thus,
microspheres containing different factors implicated
in folliculogenesis, factors mitigating ischemic damage, as well as factors involved in angiogenesis can be
tested.
Vascularization
Vascularization is a crucial element in the success of
a scaffold, required to supply oxygenation and nutrients to grafted cells and follicles and remove metabolic
waste. However, formation of new capillaries in the
material is very challenging, as it necessitates interaction of different design properties, such as choice of
material, porosity and pore interconnectivity, bioactivity and biodegradation. When follicles and/or cells
are grafted with a scaffold, they are only oxygenated
by simple diffusion, which may be limited by the construct [10]. This oxygen and nutrient deprivation may
cause cellular damage and apoptosis. An additional
concern is that neovascularization may be slower
than cell proliferation and follicle development in the
grafted scaffold, creating a higher demand for oxygen
and nutrients than the new capillaries can meet, which
can also lead to follicular death.
Several strategies can be applied to promote vascularization. As for transplantation of ovarian tissue
fragments, the scaffold can be grafted to a peritoneal
window close to the ovarian vessels and fimbria. This
window should be created a few days earlier in order
to induce angiogenesis and neovascularization in the
area destined for scaffold grafting. Alternatively, the
cortex of the ovary remaining after cancer treatment
could be removed to place the scaffold in direct contact with the medulla, an extensively vascularized area
(for a review, see Donnez et al. [1]). Grafting of autologous endothelial cells may also be an option to promote vascularization in the scaffold, as well as addition of growth factors involved in neovascularization
(bFGF, VEGF, PDGF), as previously discussed (for a
review, see Bergmann and West [10]).
Biodegradability
Ovarian follicles are exceptional in that they can grow
to around 600 times their size during folliculogenesis
(the human follicle grows from 30 m in its primordial stage to 1824 mm when it is ready to ovulate). In
addition, they recruit cells and vessels to support their
development. An ideal scaffold would need to degrade
in order to allow exponential growth of follicles, formation of vessels, and proliferation of stromal cells.
Ideally, the artificial ovary should offer an appropriate
initial environment for follicles that would be replaced
by a new ovarian-like structure after a few weeks of
grafting. The degradation rate of the scaffold is thus an
essential parameter in the success of grafting.
Although biodegradation should be conceived
with follicular development in mind, other factors
should also be taken into account. The biodegradation rate cannot be faster than cell migration and proliferation or ECM synthesis and stabilization in the
scaffold, because cells would lose physiochemical fac-
Biocompatibility
Biocompatibility is one of the most important characteristics of a scaffold and it has been shown to be
related to some of the above-mentioned parameters,
such as the degradation rate and byproduct and material wettability. Material biocompatibility can be evaluated by measuring the duration of adverse variations in
the homeostatic mechanisms that determine the host
response [45].
Scaffold grafting inevitably causes damage to surrounding tissue, inducing an inflammatory response,
foreign body reaction and cascade of wound healing
[45]. Depending on the magnitude of the inflammatory response triggered by the scaffold, the migration
of cells such as macrophages and neutrophils to the
grafting site may have undesirable effects on the success of the scaffold. These cells, involved in the inflammatory reaction, can secrete enzymes that increase the
degradation rate of the scaffold, and molecules that
453
Scaffold handling
Having a scaffold lacking the mechanical strength to
be handled during surgery would not be ideal. For
instance, although hydrogels have the advantage of
mimicking the natural ECM, they usually do not have
adequate mechanical strength and may be damaged or
even destroyed during handling.
Mechanical strength is affected by scaffold composition, pore size and porosity. Therefore, while
hydrophilic and very porous materials are probably the
best choice for grafting isolated follicles, the effect of
these parameters on the mechanical properties of the
scaffold should also be taken into consideration.
454
Among the many applications of tissue engineering, the most important for artificial ovary technology is the possibility of having a ready-made substitute for the ovary by seeding the patients own cells
and follicles to a biodegradable scaffold, with a view
to tissue regeneration or construction of a new ovary.
The question of scaffold manufacture is therefore crucial. Fabrication approaches must not only replicate
the properties of the ovary at the macroscopic level, but
also recreate nanoscale details observed in native tissue at the cellular level. Different synthetic and natural
polymers can be used for this purpose. Although natural polymers, like hyaluronic acid, poly -glutamic
acid and collagen, exhibit high biocompatibility and
biodegradability, they may pose a risk of antigenicity and show variations according to batch production. On the other hand, synthetic polymers, such
as alyphatic polyesters, polyanhydrides, polypropylene fumarates and polyphosphazenes, offer outstanding reproducibility, the ability to control degradation
kinetics, and high mechanical strength [47]. However,
they may show low biocompatibility compared to natural substances.
To process the different materials into porous scaffolds, several techniques have been developed [46, 48
50]:
r Gas foaming A biodegradable polymer is
saturated with carbon dioxide (CO2 ) at high
pressures. The solubility of the gas in the polymer
is then decreased rapidly by bringing the CO2
pressure back to atmospheric levels. This results in
nucleation and growth of gas bubbles.
r Fiber bonding/fiber meshes This technique
increases the mechanical properties of a scaffold
by dissolving PLA and casting it over a PGA mesh,
for example. The solvent is allowed to evaporate
and the construct is then heated to exceed the
melting point of PGA. Once the PLAPGA
construct has cooled, the PLA is removed by
dissolving it again. This treatment results in a
mesh of PGA fibers joined at the crosspoint.
r Phase separation The polymer solution
separates into two phases, a polymer-rich phase
and a polymer-lean phase. After the solvent is
removed, the polymer-rich phase solidifies.
Biologically active molecules can be added to the
polymer solution.
r Melt molding One of the techniques involved in
this process involves filling a Teflon mold with
Prototype failure
Prototype failure
ANIMAL
MODEL
IN VITRO
STUDIES
Scaffold
conception:
choice of
material and
fabrication
method
Prototype
Biocompatibility
tests
Toxicity testing
for follicles and
ovarian cells
Prototype
adaptation
(if necessary)
Biodegradation
tests
Follicular survival
and development
Biodegradation
tests
Neovascularization
studies
CLINICAL
TESTS
Figure 38.1 Schematic approach to the development of an artificial ovary.
455
Conclusion
Having provided a comprehensive description of the
assembly of an artificial ovary and the involvement of
tissue engineering strategies, we find ourselves at the
starting point of a new technology that may be termed
ovarian tissue engineering. The aim of this strategy is to replace not only the structure of lost tissue,
but rather its function, to potentially allow endocrine
activity and fertility to be restored in cancer patients.
Ovarian tissue engineering research should explore
different alternatives, involving a variety of materials that can be tested alone or in combination, techniques that can turn these materials into functioning
scaffolds, and many other factors that may be combined to give these scaffolds optimal bioactive properties. This will require numerous in vitro and in vivo
experiments on animal models, with data quantifiable
by functional and structural endpoints (Figure 38.1).
It is therefore likely that this innovative approach will
prove very challenging, probably taking many years
or even decades to achieve successful results. However, since it may represent a viable option for reestablishing fertility in cancer patients, it should be
extensively investigated.
References
1. Donnez J, Jadoul P, Squifflet J et al. Ovarian tissue
cryopreservation and transplantation in cancer
patients. Best Pract Res Clin Obstet Gynaecol 2010;
24(1): 87100.
2. Meirow D, Hardan I, Dor J et al. Searching for
evidence of disease and malignant cell contamination
456
457
458
Section 9
Chapter
39
Future technologies
Introduction
One of the consequences of cancer therapies, including radiation and chemotherapy, is gonadotoxicity. As
effective treatments have rendered a number of malignancies curable, or have delivered long-term survival,
post-treatment fertility has emerged as an important consideration for patients and their healthcare
providers. Unfortunately, there are currently no definitive ways to limit the injurious effects of these treatments on gonadal function, other than shielding the
gonads from direct exposure to ionizing radiation.
Suppression of gonadotropin secretion may have a
protective effect in some populations, but the efficacy
of this intervention for preserving fertility remains to
be conclusively established.
The banking of germ cells or embryos prior
to treatment represent options for preservation of
fertility. Sperm cryopreservation is a longstanding
option for sexually mature males, and embryo cryopreservation is an option for some women and couples. Though recent successes with cryopreservation
of ovarian cortex or oocytes are encouraging, options
for gamete or embryo preservation are more complicated for women, and entail procedural risks and
expense. Moreover the technologies have not advanced
to the point that female options for fertility preservation are as successful in outcomes as pre-treatment
sperm cryopreservation. The decision to pursue ovarian cortex or oocyte banking is complicated by the fact
that the gonadal response to radiation and chemotherapy varies among the population, and there is, at
present, no precise way to determine who will suffer irreversible damage and who will emerge from
treatment with fertility intact, obviating the need for
pre-treatment interventions. Additionally, preserving
the ovarian germ cell complement per se, while an
important determinant of fertility, does not necessarily insure it. Furthermore, the inability to make predictions regarding the extent of post-treatment gonadal
function impacts the design of research on interventions to spare fertility. Evaluations of such interventions could be conducted more efficiently with
smaller sample sizes based on a more precise knowledge of subject risk for significant post-treatment
gonadal dysfunction. In this chapter, we provide a
framework for thinking about factors that can predict gonadal function post-cancer therapy, focusing
on genetic contributions and reviewing outstanding
issues that need to be addressed in future research
with the aim of developing patient-specific algorithms that are predictive of post-treatment fertility or
infertility.
Predicting the weather requires knowing what the
current weather conditions are in the specified location such as air temperature and humidity, what fronts
and air masses are approaching and what other conditions might affect them. Local geography and previous
weather patterns must also be taken into consideration. These are all elements that a meteorologist must
assess before predicting future weather conditions.
Using the weather prediction rubric, we can identify
four general requirements for accurately predicting a
future biological event such as preservation of ovarian
function after cancer therapy:
1. The stability or flux of the current condition must
be known (i.e. baseline fertility, germ cell
complement and rate of germ cell depletion).
2. Knowledge of what events or conditions could
change the stability or rate of flux (i.e. types of
treatment, dosing, duration).
3. The likelihood of those events or conditions
happening (i.e. epidemiological data on
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
459
460
the majority of follicles are in a resting state at the primordial stage of development. The complement of primordial follicles has been called the resting pool [2]
and represents ovarian reserve or the population of
oocytes that have not yet become committed to the
path of growth and ovulation. Since follicles (and eggs)
are progressively lost from the resting pool over time
by entering the growing pool or undergoing apoptosis,
the ovarian reserve is constantly declining over time in
women. In general, women with a diminished ovarian
reserve are considered to have reduced fecundity [3].
Decay curves modeling the decline of the resting
pool of follicles have been presented in the literature
in mice and humans [4, 5]. In women a bimodal decay
was described, with a sharp increase in the rate of follicle loss at an average age of 38 years. However, a
more recent model suggests that the rate of follicle loss
or resting pool decay is one of slight constant acceleration throughout the perimenopause [6]. Recently,
investigators have explored the impact that radiation
and chemotherapy have on shifting this follicle population decay curve to the left [7]. This model describes
diminished ovarian reserve ensuing at an earlier age
after the toxic treatments.
The ability to predict the consequences of decay
in the germ cell population depends upon determining a womans current ovarian reserve, and there is
presently no reliable method to accomplish this task.
Antral follicle counts can vary in the hands of different
observers, and biomarkers such as anti-Mullerian hormone and inhibin B suffer from the lack of standardized assays and insufficient normative data. Moreover,
predicting how that reserve might change over time is
even more challenging.
There are several environmental and genetic
conditions with clearly defined deleterious effects
on ovarian reserve, including exposure to environmental/occupational toxins like 4-vinylcyclohexene
diepoxide, heavy tobacco use, specific X-chromosome
deletions and genetic variants (e.g. FMR1 permutations). When present, these might reasonably be
expected to increase risk of ovarian failure following
exposure to a gonadotoxic therapy. However, there
are large number of genes involved in human ovarian
development and follicular growth and their potential
contributions to variability in response to cancer
therapies are largely unknown. There are also other
factors that could affect ovarian function when compounded with a gonadotoxic cancer therapy, whose
impact is less well understood including exposure to
r
r
r
r
r
r
r
BMP-4
Smad1
Fragilis
Stella
GATA4
PUM2
DAZLA
c-kit
kit ligand (SCF)
SOX3
BMP/GDF-9
TGF
TNF
LIF
SDF-1
CXCR4
Laminin
Fibronectin
POG
TGF
TIAR
FMR1
PIN1
TIAR
LIM (Lhx9)
SF1
WT1
DAX
FIG
FOXL2
Cadherin
1 integrin
Nobox
IGF2bp2
Maelstrom
PTEN
TSC1
Foxo3a
P27
ZP 13
Follicle growth
r
r
r
r
r
r
r
r
r
r
r
r
r
r
r
r
r
r
Slc18a2
Smad3
FSHR
HSD17b1
Cyp11a1
Hsd3b1
Ihna
Dax1
Greb1
LHx8
Nrf2
Gdf-9
Mater
AMH/MIS
Activin
BMP-15
Fog2
ESR2
461
462
Outstanding questions
In order to build a clinically useful patient specific
prediction algorithm for post-treatment infertility, we
Autosomal
X-linked
FOXL2
FMR1
NOBOX
GDF-9
FSHR
FMR2
AIRE
DIAPH2
ATM
XPNPEP2
POLG
BMP-15
NR5A1
MSH5
NOG
INHA
FOXEI
B-glycan
PTHB1
AR
DMC1
ESR1
PCMT1
MCKDHB
ASCL6
PGRMC1
FIGLA
DMCI
SALL4
PTEN
TGFMR3
with fertility differences, suggests that genomic analysis might be useful in fertility prediction.
Although there are genes and markers that are
associated with early ovarian senescence, there is limited data about the predictive value of testing for such
information. But the discrete limit of ovarian function
does create the desire to predict its course over time.
This academic pursuit has had a renewed driving force
from cancer survivors that want to predict accurately
if they will be able to have children after their therapy.
Should they delay cancer treatment to undergo expensive and unproven therapies to hold on to the possibility of future reproduction? Or should they get on with
their therapy and know that in all likelihood they will
continue to have ovarian function after chemotherapy
or radiation?
The complexities of the events of fertility go beyond
ovarian function. External factors such as individual
choices in fertility management or choice of a partner
also affect ultimate fecundity and fertility rates [32], as
well as factors such as fertilization opportunities, uterine function and implantation events [33]. Since predicting fertility is fraught with confounding factors, it
may be more practical clinically to attempt to predict
the likelihood of infertility after agents destructive to
oocytes are administered.
463
5q
35.2
5q
14.115
6p
24.2
9q
21.3
13q
34
19q
13.42
20p
12.3
464
References
1. Lee SJ, Schover LR, Partridge AH et al. American
Society of Clinical Oncology recommendations on
465
37. Moron FJ, Ruiz A and Galan JJ. Genetic and genomic
insights into age at natural menopause. Genome Med
2009; 1(8): 76.
466
Section 10
Chapter
40
Introduction
Patients whose fertility would be impaired due to
treatment for their medical condition face a daunting
task and need timely, accurate information about their
options. They need to know the short and long-term
side effects of medical treatment on their reproductive
system prior to initiating adjuvant chemotherapy. They
may require help finding fertility specialists. Many of
the currently available strategies to sustain fertility
are far from certain to succeed and fertility-sparing
options may carry their own risks. Cancer patients
may need psychological support for emotional distress
occurring because of the cancer diagnosis and/or for
psychological distress which exacerbates prior emotional difficulties. This chapter will focus on what we
know about the emotional needs of cancer patients and
others for whom fertility preservation is an option, as
well as the implications for cancer care providers. We
will discuss how to recognize psychosocial distress and
the type of communication skills that are necessary
when counseling patients. Finally, we will describe the
ethical implications of fertility preservation techniques
and provide resources for the healthcare provider.
studies on patients experiences with fertility preservation have explored the issue in the context of sperm
banking [12]. At present, no studies exist to explore
systematically the emotional impact and effects of procedures such as oocyte freezing and ovarian or testicular cryopreservation [1, 36]. We do know that
fertility preservation is an important issue for cancer
patients [12, 710]. We also know that health professionals, patients and parents consider fertility preservation an important option for young cancer patients
[12, 1012]. Nevertheless, all parties involved in cancer care have information deficits and, despite the need
for counseling patients about fertility preservation, it is
not routinely offered to all patients needing it [1, 2, 8,
1318].
Individuals diagnosed with cancer during their
reproductive life span experience a life crisis in two
respects. Firstly, the diagnosis in itself may evoke emotional distress [19, 20]. Secondly, cancer treatments
may impair fertility and have profound implications
for the patients quality of life after treatment. We
know from investigations of infertility in non-cancer
patients that infertility in itself can be associated
with grief and depression, increased anxiety [21] and
reduced life satisfaction [2224]. Some patients may
experience infertility with the same emotional distress
as is typical of an AIDS diagnosis [25]. Although cancer survivors may be able to become parents though
egg and embryo donation, gestational surrogacy or
adoption, these paths to parenthood may not be available or may be prohibitively expensive. In the case of
adoption, survivors may face discrimination in their
efforts to adopt, particularly with international adoption, because of their history of cancer [26]. Many
countries, for instance, China, will not place a child
with an individual or family who has had a history of
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
467
Women
A young woman with breast cancer must time her
appointments with a fertility specialist to fit with her
menstrual cycle and is often seen by her reproductive physician between surgery and chemotherapy. She
may have feelings about surgery in itself, fear losing
her life and dread losing her hair or suffer other worries about her body. She may experience the pain of
reduced self-esteem, decreased sexual feelings and she
may fear rejection from her current or future partner
[2931]. She may have had difficulty locating a reproductive endocrinologist. She must find the funds to pay
for fertility treatment along with the money needed for
cancer care, and she has no guarantee that the fertilitysparing treatments will work. A young woman without a partner may have to decide between freezing eggs
or embryos. If she chooses embryos, there is the additional issue of choosing a sperm donor and, if she has
a boyfriend, the complexity of anticipating the impact
of her choices on her current and future life. She may
not want to pick the father of her future children during this stressful time. She may worry about how pregnancy will affect her life (e.g. if she has hormonally
sensitive cancer) or the health of her children. Should
she see a genetic counselor? She may feel isolated and
alone, not part of the reproductive community and
too young for such a serious illness. If single, she may
worry about discussing her fertility status with a desirable mate, fearing rejection from her current or future
boyfriend and his family [29]. Cancer diagnosed during a young womans reproductive life may occur just
when she has worries about ever finding Mr. Right
accompanied by questions such as Whats wrong with
me? Potential loss of fertility can add to her already
shaky self-esteem and worries about her desirability
as a mate. Friends and relatives may try to reassure
with platitudes, Just be glad youre alive or You can
always adopt, not understanding the emotional complexity she is suffering. If she has children, she may
have work or have child-care pressures and may find
the many medical appointments to be a burden. She
468
may worry that she is not able to take care of her family as she would wish. For some women, the possibility
of not being able to complete her desired family size
hurts, and comments to the effect that You should
be glad you already have a child are very painful.
Although cancer usually brings couples closer, cancer
can occur during a stressful time in a couples life. If a
couple is in the midst of a stressful period, they must
decide whether or not to freeze oocytes or embryos or
both, and will this decision exacerbate their stress?
Men
In addition to anxiety and distress about cancer itself
[19], male cancer patients may worry about how
treatment will affect their masculinity, role, identity
and sexuality [29, 32]. If the patient anticipates role
changes because of cancer treatment (e.g. his partner must take care of him) these changes in domestic arrangements may affect how the patient feels
about himself and may exacerbate pre-existing couple conflict as well as negatively impacting on sexuality. Younger couples may find it particularly difficult to adapt to new domestic roles and to experiencing the many life and financial issues associated with
medical treatment [29]. Even under the best of times,
some couples find it difficult to discuss their intimate
lives with each other, and may find it especially challenging to discuss these concerns with their physicians
and medical staff. If treatment will cause infertility,
many men worry about the impact of infertility on
their present and future lives and fear for their childrens health [29].
Adolescents
Adolescence is challenging time to help young men
and women make appropriate decisions regarding fertility preservation and sperm or oocyte freezing. The
caregiver must be sensitive to the impact of the cancer on the patient. An adolescent may be concerned
about his or her body, appearance and sexuality [29].
In addition, this is a period where relationships with
peers and parents are changing and identity formation is occurring. Cancer may affect the patients sense
of value of him- or herself [28, 29]. Adolescents may
be pessimistic about their future, disillusioned with
parenthood and afraid of transmitting cancer to their
offspring [28, 29]. Balancing parental desires and the
authentic needs of the patient is a challenge. Is the
patient feeling embarrassed, afraid of failure, rejecting
Children
The challenges associated with fertility preservation
are even more pronounced with children. We know
parents usually want to preserve fertility for their children. Van Den Berg et al. interviewed the parents of
318 boys at the time of diagnosis and 2 years later [34].
They found that 61% of the parents would approve of
spermatogonial stem cell cryopreservation collected at
time of diagnosis. However, children may be too young
to comprehend the fertility implications of their cancer
treatment and parents may want to shelter their children from such discussions [35]. In addition, parents
may not know how to find specialized centers working with ethics board-approved research protocols for
fertility preservation involving tissue cryopreservation
for children. Presently, it is difficult for the caregiver to
counsel parents about the future efficacy of such experimental procedures.
469
initiated discussions about fertility preservation themselves [44]. Partridge et al. found that 26% of women in
a web-based survey of 657 young breast cancer patients
felt that their concerns about fertility and reproduction
had not been adequately addressed at the time of diagnosis [8]. Oosterhuis et al. surveyed 97 parents of pediatric patients and 37 adolescent patients and found
that only 29% of the parents and adolescent patients
were satisfied with the information they received about
fertility-sparing options [42].
There is a debate about the meaning of lack of
recall about having been provided information (or
not) about fertility preservation, with evidence on each
side of the debate. Most simply, a lack of recall of any
discussion may indicate that information was not provided. In Duffy et al.s investigation, 34% recalled discussion about fertility-sparing options and 100% of
the patients recalled discussions about the impact of
chemotherapy on their everyday lives [15]. If the difference in recall is due to information given (but not
recalled), why do 100% of the patients recall discussing
the effects of chemotherapy on their everyday lives?
On the other side of the debate, Van Den Berg et al.
investigated 202 parents of 117 male childhood cancer
patients who had been informed about fertility during
consent [43]. Only 50% recalled statements about fertility and 36% denied receiving any information [43].
We know that anxiety and emotional distress can
impair the ability to register information [45]. We also
know that anxiety can inhibit information processing
[46, 47]. In addition, lack of information may lead to
emotional distress [4850]. Thus, a patients lack of
recall may be due to many factors, not simply due to
provider omission. Nevertheless, we know that there
are challenges to effective communication between
cancer care specialists and their patients. Quinn et al.
found that the physicians in their investigation usually
discuss fertility loss as a side effect of treatment for cancer [14, 51]. However, few actually provided specific
information for their patients to preserve their fertility.
Reasons cited by Quinn et al. [14, 51] and others [e.g.
8, 17, 52] include a lack of knowledge of options for
fertility preservation, a challenging medical setting, a
lack of time for the discussions and a lack of training in
how to discuss fertility issues [17]. Providers also indicated that gender and cancer site as well as the costs
of fertility preservation procedures and access to the
necessary resources were factors that influenced their
decisions regarding discussion about the full range of
options available [14, 51, 52].
470
At this point, there is general consensus that fertility preservation is very important to patients and
that patients affected by cancer during or prior to their
reproductive life span should be informed about possible fertility impairment due to cancer treatment [53
57]. Adult patients, parents of minors and (if possible)
minors should be provided up-to-date realistic expectations about the success rates of fertility preservation,
the cost of these procedures and referral to appropriate facilities. Parents of minors should be referred to
appropriate specialized research centers. The discussion of fertility-sparing options should address common patient concerns about whether or not fertility
preservation will decrease successful cancer treatment
or be harmful to the patient or to the patients offspring, short and long-term side effects of treatment on
the reproductive system, pregnancy risks and concerns
about the possibility of genetic risks to the offspring. In
addition, physicians and the cancer team should discuss the option of deciding not to preserve fertility.
We know that physician recommendations about
fertility preservation are very influential. Schover
et al. found that physicians encouragement to bank
sperm was almost as strong a predictor of whether
or not a patient banks sperm as the patients own
desire for future children [10]. Schover et al. noted
that the influence of provider recommendations is
consistent with what is known about the effectiveness of physician recommendations about smoking
and early cancer screening [10]. Saito et al. found
results consistent with Schover et al.s findings [41].
They interviewed 25 cancer survivors who had banked
sperm at their own initiative and 26 patients who
had banked sperm with their physicians recommendation. Patients who banked sperm with their physicians
instructions felt better about having cyropreserved
sperm. These results underscore the importance of
the cancer care teams communication and encouragement in exploring options to preserve the capacity for
biological parenthood.
investigated the impact of patient-rated provider communication skills with 990 cancer patients [60]. They
found that 45% of these patients reported that some
aspect of communication with their physicians was
unclear and 59% of the patients wanted to speak to
their medical team more frequently. Age was a factor in patient satisfaction. Patients under 50 years of
age rated social and psychological help as more important than medical information. Patients who reported
that information was incomprehensible or incomplete
described significantly worse quality of life on 17 variables up to 4 years after diagnosis.
A number of studies have found that effective
communication significantly impacts patients satisfaction and quality of life [6170]. For instance,
Roberts et al. investigated the types of communication cancer patients find helpful [71]. They found
that the best communication and psychological adjustment occurred when physicians had a caring attitude, demonstrated empathy and spent sufficient time
with their patients. Patients found it very important that their physician engaged them in collaborative decision-making [71]. Common communication pitfalls are lecturing, stopping the patients from
addressing their own concerns, depending on routine
procedures and discussions and premature reassurance. Back et al. and others recommend assessing a
patients pre-existing knowledge and questions, providing information in small units, followed by understanding the patients comprehension and evaluation
of the information provided [72].
Thus, while discussions of response to treatment
and the types and severity of side effects is important, patients also need attention to their coping and
psychological well-being. They should be encouraged
to speak about the complex psychosocial issues they
are facing because of the cancer diagnosis and fertility treatments. Addressing these psychosocial issues is
important for achieving treatment goals and engaging
patients in their own care. Patients should be encouraged to bring trusted friends and family to provide
emotional support during and after the visit. Given the
anxiety and potential inability to register information,
friends and family can also help to process information and, if necessary, ask relevant questions. Patients
should also be provided a list of clearly written instructions and resources to help reinforce the information
given during an office visit and enhance treatment
adherence. Because of the time-sensitive nature of fertility treatments, patients should be helped in finding
471
472
Conclusion
Fertility preservation is a complex field involving many
specialists from different disciplines. Psychologists or
other mental health providers should be included in a
team approach to fertility preservation. Many cancer
patients would benefit from referral to a mental health
provider for additional attention and support. Mental
health providers can counsel patients about the complex decisions they are facing when considering fertility preservation. Patients may feel more at ease discussing mood changes, sexual difficulties, self-esteem
problems and other life stressors with mental health
providers than with their oncology physician or surgeon. Psychologists and social workers may have training, or know how to get information, about CBRC
and be aware of the issues involved in cultural competency. Thus, a team approach allows each care provider
the ability to stay within their professional scope of
practice. Most importantly, when the emotional needs
of patients are addressed, normalized and considered
standard of care, cancer patients may feel understood
and most fully cared for.
References
1. Tschudin S and Bitzer J. Psychological aspects of
fertility preservation in men and women affected by
cancer and other life-threatening diseases. Hum
Reprod Update 2009; 1(1): 111.
2. Rosen A, Rodriguez-Wallberg KA and Rosenzweig L.
Psychosocial distress in young cancer survivors. Semin
Oncol Nurs 2009; 25(4): 26877.
3. Donnez J, Dolmans MM, Demylle D et al. Livebirth
after orthotopic transplantation of cryopreserved
ovarian tissue. Lancet 2004; 364: 40510.
4. Donnez J, Martinez-Madrid B, Jadoul P et al.
Ovarian tissue cryopreservation and transplantation: a
review. Hum Reprod Update 2006; 12: 51935.
5. Davis VJ. Female gamete preservation. Cancer 2006;
107 (Suppl. 7): 16904.
473
474
475
476
477
478
Section 10
Chapter
41
Fertility preservation
Ethical considerations
Pasquale Patrizio and A. L. Caplan
Established
Ovarian translocation (oophoropexy) to avoid a radiation field
Radical trachelectomy for cervical cancer
Cryopreservation of spermatozoa
Cryopreservation of embryos
Experimental
Cryopreservation of oocytes
Cryopreservation of ovarian tissue
Cryopreservation of whole ovary
Folliculogenesis in vitro
In-vitro oocyte maturation
Cryopreservation testicular tissue
Cryopreservation spermatogonial cells
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
479
Adults
From an ethical standpoint, the key reason for pursuing fertility protection is to restore personal autonomy to those who are unable to conceive [4]. However, since many of the technologies are innovative but
yet highly experimental, it is difficult to design clinical trials: how to provide a proper informed consent
and insure respect for autonomy. Who to include or
exclude in trials of innovative techniques and how best
to recruit them? The presentation of risk information
is complicated by the fact that both the adult and their
offspring may be involved. A core principle of medical
ethics is to do no harm. If ovarian tissue or testicular
tissue cryopreservation is to be tested, then the level of
risk that can be tolerated should require essential careful animal studies and close oversight of research by
review committees. It is reasonable in the absence of
grant funds to seek reimbursement from patients to
cover the expenses of the research, but there should
be no charge for clinical fees until the experimental
options have been proved safe and effective.
Ideally the decision about who is candidate for
fertility preservation should be rendered by a team
including a medical oncologist, reproductive endocrinologist, a pathologist and a psychologist, all guided
by written protocols which can be shared with patients
[4]. Patients should not be provided with false hopes,
and alternative plans including no intervention with
the prospect of adoption or childlessness should also
be part of the discussion. Equity or ownership interests in novel technologies utilized in research must be
disclosed to potential subjects.
Children
Impaired future fertility is another possible consequence of exposure to cancer therapies even for
480
481
482
be anticipated. First, there is no opportunity to perform PGD without further intervention. Embryo and
oocyte cryopreservation can both provide an inherent option for PGD. As identifiable genes for diseases and phenotypic traits continue to be uncovered,
the option of PGD may become increasingly desirable. Though IVF could be performed and PGD could
be done post-transplantation of ovarian tissue, this
would require an extra step and an extra invasive
procedure.
The option of designing ones child may overshadow the positive aspects of ovarian cryopreservation and re-transplantation, especially if there is a family history of genetically transmitted disease. Ovarian
cryopreservation and re-transplantation could be considered unpredictable regarding which traits the child
inherits.
Second, ovarian tissue cryopreservation and retransplantation presents a multifaceted potential for
false hope. False hope in medicine can be defined as
based on a set of unrealistic expectations, encouraged through incomplete or faulty information or by
a patients unwillingness to acknowledge the limits
of medicine [25]. Currently, there is no guarantee
that ovarian cryopreservation and re-transplantation
will result in future offspring, just as in embryo and
oocyte cryopreservation. A reasonable success rate
should be established prior to the mainstream offering of this technique as non-experimental. The procedure should only be offered with a clear statement
of risks and benefits [26]. As this technology is further developed, a romanticized view of forever preserving fertility is imaginable, which may place unrealistic expectations on this technology. This reality must
be clearly indicated in order to avoid major disappointments. Though any new procedure has the potential for false hope, it is important to acknowledge the
limitations of the procedure to the best of ones ability, especially when the hope is for the creation of a
new life.
483
484
Conclusions
If they have lost their reproductive function, cancer survivors may wish to become parents by using
previously stored gametes or gonadal tissue. Fertility preservation serves such a wide range of medicosocial circumstances, some quite unique, that patient
care requires an individualized and multidisciplinary
approach. In particular, fertility specialists offering fertility preservation options to cancer patients should
be properly trained and knowledgeable to discuss
patients treatment plan, prognosis, as well as unusual
health risks for future offspring and the potential
harmful effects of pregnancy.
485
486
References
1. Gosden RG. Fertility preservation definition, history
and prospect. Semin Reprod Med 2009; 27(6):
4337.
2. Horner MJ, Ries LAG, Krapcho M et al. (eds.). SEER
Cancer Statistics Review 19752006. Bethesda MD:
National Cancer Institute. http://seer.cancer.
gov/csr/1975 2006/.
3. Bromer JG and Patrizio P. Gynecologic management
of fertility. In: Wingardc JR, Gastineau D, Leather H,
Snyder EL and Szczepiorkowski ZM (eds.),
Hematopoietic Stem Cell Transplantation: A Clinicians
Handbook. Bethesda MD: AABB Press, 2009: pp.
58394.
4. Patrizio P, Butt S and Caplan A. Ovarian tissue
preservation and future fertility: emerging
technologies and ethical considerations. J Natl Cancer
Inst Monogr 2005; 34: 10710.
5. Hirtz DG and Fitzsimmons LG. Regulatory and
ethical issues in the conduct of clinical research
involving children. Curr Opin Pediatr 2002: 14;
66975.
6. Grundy R, Larcher V, Gosden RG et al. Fertility
preservation for children treated for cancer (2): ethics
of consent for gamete storage and experimentation.
Arch Dis Child 2001; 84: 3602.
7. Thompson A, Critchley H, Kelnar C and Wallace
WHB. Late reproductive sequelae following treatment
of childhood cancer and options for fertility
preservation. Best Pract Res Clin Endocrinol Metab
2002; 16: 31134.
8. Burns J. Research in children. Crit Care Med 2003;
31(Suppl.): S1316.
9. Grundy R, Larcher V, Gosden RG et al. Fertility
preservation for children treated for cancer (1):
scientific advances and research dilemmas. Arch Dis
Child 2001; 84: 3559.
10. Bahadur G. Ethics of testicular stem cells medicine.
Hum Reprod 2004; 19: 270210.
11. Klock S, Jacob M and Maier D. A prospective
study of donor insemination of recipients: secrecy,
privacy and disclosure. Fertil Steril 1994; 62(3):
47784.
12. Mc Gee G, Vaughan Brakman S and Gurmankin A.
Disclosure to children conceived with donor gametes
is not optional. Hum Reprod 2001; 16: 20335.
13. Patrizio P, Mastroianni A and Mastroianni L.
Disclosure to children conceived with donor gametes
should be optional. Hum Reprod 2001; 16: 20368.
14. Blyth E and Cameron C. The welfare of the child: an
emerging issue in the regulation of assisted
conception. Hum Reprod 1998; 13: 233942.
487
Section 10
Chapter
42
Introduction
Give me children or else I die.
(Genesis 30:1)
This quote is even more poignant when discussing the
reproductive planning and options of a cancer patient
or cancer pre-vivor. For purposes of this chapter the
term pre-vivor refers to one who does not yet have
cancer but has an increased risk or predisposition to
having cancer, such as an individual who is identified
as having a BRCA1 or BRCA2 mutation. The imperative to have children is something that can be both psychological and biological, and it may be further exacerbated by ones own confrontation of mortality. Recent
scientific advances have made the once remote possibility of conception following cancer treatment more
tangible; however, these advances are not without risks
including legal and ethical risks.
Learning that one or ones child has cancer or that
one is at a greatly increased risk of developing cancer is life altering. Parents, patients and/or caregivers
are bombarded with innumerable treatment decisions,
many of which need to be made urgently with little time for deliberation. As such, meaningful discussion about fertility preservation often does not occur.
In fact a study published in the Clinical Journal of
Oncology Nursing of nurses attitudes regarding fertility preservation revealed that patients may be strongly
focused on survival and their cure and may not want
to think about other issues . . . [1]. According to a survey by Carlson, the vast majority of surveyed physicians discuss the potential for infertility with patients
receiving cancer treatment; however, fewer than 25%
responded that they made a referral to a reproductive specialist or provided educational information [2].
Even more striking in the survey was that only 38% of
respondents knew that the American Society of Clinical Oncology (ASCO) has developed guidelines on fertility preservation [2]. Another study, published in the
Journal of Clinical Oncology found that fewer than 25%
of men bank sperm and the most common reason for
not doing so was that the option was not made known
to them [3]. Nearly one half of the men in the study
had no recollection of any discussion about infertility before beginning their cancer treatment [3]. What
is not clear is whether their lack of recollection is the
result of not being provided with the information or
the result of the stress and strain of having to cope
with a cancer diagnosis and make immediate decisions directly impacting their cancer treatment. This
research does suggest, however, that more education
of and communication with patients about potential
infertility resulting from treatment as well as fertility
preservation options is necessary.
Advances in reproductive technology and oncology have made what was once impossible, possible,
by allowing young cancer patients extended periods
of survival and thus enabling them to contemplate
a future with children. Cryopreservation of sperm
has been an available option for men since the mid
twentieth century [4], but cryopreservation of oocytes
has only become an option for women in the last
decade or so and is still considered largely experimental despite the proliferation of publicity regarding
it. According to the American Society for Reproductive Medicine (ASRM), [a]lthough currently investigational, . . . oocyte cryopreservation holds promise for
future female fertility preservation, . . . [5]. Unlike
collection of sperm for cryopreservation, collection
of oocytes carries more risk and requires more time
potentially delaying cancer treatment.
Numerous other treatments or experimental procedures are also available. According to the ASCO,
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
488
Informed consent
Informed consent is the embodiment of respect for
persons and reflects both a bioethical and a legal construct, applying to both treatment and research. The
essential elements of informed consent are the same in
both the treatment and research context, and include a
discussion with ones physician or investigator regarding the risks, benefits and alternatives to a particular
treatment or protocol. Informed consent is not merely
a signed document but rather an ongoing dialogue, the
purpose of which recognizes that adults are entitled to
accept or reject healthcare interventions on the basis of
their own personal values and in furtherance of their
own personal goals [7].
For those unable or incapable of this level of selfdetermination, such as minors, parents, acting in the
childs best interest, will be called upon to make such
determinations. The sections that follow will discuss
the issues of informed consent in both research and
treatment for both adults and minors undergoing can-
ASCO guidelines
In 2006, ASCO recommended that:
As part of education and informed consent before cancer therapy,
oncologists should address the possibility of infertility with patients
treated during their reproductive years and be prepared to discuss
possible fertility preservation options or refer appropriate and interested patients to reproductive specialists [6].
Treatment adults
The legal principle of informed consent was solidified
nearly 40 years ago by the US Court of Appeals for
the District of Columbia in the seminal informed consent case Canterbury v. Spence, which delineated what
ought to be disclosed to patients. In that case the court
held that:
True consent to what happens to ones self is the informed exercise of
a choice, and that entails an opportunity to evaluate knowledgeably
the options available and the risks attendant upon each. The average patient has little or no understanding of the medical arts, and
ordinarily has only his physician to whom he can look for enlightenment with which to reach an intelligent decision. From these
almost axiomatic considerations springs the need, and in turn the
489
490
Treatment children
As mentioned above, parents are responsible for making medical decisions for their children. The general
rule seems to be that, unless there exists an emergency,
which prevents any delay, or other exceptional circumstances, a surgeon who performs an operation upon a
minor without the consent of his parents or guardian
is guilty of a trespass and battery [14]. This decisionmaking authority is part and parcel of the constitutionally recognized right of parents to the care, control
and custody of their children [15]. An historic rationale for giving parents this authority was based on the
notion that if anything went wrong with the medical
procedure the parents would be responsible for raising
the child and thus should make the decision with this
awareness [14, 16]. Still another rationale for bestowing the obligation upon parents is that parents are in
the best position to protect their childs welfare and to
consider what would be in the best interests of the child
[17].
Under some circumstances, however, minors can
make their own healthcare decisions, particularly
around issues of reproduction. States have exceptions
for emancipated minors, mature minors and for specific healthcare services [16]. For example, the US
Supreme Court in a series of cases found that minors
have a right to determine whether to bear a child and
the decision to do so should not be vetoed by another;
however, some limitation is considered permissible
[18, 19]. In addition, legislation exists which allows
minors to make treatment decisions regarding sexually transmitted infections without parental consent
or notification [20]; obtain information about contraception [21]; and to obtain medical treatment for
alleged sexual assault without parental consent [22].
These statutes are in keeping with the general understanding that the right to reproduce is a fundamental
one. Further supporting this is the recognition by most
states that parents cannot make the decision to sterilize
a minor without independent court review [23]. Sterilization touches upon the individuals right of privacy
and the fundamental right to procreate [24]. While
parental failure to consent to the pursuit of fertility
preservation likely is not the same deliberative process as consenting to the sterilization of ones child, the
consequence for the child might be the same and thus
Research adults
Several fertility preservation techniques are still in the
experimental phase, and thus the elements of informed
consent for research should be adhered to regardless
of the funding source for such research. The goals of
research and practice differ and this must be reflected
in the consent process. The goal of treatment is to benefit an individual patient whereas the goal of research
is to contribute to generalizable knowledge [28]. Based
on this distinction, more protections are required of
human participants in research. One such protection
includes the Code of Federal Regulations requirement
that research involving human subjects be reviewed by
an Institutional Review Board (IRB) [29].
The role of the IRB is to determine that the risks
to patients are minimized; that the benefits of those
participating in the research will outweigh the risks;
that selection of participants is equitable; and that
informed consent is sought from the participant or
his or her representative [30]. The IRB review will
include a review of consent documents as well as the
consent process to determine whether all of the consent elements required by the Regulations are satisfied. Section 46.116 sets forth the general requirements
for informed consent. These requirements include,
among other things, an explanation of the purpose of
491
Research children
Standard of care?
492
493
494
Conclusion
As cancer patients and cancer pre-vivors continue
to have better prognosis for long-term survival and
References
1. King L, Quinn GP, Vandaparampil ST et al.
Oncology nurses perceptions of barriers to discussion
of fertility preservation with patients with cancer. Clin
J Oncol Nurs 2008; 12: 467.
2. Carlson RH. Survey: little awareness of the ASCO
fertility-preservation guidelines by cancer specialists.
Oncol Times 2009; 31: 2630.
3. Schover LR, Brey K, Lichtin A, Lipshultz LI and
Jeha S. Oncologists attitudes and practices regarding
banking sperm before cancer treatment. J Clin Oncol
2002; 20: 18907.
4. Walters EM, Benson JD, Woods EJ and Crister JK.
The history of sperm cryopreservation. In: Pacey AA
and Tomlinson MJ (eds.), Sperm Banking Theory and
Practice. Cambridge: Cambridge University Press,
2009: pp. 117.
5. Practice Committee of the American Society for
Reproductive Medicine and the Practice Committee
of the Society for Assisted Reproductive Technology.
Ovarian tissue and oocyte cryopreservation. Fertil
Steril 2008; 90: S2416.
495
496
Section 10
Chapter
43
The purpose of this chapter is to provide an interpretive framework for examining how Christian theological tenets and convictions might inform an ethical assessment of fertility preservation. In undertaking
this task I make no attempt to survey various religions,
other than to note that their respective beliefs inspire
disparate moral assessments of the medical procedures
deployed in preserving fertility. Subsequent inquiry in
comparative religious ethics and interfaith dialogue
are certainly required given the diverse character of
contemporary society, but will need to be pursued by
scholars with greater expertise. I also do not attempt
to make a systematic or normative assessment of fertility preservation in general, nor do I offer any detailed
evaluation of the discrete ethical issues accompanying
the particular medical procedure described in the previous chapters. Again, these are important tasks, but
beyond the limited scope of this chapter.
My intent, to invoke a crude analogy, is to visit
some important theological landmarks along the
moral turf in which fertility preservation is embedded.
My description of these landmarks is both informed
and limited by my perspective as a Christian theologian, but I use terminology which I hope will prove
informative to broader religious, secular and professional audiences. In most respects we will be revisiting
familiar landmarks, for many of the ethical issues associated with fertility preservation are similar or identical to earlier and ongoing disputes over the ethics
of assisted reproductive technology (ART) generally
(e.g. [13]). The principal contextual difference in this
instance is that fertility is being preserved, and thereby
assisted, in response to largely non-controversial therapies rather than treating infertility. Given the wide
variety of Christian churches and their respective
teachings and theological convictions, no universal
ethical position on the ethics of fertility preservation in
general or the associated medical treatments in particular can be formulated. Consequently, in conducting
this inquiry I examine four pertinent theological and
biblical precepts; describe four general moral stances
along a spectrum of options that may be derived from
these precepts; and identify a principal strength and
weakness, respectively, of each stance.
Principles and Practice of Fertility Preservation, eds. Jacques Donnez and S. Samuel Kim.
C Cambridge University Press 2011.
Published by Cambridge University Press.
497
498
ties, costs and accessibility. Given the general healthcare needs of civil community, does preserving the fertility of relatively few individuals justify the allocation
of scarce medical funding and personnel in achieving
this goal?
The third precept is derived from the doctrine
of the incarnation. The Gospel according to St John
asserts that the Word became flesh, a reference to Jesus
Christ (see John 1.118). The central tenet of this doctrinal teaching is that in Christ, as the second person
of the Trinity, God became a human being. This act
in turn affirms the embodied nature of human beings
and consequently the finitude of the human condition. Bodily health is therefore not a matter of indifference. Subsequent theological reflection has affirmed
the goodness of the body despite frequent heretical
attempts to disregard or malign the body as a mere
vessel, or worse, a prison of the soul. In Beth Felker
Jones evocative words, In the Christian tradition, the
temptation to denigrate the body has been continually
reasserted and consistently rejected[8].
The doctrine of the incarnation inspires a Christian
affirmation of the human body, but no obvious ethical
stance is forthcoming regarding modern health care in
general, or assisted reproduction and fertility preservation in particular. Although the life of embodied
human beings is highly valued and respected, efforts
to preserve or reproduce life are relative rather than
absolute [9]. For example, there is no corresponding moral duty to use every means possible to extend
the life of dying individuals for as long as possible
should such efforts prove unduly onerous or futile,
though euthanasia and assisted suicide are generally
proscribed [10, 11]. Similarly, the biological means of
reproducing human life is held in high esteem, but
it is again a relative rather than absolute good to be
pursued. An infertile couple, for instance, incurs no
moral obligation to employ every available medical
treatment. As noted above, a Catholic couple would
be prohibited from utilizing ART and could fulfill
their natural parental desires through such alternatives as foster care or adoption [12, 13]. Many Protestant couples, however, believe they are free to utilize
or refrain from utilizing ART in treating their infertility, which would presumably extend to attempting to
preserve or not preserve fertility as well. This expansive range of options is derived from the theological teaching of the incarnation which while affirming embodiment, and derivatively reproduction, also
indicates that with the birth of Jesus the urgency
Moral stances
The following four ethical stances are derived from the
theological and biblical precepts summarized in the
preceding section. Each stance demonstrates how religious beliefs and convictions might inform particular ethical assessments of fertility preservation. These
stances do not reflect arguments promulgated by a
specific church or theologian. Rather, they serve as
heuristic devices that demonstrate varying and often
conflicting assessments. Furthermore, these stances
disclose how theological and biblical precepts may be
interpreted and applied in a variety of imaginative
ways in constructing a moral argument. As will be
seen, there is no given correspondence between a particular precept and a particular stance regarding the
ethics of fertility preservation. It should also be noted
that these stances do not exhaust the possible options
that could be formulated, but rather serve as examples
along a spectrum ranging from prohibition to encouragement.
In each instance I make the following assumptions: An individual is facing the prospect of a therapeutic application that may result in the loss of fertility. The various treatments that could be applied
in attempting to preserve fertility are accessible, relatively safe and potentially efficacious and adequate
funding is readily available. All legal safeguards and
recognized ethical practices and procedures are followed such as informed consent, protection of minors
and proper authorization and oversight of experimental protocols. In addition, I make no attempt to
assess the adequacy of the various moral stances, especially in regard to their respective interpretations and
applications of theological and biblical precepts and
coherency of argument, nor do I offer any counter
interpretations or arguments. Rather, I allow each
stance to stand in its own right in order to identify
various points along a spectrum of options concerning the ethics of fertility preservation that are based
upon selected religious beliefs and convictions. Furthermore, it should be noted that in some instances
these arguments do not accept medical designations
of embryonic development, for example, the difference between pre-embryos and embryos, as implying any inherent normative content. It is important
499
for medical practitioners to keep these potential discrepancies in mind when dealing with some patients
who may exhibit some reluctance in pursuing fertility
preservation. In short, when doctors and patients refer
to an embryo, they may not, in some instances, be
referring to a common perception. Although the religious reservations of these patients need to be honored, they do not necessarily call into the question
either the morality of the procedures or humane intentions motivating their development and deployment.
The first stance may be characterized as preserving the natural reproductive process. This stance draws
heavily upon the precepts of the incarnation and
the procreative mandate. The divine affirmation of
the embodied nature of human existence necessarily
entails the need of human beings to perpetuate themselves from one generation to the next; hence Gods
command to be fruitful and multiply. This affirmation and command, however, imposes constraints in
exercising an accompanying stewardship. In affirming
their embodiment and obeying the command to procreate, individuals are not free to do whatever they
might want. The affirmation and mandate acknowledges and consents to the frailty and limitations of the
human body, otherwise stewardship is distorted into a
form of mastery that rightfully belongs only to God.
Although this stance does not reject modern medicine
in general, indeed it is seen as an important means of
excising the stewardship of Gods creation and expressing the love of neighbor, it too is limited to restoring
the natural health of the body.2
Given these religious presuppositions, the morality
of fertility preservation is assessed largely, though not
exclusively, in respect to whether or not it preserves
or violates the natural reproductive process. Consequently, fertility preservation should not be used in
many, if not most, instances because the treatments
employed violate this natural integrity. Similar to the
objections against ART, gamete and embryo storage
are illicit because of the means required to procure the
gametes and embryos. Although such medical treatments are motivated by the legitimate desire to treat
infertility resulting from other therapeutic applications, this motive does not justify bypassing the nat2
The description of the basic religious principles underlying
this stance draws upon selected themes explicated in relevant
Catholic social teaching. See, e.g., Donum Vitae and Paul VI
[20]. For related Protestant themes see, e.g., Mitchell et al.
[21].
500
from what may be described as Christian environmentalism. See, e.g., Fern [28] and Scott [29]. In respect to setting
social priorities governing the development and distribution
of medical resources, see, e.g., Cahill [30].
6
This discouragement could be accomplished, e.g., through
a combination of informal social and religious disapproval,
refusal by public and private insurance carriers to cover costs
and other economic disincentives.
7
The same objection could also be raised against ART in
general. An individual holding this stance could also be dedicated to preserving the integrity of the natural reproductive
process as described in the previous moral stance.
501
502
503
Concluding remarks
As the preceding section demonstrates, a singular or
universal Christian assessment of fertility preservation cannot be formulated. There are, rather, multiple assessments. This wide variety should not be surprising given the historical divisions among Christian churches in respect to doctrine and practice. Nor
should it be presumed that these divisions are uniquely
manifested in assessing the ethics of fertility preservation. Rather, ethical concerns over fertility preserva-
504
Acknowledgements
I am indebted to my research assistant, Jason Gill, for
his highly efficient and invaluable assistance.
References
1. Cahill, LS. Theological Bioethics: Participation, Justice,
Change. Washington DC: Georgetown University
Press, 2005: pp. 169210.
2. Meilaender, G. Bioethics: A Primer for Christians.
Grand Rapids MI: Eerdmans, 1996: pp. 1125.
13
For an overview of religious contributions in the early history of bioethics, see Jonsen [56].
505
506
Index
anastomosis 251
end-to-side 384
microvascular 3834
sleeve 384
whole ovary transplantation 3389
advantages and
disadvantages 339
androgens 121
Angelmans syndrome 102
angiogenesis 449
angiogenic switch 41
angiogenin 449
angiopoeitin 333
anonymous donation
oocytes 317, 318
sperm 2313
anthracyclines 65
antibody dependent cellular
cytotoxicity (ADCC) 44
anti-emetics 64
anti-growth signaling 401
anti-Mullerian hormone (AMH) 13,
25, 115, 116, 329
antral follicle count (AFC) 13
apoptosis 38, 101, 110
evasion by cancer cells 41
germ line 103
ability 105
DNA damage 1035
prosurvival factors in
spermatozoa 10910
spermiogenesis 1056
chromatin remodeling 1067
importance 1079
Aristotle 1
aromatase inhibitors
teratogenicity 64
artificial bladder 415
artificial ovary 308, 44856
assembly 450
bioactivity 452
biocompatibility 4534
biodegradability 453
crystallinity 451
interaction with
cells/follicles 4501
morphology 451
natural ovary 450
physical support 452
porosity 452
requirements 450
scaffold handling 454
surface 4512
vascularization 4523
fertility re-establishment
strategy 448
advantages 449
follicle number and quality 450
follicular development 449
follicular growth 44950
follicular survival 449
tissue engineering 4546
ascorbic acid 379
Asperts syndrome 103
assisted conception 1
assisted reproductive technology
(ART) 1, 1012
cryocooling 132
cryopreserved embryos 138
ataxia telangiectasia 40
Austin, Colin Bunny 3
autoimmune diseases 289
autotransplantation 201, 213
5-azacytidine 442
azoospermia 18
chemotherapy 75, 76
BAD 109
base excision repair (BER) 40
basic fibroblast growth factor
(bFGF) 115, 202, 333, 411, 449
BEACOPP regimen (bleomycin,
etoposide, doxorubicin,
cyclophosphamide, vincristine,
procarbazine and
prednisolone) 14, 18
BeckwithWiedmann syndrome 102
benign tumors 35
benzo[a]pyrenes 103
BEP regimen (bleomycin, etoposide
and cisplatin) 274
betacellulin 121, 423
bilateral salpingo-oophorectomy 269,
270
biocompatibility 452
biparental androgenones 443
biparental gynogenones 443
birth defects 102
507
Index
blastoma 35
bone density and GnRH agonist
use 242
bone marrow transplantation
(BMT) 14
ovarian failure rates 15
bone morphogenic proteins
(BMPs) 115
BMP4 116, 410, 411
BMP7 116, 410, 411
BMP15 117, 118
borderline ovarian tumors
(BOTs) 2667
fertility results 26970
limits of surgery 2689
disease stage 268
histological subtype 268
serous tumor with MP
pattern 268
ovarian induction or IVF 270
recurrence rates 269
surgery and outcomes 267
survival following surgery 2678
Bourn Hall Clinic 5
brain tumors 91
BRCA mutations 62, 63
breast cancer 49, 57, 69
adjuvant therapy
newer treatments and additional
benefits 512
risk of recurrence and
reduction 50
amenorrhea 65
amenorrhea and fertility loss 545
amenorrhea and survival 501
ART
time between treatment and
pregnancy 311
contraception in aftermath 68
endocrine therapy and reproductive
function 66
fertility preservation 66
adjuvant therapy delay 56
barriers to 55
controlled ovarian stimulation
(COS) 56
cost 55
donor eggs and surrogacy 67
embryo cryopreservation 667
oocyte preservation 67
ovarian harvest 56
ovarian tissue
cryopreservation 67
suppression of ovarian
function 678
incidence 62
lactation in aftermath 69
occurrence 49
ovarian failure rates 15
508
differentiation aberrations 39
DNA damage repair system
failures 39
growth factor signaling 378
signal transduction molecules 38
origins
cancer stem cell theory
(CSC) 367
oncogenes and tumor suppression
genes 356
stepwise accumulation of
mutations 36
transformation of normal cells 35
carbohydrates for
cryopreservation 178
carboplatin 52, 55
carcinoembryonic antigen (CEA) 44
carcinoma 35
Cut Standard Straw (CSS) 186
-catenin 119
CEF regimen (cyclophosphamide,
epirubicin and
fluorouracil) 51, 65
cell cycle abnormalities 38
cell death defects 389
cell-mediated immune response 43
cervical cancer
adolescents and young adults 956
surgery 2578, 263
assessment 2623
experience of surgical team 2612
experience worldwide 262
technique 25861
cervical intraepithelial neoplasia 262
Chang, M. C. 3
chemoprotection 18
chemotherapy
breast cancer 50, 64
ovarian damage
clinical detection 1213
nature of damage 1314
risk of ovarian failure 14, 15
ovaries 734
testes 756
childhood cancers 11
epidemiology 836
international classification 84
children from oocyte donation
fears of parents 319
children from sperm donation 231
disclosure 2335
age of disclosure 235
religious and cultural
influences 234
shame 233
chlorambucil 18, 25
azoospermia 75
chlortetracycline (CTC)Hoechst
33258 staining 181
Index
509
Index
epithelialmesenchymal transition
(EMT) 42
epitopes 391
EpsteinBarr virus (EPV) 92
equilibrium cryopreservation 130
equilibrium vitrification 133
equilirium slow freezing 130
estradiol levels 29
estradiol patches 322
estrogen receptor-positive tumors 426
estrogen receptors (ERs) 50
1,N6-ethenoadenosine 104
1,N2-ethenoguanosine 104
ethical concerns 4856
ART clinics 485
embryo freezing 31
future use following death of
parent 280
fertility preservation 47980
adults 480
children 4801
financial compensation for
donors 316, 317
future trends 479, 4835
legality of donation 316
ovarian cryopreservation and
re-transplantation 483
ownership of frozen oocytes 297
pre-implantation genetic diagnosis
(PGD) 482
spermatogonia, transplantation of
cryopreserved 206
stem cell research 4835
embryo research 484
legality 484
moral status of embryo 484
spare embryos 485
testicular cell and tissue
transplantation 219
use of donor gametes 4812
disclosure to children 4812
international variations 482
ethylene glycol (EG) 151
Ewings sarcoma 75
extracellular matrix (ECM) 41, 409,
414, 449
extravasation 42
extremely low-frequency
electromagnetic fields
(ELF-MF) 901
FAC regimen (fluorouracil, adriamycin
and cyclophosphamide) 51, 66
FaddyGosden model for natural
oocyte decline 16
Fallopian tubes 115
false hope potential of therapies 483
familial cancers 40
family members as sperm donors 232
510
Foxo3 115
Foxo3a 116
fracturing in vitrification solutions
154
freedom of choice 502
frozenthawed ovarian tissue 7, 26
frozenthawed embryo (FET) 433
gamete intrafallopian transfer
(GIFT) 1, 4, 6
GATA binding protein 4 (GATA4) 202
GDNF family receptor alpha-1
(GFR1) 202
genes and predictions 460
genomic integrity 391
germ stem cells in adults 11
germinal vesicle 119, 296
germinal vesicle breakdown 119
Gibbs free energy 148
glass transition temperature 132, 150
glial cell line-derived neurotrophic
factor (GDNF) 202, 203
glutathione transferase 246
glycerol 178, 284
GnRH agonists 6, 23, 27, 29, 239
breast cancer 51
clinical data about ovarian
reserve 242, 243, 245
control group studies 2424
clinical use
dose 241
flare response 2401
ovarian reserve 2412
side effects 241
direct and indirect effects 245
immunological effect 247
mechanism of action 23940
ovarian impact 244
ovarian protection, hypothetical
mechanisms 247
ovarian stromal blood flow 2445
suppression of ovarian function 67
GnRH analogues 18
gonadal medical protection 27
gonadotrophin deficiency 16
gonadotrophin-releasing hormone
(GnRH) 66
ovarian expression 2456
ovarian steroidogenesis 239
gonadotrophins 119
goserelin 51, 53, 244
Graff, Reiner de 2
Graffian follicles 410
granulocyte-colony stimulating
factors 64
granulosa cells (GCs) 12, 13, 24, 114
poliferation 352
green fluorescent protein (GFP) 200,
443
Index
511
Index
512
Index
chemotherapy 24
disease modifying antirheumatic
drugs (DMARDS) 25
non-steroidal anti-inflammatory
drugs (NSAIDs) 25
radiotherapy 25
rheumatic disease treatments 25
surgery 25
non-epithelial ovarian cancer 274
malignant germ cell tumors
(MGCTs) 274
sex cord stromal tumors
(SCSTs) 274
non-equilibrium slow freezing 131
non-growing follicles (NGFs) 11
non-homologous endjoining
(NHEJ) 40
non-permeating cryoprotective
agents 130, 178, 284, 348
non-specific immunotherapy 44
non-steroidal anti-inflammatory drugs
(NSAIDs) 25
nucleotide excision repair (NER) 40
obesity and GnRH agonist
effectiveness 241
odds ratios (ORs) 433
oligomenorrhea 14
oncogenes 356
oocyte cryopreservation 136, 283
chemical and physical aspects of
freezing 2835
cell volume change 284
equations explaining cell
damage 283
osmotic effects 284
immature 137
mature 1367
morphological variables 2857
slow-cooling protocols and
outcomes 2879
slow freezerapid thaw
program 288
oocyte cumulus complexes (OCC)
346
oocyte donation for cancer
survivors 310, 31516, 3234
ovarian function testing 31213
ART
ovarian stimulation 313
ovarian stimulation, hormonally
sensitive cancers 31415
clinical practice of donation 31623
anonymous versus known
donation 317
care of donor 319
donor recruitment 31617
endometrium preparation and
synchronization 3223
genetic screening of
donors 31718
infectious screening of
donors 317
matching donors and
recipients 319
oocyte retrieval 3212
ovarian stimulation of donor 320
psychological screening 318
psychological screening of
donors 31819
psychological screening of
recipient 319
recipient screening 322
evaluation for pregnancy 310
chronic health problems 310
gestational carrier 312
prognosis 310
time between treatment and
pregnancy 31112
uterine evaluation 312
oocyte preservation 67
oocyte recovery with tubal
insemination (ORTI) 4
oocyte retrieval 56
complications
intraperitoneal bleeding 321
pelvic abscess 321
pelvic infection 321
oocyte secreted factors (OSFs) 117
oocyte vitrification 23, 26, 29,
2934
clinical use 295
continuing evolution 301
establishing and troubleshooting
oocyte cryostorage 298301
Kuwayama method 156
outcomes 297
post-warming survival 297
protocol improvement 2978
typical media 298
purpose 2945
relevance 2957
oocytes
development 114
see also follicular development
meiosis 115
meiosis 119
number at birth 73
number of mature eggs released 73
oophorectomy 270
Open Pulled Straw (OPS) method 132,
186
orthotopic autotransplantation 334
oscillating heat pipe (OHP) 133
ovarian borderline tumors 301
ovarian cryobanking 328, 339
clinical guidelines 329
current status 334
heterotopic
autotransplantation 336, 3347
orthotopic
autotransplantation 334
xenotransplantation 338, 3378
historical perspective 3289
issues and concerns 330, 330
cancer cell transmission 3334
cryoinjury 3302
ischemic injury 3323
whole ovary transplantation by
vascular anastomosis 3389
advantages and
disadvantages 339
ovarian cryopreservation, whole 367,
373
challenges 3678
human ovary 3713
summary of experiments 372
viability evaluation 372
rat model 368
sheep model 368
slow cooling 36871
vitrification 371
ovarian development 1112
ovarian function (OVF) 377, 378
ovarian hyperstimulation syndrome
(OHSS) 320, 421, 434
IVM rescue 4256
ovarian malignant tumors,
conservative management 266,
274
borderline ovarian tumors
(BOTs) 2667
fertility results 26970
limits of surgery 2689
surgery and outcomes 267
survival following surgery 2678
epithelial ovarian cancer (EOC)
fertility results 272
indications for surgery 2702
surgical procedure 272
literature reviews
EOC 271
MGCTs 273
ovarian induction or IVF after
BOTs 270
recurrence rates of BOTs 269
non-epithelial ovarian cancer 274
malignant germ cell tumors
(MGCTs) 274
sex cord stromal tumors
(SCSTs) 274
ovarian metastasis 334
ovarian reserve 11, 2412
clinical data from GnRH agonist
use 242, 243, 245
control group studies 2424
oocyte numbers 241
513
Index
514
risk factors 96
adolescents and young
adults 956
extremely low-frequency
electromagnetic fields 901
infections 92
ionizing radiation 90
parental lifestyle factors 925
parental occupational
exposures 912
risk of infertility 767
role of physician 79
testes
chemotherapy 756
normal physiology 75
radiotherapy 76
pelvic abscess 321
pelvic infection 321
pelvic malignancies, incidence
250
peritubular myoid cells 17
permeating cryoprotective agents 130,
178, 284, 347
rate of penetration 348
pesticides and cancer 91
phagocytosis 109
phosphatase and tensin homolog
(PTEN) 115
phosphatidylinositol-3-kinase (PI3K)
pathway 115, 203
phosphatidylserine translocation
(PST) 189
phosphodiesterase 3A (PDE3A) 120
pituitary gonadotrophin 114
platelet derived growth factor
(PDGF) 115
platinum compounds
azoospermia 76
pluripotent stem cells 440
epigenic regulation 4412
germ cell development 4423
germ cells 443, 4445
induced 441
germ cell differentiation 445
male germ cells 444
oocytes 4434
potential applications and future
prospects 445
stem cells 440
embryonic (ESC) 440
testis and ovary 440
Poisson ratio 153
PolscopeR 286
poly (ADP-ribose) (PAR) 105
poly (ADP-ribose) polymerase
(PARP) 40
polycyclic aromatic
hydrocarbonDNA
adducts 104
Index
non-steroidal anti-inflammatory
drugs (NSAIDs) 25
sarcoma 35
senescence 41
sentinel lymph node biopsy 63
sentinel node 258
serine proteases 122
serinethreonine kinase family
(AKT) 203
Sertoli cells 13, 17, 75, 165
severe combined immunodeficient
(SCID) mice 308, 337
sex cord stromal tumors (SCSTs) 274
signal transduction molecules 38
sleeve anastomosis 384
Smad2 and Smad3 117
smoking see tobacco smoking
social priorities 501
solution effects 296
Spallanzani, Lazzaro 2
spare embryos 485
sperm banking 2256
sperm chromatin structure assay
(SCSA) 102
sperm cryopreservation 78
Sperm DNA Degradation (SDD) 109
sperm donation and ART 225, 2301
anonymous versus known
donation 2313
disclosure to children 2335
religious and cultural
influences 234
sperm preparation medium (SPM) 180
spermatogenesis 101
spermatogonia (type A dark) 209
spermatogonia (type A pale) 209
spermatogonia, transplantation of
cryopreserved 199200, 206
cryopreservation 2045
culture of human cells 2034
development 2001
ethical concerns 206
isolation, purification and culture of
murine cells 2023
morphological identification 2012
removal of malignant cells 205
spermatogonial stem cells (SSCs) 199
spermatozoa 101, 110
apoptosis and prosurvival
factors 10910
apoptosis in germ line 103
ability 105
DNA damage 1035
cryopreservation 1356, 176, 193
fertilization properties 187
DNA damage 1012
long-term storage 1923
male age and progeny disease 1023
515
Index
spermatozoa (cont.)
mammalian sperm head size 180
smoking and progeny disease 103
spermiogenesis 1056
chromatin remodeling 1067
importance of apoptosis 1079
vitrification 1767
IVF, ICSI and
insemination 18992
optimal cooling rates 180
suitability 1779
technique 17989
sphingosine-1-phosphate 210
squamous cell carcinoma of cervix 364
Src family kinase 203
stem cell factor (SCF) 115
stem cells 440
embryonic (ESC) 440
germ cells 443, 4445
male germ cells 444
oocytes 4434
testis and ovary 440
Steptoe, Patrick 3
Bourn Hall Clinic 5
sterilization of a minor 490
stewardship of the planet 498
StolesEinstein equation 154
strain 153
stress 153
stromal cells 409, 416
follicle co-culture systems 41415
organ culture and primordial follicle
activation 412
role in follicle development 41011
studies
follicle-to-stroma paracrine
signaling 411
stroma-to-follicle paracrine
signaling 41112
three-dimensional follicle culture
systems 41214
sulfasalazine 25
supercooled solutions 147, 149
supercooled water 134
surface free energy 451
surrogacy 67
systemic lupus erythematosus
(SLE) 14
ovarian failure rates 15
premature ovarian failure (POF) 27
T lymphocytes 43
T regimen (taxotere) 51
T regulatory cells (Tregs) 44, 45
TAC regimen (taxotere, adriamycin
and cyclophosphamide) 51, 66
tamoxifen 50, 52, 53, 55, 244, 246
breast cancer during pregnancy 64
hormonal stimulation 67
516
irregular menses 66
ovarian stimulation 280
teratogenicity 64
taxanes 52, 55, 65, 66
taxotere 55
technological affirmation of life 503
telomerase 41
telomeres 41
alternative lengthening 41
tensile stress, maximum 154
testes
chemotherapy 1718, 756
function 17
hormonal suppression, effect of 165
immature testicular tissue
cryopreservation 210
cell suspensions 210
tissue pieces 21011
whole testis 211
normal physiology 75
radiotherapy 18, 76
undescended 102
testicular cancer
adolescents and young adults 95
testicular sperm extraction (TESE) 7
testosterone, low levels 18
5, 5
, 6, 6
-tetrachloro-1, 1
, 3, 3
tetraethylbenzamidazolocarbocyanin iodide 183, 189
thalassemia screening 318
theca cell organizer 411
thecal cells (TCs) 114, 115
three-dimensional follicle culture
systems 412
thrombocytopenia 242
thyroid cancer
adolescents and young adults 96
timetemperature transformation
(TTT) diagram 132
tissue engineering 41516
artificial ovary 4546
tobacco smoking, parental 934
DNA damage in spermatozoa 103
total body irradiation (TBI) 14
effect on ovaries 75
effect on testes 18
effect on uterus 17
transforming growth factor beta
(TGF) 40, 42, 333
superfamily 116, 117
trastuzamab 51, 64
tuberous sclerosis complexes
(TSCs) 116
tubulin 285
depolymerization of dimers 285
tumor-associated antigens (TAAs) 44
tumor-causing viruses 35
tumor-infiltrating lymphocytes
(TILs) 43, 44
Index
Warnock Report 7
wettability 451
wine, protection against cancer 94
xeroderma pigmentosum 40
xenografting 352
xenotransplantation 201, 213, 214,
338, 3378
fresh human testicular tissue 216
frozen human testicular tissue 216
517