SlideShare a Scribd company logo
Current Treatment Options in Oncology
DOI 10.1007/s11864-012-0183-7
Oncofertility (JS Jeruss, Section Editor)
Pediatric and Young Adult
Patients and Oncofertility
Katherine E. Dillon, BA
Clarisa R. Gracia, MD, MSCE*
Address
*University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA
19104, USA
Email: cgracia@obgyn.upenn.edu
* Springer Science+Business Media, LLC 2012
Keywords Oncofertility I Reproductive risks I Fertility Preservation I Gonads
Opinion statement
With improving survival rates for pediatric and young adult cancer patients, consider-
ations regarding the long-term effects of therapy have become more important. Cancer
therapies are known to pose reproductive risks, though the effects may be unpredict-
able. All at-risk patients should have a discussion about potential treatment-related
infertility before the onset of cancer therapy, and should be offered appropriate fertil-
ity preservation options. Embryo and sperm cryopreservation are considered standard
therapy, though oocyte cryopreservation is gaining acceptance. Ovarian tissue cryo-
preservation, while still experimental, is showing great promise. It is the only option
currently available to prepubertal girls. No fertility preservation options exist for pre-
pubertal boys though some institutions may offer experimental testicular tissue cryo-
preservation.
Introduction
With advancements in the diagnosis and treatment of
cancer in pediatric and adolescent patients, increasing
numbers of survivors in this age group are looking to-
ward a future as productive adults. For many this
includes consideration of future fertility. Given that
cancer treatments have been shown to impair normal
pubertal development and may lead to infertility, it is
imperative that oncologists discuss the issue of fertility
preservation with patients prior to the onset of cancer
treatment. This requires that clinicians be informed
about the preservation options available and the re-
productive challenges patients may face.
This review article aims to use the most up-to-date
data to inform the clinician about the effects of various
cancer treatments on reproductive potential and the
options for fertility preservation prior to cancer thera-
py. It will focus on important recent advancements
made in the field of oncofertility and will highlight fu-
ture directions for fertility preservation technologies.
Cancer in children and young adults
The most common cancers in children are leuke-
mias, followed by central nervous system malignan-
cies, lymphomas, soft tissue sarcomas, renal cancer
(Wilm’s Tumor) and bone tumors, in that order
[1]. Treatments often involve a combination of sur-
gery, radiation therapy, chemotherapy with alkylat-
ing agents, or allogenic bone marrow transplant.
Late effects of cancer are gaining more attention
since over 80% of childhood cancer patients survive
at least five years [2] and an estimated one in 640
adults ages 20–39 is currently a long-term child-
hood cancer survivor [1]. However, it is a challenge
to balance curative therapy while reducing the risks
to fertility. This is especially difficult due to ever-
changing cancer regimens and a lack of prospective
data concerning their effects on fertility outcomes.
One of the recognized challenges in studying the
reproductive risks of cancer therapy is that outcomes
such as fertility are difficult to capture. An individual’s
fertility depends on many factors including the avail-
ability of a fertile partner of the opposite sex, and a de-
sire to conceive. The gold standard epidemiologic
method to study fertility is to conduct prospective
“time to pregnancy” studies. Such studies are generally
not feasible in conditions where the exposure (cancer
therapy) is relatively rare. Therefore, most studies have
assessed other related outcomes such as menstrual
function or reported pregnancy after cancer treatment.
In females, measures of ovarian reserve, such antral folli-
cle counts and ovarian volume assessed by ultrasound,
and serum measures of inhibin B, anti-Mullerian hor-
mone (AMH), estradiol, and follicle-stimulating hor-
mone (FSH) have also been useful proxy outcomes.
In the field of pediatric oncofertility most of the
large-scale studies with epidemiologic data stem from
findings from the childhood cancer survivorship study
(CCSS), a large retrospective cohort study assessing a
variety of late effects of childhood cancer. The CCSS
is the largest cohort study of childhood and adolescent
cancer survivors with 20,720 previously untreated
patients with a new diagnosis before the age of 21 en-
rolled. It is important to recognize that the effect of
cancer therapies on an individual’s reproductive func-
tion can be unpredictable. Nonetheless, the available
data on the reproductive risks of cancer therapies will
be reviewed, and is summarized in Table 1.
Treatment effects in females
Pelvic radiation
The ovaries are particularly sensitive to damage from cancer treatments due
to the finite number of un-renewable germ cells (follicles) present in the
ovary. The ovary begins producing oocytes before birth and oocyte count
peaks at 20 weeks gestation. After this peak, progressive follicular atresia
occurs. Radiotherapy can cause ovarian failure by accelerating follicular
atresia and destruction of the follicular pool. Earlier studies have shown that
females treated with pelvic radiation doses in excess of 2 gray (Gy) are at high
risk of permanent ovarian failure [3]. Using this data, Wallace et. al devel-
oped a mathematical model that estimates the dose of ovarian radiation
necessary to cause ovarian failure taking into account age at treatment. For
example, the estimated mean radiation dose to the ovaries needed to cause
sterilization is 16 Gy at age 15, while a dose of only 12 Gy is required at age
30 [4]. Radiotherapy can also have deleterious effects on the uterus as it
causes scarring and diminished uterine blood flow in a dose-dependent
fashion [5], resulting in decreased uterine volume [6, 7].
Recent studies have examined the reproductive risks associated with pel-
vic radiotherapy in pediatric and adolescent cancer patients and have
reported more relevant reproductive outcomes such as reports of pregnancy.
For example, data from the CCSS demonstrated that the relative risk of
achieving pregnancy at least five years post-treatment in female childhood
cancer survivors exposed to 5 to 10 Gy of radiation to the pelvis was 0.56,
and dropped to 0.18 for exposure greater than 10 Gy when survivors were
compared to a similar-age sibling cohort [8••]. While the relative risks
reported in this study clearly highlight the adverse effects of radiation
Oncofertility (JS Jeruss, Section Editor)
exposure, it is important to recognize the limitations of the data when
counseling patients. This study examined rates of conceiving a pregnancy 5 years
or more after diagnosis compared to a sibling cohort so as to match for socio-
economic status and education. It is possible that having a cancer diagnosis itself
influenced the decisions to pursue pregnancy or the timing of pregnancy in
survivors. Information about whether patients attempted pregnancy was not in
the study, though the analysis did control for marital status. Moreover, infor-
mation on menstruation cycles or reported infertility was not available.
A retrospective French study of childhood cancer survivors over the age of
18 (median age 27 years) found that direct pelvic irradiation was the most
significant risk factor for subfertility, defined as no spontaneous menstrua-
tion or pregnancies. The odds of subfertility were 17.6 in the exposed com-
pared to the unexposed group [9•]. When controlling for adjuvant therapy,
radiation doses to the reproductive organs of less than 4 Gy were not sig-
nificantly associated with subfertility [9•]. The study also suggests that
ovarian transposition or shielding may protect reproductive function. Limi-
tations of this study include the convenience sample with small subgroup
numbers and the suboptimal definition of subfertility [10, 11].
Not only does exposure to pelvic radiation increase risk of infertility, pel-
vic irradiation is also associated with adverse pregnancy outcomes. Another
CCSS study of 1281 female survivors demonstrated an association between
pelvic irradiation exposure prior to menarche and subsequent stillbirth or
neonatal death [12••]. Compared to survivors who did not receive radio-
therapy, those exposed to 2.5–9.9 Gy of radiotherapy had a relative risk of
5.8 for stillbirth or neonatal death while those exposed to more than 10 Gy
had a relative risk of 19. A previous study also demonstrated that pelvic
irradiation increased the risk of preterm birth for female childhood cancer
survivors [13]. Patients with a history of pelvic radiation therapy should be
counseled about the perinatal risks of pregnancy and may consider using a
gestational carrier if pregnancy is unsuccessful.
Hypothalamic pituitary radiation
Hypothalamic-pituitary radiation causes destruction and fibrosis of the
pituitary and can lead to significant hypothalamic-pituitary dysfunction.
Table 1. Reproductive risks of cancer therapy in pediatric and young adult patients
Risk Females Males
High 910 Gy Pelvic Radiation Testicular Radiation
Hypothalamic Pituitary Radiation Hypothalamic Pituitary Radiation
Hematopoetic Cell Transplantion Hematopoetic Cell Transplantation
Total Body Irradiation Total Body Irradiation
Chemotherapy with Alkylating Agent Dose ≥2a
Chemotherapy with Alkylating Agent Dose ≥2a
Medium 5–10 Gy Pelvic Radiation Platinum-containing drugs
Chemotherapy with Alkylating Dose 90a
Chemotherapy with Alkylating Dose 90a
Low G4 Gy Pelvic Radiation Non-Alkylating Chemotherapy
Non-Alkylating Chemotherapy Anti-Metabolites
Anti-metabolites
a
Alkylating Agent Dose calculated according to Green et. al. [8••].
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
With respect to reproduction, an intact hypothalamic-pituitary axis is
required for ovulation and unassisted conception. Signs of hypothalamic
dysfunction include amenorrhea, elevated prolactin and suppressed levels
of gonadotropins. It has previously been reported that the relative risk
for a survivor achieving pregnancy after receiving a radiation dose of
3 Gy or more to the hypothalamic-pituitary axis (HPT) was 0.61 when
compared to a similar-age sibling cohort [8••]. A more recent report
which included patients who were not exposed to ovarian radiation and
controlled for relevant demographic variables including age at diagnosis
and marital status, suggests that even lower doses pose reproductive
risks. The hazard ratio of achieving pregnancy after 2.2–2.7 Gy to the
HPT was 0.67 compared to unexposed survivors [14•]. This study dem-
onstrates that even very low levels of radiation to the pituitary impair
fertility. Importantly, pituitary irradiation does not appear to be associated with
adverse perinatal outcomes [12••].
Chemotherapy
Chemotherapeutic agents, particularly alkylating agents, cause damage to the
ovary by cross-linking DNA and introducing single-stranded DNA breaks. His-
tologic evaluation of the ovaries in patients exposed to chemotherapy after
treatment demonstrate a range of findings, from decreased numbers of follicles
to absent follicles and stromal fibrosis [15]. Age is one of the most important
factors in determining the gonadotoxicity of treatment since ovarian reserve and
follicle number decrease with age. At older ages lower doses of chemotherapy
are required to produce ovarian failure. Independent of age, there is no evidence
that the prepubertal ovary is protected from damage from chemotherapy.
It has been demonstrated that the ovary is very susceptible to chemother-
apeutic treatments, particularly alkylating agents, in a dose-dependent fash-
ion. In one study, the relative risk of a female survivor of childhood cancer
reporting pregnancy was inversely related to the cumulative alkylator expo-
sure (typically summarized as alkylating agent dose, AAD) [8••]. For exam-
ple, an AAD score of three had a relative risk of 0.72 of pregnancy and an
AAD score of four had a relative risk of 0.65 of pregnancy compared to
survivors who received no alkylators. Additionally, the chemotherapeutic
agents CCNU and cyclophosphamide were independently associated with
impaired fertility in a dose-dependent fashion [8••].
It is clear that not all treatments are equally gonadotoxic. A study of
pediatric patients treated for Hodgkin’s lymphoma (HL) demonstrated that
measures of ovarian reserve, such as AMH, were significantly lower in
patients treated with ABVD or EBVD and MOPP therapy compared to ABVD
or EBVD without MOPP therapy [16]. Additionally, a current study in Europe
is comparing procarbazine and dacarbazine for treatment of HL in pediatric
patients with the hypothesis that dacarbazine is less harmful to the ovary but
just as efficacious as procarbazine [17].
It does not appear that alkylating drugs increase the risk of stillbirth
or neonatal death among the children of female survivors, even in those
who received the highest doses [12••]. Thus, there is currently no evi-
dence that having received previous chemotherapy results in genetic
defects in offspring.
Oncofertility (JS Jeruss, Section Editor)
Bone marrow transplantation
Hematopoietic cell transplantation (HCT) poses especially high reproductive
risks since conditioning regimens often require high-dose chemotherapy
and/or total body irradiation (TBI). In post-pubertal females, ovarian failure
has been observed in 65–84% of pediatric transplant recipients [18, 19]. In
pre-pubertal females, incomplete pubertal development or pubertal failure
has been reported in approximately 57% of females following HCT [20]. The
reproductive risks appear to be higher in conditioning regimens which
include cyclophosphamide with busulfan, and those that require TBI [21].
Treatment effects in males
Testicular radiation
The male testes are responsible for spermatogenesis and steroidogenesis.
Within the testes spermatogonial germ cells, supported by Sertoli cells,
generate sperm while the Leydig cells produce testosterone. With respect to
cancer therapies, it is important to understand that spermatogonia are more
sensitive to insults than Leydig cells. Therefore, male cancer survivors are
more likely to experience infertility than problems with pubertal develop-
ment or sexual function.
Testicular tissue is extremely sensitive to radiation, even more so than the
ovary. Radiation doses as low as 0.1 Gy can cause oligospermia, and re-
versible azospermia has been identified at doses as low as 0.35 Gy [22].
Though the testes are rarely directly radiated, even exposure due to scatter
radiation can have reproductive consequences. As mentioned, the Leydig
cells are somewhat less sensitive, and doses of at least 12 Gy are necessary to
impair pubertal development and sexual function [23]. Thus after radiation,
puberty may proceed normally even in the setting of oligospermia and is not
a reliable marker of recovered fertility.
Data from the CCSS demonstrate that doses of radiation over 7.5 Gy are
associated with a significantly lower likelihood of fathering a child, with a
hazard ratio of 0.12 compared with sibling controls [24••]. This study is
limited by the assessment of reproductive choices.
There is some theoretical concern that cancer therapies have the potential
to cause mutagenesis in the testes and therefore may impact the health of
offspring. The CCSS has not found evidence of increased genetic problems,
stillbirth, or neonatal death in men treated with gonadal radiation as
children [12••].
Chemotherapy
Chemotherapy, especially alkylating agents, also causes significant damage to
testicular tissue in an agent-specific and dose-dependent manner. Antime-
tabolite therapy such as methotrexate and mercaptopurine appear to have no
effect on male fertility, while cisplatin-based regimens temporarily impair
spermatogenesis with recovery shown in a significant number of patients
[25].
Data from the CCSS demonstrated that males with Hodgkin’s lymphoma
were the least likely to father a child (hazard ratio (HR) 0.34, confidence
interval (CI) 0.28–0.41) compared to other types of cancer, while those
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
diagnosed with Wilms tumor or neuroblastoma were as likely as healthy
siblings to father a pregnancy. Moreover, there was a dose–response effect
between the dose of alkylating agent and reduced likelihood to father a child
(HR 0.67, CI 0.51–0.88 for an AAD score of 2 and a HR 0.16, CI 0.08–0.32
for an AAD score of 6–11) [24••]. Specific chemotherapeutic agents appear
to impair fertility, including procarbazine in the second or third dose
tertile and cyclophosphamide in the third dose tertile [24••].
It is important to note that pregnancies resulting from assisted reproduc-
tive technologies were not included in this study. Therefore it is possible that
higher pregnancy rates may be achieved utilizing techniques such as in vitro
fertilization and intracytoplasmic sperm injection. Additionally, this study
examined rates of fathering a pregnancy five years or more after diagnosis
compared to sibling controls. It is possible that having a cancer diagnosis
influenced the decisions surrounding whether to pursue pregnancy and
timing of pregnancy. Information about this decision-making was not col-
lected in the study, though the analysis was controlled for marital status.
It is also important to note that male survivors without high-risk expo-
sures (including radiotherapy to the testes greater than 7.5 Gy, AAD score
greater than or equal to 2, or treatment with procarbazine or the third tertile
of cyclophosphamide) were just as likely as their siblings to father a child
[24••].
Pregnancies in partners of male cancer survivors appear to be reassuring.
The CCSS study has not found an increase in stillbirths or neonatal
deaths among children of males who received chemotherapeutic agents, even
when examining higher tertile doses of the drugs [12••].
Bone marrow transplantation
As in females, hematopoietic cell transplantation (HCT) poses especially
high risks to fertility, including delayed puberty, as it requires exposure to
high-dose chemotherapy and/or total body irradiation before the procedure.
Azoospermia in post-pubertal boys has been shown in 48–85% of those
undergoing HCT [21, 26, 27]. Incomplete pubertal development or pubertal
failure has been reported to occur in approximately 53% of prepubescent
males exposed to HCT [20]. Similar to females, the risk of delayed puberty is
dependent on the conditioning regimen administered, with the highest risk
protocols including a combination of cyclophosphamide and busulfan or
TBI [21]
The need for fertility preservation counseling
Given the clear risks to fertility associated with many therapies, it is imper-
ative that clinicians discuss the fertility risks and preservation options
with patients and families prior to the onset of treatment. Often clinicians
caring for cancer patients are hesitant to initiate such conversations, citing
reasons such as insufficient knowledge, lack of time to discuss the issue with
patients, or perceptions that the patient cannot delay treatment [28]. A recent
study in women age 18 or older found that pretreatment infertility
counseling by a fertility specialist and an oncologist resulted in lower regret
than counseling by an oncologist alone [29]. These issues are even more
Oncofertility (JS Jeruss, Section Editor)
important for the pediatric and adolescent population who often feel that
they are not given enough say in their medical choices. Importantly, the
ability to have biological children greatly effects the quality of life of
childhood cancer survivors in adulthood; it has been demonstrated that
a perceived loss of fertility is associated with lower marriage rates and
increased divorce rates in this population [30].
Furthermore, the American Society of Clinical Oncology (ASCO) and the
American Society of Reproductive Medicine (ASRM) have both issued
guidelines in recent years. The ASRM guidelines stipulate that ‘physicians
should inform cancer patients about options for fertility preservation and
future reproduction prior to treatment’, and specifically that ‘parents may act
to preserve the fertility of cancer patients who are minors if the child assents
and the intervention is likely to provide net benefits to the child [31]. These
guidelines were reinforced one year later when ASCO recommended that
clinicians should address issues of infertility at the earliest possible oppor-
tunity [32]. A schematic of the fertility preservation options available is
presented in Fig. 1.
Fertility preservation options
Females
Embryo cryopreservation
The options for pediatric and adolescent female cancer patients are
somewhat limited compared to their adult counterparts. Overall, the
established and most successful method of fertility preservation is em-
bryo cryopreservation. However, this requires that the patient be post-
pubescent and either have a partner or be willing to use donor sperm.
Figure 1. Fertility preservation options for children and young adults diagnosed with cancer a Not an option if there is a high
risk of ovarian metastases. b Experimental—only performed as part of a clinical study approved by an IRB. Used with permission
from SaveMyFertility.org.
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
Ovarian stimulation and oocyte retrieval can take 2–5 weeks and may
delay treatment of some malignancies. While embryo cryopreservation
has a success rate of 30–40% per transfer [33, 34], this may not be a
realistic approach for girls and young single women.
Oocyte cryopreservation
Though still considered experimental, oocyte cryopreservation is an-
other option that can be offered and conducted under a research
protocol overseen by an Institutional Review Board. Oocyte cryo-
preservation has the advantage of eliminating the need for a partner
or donor sperm, offering females reproductive autonomy. However,
it still requires that the patient be post-pubertal and undergo ovarian
stimulation and oocyte retrieval. Over 900 live births have been
reported from oocyte cryopreservation and outcomes appear gener-
ally reassuring [35]. Often adolescent patients present with cancers
such as leukemia and generally need immediate cancer therapy.
However, in post-pubescent females with available time, oocyte
cryopreservation may be the preferred method of fertility preserva-
tion. While historically success rates of oocyte cryopreservation have
been lower than embryo cryopreservation, recent improvements in
cryopreserved techniques such as vitrification have resulted in dra-
matic improvements in success. Indeed some studies suggest that
pregnancy rates are approaching those for embryo cryopreservation
and even fresh IVF [35–37]. There is evidence that more than 50% of
fertility clinics in the United States are offering oocyte cryopreservation
[36].
Ovarian tissue cryopreservation
Ovarian tissue cryopreservation is an experimental technique that
shows great promise. The procedure can be performed emergently,
minimizing delay in cancer treatment, and it is the only option
available for prepubescent girls. The procedure involves removing a
piece of ovarian cortical tissue or the whole ovary either laparos-
copically or through laparotomy. Typically, ovarian cortical strips are
cryopreserved and can subsequently be thawed and transplanted
later (frozen-thawed autologous tissue transplanation has resulted in
14 live births to date [38]). Alternatively, follicles extracted from
ovarian tissue may, in the future, be matured in vitro (IVM) to
produce mature oocytes that can be fertilized and transferred into
the uterus at a later time. While this latter method is still experi-
mental and has not yet resulted in any human live births, continued
investigation is being conducted in human models [39, 40]. IVM
would be preferable in cases where there is a high concern for
reseeding cancer. This is crucially important as there is a potential
risk when reimplanting tissue from a patient with a hematologic
disease. This risk appears to be significant in patients with leukemia
or other malignancies that may have spread to the ovary [41•]. A
recent publication by Dolmans et al. highlights this risk. Investiga-
tors transplanted thawed ovarian tissue from 18 patients with leu-
kemia into mice. Analyzed tissue demonstrated molecular evidence
Oncofertility (JS Jeruss, Section Editor)
of cancer in 7/12 cases of acute lymphoblastic leukemia (ALL) and
2/6 chronic myelogenous leukemia transplants. When mice were
sacrificed after six months and gross tumors were found in 4 mice,
all had received ovarian transplants from ALL patients [41•].
Given that ovarian tissue cryopreservation requires surgery under
general anesthesia, whenever possible ovarian tissue cryopreservation
should be coupled to another necessary surgical procedure, often a
central venous access procedure or tumor excision. In our own
practice, we have performed 12 cases of ovarian tissue cryopreser-
vations in female patients under the age of 20. Four had central
nervous system tumors, two had acute myeloid leukemia, two had
Ewing’s sarcoma, one had HL, one had myelodysplastic syndrome,
one had rhabdomyosarcoma, and one had sickle-cell disease. Of
these, 6 (50%) had received prior chemotherapy and were planning
for stem cell transplantation, 3 also had ovarian transposition per-
formed at the time of the biopsy, and one had already failed oocyte
retrieval. There have been no surgical or post-operative complica-
tions, but one patient has died from disease recurrence. No patients
have yet requested that the tissue be used clinically.
Oophoropexy
Other options for females include oophoropexy or fertility-sparing
cancer surgery. Oophoropexy involves surgically displacing the ova-
ries out of the radiation field (often above the pelvic brim or behind
the uterus) to protect against damage from radiation exposure. This
requires that the reproductive endocrinologist work closely with the
radiation oncologist to determine placement that minimizes the ra-
diation exposure. Potential complications include pelvic pain and
fallopian tube damage [42].
Ovarian suppression
Temporary suppression of ovarian function during chemotherapy
using gonadotropin-releasing hormone analogues (GnRH) has been
shown to reduce damage in animal models [43]. However, the exact
mechanism is not understood. Human studies suggest that menstrual
cycles may be more likely to resume in patients treated with GnRH
as compared to those not. However, studies are limited by small
samples and inappropriate comparison populations. Large prospec-
tive trials are lacking and randomized controlled trials are needed to
address this controversial issue.
Hormone replacement therapy
Prepubertal cancer patients with delayed pubertal development due
to ovarian failure should receive a physiologic regimen of hormone
replacement therapy to ensure optimal development of secondary sex
characteristics and adult stature [20]. After menarche, early loss of
ovarian function is associated with menopausal symptoms and long
term health risks including cardiovascular disease and osteoporosis.
While the optimal method of hormone replacement therapy is not
clear, some form ofhormone therapy is recommended since it effec-
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
tively treats menopausal symptoms and improves bone mineral
density [44, 45].
Males
Sperm cryopreservation
For males, sperm cryopreservation through masturbation is an established
and successful method of fertility preservation prior to cancer treatment. It is
non-invasive, relatively inexpensive, and can be performed without any delay
in the initiation of cancer therapy. All adolescent and young adult males
should be offered sperm cryopreservation prior to the onset of treatment.
Because sperm production begins around the age of 12 or 13 in males, ado-
lescent boys who are unable to produce semen through masturbation may
undergo penile vibratory stimulation or rectal electrostimulation under an-
esthesia [46]. Additionally, sperm may be collected via testicular or epididy-
mal aspiration under anesthesia if spermatogenesis is established. After
cryopreservation, stored sperm may be used for insemination or in vitro fer-
tilization (with intracytoplasmic sperm injection if counts are low) [47, 48].
Testicular tissue cryopreservation
For pre-pubescent males, the only options available are experimental and
require further development through basic science research. Cryopreserva-
tion and subsequent in vitro maturation or transplantation of spermatogo-
nial stem cells and testicular tissue has theoretical potential but cannot
currently be used in a clinical setting. A pilot study at The Children’s Hospital
of Philadelphia reports evidence ofacceptabilityand no cases of peri- or post-
operative complications from testicular tissue biopsy and cryopreservation
[49]. There is no conclusive evidence that GnRH analogues are effective in
protecting against testicular damage from chemotherapy in males [32].
Future directions
Oncofertility in the pediatric and adolescent populations continues to
evolve. We must work together in the basic, translational, and clinical science
communities to make progress in minimizing the gonadotoxicity of cancer
treatments, improving and developing novel fertility preservation techni-
ques, prospectively monitoring and detecting subfertility in cancer survivors,
and providing appropriate counseling to patients and their families about
these issues. This demands a comprehensive team approach including
oncologists and reproductive endocrinologists working together to provide
the best possible outcome for each young patient.
Disclosure
No potential conflicts of interest relevant to this article were reported.
Oncofertility (JS Jeruss, Section Editor)
References and Recommended Reading
Papers of particular interest, published recently, have been
highlighted as:
• Of importance
•• Of major importance
1. Stat bite: Childhood cancer survival, 1975–2003. J
Natl Cancer Inst. 2006;98:883.
2. Hewitt M, Rowland JH, Yancik R. Cancer survivors in
the United States: age, health, and disability. J Gerontol
Biol Sci Med Sci. 2003;58:82–91.
3. Wallace WH, Thomson AB, Kelsey TW. The radio-
sensitivity of the human oocyte. Hum Reprod.
2003;18:117–21.
4. Wallace WH, Thomson AB, Saran F, Kelsey TW. Pre-
dicting age of ovarian failure after radiation to a field
that includes the ovaries. Int J Radiat Oncol Biol
Phys. 2005;62:738–44.
5. Critchley HO, Wallace WH. Impact of cancer treat-
ment on uterine function. J Natl Cancer Inst Monogr.
2005;34:64–8.
6. Holm K, Nysom K, Brocks V, Hertz H, Jacobsen N,
Muller J. Ultrasound B-mode changes in the uterus
and ovaries and Doppler changes in the uterus after
total body irradiation and allogeneic bone marrow
transplantation in childhood. Bone Marrow Trans-
plant. 1999;23:259–63.
7. Wo JY, Viswanathan AN. Impact of radiotherapy on
fertility, pregnancy, and neonatal outcomes in female
cancer patients. Int J Radiat Oncol Biol Phys.
2009;73:1304–12.
8.•• Green DM, Kawashima T, Stovall M, Leisenring W,
Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison
LL. Fertility of female survivors of childhood cancer: a
report from the childhood cancer survivor study. J Clin
Oncol. 2009;27:2677–85.
This study from the CCSS cohort presents excellent epide-
miologic data on the relative risk of females achieving
pregnancy after receiving cancer therapy in childhood. The
data examined included all doses and volumes of radiation
therapy received and all chemotherapeutic agents including
cumulative doses of alkylating agents.
9.• Sudour H, Chastagner P, Claude L, Desandes E, Klein
M, Carrie C, Bernier V. Fertility and pregnancy out-
come after abdominal irradiation that included or
excluded the pelvis in childhood tumor survivors. Int
J Radiat Oncol Biol Phys. 2010;76:867–73.
An important recent retrospective study demonstrating a
dose–response effect of pelvic irradiation on subfertility in a
female childhood cancer survivor population.
10. Larsen EC, Muller J, Schmiegelow K, Rechnitzer C,
Andersen AN. Reduced ovarian function in long-term
survivors of radiation- and chemotherapy-treated
childhood cancer. J Clin Endocrinol Metab.
2003;88:5307–14.
11. Letourneau JM, Ebbel EE, Katz PP, Oktay KH, McCulloch
CE, Ai WZ, Chien AJ, Melisko ME, Cedars MI, Rosen MP.
Acute ovarian failure underestimates age-specific repro-
ductive impairment for young women undergoing che-
motherapy for cancer. Cancer. 2011.
12.•• Signorello LB, Mulvihill JJ, Green DM, Munro HM,
Stovall M, Weathers RE, Mertens AC, Whitton JA,
Robison LL, Boice Jr JD. Stillbirth and neonatal death
in relation to radiation exposure before conception: a
retrospective cohort study. Lancet. 2010;376:624–30.
An important study from CCSS cohort demonstrating that
pelvic irradiation in females is associated with adverse
pregnancy outcomes, while pituitary irradiation, irradiation
of the testes, and chemotherapy with alkylating agents were
not found to be risk factors.
13. Signorello LB, Cohen SS, Bosetti C, Stovall M, Kasper
CE, Weathers RE, Whitton JA, Green DM, Donaldson
SS, Mertens AC, Robison LL, Boice Jr JD. Female
survivors of childhood cancer: preterm birth and low
birth weight among their children. J Natl Cancer Inst.
2006;98:1453–61.
14.• Green DM, Nolan VG, Kawashima T, Stovall M,
Donaldson SS, Srivastava D, Leisenring W, Robison
LL, Sklar CA. Decreased fertility among female
childhood cancer survivors who received 22–27 Gy
hypothalamic/pituitary irradiation: a report from the
Childhood Cancer Survivor Study. Fertil Steril.
2011;95:1922–7, 1927.e1.
This important research presents data demonstrating that
even very low levels of irradiation to the pituitary may im-
pair fertility in females.
15. Familiari G, Caggiati A, Nottola SA, Ermini M, Di
Benedetto MR, Motta PM. Ultrastructure of human
ovarian primordial follicles after combination che-
motherapy for Hodgkin’s disease. Hum Reprod.
1993;8:2080–7.
16. van Beek RD, van den Heuvel-Eibrink MM, Laven JS, de
Jong FH, Themmen AP, Hakvoort-Cammel FG, van
den Bos C, van den Berg H, Pieters R. de Muinck Keizer-
Schrama SM. Anti-Mullerian hormone is a sensitive
serum marker for gonadal function in women treated
for Hodgkin’s lymphoma during childhood. J Clin
Endocrinol Metab. 2007;92:3869–74.
17. Wallace WH. Oncofertility and preservation of re-
productive capacity in children and young adults.
Cancer. 2011;117:2301–10.
18. Jadoul P, Anckaert E, Dewandeleer A, Steffens M,
Dolmans MM, Vermylen C, Smitz J, Donnez J, Maiter
D. Clinical and biologic evaluation of ovarian func-
tion in women treated by bone marrow transplan-
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
tation for various indications during childhood or
adolescence. Fertil Steril. 2011;96:126–133.e3.
19. Thibaud E, Rodriguez-Macias K, Trivin C, Esperou H,
Michon J, Brauner R. Ovarian function after bone
marrow transplantation during childhood. Bone
Marrow Transplant. 1998;21:287–90.
20. Sanders JE. Growth and development after hemato-
poietic cell transplant in children. Bone Marrow
Transplant. 2008;41:223–7.
21. Sanders JE, Hawley J, Levy W, Gooley T, Buckner CD,
Deeg HJ, Doney K, Storb R, Sullivan K, Witherspoon
R, Appelbaum FR. Pregnancies following high-dose
cyclophosphamide with or without high-dose bu-
sulfan or total-body irradiation and bone marrow
transplantation. Blood. 1996;87:3045–52.
22. Ash P. The influence of radiation on fertility in man.
Br J Radiol. 1980;53:271–8.
23. Sklar CA, Robison LL, Nesbit ME, Sather HN,
Meadows AT, Ortega JA, Kim TH, Hammond GD.
Effects of radiation on testicular function in long-
term survivors of childhood acute lymphoblastic
leukemia: a report from the Children Cancer Study
Group. J Clin Oncol. 1990;8:1981–7.
24.•• Green DM, Kawashima T, Stovall M, Leisenring W,
Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison
LL. Fertility of male survivors of childhood cancer: a
report from the Childhood Cancer Survivor Study. J
Clin Oncol. 2010;28:332–9.
This study from the CCSS cohort presents excellent epide-
miologic data on the relative risk of males siring offspring
after receiving cancer therapy in childhood. The data exam-
ined included all doses and volumes of radiation therapy
received and all chemotherapeutic agents including cumu-
lative doses of alkylating agents.
25. Hansen PV, Hansen SW. Gonadal function in men with
testicular germ cell cancer: the influence of cisplatin-
based chemotherapy. Eur Urol. 1993;23:153–6.
26. Howell SJ, Shalet SM. Spermatogenesis after cancer
treatment: damage and recovery. J Natl Cancer Inst
Monogr. 2005;34:12–7.
27. Anserini P, Chiodi S, Spinelli S, Costa M, Conte N,
Copello F, Bacigalupo A. Semen analysis following
allogeneic bone marrow transplantation. Additional
data for evidence-based counselling. Bone Marrow
Transplant. 2002;30:447–51.
28. Quinn GP, Vadaparampil ST, Lee JH, Jacobsen PB,
Bepler G, Lancaster J, Keefe DL, Albrecht TL. Physi-
cian referral for fertility preservation in oncology
patients: a national study of practice behaviors. J Clin
Oncol. 2009;27:5952–7.
29. Letourneau JM, Ebbel EE, Katz PP, Katz A, Ai WZ,
Chien AJ, Melisko ME, Cedars MI, Rosen MP. Pre-
treatment fertility counseling and fertility preserva-
tion improve quality of life in reproductive age
women with cancer. Cancer. 2011.
30. Janson C, Leisenring W, Cox C, Termuhlen AM,
Mertens AC, Whitton JA, Goodman P, Zeltzer L,
Robison LL, Krull KR, Kadan-Lottick NS. Predictors of
marriage and divorce in adult survivors of childhood
cancers: a report from the Childhood Cancer Survi-
vor Study. Cancer Epidemiol Biomarkers Prev.
2009;18:2626–35.
31. Ethics Committee of the American Society for Re-
productive Medicine. Fertility preservation and re-
production in cancer patients. Fertil Steril.
2005;83:1622–8.
32. Lee SJ, Schover LR, Partridge AH, Patrizio P, Wallace
WH, Hagerty K, Beck LN, Brennan LV, Oktay K.
American Society of Clinical Oncology. American
Society of Clinical Oncology recommendations on
fertility preservation in cancer patients. J Clin Oncol.
2006;24:2917–31.
33. Lobo RA. Potential options for preservation of fer-
tility in women. N Engl J Med. 2005;353:64–73.
34. Ginsburg ES, Yanushpolsky EH, Jackson KV. In vitro
fertilization for cancer patients and survivors. Fertil
Steril. 2001;75:705–10.
35. Noyes N, Porcu E, Borini A. Over 900 oocyte cryo-
preservation babies born with no apparent increase
in congenital anomalies. Reprod Biomed Online.
2009;18:769–76.
36. Rudick B, Opper N, Paulson R, Bendikson K, Chung
K. The status of oocyte cryopreservation in the United
States. Fertil Steril. 2010;94:2642–6.
37. Cobo A, Meseguer M, Remohi J, Pellicer A. Use of
cryo-banked oocytes in an ovum donation
programme: a prospective, randomized, controlled,
clinical trial. Hum Reprod. 2010;25:2239–46.
38. Donnez J, Silber S, Andersen CY, Demeestere I, Piver
P, Meirow D, Pellicer A, Dolmans MM. Children
born after autotransplantation of cryopreserved
ovarian tissue. a review of 13 live births. Ann Med.
2011;43:437–50.
39. Ting AY, Yeoman RR, Lawson MS, Zelinski MB. In
vitro development of secondary follicles from cryo-
preserved rhesus macaque ovarian tissue after slow-
rate freeze or vitrification. Hum Reprod.
2011;26:2461–72.
40. Xu M, Barrett SL, West-Farrell E, Kondapalli LA,
Kiesewetter SE, Shea LD, Woodruff TK. In vitro grown
human ovarian follicles from cancer patients support
oocyte growth. Hum Reprod. 2009;24:2531–40.
41.• Dolmans MM, Marinescu C, Saussoy P, Van
Langendonckt A, Amorim C, Donnez J. Reim-
plantation of cryopreserved ovarian tissue from
patients with acute lymphoblastic leukemia is
potentially unsafe. Blood. 2010;116:2908–14.
Demonstrated that reimplanted ovarian tissue from patients
with hematologic malignancies may potentially lead to
reseeding a cancer recurrence.
42. Morice P, Juncker L, Rey A, El-Hassan J, Haie-Meder
C, Castaigne D. Ovarian transposition for patients
with cervical carcinoma treated by radiosurgical
combination. Fertil Steril. 2000;74:743–8.
Oncofertility (JS Jeruss, Section Editor)
43. Chen H, Li J, Cui T, Hu L. Adjuvant gonadotropin-
releasing hormone analogues for the prevention of
chemotherapy induced premature ovarian failure in
premenopausal women. Cochrane Database Syst
Rev. 2011;11:CD008018.
44. Piccioni P, Scirpa P, D’Emilio I, Sora F, Scarciglia M,
Laurenti L, De Matteis S, Sica S, Leone G, Chiusolo P.
Hormonal replacement therapy after stem cell
transplantation. Maturitas. 2004;49:327–33.
45. Castelo-Branco C, Rovira M, Pons F, Duran M, Sierra
J, Vives A, Balasch J, Fortuny A, Vanrell J. The effect of
hormone replacement therapy on bone mass in
patients with ovarian failure due to bone marrow
transplantation. Maturitas. 1996;23:307–12.
46. Schmiegelow ML, Sommer P, Carlsen E, Sonksen JO,
Schmiegelow K, Muller JR. Penile vibratory stimula-
tion and electroejaculation before anticancer therapy
in two pubertal boys. J Pediatr Hematol Oncol.
1998;20:429–30.
47. Nicopoullos JD, Gilling-Smith C, Almeida PA, Ramsay
JW. The results of 154 ICSI cycles using surgically re-
trieved sperm from azoospermic men. Hum Reprod.
2004;19:579–85.
48. Levron J, Madgar I, Shefi S, Meirow D, Wiser A, Bider
D, Dor J, Raviv G. IVF outcome with cryopreserved
testicular sperm. Andrologia. 2011;43:48–51.
49. Ginsberg JP, Carlson CA, Lin K, Hobbie WL, Wigo
E, Wu X, Brinster RL, Kolon TF. An experimental
protocol for fertility preservation in prepubertal
boys recently diagnosed with cancer: a report of
acceptability and safety. Hum Reprod.
2010;25:37–41.
Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia

More Related Content

What's hot

Living with Advanced Breast Cancer: Challenges and Opportunities
Living with Advanced Breast Cancer: Challenges and OpportunitiesLiving with Advanced Breast Cancer: Challenges and Opportunities
Living with Advanced Breast Cancer: Challenges and Opportunities
bkling
 
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
Anil Gupta
 
Report Back from SGO: What’s the Latest in Uterine Cancer?
Report Back from SGO: What’s the Latest in Uterine Cancer?Report Back from SGO: What’s the Latest in Uterine Cancer?
Report Back from SGO: What’s the Latest in Uterine Cancer?
bkling
 
Topic-Driven Round Table on DCIS: What are Your Radiation Options?
Topic-Driven Round Table on DCIS: What are Your Radiation Options?Topic-Driven Round Table on DCIS: What are Your Radiation Options?
Topic-Driven Round Table on DCIS: What are Your Radiation Options?
bkling
 
Ovarian Cancer: What's New?
Ovarian Cancer: What's New?Ovarian Cancer: What's New?
Ovarian Cancer: What's New?
bkling
 
OVARIAN CANCER & NULLIPARITY
 OVARIAN CANCER & NULLIPARITY OVARIAN CANCER & NULLIPARITY
OVARIAN CANCER & NULLIPARITYNour Matar
 
Navigating Nutrition During Cancer and COVID-19
Navigating Nutrition During Cancer and COVID-19Navigating Nutrition During Cancer and COVID-19
Navigating Nutrition During Cancer and COVID-19
bkling
 
Uterine Cancer Recurrence: All You Need To Know
Uterine Cancer Recurrence: All You Need To KnowUterine Cancer Recurrence: All You Need To Know
Uterine Cancer Recurrence: All You Need To Know
bkling
 
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
bkling
 
Senology Newsletter - June 4, 2013
Senology Newsletter - June 4, 2013Senology Newsletter - June 4, 2013
Senology Newsletter - June 4, 2013
Senology.org
 
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
bkling
 
Optimising maternal-fetal outcomes in preterm labour: a decision analysis
Optimising maternal-fetal outcomes in preterm labour: a decision analysisOptimising maternal-fetal outcomes in preterm labour: a decision analysis
Optimising maternal-fetal outcomes in preterm labour: a decision analysis
Võ Tá Sơn
 
Communities of Color and Participation in Breast Cancer Research
Communities of Color and Participation in Breast Cancer ResearchCommunities of Color and Participation in Breast Cancer Research
Communities of Color and Participation in Breast Cancer Research
bkling
 
Am. j. epidemiol. 2016-robinson-388-99
Am. j. epidemiol. 2016-robinson-388-99Am. j. epidemiol. 2016-robinson-388-99
Am. j. epidemiol. 2016-robinson-388-99
Esposito Evelyn
 
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MDOvarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research Fund Alliance
 
When Cancer Comes Back, Sarah Adams, MD
When Cancer Comes Back, Sarah Adams, MDWhen Cancer Comes Back, Sarah Adams, MD
When Cancer Comes Back, Sarah Adams, MD
Ovarian Cancer Research Fund Alliance
 
Breast cancer handout
Breast cancer handoutBreast cancer handout
Breast cancer handout
หนู แฮมส์
 
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
bkling
 
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
bkling
 

What's hot (20)

Living with Advanced Breast Cancer: Challenges and Opportunities
Living with Advanced Breast Cancer: Challenges and OpportunitiesLiving with Advanced Breast Cancer: Challenges and Opportunities
Living with Advanced Breast Cancer: Challenges and Opportunities
 
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
Why HPV Vaccination for Preventing Cervical Cancer in India Should Become a P...
 
Report Back from SGO: What’s the Latest in Uterine Cancer?
Report Back from SGO: What’s the Latest in Uterine Cancer?Report Back from SGO: What’s the Latest in Uterine Cancer?
Report Back from SGO: What’s the Latest in Uterine Cancer?
 
Topic-Driven Round Table on DCIS: What are Your Radiation Options?
Topic-Driven Round Table on DCIS: What are Your Radiation Options?Topic-Driven Round Table on DCIS: What are Your Radiation Options?
Topic-Driven Round Table on DCIS: What are Your Radiation Options?
 
Ovarian Cancer: What's New?
Ovarian Cancer: What's New?Ovarian Cancer: What's New?
Ovarian Cancer: What's New?
 
OVARIAN CANCER & NULLIPARITY
 OVARIAN CANCER & NULLIPARITY OVARIAN CANCER & NULLIPARITY
OVARIAN CANCER & NULLIPARITY
 
Navigating Nutrition During Cancer and COVID-19
Navigating Nutrition During Cancer and COVID-19Navigating Nutrition During Cancer and COVID-19
Navigating Nutrition During Cancer and COVID-19
 
Uterine Cancer Recurrence: All You Need To Know
Uterine Cancer Recurrence: All You Need To KnowUterine Cancer Recurrence: All You Need To Know
Uterine Cancer Recurrence: All You Need To Know
 
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
SHARE Presentation: New Developments in the Medical Treatment of Breast Cance...
 
Senology Newsletter - June 4, 2013
Senology Newsletter - June 4, 2013Senology Newsletter - June 4, 2013
Senology Newsletter - June 4, 2013
 
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
SHARE: Report Back from Annual Meeting of American Association of Cancer Rese...
 
Breakout: Side Effects of Cancer Treatment: Robert Morgan MD
Breakout: Side Effects of Cancer Treatment: Robert Morgan MD Breakout: Side Effects of Cancer Treatment: Robert Morgan MD
Breakout: Side Effects of Cancer Treatment: Robert Morgan MD
 
Optimising maternal-fetal outcomes in preterm labour: a decision analysis
Optimising maternal-fetal outcomes in preterm labour: a decision analysisOptimising maternal-fetal outcomes in preterm labour: a decision analysis
Optimising maternal-fetal outcomes in preterm labour: a decision analysis
 
Communities of Color and Participation in Breast Cancer Research
Communities of Color and Participation in Breast Cancer ResearchCommunities of Color and Participation in Breast Cancer Research
Communities of Color and Participation in Breast Cancer Research
 
Am. j. epidemiol. 2016-robinson-388-99
Am. j. epidemiol. 2016-robinson-388-99Am. j. epidemiol. 2016-robinson-388-99
Am. j. epidemiol. 2016-robinson-388-99
 
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MDOvarian Cancer Research and Prevention, Andrew Berchuck, MD
Ovarian Cancer Research and Prevention, Andrew Berchuck, MD
 
When Cancer Comes Back, Sarah Adams, MD
When Cancer Comes Back, Sarah Adams, MDWhen Cancer Comes Back, Sarah Adams, MD
When Cancer Comes Back, Sarah Adams, MD
 
Breast cancer handout
Breast cancer handoutBreast cancer handout
Breast cancer handout
 
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
Invasive Lobular Carcinoma — Highlights from the First Ever ILC Symposium
 
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
SHARE Webinar: Latest Research on Metastatic Breast Cancer from SABCS 2015
 

Similar to Pediatric and young_adult_patients_and_oncofertility

Pp module content outline
Pp module content outlinePp module content outline
Pp module content outline
npenwill
 
Fertility drugs & oa ca ksa fv1
Fertility drugs & oa ca ksa fv1Fertility drugs & oa ca ksa fv1
Fertility drugs & oa ca ksa fv1
Basalama Ali
 
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
Amer Raza
 
Challenges in breast conserving surgery
Challenges in breast conserving surgeryChallenges in breast conserving surgery
Challenges in breast conserving surgeryDr./ Ihab Samy
 
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
alamocitycancercouncil
 
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
CrimsonPublishers-SBB
 
PREGNANCY ASSOCIATED BREAST CANCER
PREGNANCY ASSOCIATED BREAST CANCERPREGNANCY ASSOCIATED BREAST CANCER
PREGNANCY ASSOCIATED BREAST CANCER
snowhiteheart
 
10.1177 1758834013494988
10.1177 175883401349498810.1177 1758834013494988
10.1177 1758834013494988
Indra Wsd
 
Pdf of print ahead
Pdf of print aheadPdf of print ahead
Pdf of print aheadCh Rao
 
effective health care review
effective health care revieweffective health care review
effective health care reviewAdina Chen Bar
 
A prospective study of breast lump andclinicopathologicalanalysis in relation...
A prospective study of breast lump andclinicopathologicalanalysis in relation...A prospective study of breast lump andclinicopathologicalanalysis in relation...
A prospective study of breast lump andclinicopathologicalanalysis in relation...
iosrjce
 
Diagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docxDiagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docx
duketjoy27252
 
Diagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docxDiagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docx
lynettearnold46882
 
How to have quality of life in Advanced ovarian malignancy
How to have quality of life in Advanced ovarian malignancyHow to have quality of life in Advanced ovarian malignancy
How to have quality of life in Advanced ovarian malignancy
Rajesh Gajbhiye
 
Vorinostat Case Study
Vorinostat Case StudyVorinostat Case Study
Vorinostat Case Study
Ashley Jean
 
Breast Cancer in Young Women and its Impact on Reproductive Function
Breast Cancer in Young Women and its Impact on Reproductive FunctionBreast Cancer in Young Women and its Impact on Reproductive Function
Breast Cancer in Young Women and its Impact on Reproductive Function
Apollo Hospitals
 
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdfGuidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
Fernanda
 

Similar to Pediatric and young_adult_patients_and_oncofertility (20)

Pp module content outline
Pp module content outlinePp module content outline
Pp module content outline
 
Fertility drugs & oa ca ksa fv1
Fertility drugs & oa ca ksa fv1Fertility drugs & oa ca ksa fv1
Fertility drugs & oa ca ksa fv1
 
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
SIP 66 BJOG - 2021 - Manchanda - Risk‐Reducing Salpingo‐Oophorectomy and the ...
 
Challenges in breast conserving surgery
Challenges in breast conserving surgeryChallenges in breast conserving surgery
Challenges in breast conserving surgery
 
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
Breast Density, Breast Cancer Risk, and California Breast Density Notificatio...
 
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
Crimson Publishers_Oral Contraceptives and Breast Cancer Risk: A Study among ...
 
PREGNANCY ASSOCIATED BREAST CANCER
PREGNANCY ASSOCIATED BREAST CANCERPREGNANCY ASSOCIATED BREAST CANCER
PREGNANCY ASSOCIATED BREAST CANCER
 
10.1177 1758834013494988
10.1177 175883401349498810.1177 1758834013494988
10.1177 1758834013494988
 
Pdf of print ahead
Pdf of print aheadPdf of print ahead
Pdf of print ahead
 
effective health care review
effective health care revieweffective health care review
effective health care review
 
A prospective study of breast lump andclinicopathologicalanalysis in relation...
A prospective study of breast lump andclinicopathologicalanalysis in relation...A prospective study of breast lump andclinicopathologicalanalysis in relation...
A prospective study of breast lump andclinicopathologicalanalysis in relation...
 
Diagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docxDiagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docx
 
Diagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docxDiagnosed with breast cancer while on a family historyscreen.docx
Diagnosed with breast cancer while on a family historyscreen.docx
 
Uzma
UzmaUzma
Uzma
 
Gnurpptfinal
GnurpptfinalGnurpptfinal
Gnurpptfinal
 
How to have quality of life in Advanced ovarian malignancy
How to have quality of life in Advanced ovarian malignancyHow to have quality of life in Advanced ovarian malignancy
How to have quality of life in Advanced ovarian malignancy
 
Vorinostat Case Study
Vorinostat Case StudyVorinostat Case Study
Vorinostat Case Study
 
Breast Cancer in Young Women and its Impact on Reproductive Function
Breast Cancer in Young Women and its Impact on Reproductive FunctionBreast Cancer in Young Women and its Impact on Reproductive Function
Breast Cancer in Young Women and its Impact on Reproductive Function
 
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdfGuidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
Guidelines for Diagnostic Imaging During Pregnancy and Lactation.pdf
 
Empty
EmptyEmpty
Empty
 

More from Asociatia SOS Infertilitatea - www.vremcopii.ro

SOS Infertility Association Projects 2023.pdf
SOS Infertility Association Projects 2023.pdfSOS Infertility Association Projects 2023.pdf
SOS Infertility Association Projects 2023.pdf
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdf
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdfProiecte ale Asociatiei SOS Infertilitatea 2023.pdf
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdf
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Brosura OSPV 28 copii.pdf
Brosura OSPV 28 copii.pdfBrosura OSPV 28 copii.pdf
Brosura Acesti copii care altfel nu ar fi existat.pdf
Brosura Acesti copii care altfel nu ar fi existat.pdfBrosura Acesti copii care altfel nu ar fi existat.pdf
Brosura Acesti copii care altfel nu ar fi existat.pdf
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdfNr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Partnership „What do you know about YOUR FERTILITY?” magazine
Partnership „What do you know about YOUR FERTILITY?” magazinePartnership „What do you know about YOUR FERTILITY?” magazine
Partnership „What do you know about YOUR FERTILITY?” magazine
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolutionCe stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolutionCe stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Articol infertilitate revista ELLE
Articol infertilitate revista ELLEArticol infertilitate revista ELLE
Articol infertilitate revista ELLE
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Revista Ce stii despre FERTILITATEA TA?
Revista Ce stii despre FERTILITATEA TA?Revista Ce stii despre FERTILITATEA TA?
Revista Ce stii despre FERTILITATEA TA?
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Detalii Subprogram FIV MS 2018
Detalii Subprogram FIV MS 2018Detalii Subprogram FIV MS 2018
Detalii Subprogram FIV MS 2018
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Statistici subprogram FIV 2019
Statistici subprogram FIV 2019Statistici subprogram FIV 2019
Statistici subprogram FIV 2019
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Rate de succes Subprogram FIV MS 2011 - 2019
Rate de succes Subprogram FIV MS 2011 -  2019Rate de succes Subprogram FIV MS 2011 -  2019
Rate de succes Subprogram FIV MS 2011 - 2019
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ongCadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
Asociatia SOS Infertilitatea - www.vremcopii.ro
 
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspectiveNicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
Asociatia SOS Infertilitatea - www.vremcopii.ro
 

More from Asociatia SOS Infertilitatea - www.vremcopii.ro (20)

SOS Infertility Association Projects 2023.pdf
SOS Infertility Association Projects 2023.pdfSOS Infertility Association Projects 2023.pdf
SOS Infertility Association Projects 2023.pdf
 
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdf
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdfProiecte ale Asociatiei SOS Infertilitatea 2023.pdf
Proiecte ale Asociatiei SOS Infertilitatea 2023.pdf
 
Brosura OSPV 28 copii.pdf
Brosura OSPV 28 copii.pdfBrosura OSPV 28 copii.pdf
Brosura OSPV 28 copii.pdf
 
Brosura Acesti copii care altfel nu ar fi existat.pdf
Brosura Acesti copii care altfel nu ar fi existat.pdfBrosura Acesti copii care altfel nu ar fi existat.pdf
Brosura Acesti copii care altfel nu ar fi existat.pdf
 
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdfNr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
Nr 4 Ce stii despre FERTULITATEA TA noiembrie 2021.pdf
 
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
Revista „Ce stii despre FERTILITATEA TA?” nr 5, June 2022
 
Partnership „What do you know about YOUR FERTILITY?” magazine
Partnership „What do you know about YOUR FERTILITY?” magazinePartnership „What do you know about YOUR FERTILITY?” magazine
Partnership „What do you know about YOUR FERTILITY?” magazine
 
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
Apel la parteneriat revista „Ce stii despre fertilitatea ta” nr. 4, 1 nov 2021
 
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolutionCe stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, high resolution
 
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolutionCe stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
Ce stii despre FERTILITATEA TA nr 3, iunie 2021, low resolution
 
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
Presentation „What do you know about YOUR FERTILITY?” magazine, Romania
 
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
Apel la parteneriat revista „Ce știi despre FERTILITATEA TA?” nr 3, iunie 2021
 
Articol infertilitate revista ELLE
Articol infertilitate revista ELLEArticol infertilitate revista ELLE
Articol infertilitate revista ELLE
 
Revista Ce stii despre FERTILITATEA TA?
Revista Ce stii despre FERTILITATEA TA?Revista Ce stii despre FERTILITATEA TA?
Revista Ce stii despre FERTILITATEA TA?
 
Detalii Subprogram FIV MS 2018
Detalii Subprogram FIV MS 2018Detalii Subprogram FIV MS 2018
Detalii Subprogram FIV MS 2018
 
Statistici subprogram FIV 2019
Statistici subprogram FIV 2019Statistici subprogram FIV 2019
Statistici subprogram FIV 2019
 
Rate de succes Subprogram FIV MS 2011 - 2019
Rate de succes Subprogram FIV MS 2011 -  2019Rate de succes Subprogram FIV MS 2011 -  2019
Rate de succes Subprogram FIV MS 2011 - 2019
 
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ongCadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
Cadru fiscal-sponsorizari-decembrie-2019-si-regimul-fiscal-al-veniturilor-ong
 
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspectiveNicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
Nicoleta Cristea Brunel ESHRE Campus 2019 OHSS patients perspective
 
Globuri barza cuplu
Globuri barza cupluGloburi barza cuplu
Globuri barza cuplu
 

Recently uploaded

Penicillin...........................pptx
Penicillin...........................pptxPenicillin...........................pptx
Penicillin...........................pptx
Cherry
 
Structures and textures of metamorphic rocks
Structures and textures of metamorphic rocksStructures and textures of metamorphic rocks
Structures and textures of metamorphic rocks
kumarmathi863
 
general properties of oerganologametal.ppt
general properties of oerganologametal.pptgeneral properties of oerganologametal.ppt
general properties of oerganologametal.ppt
IqrimaNabilatulhusni
 
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
Health Advances
 
NuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final versionNuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final version
pablovgd
 
filosofia boliviana introducción jsjdjd.pptx
filosofia boliviana introducción jsjdjd.pptxfilosofia boliviana introducción jsjdjd.pptx
filosofia boliviana introducción jsjdjd.pptx
IvanMallco1
 
Orion Air Quality Monitoring Systems - CWS
Orion Air Quality Monitoring Systems - CWSOrion Air Quality Monitoring Systems - CWS
Orion Air Quality Monitoring Systems - CWS
Columbia Weather Systems
 
justice-and-fairness-ethics with example
justice-and-fairness-ethics with examplejustice-and-fairness-ethics with example
justice-and-fairness-ethics with example
azzyixes
 
Hemoglobin metabolism_pathophysiology.pptx
Hemoglobin metabolism_pathophysiology.pptxHemoglobin metabolism_pathophysiology.pptx
Hemoglobin metabolism_pathophysiology.pptx
muralinath2
 
Lateral Ventricles.pdf very easy good diagrams comprehensive
Lateral Ventricles.pdf very easy good diagrams comprehensiveLateral Ventricles.pdf very easy good diagrams comprehensive
Lateral Ventricles.pdf very easy good diagrams comprehensive
silvermistyshot
 
Anemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditionsAnemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditions
muralinath2
 
Richard's entangled aventures in wonderland
Richard's entangled aventures in wonderlandRichard's entangled aventures in wonderland
Richard's entangled aventures in wonderland
Richard Gill
 
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdfSCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SELF-EXPLANATORY
 
erythropoiesis-I_mechanism& clinical significance.pptx
erythropoiesis-I_mechanism& clinical significance.pptxerythropoiesis-I_mechanism& clinical significance.pptx
erythropoiesis-I_mechanism& clinical significance.pptx
muralinath2
 
Viksit bharat till 2047 India@2047.pptx
Viksit bharat till 2047  India@2047.pptxViksit bharat till 2047  India@2047.pptx
Viksit bharat till 2047 India@2047.pptx
rakeshsharma20142015
 
Cancer cell metabolism: special Reference to Lactate Pathway
Cancer cell metabolism: special Reference to Lactate PathwayCancer cell metabolism: special Reference to Lactate Pathway
Cancer cell metabolism: special Reference to Lactate Pathway
AADYARAJPANDEY1
 
Seminar of U.V. Spectroscopy by SAMIR PANDA
 Seminar of U.V. Spectroscopy by SAMIR PANDA Seminar of U.V. Spectroscopy by SAMIR PANDA
Seminar of U.V. Spectroscopy by SAMIR PANDA
SAMIR PANDA
 
RNA INTERFERENCE: UNRAVELING GENETIC SILENCING
RNA INTERFERENCE: UNRAVELING GENETIC SILENCINGRNA INTERFERENCE: UNRAVELING GENETIC SILENCING
RNA INTERFERENCE: UNRAVELING GENETIC SILENCING
AADYARAJPANDEY1
 
plant biotechnology Lecture note ppt.pptx
plant biotechnology Lecture note ppt.pptxplant biotechnology Lecture note ppt.pptx
plant biotechnology Lecture note ppt.pptx
yusufzako14
 
insect taxonomy importance systematics and classification
insect taxonomy importance systematics and classificationinsect taxonomy importance systematics and classification
insect taxonomy importance systematics and classification
anitaento25
 

Recently uploaded (20)

Penicillin...........................pptx
Penicillin...........................pptxPenicillin...........................pptx
Penicillin...........................pptx
 
Structures and textures of metamorphic rocks
Structures and textures of metamorphic rocksStructures and textures of metamorphic rocks
Structures and textures of metamorphic rocks
 
general properties of oerganologametal.ppt
general properties of oerganologametal.pptgeneral properties of oerganologametal.ppt
general properties of oerganologametal.ppt
 
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...The ASGCT Annual Meeting was packed with exciting progress in the field advan...
The ASGCT Annual Meeting was packed with exciting progress in the field advan...
 
NuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final versionNuGOweek 2024 Ghent - programme - final version
NuGOweek 2024 Ghent - programme - final version
 
filosofia boliviana introducción jsjdjd.pptx
filosofia boliviana introducción jsjdjd.pptxfilosofia boliviana introducción jsjdjd.pptx
filosofia boliviana introducción jsjdjd.pptx
 
Orion Air Quality Monitoring Systems - CWS
Orion Air Quality Monitoring Systems - CWSOrion Air Quality Monitoring Systems - CWS
Orion Air Quality Monitoring Systems - CWS
 
justice-and-fairness-ethics with example
justice-and-fairness-ethics with examplejustice-and-fairness-ethics with example
justice-and-fairness-ethics with example
 
Hemoglobin metabolism_pathophysiology.pptx
Hemoglobin metabolism_pathophysiology.pptxHemoglobin metabolism_pathophysiology.pptx
Hemoglobin metabolism_pathophysiology.pptx
 
Lateral Ventricles.pdf very easy good diagrams comprehensive
Lateral Ventricles.pdf very easy good diagrams comprehensiveLateral Ventricles.pdf very easy good diagrams comprehensive
Lateral Ventricles.pdf very easy good diagrams comprehensive
 
Anemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditionsAnemia_ different types_causes_ conditions
Anemia_ different types_causes_ conditions
 
Richard's entangled aventures in wonderland
Richard's entangled aventures in wonderlandRichard's entangled aventures in wonderland
Richard's entangled aventures in wonderland
 
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdfSCHIZOPHRENIA Disorder/ Brain Disorder.pdf
SCHIZOPHRENIA Disorder/ Brain Disorder.pdf
 
erythropoiesis-I_mechanism& clinical significance.pptx
erythropoiesis-I_mechanism& clinical significance.pptxerythropoiesis-I_mechanism& clinical significance.pptx
erythropoiesis-I_mechanism& clinical significance.pptx
 
Viksit bharat till 2047 India@2047.pptx
Viksit bharat till 2047  India@2047.pptxViksit bharat till 2047  India@2047.pptx
Viksit bharat till 2047 India@2047.pptx
 
Cancer cell metabolism: special Reference to Lactate Pathway
Cancer cell metabolism: special Reference to Lactate PathwayCancer cell metabolism: special Reference to Lactate Pathway
Cancer cell metabolism: special Reference to Lactate Pathway
 
Seminar of U.V. Spectroscopy by SAMIR PANDA
 Seminar of U.V. Spectroscopy by SAMIR PANDA Seminar of U.V. Spectroscopy by SAMIR PANDA
Seminar of U.V. Spectroscopy by SAMIR PANDA
 
RNA INTERFERENCE: UNRAVELING GENETIC SILENCING
RNA INTERFERENCE: UNRAVELING GENETIC SILENCINGRNA INTERFERENCE: UNRAVELING GENETIC SILENCING
RNA INTERFERENCE: UNRAVELING GENETIC SILENCING
 
plant biotechnology Lecture note ppt.pptx
plant biotechnology Lecture note ppt.pptxplant biotechnology Lecture note ppt.pptx
plant biotechnology Lecture note ppt.pptx
 
insect taxonomy importance systematics and classification
insect taxonomy importance systematics and classificationinsect taxonomy importance systematics and classification
insect taxonomy importance systematics and classification
 

Pediatric and young_adult_patients_and_oncofertility

  • 1. Current Treatment Options in Oncology DOI 10.1007/s11864-012-0183-7 Oncofertility (JS Jeruss, Section Editor) Pediatric and Young Adult Patients and Oncofertility Katherine E. Dillon, BA Clarisa R. Gracia, MD, MSCE* Address *University of Pennsylvania, 3701 Market Street, Suite 800, Philadelphia, PA 19104, USA Email: cgracia@obgyn.upenn.edu * Springer Science+Business Media, LLC 2012 Keywords Oncofertility I Reproductive risks I Fertility Preservation I Gonads Opinion statement With improving survival rates for pediatric and young adult cancer patients, consider- ations regarding the long-term effects of therapy have become more important. Cancer therapies are known to pose reproductive risks, though the effects may be unpredict- able. All at-risk patients should have a discussion about potential treatment-related infertility before the onset of cancer therapy, and should be offered appropriate fertil- ity preservation options. Embryo and sperm cryopreservation are considered standard therapy, though oocyte cryopreservation is gaining acceptance. Ovarian tissue cryo- preservation, while still experimental, is showing great promise. It is the only option currently available to prepubertal girls. No fertility preservation options exist for pre- pubertal boys though some institutions may offer experimental testicular tissue cryo- preservation. Introduction With advancements in the diagnosis and treatment of cancer in pediatric and adolescent patients, increasing numbers of survivors in this age group are looking to- ward a future as productive adults. For many this includes consideration of future fertility. Given that cancer treatments have been shown to impair normal pubertal development and may lead to infertility, it is imperative that oncologists discuss the issue of fertility preservation with patients prior to the onset of cancer treatment. This requires that clinicians be informed about the preservation options available and the re- productive challenges patients may face. This review article aims to use the most up-to-date data to inform the clinician about the effects of various cancer treatments on reproductive potential and the options for fertility preservation prior to cancer thera- py. It will focus on important recent advancements made in the field of oncofertility and will highlight fu- ture directions for fertility preservation technologies. Cancer in children and young adults The most common cancers in children are leuke- mias, followed by central nervous system malignan- cies, lymphomas, soft tissue sarcomas, renal cancer (Wilm’s Tumor) and bone tumors, in that order [1]. Treatments often involve a combination of sur- gery, radiation therapy, chemotherapy with alkylat-
  • 2. ing agents, or allogenic bone marrow transplant. Late effects of cancer are gaining more attention since over 80% of childhood cancer patients survive at least five years [2] and an estimated one in 640 adults ages 20–39 is currently a long-term child- hood cancer survivor [1]. However, it is a challenge to balance curative therapy while reducing the risks to fertility. This is especially difficult due to ever- changing cancer regimens and a lack of prospective data concerning their effects on fertility outcomes. One of the recognized challenges in studying the reproductive risks of cancer therapy is that outcomes such as fertility are difficult to capture. An individual’s fertility depends on many factors including the avail- ability of a fertile partner of the opposite sex, and a de- sire to conceive. The gold standard epidemiologic method to study fertility is to conduct prospective “time to pregnancy” studies. Such studies are generally not feasible in conditions where the exposure (cancer therapy) is relatively rare. Therefore, most studies have assessed other related outcomes such as menstrual function or reported pregnancy after cancer treatment. In females, measures of ovarian reserve, such antral folli- cle counts and ovarian volume assessed by ultrasound, and serum measures of inhibin B, anti-Mullerian hor- mone (AMH), estradiol, and follicle-stimulating hor- mone (FSH) have also been useful proxy outcomes. In the field of pediatric oncofertility most of the large-scale studies with epidemiologic data stem from findings from the childhood cancer survivorship study (CCSS), a large retrospective cohort study assessing a variety of late effects of childhood cancer. The CCSS is the largest cohort study of childhood and adolescent cancer survivors with 20,720 previously untreated patients with a new diagnosis before the age of 21 en- rolled. It is important to recognize that the effect of cancer therapies on an individual’s reproductive func- tion can be unpredictable. Nonetheless, the available data on the reproductive risks of cancer therapies will be reviewed, and is summarized in Table 1. Treatment effects in females Pelvic radiation The ovaries are particularly sensitive to damage from cancer treatments due to the finite number of un-renewable germ cells (follicles) present in the ovary. The ovary begins producing oocytes before birth and oocyte count peaks at 20 weeks gestation. After this peak, progressive follicular atresia occurs. Radiotherapy can cause ovarian failure by accelerating follicular atresia and destruction of the follicular pool. Earlier studies have shown that females treated with pelvic radiation doses in excess of 2 gray (Gy) are at high risk of permanent ovarian failure [3]. Using this data, Wallace et. al devel- oped a mathematical model that estimates the dose of ovarian radiation necessary to cause ovarian failure taking into account age at treatment. For example, the estimated mean radiation dose to the ovaries needed to cause sterilization is 16 Gy at age 15, while a dose of only 12 Gy is required at age 30 [4]. Radiotherapy can also have deleterious effects on the uterus as it causes scarring and diminished uterine blood flow in a dose-dependent fashion [5], resulting in decreased uterine volume [6, 7]. Recent studies have examined the reproductive risks associated with pel- vic radiotherapy in pediatric and adolescent cancer patients and have reported more relevant reproductive outcomes such as reports of pregnancy. For example, data from the CCSS demonstrated that the relative risk of achieving pregnancy at least five years post-treatment in female childhood cancer survivors exposed to 5 to 10 Gy of radiation to the pelvis was 0.56, and dropped to 0.18 for exposure greater than 10 Gy when survivors were compared to a similar-age sibling cohort [8••]. While the relative risks reported in this study clearly highlight the adverse effects of radiation Oncofertility (JS Jeruss, Section Editor)
  • 3. exposure, it is important to recognize the limitations of the data when counseling patients. This study examined rates of conceiving a pregnancy 5 years or more after diagnosis compared to a sibling cohort so as to match for socio- economic status and education. It is possible that having a cancer diagnosis itself influenced the decisions to pursue pregnancy or the timing of pregnancy in survivors. Information about whether patients attempted pregnancy was not in the study, though the analysis did control for marital status. Moreover, infor- mation on menstruation cycles or reported infertility was not available. A retrospective French study of childhood cancer survivors over the age of 18 (median age 27 years) found that direct pelvic irradiation was the most significant risk factor for subfertility, defined as no spontaneous menstrua- tion or pregnancies. The odds of subfertility were 17.6 in the exposed com- pared to the unexposed group [9•]. When controlling for adjuvant therapy, radiation doses to the reproductive organs of less than 4 Gy were not sig- nificantly associated with subfertility [9•]. The study also suggests that ovarian transposition or shielding may protect reproductive function. Limi- tations of this study include the convenience sample with small subgroup numbers and the suboptimal definition of subfertility [10, 11]. Not only does exposure to pelvic radiation increase risk of infertility, pel- vic irradiation is also associated with adverse pregnancy outcomes. Another CCSS study of 1281 female survivors demonstrated an association between pelvic irradiation exposure prior to menarche and subsequent stillbirth or neonatal death [12••]. Compared to survivors who did not receive radio- therapy, those exposed to 2.5–9.9 Gy of radiotherapy had a relative risk of 5.8 for stillbirth or neonatal death while those exposed to more than 10 Gy had a relative risk of 19. A previous study also demonstrated that pelvic irradiation increased the risk of preterm birth for female childhood cancer survivors [13]. Patients with a history of pelvic radiation therapy should be counseled about the perinatal risks of pregnancy and may consider using a gestational carrier if pregnancy is unsuccessful. Hypothalamic pituitary radiation Hypothalamic-pituitary radiation causes destruction and fibrosis of the pituitary and can lead to significant hypothalamic-pituitary dysfunction. Table 1. Reproductive risks of cancer therapy in pediatric and young adult patients Risk Females Males High 910 Gy Pelvic Radiation Testicular Radiation Hypothalamic Pituitary Radiation Hypothalamic Pituitary Radiation Hematopoetic Cell Transplantion Hematopoetic Cell Transplantation Total Body Irradiation Total Body Irradiation Chemotherapy with Alkylating Agent Dose ≥2a Chemotherapy with Alkylating Agent Dose ≥2a Medium 5–10 Gy Pelvic Radiation Platinum-containing drugs Chemotherapy with Alkylating Dose 90a Chemotherapy with Alkylating Dose 90a Low G4 Gy Pelvic Radiation Non-Alkylating Chemotherapy Non-Alkylating Chemotherapy Anti-Metabolites Anti-metabolites a Alkylating Agent Dose calculated according to Green et. al. [8••]. Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
  • 4. With respect to reproduction, an intact hypothalamic-pituitary axis is required for ovulation and unassisted conception. Signs of hypothalamic dysfunction include amenorrhea, elevated prolactin and suppressed levels of gonadotropins. It has previously been reported that the relative risk for a survivor achieving pregnancy after receiving a radiation dose of 3 Gy or more to the hypothalamic-pituitary axis (HPT) was 0.61 when compared to a similar-age sibling cohort [8••]. A more recent report which included patients who were not exposed to ovarian radiation and controlled for relevant demographic variables including age at diagnosis and marital status, suggests that even lower doses pose reproductive risks. The hazard ratio of achieving pregnancy after 2.2–2.7 Gy to the HPT was 0.67 compared to unexposed survivors [14•]. This study dem- onstrates that even very low levels of radiation to the pituitary impair fertility. Importantly, pituitary irradiation does not appear to be associated with adverse perinatal outcomes [12••]. Chemotherapy Chemotherapeutic agents, particularly alkylating agents, cause damage to the ovary by cross-linking DNA and introducing single-stranded DNA breaks. His- tologic evaluation of the ovaries in patients exposed to chemotherapy after treatment demonstrate a range of findings, from decreased numbers of follicles to absent follicles and stromal fibrosis [15]. Age is one of the most important factors in determining the gonadotoxicity of treatment since ovarian reserve and follicle number decrease with age. At older ages lower doses of chemotherapy are required to produce ovarian failure. Independent of age, there is no evidence that the prepubertal ovary is protected from damage from chemotherapy. It has been demonstrated that the ovary is very susceptible to chemother- apeutic treatments, particularly alkylating agents, in a dose-dependent fash- ion. In one study, the relative risk of a female survivor of childhood cancer reporting pregnancy was inversely related to the cumulative alkylator expo- sure (typically summarized as alkylating agent dose, AAD) [8••]. For exam- ple, an AAD score of three had a relative risk of 0.72 of pregnancy and an AAD score of four had a relative risk of 0.65 of pregnancy compared to survivors who received no alkylators. Additionally, the chemotherapeutic agents CCNU and cyclophosphamide were independently associated with impaired fertility in a dose-dependent fashion [8••]. It is clear that not all treatments are equally gonadotoxic. A study of pediatric patients treated for Hodgkin’s lymphoma (HL) demonstrated that measures of ovarian reserve, such as AMH, were significantly lower in patients treated with ABVD or EBVD and MOPP therapy compared to ABVD or EBVD without MOPP therapy [16]. Additionally, a current study in Europe is comparing procarbazine and dacarbazine for treatment of HL in pediatric patients with the hypothesis that dacarbazine is less harmful to the ovary but just as efficacious as procarbazine [17]. It does not appear that alkylating drugs increase the risk of stillbirth or neonatal death among the children of female survivors, even in those who received the highest doses [12••]. Thus, there is currently no evi- dence that having received previous chemotherapy results in genetic defects in offspring. Oncofertility (JS Jeruss, Section Editor)
  • 5. Bone marrow transplantation Hematopoietic cell transplantation (HCT) poses especially high reproductive risks since conditioning regimens often require high-dose chemotherapy and/or total body irradiation (TBI). In post-pubertal females, ovarian failure has been observed in 65–84% of pediatric transplant recipients [18, 19]. In pre-pubertal females, incomplete pubertal development or pubertal failure has been reported in approximately 57% of females following HCT [20]. The reproductive risks appear to be higher in conditioning regimens which include cyclophosphamide with busulfan, and those that require TBI [21]. Treatment effects in males Testicular radiation The male testes are responsible for spermatogenesis and steroidogenesis. Within the testes spermatogonial germ cells, supported by Sertoli cells, generate sperm while the Leydig cells produce testosterone. With respect to cancer therapies, it is important to understand that spermatogonia are more sensitive to insults than Leydig cells. Therefore, male cancer survivors are more likely to experience infertility than problems with pubertal develop- ment or sexual function. Testicular tissue is extremely sensitive to radiation, even more so than the ovary. Radiation doses as low as 0.1 Gy can cause oligospermia, and re- versible azospermia has been identified at doses as low as 0.35 Gy [22]. Though the testes are rarely directly radiated, even exposure due to scatter radiation can have reproductive consequences. As mentioned, the Leydig cells are somewhat less sensitive, and doses of at least 12 Gy are necessary to impair pubertal development and sexual function [23]. Thus after radiation, puberty may proceed normally even in the setting of oligospermia and is not a reliable marker of recovered fertility. Data from the CCSS demonstrate that doses of radiation over 7.5 Gy are associated with a significantly lower likelihood of fathering a child, with a hazard ratio of 0.12 compared with sibling controls [24••]. This study is limited by the assessment of reproductive choices. There is some theoretical concern that cancer therapies have the potential to cause mutagenesis in the testes and therefore may impact the health of offspring. The CCSS has not found evidence of increased genetic problems, stillbirth, or neonatal death in men treated with gonadal radiation as children [12••]. Chemotherapy Chemotherapy, especially alkylating agents, also causes significant damage to testicular tissue in an agent-specific and dose-dependent manner. Antime- tabolite therapy such as methotrexate and mercaptopurine appear to have no effect on male fertility, while cisplatin-based regimens temporarily impair spermatogenesis with recovery shown in a significant number of patients [25]. Data from the CCSS demonstrated that males with Hodgkin’s lymphoma were the least likely to father a child (hazard ratio (HR) 0.34, confidence interval (CI) 0.28–0.41) compared to other types of cancer, while those Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
  • 6. diagnosed with Wilms tumor or neuroblastoma were as likely as healthy siblings to father a pregnancy. Moreover, there was a dose–response effect between the dose of alkylating agent and reduced likelihood to father a child (HR 0.67, CI 0.51–0.88 for an AAD score of 2 and a HR 0.16, CI 0.08–0.32 for an AAD score of 6–11) [24••]. Specific chemotherapeutic agents appear to impair fertility, including procarbazine in the second or third dose tertile and cyclophosphamide in the third dose tertile [24••]. It is important to note that pregnancies resulting from assisted reproduc- tive technologies were not included in this study. Therefore it is possible that higher pregnancy rates may be achieved utilizing techniques such as in vitro fertilization and intracytoplasmic sperm injection. Additionally, this study examined rates of fathering a pregnancy five years or more after diagnosis compared to sibling controls. It is possible that having a cancer diagnosis influenced the decisions surrounding whether to pursue pregnancy and timing of pregnancy. Information about this decision-making was not col- lected in the study, though the analysis was controlled for marital status. It is also important to note that male survivors without high-risk expo- sures (including radiotherapy to the testes greater than 7.5 Gy, AAD score greater than or equal to 2, or treatment with procarbazine or the third tertile of cyclophosphamide) were just as likely as their siblings to father a child [24••]. Pregnancies in partners of male cancer survivors appear to be reassuring. The CCSS study has not found an increase in stillbirths or neonatal deaths among children of males who received chemotherapeutic agents, even when examining higher tertile doses of the drugs [12••]. Bone marrow transplantation As in females, hematopoietic cell transplantation (HCT) poses especially high risks to fertility, including delayed puberty, as it requires exposure to high-dose chemotherapy and/or total body irradiation before the procedure. Azoospermia in post-pubertal boys has been shown in 48–85% of those undergoing HCT [21, 26, 27]. Incomplete pubertal development or pubertal failure has been reported to occur in approximately 53% of prepubescent males exposed to HCT [20]. Similar to females, the risk of delayed puberty is dependent on the conditioning regimen administered, with the highest risk protocols including a combination of cyclophosphamide and busulfan or TBI [21] The need for fertility preservation counseling Given the clear risks to fertility associated with many therapies, it is imper- ative that clinicians discuss the fertility risks and preservation options with patients and families prior to the onset of treatment. Often clinicians caring for cancer patients are hesitant to initiate such conversations, citing reasons such as insufficient knowledge, lack of time to discuss the issue with patients, or perceptions that the patient cannot delay treatment [28]. A recent study in women age 18 or older found that pretreatment infertility counseling by a fertility specialist and an oncologist resulted in lower regret than counseling by an oncologist alone [29]. These issues are even more Oncofertility (JS Jeruss, Section Editor)
  • 7. important for the pediatric and adolescent population who often feel that they are not given enough say in their medical choices. Importantly, the ability to have biological children greatly effects the quality of life of childhood cancer survivors in adulthood; it has been demonstrated that a perceived loss of fertility is associated with lower marriage rates and increased divorce rates in this population [30]. Furthermore, the American Society of Clinical Oncology (ASCO) and the American Society of Reproductive Medicine (ASRM) have both issued guidelines in recent years. The ASRM guidelines stipulate that ‘physicians should inform cancer patients about options for fertility preservation and future reproduction prior to treatment’, and specifically that ‘parents may act to preserve the fertility of cancer patients who are minors if the child assents and the intervention is likely to provide net benefits to the child [31]. These guidelines were reinforced one year later when ASCO recommended that clinicians should address issues of infertility at the earliest possible oppor- tunity [32]. A schematic of the fertility preservation options available is presented in Fig. 1. Fertility preservation options Females Embryo cryopreservation The options for pediatric and adolescent female cancer patients are somewhat limited compared to their adult counterparts. Overall, the established and most successful method of fertility preservation is em- bryo cryopreservation. However, this requires that the patient be post- pubescent and either have a partner or be willing to use donor sperm. Figure 1. Fertility preservation options for children and young adults diagnosed with cancer a Not an option if there is a high risk of ovarian metastases. b Experimental—only performed as part of a clinical study approved by an IRB. Used with permission from SaveMyFertility.org. Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
  • 8. Ovarian stimulation and oocyte retrieval can take 2–5 weeks and may delay treatment of some malignancies. While embryo cryopreservation has a success rate of 30–40% per transfer [33, 34], this may not be a realistic approach for girls and young single women. Oocyte cryopreservation Though still considered experimental, oocyte cryopreservation is an- other option that can be offered and conducted under a research protocol overseen by an Institutional Review Board. Oocyte cryo- preservation has the advantage of eliminating the need for a partner or donor sperm, offering females reproductive autonomy. However, it still requires that the patient be post-pubertal and undergo ovarian stimulation and oocyte retrieval. Over 900 live births have been reported from oocyte cryopreservation and outcomes appear gener- ally reassuring [35]. Often adolescent patients present with cancers such as leukemia and generally need immediate cancer therapy. However, in post-pubescent females with available time, oocyte cryopreservation may be the preferred method of fertility preserva- tion. While historically success rates of oocyte cryopreservation have been lower than embryo cryopreservation, recent improvements in cryopreserved techniques such as vitrification have resulted in dra- matic improvements in success. Indeed some studies suggest that pregnancy rates are approaching those for embryo cryopreservation and even fresh IVF [35–37]. There is evidence that more than 50% of fertility clinics in the United States are offering oocyte cryopreservation [36]. Ovarian tissue cryopreservation Ovarian tissue cryopreservation is an experimental technique that shows great promise. The procedure can be performed emergently, minimizing delay in cancer treatment, and it is the only option available for prepubescent girls. The procedure involves removing a piece of ovarian cortical tissue or the whole ovary either laparos- copically or through laparotomy. Typically, ovarian cortical strips are cryopreserved and can subsequently be thawed and transplanted later (frozen-thawed autologous tissue transplanation has resulted in 14 live births to date [38]). Alternatively, follicles extracted from ovarian tissue may, in the future, be matured in vitro (IVM) to produce mature oocytes that can be fertilized and transferred into the uterus at a later time. While this latter method is still experi- mental and has not yet resulted in any human live births, continued investigation is being conducted in human models [39, 40]. IVM would be preferable in cases where there is a high concern for reseeding cancer. This is crucially important as there is a potential risk when reimplanting tissue from a patient with a hematologic disease. This risk appears to be significant in patients with leukemia or other malignancies that may have spread to the ovary [41•]. A recent publication by Dolmans et al. highlights this risk. Investiga- tors transplanted thawed ovarian tissue from 18 patients with leu- kemia into mice. Analyzed tissue demonstrated molecular evidence Oncofertility (JS Jeruss, Section Editor)
  • 9. of cancer in 7/12 cases of acute lymphoblastic leukemia (ALL) and 2/6 chronic myelogenous leukemia transplants. When mice were sacrificed after six months and gross tumors were found in 4 mice, all had received ovarian transplants from ALL patients [41•]. Given that ovarian tissue cryopreservation requires surgery under general anesthesia, whenever possible ovarian tissue cryopreservation should be coupled to another necessary surgical procedure, often a central venous access procedure or tumor excision. In our own practice, we have performed 12 cases of ovarian tissue cryopreser- vations in female patients under the age of 20. Four had central nervous system tumors, two had acute myeloid leukemia, two had Ewing’s sarcoma, one had HL, one had myelodysplastic syndrome, one had rhabdomyosarcoma, and one had sickle-cell disease. Of these, 6 (50%) had received prior chemotherapy and were planning for stem cell transplantation, 3 also had ovarian transposition per- formed at the time of the biopsy, and one had already failed oocyte retrieval. There have been no surgical or post-operative complica- tions, but one patient has died from disease recurrence. No patients have yet requested that the tissue be used clinically. Oophoropexy Other options for females include oophoropexy or fertility-sparing cancer surgery. Oophoropexy involves surgically displacing the ova- ries out of the radiation field (often above the pelvic brim or behind the uterus) to protect against damage from radiation exposure. This requires that the reproductive endocrinologist work closely with the radiation oncologist to determine placement that minimizes the ra- diation exposure. Potential complications include pelvic pain and fallopian tube damage [42]. Ovarian suppression Temporary suppression of ovarian function during chemotherapy using gonadotropin-releasing hormone analogues (GnRH) has been shown to reduce damage in animal models [43]. However, the exact mechanism is not understood. Human studies suggest that menstrual cycles may be more likely to resume in patients treated with GnRH as compared to those not. However, studies are limited by small samples and inappropriate comparison populations. Large prospec- tive trials are lacking and randomized controlled trials are needed to address this controversial issue. Hormone replacement therapy Prepubertal cancer patients with delayed pubertal development due to ovarian failure should receive a physiologic regimen of hormone replacement therapy to ensure optimal development of secondary sex characteristics and adult stature [20]. After menarche, early loss of ovarian function is associated with menopausal symptoms and long term health risks including cardiovascular disease and osteoporosis. While the optimal method of hormone replacement therapy is not clear, some form ofhormone therapy is recommended since it effec- Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
  • 10. tively treats menopausal symptoms and improves bone mineral density [44, 45]. Males Sperm cryopreservation For males, sperm cryopreservation through masturbation is an established and successful method of fertility preservation prior to cancer treatment. It is non-invasive, relatively inexpensive, and can be performed without any delay in the initiation of cancer therapy. All adolescent and young adult males should be offered sperm cryopreservation prior to the onset of treatment. Because sperm production begins around the age of 12 or 13 in males, ado- lescent boys who are unable to produce semen through masturbation may undergo penile vibratory stimulation or rectal electrostimulation under an- esthesia [46]. Additionally, sperm may be collected via testicular or epididy- mal aspiration under anesthesia if spermatogenesis is established. After cryopreservation, stored sperm may be used for insemination or in vitro fer- tilization (with intracytoplasmic sperm injection if counts are low) [47, 48]. Testicular tissue cryopreservation For pre-pubescent males, the only options available are experimental and require further development through basic science research. Cryopreserva- tion and subsequent in vitro maturation or transplantation of spermatogo- nial stem cells and testicular tissue has theoretical potential but cannot currently be used in a clinical setting. A pilot study at The Children’s Hospital of Philadelphia reports evidence ofacceptabilityand no cases of peri- or post- operative complications from testicular tissue biopsy and cryopreservation [49]. There is no conclusive evidence that GnRH analogues are effective in protecting against testicular damage from chemotherapy in males [32]. Future directions Oncofertility in the pediatric and adolescent populations continues to evolve. We must work together in the basic, translational, and clinical science communities to make progress in minimizing the gonadotoxicity of cancer treatments, improving and developing novel fertility preservation techni- ques, prospectively monitoring and detecting subfertility in cancer survivors, and providing appropriate counseling to patients and their families about these issues. This demands a comprehensive team approach including oncologists and reproductive endocrinologists working together to provide the best possible outcome for each young patient. Disclosure No potential conflicts of interest relevant to this article were reported. Oncofertility (JS Jeruss, Section Editor)
  • 11. References and Recommended Reading Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance 1. Stat bite: Childhood cancer survival, 1975–2003. J Natl Cancer Inst. 2006;98:883. 2. Hewitt M, Rowland JH, Yancik R. Cancer survivors in the United States: age, health, and disability. J Gerontol Biol Sci Med Sci. 2003;58:82–91. 3. Wallace WH, Thomson AB, Kelsey TW. The radio- sensitivity of the human oocyte. Hum Reprod. 2003;18:117–21. 4. Wallace WH, Thomson AB, Saran F, Kelsey TW. Pre- dicting age of ovarian failure after radiation to a field that includes the ovaries. Int J Radiat Oncol Biol Phys. 2005;62:738–44. 5. Critchley HO, Wallace WH. Impact of cancer treat- ment on uterine function. J Natl Cancer Inst Monogr. 2005;34:64–8. 6. Holm K, Nysom K, Brocks V, Hertz H, Jacobsen N, Muller J. Ultrasound B-mode changes in the uterus and ovaries and Doppler changes in the uterus after total body irradiation and allogeneic bone marrow transplantation in childhood. Bone Marrow Trans- plant. 1999;23:259–63. 7. Wo JY, Viswanathan AN. Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients. Int J Radiat Oncol Biol Phys. 2009;73:1304–12. 8.•• Green DM, Kawashima T, Stovall M, Leisenring W, Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison LL. Fertility of female survivors of childhood cancer: a report from the childhood cancer survivor study. J Clin Oncol. 2009;27:2677–85. This study from the CCSS cohort presents excellent epide- miologic data on the relative risk of females achieving pregnancy after receiving cancer therapy in childhood. The data examined included all doses and volumes of radiation therapy received and all chemotherapeutic agents including cumulative doses of alkylating agents. 9.• Sudour H, Chastagner P, Claude L, Desandes E, Klein M, Carrie C, Bernier V. Fertility and pregnancy out- come after abdominal irradiation that included or excluded the pelvis in childhood tumor survivors. Int J Radiat Oncol Biol Phys. 2010;76:867–73. An important recent retrospective study demonstrating a dose–response effect of pelvic irradiation on subfertility in a female childhood cancer survivor population. 10. Larsen EC, Muller J, Schmiegelow K, Rechnitzer C, Andersen AN. Reduced ovarian function in long-term survivors of radiation- and chemotherapy-treated childhood cancer. J Clin Endocrinol Metab. 2003;88:5307–14. 11. Letourneau JM, Ebbel EE, Katz PP, Oktay KH, McCulloch CE, Ai WZ, Chien AJ, Melisko ME, Cedars MI, Rosen MP. Acute ovarian failure underestimates age-specific repro- ductive impairment for young women undergoing che- motherapy for cancer. Cancer. 2011. 12.•• Signorello LB, Mulvihill JJ, Green DM, Munro HM, Stovall M, Weathers RE, Mertens AC, Whitton JA, Robison LL, Boice Jr JD. Stillbirth and neonatal death in relation to radiation exposure before conception: a retrospective cohort study. Lancet. 2010;376:624–30. An important study from CCSS cohort demonstrating that pelvic irradiation in females is associated with adverse pregnancy outcomes, while pituitary irradiation, irradiation of the testes, and chemotherapy with alkylating agents were not found to be risk factors. 13. Signorello LB, Cohen SS, Bosetti C, Stovall M, Kasper CE, Weathers RE, Whitton JA, Green DM, Donaldson SS, Mertens AC, Robison LL, Boice Jr JD. Female survivors of childhood cancer: preterm birth and low birth weight among their children. J Natl Cancer Inst. 2006;98:1453–61. 14.• Green DM, Nolan VG, Kawashima T, Stovall M, Donaldson SS, Srivastava D, Leisenring W, Robison LL, Sklar CA. Decreased fertility among female childhood cancer survivors who received 22–27 Gy hypothalamic/pituitary irradiation: a report from the Childhood Cancer Survivor Study. Fertil Steril. 2011;95:1922–7, 1927.e1. This important research presents data demonstrating that even very low levels of irradiation to the pituitary may im- pair fertility in females. 15. Familiari G, Caggiati A, Nottola SA, Ermini M, Di Benedetto MR, Motta PM. Ultrastructure of human ovarian primordial follicles after combination che- motherapy for Hodgkin’s disease. Hum Reprod. 1993;8:2080–7. 16. van Beek RD, van den Heuvel-Eibrink MM, Laven JS, de Jong FH, Themmen AP, Hakvoort-Cammel FG, van den Bos C, van den Berg H, Pieters R. de Muinck Keizer- Schrama SM. Anti-Mullerian hormone is a sensitive serum marker for gonadal function in women treated for Hodgkin’s lymphoma during childhood. J Clin Endocrinol Metab. 2007;92:3869–74. 17. Wallace WH. Oncofertility and preservation of re- productive capacity in children and young adults. Cancer. 2011;117:2301–10. 18. Jadoul P, Anckaert E, Dewandeleer A, Steffens M, Dolmans MM, Vermylen C, Smitz J, Donnez J, Maiter D. Clinical and biologic evaluation of ovarian func- tion in women treated by bone marrow transplan- Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia
  • 12. tation for various indications during childhood or adolescence. Fertil Steril. 2011;96:126–133.e3. 19. Thibaud E, Rodriguez-Macias K, Trivin C, Esperou H, Michon J, Brauner R. Ovarian function after bone marrow transplantation during childhood. Bone Marrow Transplant. 1998;21:287–90. 20. Sanders JE. Growth and development after hemato- poietic cell transplant in children. Bone Marrow Transplant. 2008;41:223–7. 21. Sanders JE, Hawley J, Levy W, Gooley T, Buckner CD, Deeg HJ, Doney K, Storb R, Sullivan K, Witherspoon R, Appelbaum FR. Pregnancies following high-dose cyclophosphamide with or without high-dose bu- sulfan or total-body irradiation and bone marrow transplantation. Blood. 1996;87:3045–52. 22. Ash P. The influence of radiation on fertility in man. Br J Radiol. 1980;53:271–8. 23. Sklar CA, Robison LL, Nesbit ME, Sather HN, Meadows AT, Ortega JA, Kim TH, Hammond GD. Effects of radiation on testicular function in long- term survivors of childhood acute lymphoblastic leukemia: a report from the Children Cancer Study Group. J Clin Oncol. 1990;8:1981–7. 24.•• Green DM, Kawashima T, Stovall M, Leisenring W, Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison LL. Fertility of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol. 2010;28:332–9. This study from the CCSS cohort presents excellent epide- miologic data on the relative risk of males siring offspring after receiving cancer therapy in childhood. The data exam- ined included all doses and volumes of radiation therapy received and all chemotherapeutic agents including cumu- lative doses of alkylating agents. 25. Hansen PV, Hansen SW. Gonadal function in men with testicular germ cell cancer: the influence of cisplatin- based chemotherapy. Eur Urol. 1993;23:153–6. 26. Howell SJ, Shalet SM. Spermatogenesis after cancer treatment: damage and recovery. J Natl Cancer Inst Monogr. 2005;34:12–7. 27. Anserini P, Chiodi S, Spinelli S, Costa M, Conte N, Copello F, Bacigalupo A. Semen analysis following allogeneic bone marrow transplantation. Additional data for evidence-based counselling. Bone Marrow Transplant. 2002;30:447–51. 28. Quinn GP, Vadaparampil ST, Lee JH, Jacobsen PB, Bepler G, Lancaster J, Keefe DL, Albrecht TL. Physi- cian referral for fertility preservation in oncology patients: a national study of practice behaviors. J Clin Oncol. 2009;27:5952–7. 29. Letourneau JM, Ebbel EE, Katz PP, Katz A, Ai WZ, Chien AJ, Melisko ME, Cedars MI, Rosen MP. Pre- treatment fertility counseling and fertility preserva- tion improve quality of life in reproductive age women with cancer. Cancer. 2011. 30. Janson C, Leisenring W, Cox C, Termuhlen AM, Mertens AC, Whitton JA, Goodman P, Zeltzer L, Robison LL, Krull KR, Kadan-Lottick NS. Predictors of marriage and divorce in adult survivors of childhood cancers: a report from the Childhood Cancer Survi- vor Study. Cancer Epidemiol Biomarkers Prev. 2009;18:2626–35. 31. Ethics Committee of the American Society for Re- productive Medicine. Fertility preservation and re- production in cancer patients. Fertil Steril. 2005;83:1622–8. 32. Lee SJ, Schover LR, Partridge AH, Patrizio P, Wallace WH, Hagerty K, Beck LN, Brennan LV, Oktay K. American Society of Clinical Oncology. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol. 2006;24:2917–31. 33. Lobo RA. Potential options for preservation of fer- tility in women. N Engl J Med. 2005;353:64–73. 34. Ginsburg ES, Yanushpolsky EH, Jackson KV. In vitro fertilization for cancer patients and survivors. Fertil Steril. 2001;75:705–10. 35. Noyes N, Porcu E, Borini A. Over 900 oocyte cryo- preservation babies born with no apparent increase in congenital anomalies. Reprod Biomed Online. 2009;18:769–76. 36. Rudick B, Opper N, Paulson R, Bendikson K, Chung K. The status of oocyte cryopreservation in the United States. Fertil Steril. 2010;94:2642–6. 37. Cobo A, Meseguer M, Remohi J, Pellicer A. Use of cryo-banked oocytes in an ovum donation programme: a prospective, randomized, controlled, clinical trial. Hum Reprod. 2010;25:2239–46. 38. Donnez J, Silber S, Andersen CY, Demeestere I, Piver P, Meirow D, Pellicer A, Dolmans MM. Children born after autotransplantation of cryopreserved ovarian tissue. a review of 13 live births. Ann Med. 2011;43:437–50. 39. Ting AY, Yeoman RR, Lawson MS, Zelinski MB. In vitro development of secondary follicles from cryo- preserved rhesus macaque ovarian tissue after slow- rate freeze or vitrification. Hum Reprod. 2011;26:2461–72. 40. Xu M, Barrett SL, West-Farrell E, Kondapalli LA, Kiesewetter SE, Shea LD, Woodruff TK. In vitro grown human ovarian follicles from cancer patients support oocyte growth. Hum Reprod. 2009;24:2531–40. 41.• Dolmans MM, Marinescu C, Saussoy P, Van Langendonckt A, Amorim C, Donnez J. Reim- plantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood. 2010;116:2908–14. Demonstrated that reimplanted ovarian tissue from patients with hematologic malignancies may potentially lead to reseeding a cancer recurrence. 42. Morice P, Juncker L, Rey A, El-Hassan J, Haie-Meder C, Castaigne D. Ovarian transposition for patients with cervical carcinoma treated by radiosurgical combination. Fertil Steril. 2000;74:743–8. Oncofertility (JS Jeruss, Section Editor)
  • 13. 43. Chen H, Li J, Cui T, Hu L. Adjuvant gonadotropin- releasing hormone analogues for the prevention of chemotherapy induced premature ovarian failure in premenopausal women. Cochrane Database Syst Rev. 2011;11:CD008018. 44. Piccioni P, Scirpa P, D’Emilio I, Sora F, Scarciglia M, Laurenti L, De Matteis S, Sica S, Leone G, Chiusolo P. Hormonal replacement therapy after stem cell transplantation. Maturitas. 2004;49:327–33. 45. Castelo-Branco C, Rovira M, Pons F, Duran M, Sierra J, Vives A, Balasch J, Fortuny A, Vanrell J. The effect of hormone replacement therapy on bone mass in patients with ovarian failure due to bone marrow transplantation. Maturitas. 1996;23:307–12. 46. Schmiegelow ML, Sommer P, Carlsen E, Sonksen JO, Schmiegelow K, Muller JR. Penile vibratory stimula- tion and electroejaculation before anticancer therapy in two pubertal boys. J Pediatr Hematol Oncol. 1998;20:429–30. 47. Nicopoullos JD, Gilling-Smith C, Almeida PA, Ramsay JW. The results of 154 ICSI cycles using surgically re- trieved sperm from azoospermic men. Hum Reprod. 2004;19:579–85. 48. Levron J, Madgar I, Shefi S, Meirow D, Wiser A, Bider D, Dor J, Raviv G. IVF outcome with cryopreserved testicular sperm. Andrologia. 2011;43:48–51. 49. Ginsberg JP, Carlson CA, Lin K, Hobbie WL, Wigo E, Wu X, Brinster RL, Kolon TF. An experimental protocol for fertility preservation in prepubertal boys recently diagnosed with cancer: a report of acceptability and safety. Hum Reprod. 2010;25:37–41. Pediatric and Young Adult Patients and Oncofertility Dillon and Gracia