Patients with hematologic malignancies and those undergoing hematopoietic cell transplantation (HCT) face a complex set of challenges when considering options for fertility preservation (FP). There are no standard options for prepubertal children, and women with hematologic malignancies may not be eligible for standard FP options. Fortunately, initial therapies for most blood cancers are not highly gonadotoxic, affording an important opportunity for postremission counseling and referrals to fertility specialists. These patients face a high risk of relapse, and many will be referred for autologous or allogeneic HCT, which carries an extremely high risk of infertility. The expanding indications for HCT to include benign hematologic disorders as well as autoimmune diseases mandate that all hematologists are familiar with these risks. Oncofertility researchers are continually pushing the boundaries of what may be possible for our patients; in the meantime, communication and shared decision-making between hematologists and patients, as well as program-building, education, and outreach are essential to ensure that these patients, many of whom will be cured, maintain all of their options for a fulfilling life after intensive therapy.

In 2016, there were 15.5 million cancer survivors in the United States, projected to increase to 20 million in 2026. About 7% to 8% will be survivors of a hematologic malignancy.1  For children and adolescents with hematologic malignancies, 5-year survival is high: 60% for acute myeloid leukemia, to nearly 90% for acute lymphoblastic leukemia, and 98% for Hodgkin lymphoma (HL). Younger adults also enjoy good outcomes, and death rates are declining.1  There will be approximately 242 000 survivors of hematopoietic cell transplant (HCT) by 2020, which accounts for roughly 1% of all survivors.2 

The reproductive system is among the most vulnerable to damaging sequelae of cancer and its treatments; patients also experience significant psychosocial and financial harms.3,4  This review is the first to focus on the unique fertility preservation (FP) concerns faced by patients with hematologic malignancies and those undergoing HCT. Existing guidelines for FP, such as those from the American Society for Reproductive Medicine,5  the National Comprehensive Cancer Network,6  and the American Society for Clinical Oncology7,8  address the general needs of all people with cancer, relying mainly on research in patients with breast and testicular cancer. There has been 1 clinical guide to FP in patients undergoing HCT.9  Patients with hematologic malignancies demand special consideration because (1) blood cancers such as acute lymphoblastic leukemia and HL are significantly more common in children and young adults, putting FP front and center; (2) patients with blood cancers may present with medical complications that preclude standard FP options; (3) some FP techniques are contraindicated in patients with hematologic malignancies; (4) sometimes up front, and nearly always at relapse, these patients are offered HCT, which is associated with almost universal infertility. Hence, there is a significant unmet need to address FP in the unique circumstances faced by hematology patients. This review will address FP for patients with acute leukemias, HL and non-HL, chronic myeloid leukemia (CML), and those receiving HCT. Increasingly, HCT is considered in the management of some nonmalignant conditions, including sickle cell disease (SCD) and autoimmune diseases; all hematologists should therefore be familiar with these issues.

Infertility is common after cancer treatment. Compared with their siblings, male cancer survivors are twice as likely to experience infertility10  and 44% less likely to father a child11 ; female survivors are 18% less likely to report a live birth12  (Figure 1). Survivors of HCT are 36-fold less likely to conceive a pregnancy.13 

Figure 1.

Cumulative incidence of pregnancies and livebirths in cancer survivors and siblings. Incidence curves are shown with upper and lower 95% confidence intervals. (A) Male cancer survivors, first ever pregnancy sired (left) and first ever livebirth sired (right). (B) Female cancer survivors, first ever pregnancy (left) and first ever livebirth (right). Reprinted from Chow et al12  with permission from Elsevier.

Figure 1.

Cumulative incidence of pregnancies and livebirths in cancer survivors and siblings. Incidence curves are shown with upper and lower 95% confidence intervals. (A) Male cancer survivors, first ever pregnancy sired (left) and first ever livebirth sired (right). (B) Female cancer survivors, first ever pregnancy (left) and first ever livebirth (right). Reprinted from Chow et al12  with permission from Elsevier.

Close modal

The prospect of infertility looms large,14-17  and distress about infertility and regret about lack of information can lead to depression and diminished quality of life.15,18-20  Decision regret (regret after patient treatment-related decisions)21  can be reduced by pretreatment counseling,22  yet physicians inconsistently provide information to patients about fertility risks. Up to 68% of patients report that fertility concerns were not discussed as part of their conversations about therapy.23-25  Collaborative care pathways can significantly increase referrals to reproductive specialists.26-30  Counseling and engagement in FP reduce reproductive concern31 . Sociodemographic disparities have been identified: women are referred to reproductive specialists less frequently than men32  and patients who are white, childless, educated, heterosexual, and privately insured are more likely to be counseled and referred.24,33,34  Simply having a discussion about infertility risks can lower distress and improve quality of life after treatment.18,35 

Women are born with all of the oocytes they will ever have. The number of oocytes peaks at approximately 7 million in the female fetus at 20 weeks’ gestation. The ovarian follicles undergo a steady process of atresia, falling to about 1 million at birth and continuing to decline until menopause, when the “stockpile” of oocytes is depleted.36,37  This fixed number of oocytes in women stands in contrast to the virtually unlimited number of sperm produced by men (provided that spermatogonial stem cells are present). Sperm are continually produced in the walls of the seminiferous tubules beginning in puberty at the rate of 45 to 270 million sperm per day38 ; sperm production takes approximately 74 days.39 

Men

Low sperm counts are common in men with cancer even before the initiation of therapy. Although best described in men with testicular cancer, men with HL and leukemia40,41  are also affected. The testes are exquisitely sensitive to damage by cytotoxic treatments resulting from the prolific and continual production of mature spermatozoa. Oligo- and azoospermia may result from damage to the Sertoli and Leydig cells, toxicity to the stem spermatogonia, and damage to the hypothalamic-pituitary-gonadal axis.42-45  Mature sperm are relatively resistant to depletion, but sensitive to DNA damage and therefore unsuitable for sperm banking if recently exposed to cytotoxic therapy.46  This highlights the importance of effective contraception from the very beginning of treatment.

Alkylating agents, platinum, and radiation are most toxic to sperm production. Age at the time of treatment does not appear to affect the risk of infertility in adults, but the impact of age at treatment in prepubertal boys is unknown.44,47  Permanent azoospermia occurs in >90% of men with HL after treatment with alkylator-intensive regimens such as mustargen, vincristine, procarbazine, and prednisone (MOPP) and cyclophosphamide, vincristine, procarbazine, and prednisone (COPP), but doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) results in only transient oligozoospermia with return to normal sperm counts in about 18 months.48  Estimating intensity of therapy with a model using cyclophosphamide equivalents can reliably predict fertility risk.49 

Radiation has complex effects on spermatogenesis. Even low doses of radiation (<0.1 Gy) can impair sperm production. Temporary azoospermia can occur after doses as low as 2 to 3 Gy; 4 to 6 Gy or more results in high rates of azoospermia. The likelihood of recovery becomes less likely with higher doses: patients receiving >6 Gy are likely to be permanently azoospermic.50  Oligozoospermia occurs approximately 10 weeks after radiation exposure and azoospermia at about 18 weeks.51  Fractionated therapy accentuates toxicity.52  After myeloablative (MA) conditioning before HCT, men nearly universally demonstrate hypergonadotropic hypogonadism and azoospermia.

Over time, sperm counts may recover: up to 50% of men may experience a return of spermatogenesis, varying from 12 to 18 months after treatment to 5 years or more, depending on the intensity of therapy.53  Recovery may occur even after high-dose alkylating agents and total body irradiation at 12 Gy.52,54  Because recovery is variable, expert panels suggest yearly surveillance for impaired spermatogenesis, testosterone deficiency, and sexual dysfunction.50  Graft-versus-host disease is an independent risk factor for prolonged azoospermia after autologous or allogeneic HCT (alloHCT), imparting a twofold increased risk.55 

Women

It is difficult to measure fertility reliably in women. Amenorrhea is not equivalent to infertility, and conversely, women who resume cyclical menses may not have normal fertility. A better endpoint is ovarian reserve, an assessment of the quantity of oocyte-containing follicles. Estradiol, follicle-stimulating hormone, luteinizing hormone, and cyclical menses are poor measures of ovarian reserve; anti-Müllerian hormone (AMH) levels56-58  and antral follicle counts (AFC) via transvaginal ultrasound may be more accurate.59  Women do not produce additional gametes and recovery of ovarian function does not occur. A common misperception is that if patients resume normal menstrual function after cancer therapy, their ovarian reserve is intact. In fact, premature ovarian failure (POF) is a common outcome even in women who resume menses after treatment.60-62  POF is one of the most important, and underrecognized,61  consequences of cytotoxic therapy, occurring from the depletion of primordial follicles and resultant diminution of ovarian reserve.61,63  In a large study of survivors of childhood cancer who were menstruating at age 21, the risk of menopause by age 25 was increased 3.6-fold in women who received abdominal radiation, 9.2-fold with alkylator-based chemotherapy, and 27-fold in women who received both.64  These findings have been confirmed.62,65,66 

The chance of permanent amenorrhea is related to the patient’s age, chemotherapeutic drug class, and dose. Alkylating agents, especially procarbazine and busulfan, and platinum confer an excess risk of POF.67  Older women are less likely to resume menses after treatment68  (Figure 2) and if menses do resume, POF is more likely to ensue, with a 23% increased risk of POF per year of age at treatment.69 

Figure 2.

Likelihood of women treated for breast cancer resuming menses after chemotherapy based on age at the time of therapy. Reprinted from Petrek et al68  with permission.

Figure 2.

Likelihood of women treated for breast cancer resuming menses after chemotherapy based on age at the time of therapy. Reprinted from Petrek et al68  with permission.

Close modal

There are a number of mechanisms by which chemotherapy may affect ovarian health. Because chemotherapy preferentially damages dividing cells, maturing follicles are most vulnerable and are rapidly depleted. Granulosa cells, active cells that nurture the growing follicles, may also be damaged, leading to death of the oocyte within that follicle.70  Primordial follicle apoptosis,71,72  ovarian atrophy, and cortical fibrosis have also been observed.73  It may seem paradoxical that oocytes resting in a quiescent state as primordial follicles are depleted at an accelerated pace when exposed to chemotherapy. Primordial follicles are recruited out of their inactive state into more vulnerable primary or secondary follicles during chemotherapy, rendering them susceptible to cytotoxic damage.37  Several pathways, including phosphatidylinositol 3-kinase/PTEN/Akt, are involved in activating the primordial follicle, triggering irreversible and unidirectional development to a mature follicle that, under normal circumstances, leads to either ovulation or atresia. In a mouse model, cyclophosphamide resulted in upregulation of the phosphatidylinositol 3-kinase/PTEN/Akt pathway, triggering apoptosis of maturing follicles.74  The concomitant reduction in inhibitory factors normally released by maturing follicles caused increased activation of primordial follicles, with a resultant depletion of this pool.75 

Radiation is directly toxic to oocytes; the dose at which 50% of oocytes die is <2 Gy.76  The age at which permanent ovarian failure occurs depends on the patient’s age at the time of treatment as well as the dose of radiation. The sterilizing dose at age 10 is 18.4 Gy, but falls to 14.3 Gy at age 30 (Figure 3).76  This explains the observation that permanent ovarian failure in women receiving MA total body irradiation is nearly universal. After alloHCT, graft-versus-host disease can target the ovaries and cause ovarian failure.77 

Figure 3.

The effective (red) and mean (blue) sterilizing dose of radiation for female patients by age at treatment. Reprinted from Wallace et al76  with permission from Elsevier.

Figure 3.

The effective (red) and mean (blue) sterilizing dose of radiation for female patients by age at treatment. Reprinted from Wallace et al76  with permission from Elsevier.

Close modal

Novel therapies: men and women

Some novel agents are gonadotoxic. The US Food and Drug Administration updated its label for bevacizumab after a 34% incidence of ovarian failure (compared with 2% in the control arm) occurred in a phase 2 study.78  Crizotinib, a tyrosine kinase inhibitor (TKI) directed at the anaplastic lymphoma kinase protein, is associated with hypogonadism in men.79  Fertility risks associated with other new therapies are unknown.

Men

Although low sperm counts are common in men with cancer at diagnosis, sperm collection for cryopreservation should be attempted with a goal of acquiring 3 samples before initiation of therapy for future use with intrauterine inseminations or in vitro fertilization (IVF); even with oligozoospermia, intracytoplasmic sperm injection (ICSI) is possible.80,81  Alternative methods of sperm sampling, such as penile vibratory stimulation or electroejaculation,82-85  may be effective for adolescents, or when there are ethical or religious objections or psychosocial barriers to masturbation. In men with azoospermia after treatment, testicular sperm extraction may be performed.86-90  Viable sperm are found in up to 50% of testicular sperm extractions91  and may yield successful pregnancies with IVF-ICSI. Sufficient time after cytotoxic therapy must be allowed to permit nonexposed spermatogonial stem cells to develop into spermatozoa.46 

Testicular tissue cryopreservation is experimental and is the only choice for pre-pubertal boys, but may be contaminated with leukemic cells.92-95  There is no role for hypothalamic-pituitary-gonadal suppression to preserve fertility in men.96 

Women

To obtain oocytes for either embryo or oocyte cryopreservation, hormonal stimulation (controlled ovarian hyperstimulation, COH) is used to trigger the simultaneous maturation of numerous follicles. Importantly, there are protocols that are menstrual cycle-independent (random start ovarian stimulation97 ) that result in induction of follicles within 10 to 12 days. Via a transvaginal needle approach, mature oocytes are extracted and then cryopreserved or incubated with sperm (either from a partner or a donor) to create embryos, which are then frozen. There are some drawbacks to oocyte retrieval: delaying cancer-directed therapy for almost 2 weeks may be medically risky; physicians may object to exposing patients to high estrogen levels because of the risk of liver toxicity or thrombosis; and an invasive procedure to obtain oocytes may not be feasible resulting from cytopenias, infections, or cardiopulmonary compromise.

The success of these approaches is dependent on the age of the woman at the time of oocyte retrieval, the method of cryopreservation, and any underlying concomitant endocrine or gynecologic conditions. Maternal age influences oocyte quantity and quality: aneuploidy is associated with increasing age. In 2017, US live birth rates from frozen embryo transfer cycles in the general population ranged from 5% (for women >42 years) to 43% (for women <35 years).98  In a European cohort, cancer survivors experienced live birth rates of up to 44%.99  A US study demonstrated similar cumulative pregnancy rates per transfer (37% in women with cancer vs 43% in age-matched controls).100  Pregnancy rates after embryo cryopreservation in women with cancer appear comparable to those undergoing fertility treatments for other indications.

The method of oocyte cryopreservation (slow freeze or vitrification) affects pregnancy rates. Vitrification is the preferred method because oocyte and embryo survival, fertilization, and pregnancy rates are superior.101 

Cryopreservation of oocytes (as opposed to embryos) may be preferred by women without partners or those who may have ethical objections to storing embryonic tissue.102  It also offers an option for women who live in countries that have limitations on embryo cryopreservation.7,102-104  Much of the data regarding pregnancies after frozen oocytes are derived from oocyte donors and suggest comparable live birth rates compared with fresh oocytes,105-108  but a meta-analysis indicates pregnancy rates may be about 25% lower using frozen oocytes.109  Data in cancer survivors are limited, largely because of current oocyte utilization rates <20%. Nonetheless, the available data are reassuring: a single-center study of patients who cryopreserved oocytes before cancer treatment (ages 28-40) demonstrated a 44% live birth rate.110 

Ovarian tissue cryopreservation is an experimental technique111,112  in which, via laparoscopy, the entire ovary or sections are harvested and frozen. After completion of cancer therapy, these tissues are reimplanted (usually overlaid on native ovary, Fallopian tube, or peritoneum).113-115  This is the only option for prepubertal girls, and it can be performed immediately, without hormonal stimulation. Reimplantation of ovarian tissue can recapitulate normal hormonal function, preserving not just fertility but also a normal hormonal milieu that may yield other health benefits such as improved bone and cardiovascular health and sexual function. This approach has resulted in pregnancy rates of approximately 30% and endocrine recovery in >90% of women.116,117  However, in both mouse models and humans with hematologic malignancies, low levels of cancer are detectable in ovarian tissue and can result in cancer relapse after reimplantation.118-120  Hence, this approach is contraindicated in women with newly diagnosed blood cancers. The safety of ovarian tissue cryopreservation in remission, particularly for women who may have markers for minimal residual disease which permit testing before reimplantation, remains to be determined.121-124  A single case report suggests this may be feasible.125 

Intense controversy surrounds the use of hormonal manipulation, such as gonadotropin releasing hormone analogs (GnRHa), for ovarian protection. Results of some randomized studies and meta-analyses show a benefit, mainly in women with breast cancer, limiting their generalizability.8  Data in hematology patients are scant; 1 study in patients with lymphoma indicated that GnRHa are not effective in preventing chemotherapy-induced POF.126  However, small studies suggest that GnRHa may be protective when administered before HCT.127,128 

Fertility may be better preserved with reduced intensity conditioning (RIC) as opposed to MA conditioning before alloHCT.129  However, the reader is urged to recognize the optimal therapy for the patient and to counsel patients carefully regarding the choice of conditioning when weighing fertility concerns. At this time, RIC offers only a theoretical benefit regarding FP, whereas high-quality evidence favors an MA approach for some patients, such as those with acute myeloid leukemia.130  Infertility risks can influence patients’ treatment decisions,131  highlighting the essential importance of patient education and shared decision-making when engaging in treatment planning.

The hematologist must collaborate with the patient and the reproductive specialist in weighing the substantial risks and benefits to these approaches (Table 1). Decision aids can be of great assistance132  and have been developed for young women with breast cancer133  as well as others.134,135  Tools that can be adapted for patients with hematologic malignancies are under development (Nancy N. Baxter, St. Michael’s Hospital, e-mail, 25 February 2019).

Patients with blood cancers are often acutely ill at presentation. A large mediastinal mass may interfere with the patient’s ability to lay flat for oocyte retrieval or may threaten cardiopulmonary compromise. Hyperleukocytosis, thrombocytopenia, disseminated intravascular coagulation, or infection may not permit an invasive procedure or a delay in therapy. Fertility risks and options should be discussed with these patients. The hematologist’s primary responsibility is to the urgent care of the patient, and often FP must be a secondary concern. In acutely ill patients who must begin cancer-directed therapy straightaway, the only possible intervention may be hormonal, such as GnRHa.136  Menstrual suppression, a side effect of GnRHa, can be beneficial because profound thrombocytopenia may result in menorrhagia. Despite (or perhaps because of) their limited options, women with blood cancers must understand the risks of infertility as early as possible, and ideally before the initiation of cancer-directed therapy, because a strong body of literature indicates that counseling, even without subsequent utilization of a FP intervention, improves quality of life and reduces distress in survivors.137,138 

Fortunately, the regimens that are commonly used for the initial treatment of acute leukemia and HL do not usually cause immediate and permanent ovarian failure (<20%).139  High-intensity HL regimens such as bleomycin, etoposide, doxorubicin, cyclophosphamide, oncovin, procarbazine, and prednisone (BEACOPP) and alkylator-based regimens such as cyclophosphamide, doxorubicin, oncovin, and prednisone (CHOP) for non-HL are associated with greater fertility risks, and patients who do not have the time or the ability to undergo oocyte retrieval should be counseled accordingly.140,141  It is especially important to refer all women to reproductive specialists for counseling and possible FP procedures after the completion of therapy because many women will experience POF, even if they resume normal menses after therapy. Also, salvage therapy for relapse includes even more intensive chemotherapy followed almost uniformly by HCT, with its attendant substantial risk of infertility. Therefore, for many women in remission, referral for possible oocyte retrieval is of great importance as this may be their only opportunity to preserve fertility options. AMH levels56  and oocyte yields for these women are lower but sufficient, and healthy pregnancies have been reported,142-145  suggesting that this is safe and feasible.

Women with bone marrow failure syndromes, including aplastic anemia, myelodysplastic syndrome, and GATA2 haploinsufficiency, all of whom may anticipate receiving high-dose therapy including alloHCT, have successfully undergone oocyte retrieval, with a low risk of complications.146,147 

A suggested comprehensive approach to FP in women with hematologic malignancies is outlined in Figure 4.

Figure 4.

Fertility preservation approach for women (postpuberty) with newly diagnosed hematologic malignancy.

Figure 4.

Fertility preservation approach for women (postpuberty) with newly diagnosed hematologic malignancy.

Close modal

CML

About 25% of patients with CML are younger than age 50. Most patients present in chronic phase, and lifelong TKI therapy is generally necessary. Men taking imatinib likely can safely father a pregnancy; data for other TKIs are minimal.148,149  TKIs are US Food and Drug Administration pregnancy category D drugs (“positive evidence of human fetal risk”). Women with CML in chronic phase who desire pregnancy must consider several options. Retrieving oocytes before or during TKI therapy allows the greatest flexibility: if a complete molecular remission is achieved and maintained, discontinuation of therapy may be possible150  and a pregnancy could then be pursued in relatively short order. Alternatively, if an optimal response is not obtained, or the patient does not wish to discontinue TKI, her oocytes could be fertilized and a gestational carrier may be used, a complex decision that partly depends on state151  and international laws.152 

Some women with CML may elect to stop TKI without meeting “treatment-free remission” criteria for TKI discontinuation153  to permit conception and pregnancy. We offer the opinion that candidates for this approach are women who: express a typical bcr/abl transcript monitored by standard quantitative polymerase chain reaction testing, achieve at least a major molecular response (<0.1%), undergo quantitative polymerase chain reaction bcr/abl testing monthly,153  and be amenable to, and appropriate candidates for, interferon154  if a 1-log rise in bcr/abl occurs.155  Women diagnosed with CML during pregnancy may be observed or managed with interferon.149  Reproductive counseling and options for women with CML must be individualized, and formal guidelines are needed.156,157 

Although nonmalignant diseases represent a fraction of transplants performed,158  these numbers are increasing. Autologous HCT has shown promise in several autoimmune diseases,159-161  particularly multiple sclerosis162,163  and systemic sclerosis, for which HCT was recently deemed a standard of care for patients with severe disease.164,165  Compromised fertility may be less frequent in these patients,166,167  possibly because they are less likely to have significant prior exposure to gonadotoxic therapies, but data are very limited.

Reproductive considerations for patients with SCD are complex168  because they may have reduced fertility at baseline. In women, this may be due to inflammation or vaso-occlusion with resultant ovarian ischemia.169  Hypogonadism occurs in up to 24% of men with SCD,170,171  with evidence for both primary and secondary testicular failure. Putative causes include pituitary or testicular infarctions due to vasoocclusion, and iron overload.172  Oligo- and azoospermia are common in men with SCD,173  and hydroxyurea can further lower sperm counts, sometimes irreversibly,174-176  leading some investigators to advocate for sperm cryopreservation before initiating hydroxyurea. Transfusion may improve hormonal profiles and sperm counts177  and could be considered before sperm cryopreservation.

AlloHCT may cure SCD178-180  and is associated with lower health care costs over time and improved health-related quality of life.181,182  Very little is known about fertility risks after HCT for SCD; this is a key question identified in a pediatric consortium position statement.183  MA HCT in children causes high rates of infertility and POF in female patients; male patients may demonstrate either hypergonadotropic or hypogonadotropic hypogonadism, suggesting that some boys may not have achieved puberty, which is often delayed in SCD.180,184,185  A small study of children receiving RIC alloHCT for SCD demonstrates relatively preserved gonadal function.186  Some adults with SCD feel that the possibility of infertility after alloHCT is an unacceptable risk.187  Further studies to understand patient preferences may help guide providers in counseling patients with SCD who are considering transplant.

FP for women with SCD may be risky. Ovarian hyperstimulation syndrome occurs in 1% to 5% of all COH cycles and may cause effusions, renal failure, abdominal pain, and thromboembolism.188  One small series described a high rate of ovarian hyperstimulation syndrome among women with SCD undergoing oocyte retrieval,189  while a series of younger patients suggested fewer complications.190  Ovarian tissue cryopreservation is feasible; the first live birth associated with this technique was reported in a prepubertal girl receiving alloHCT for SCD.191 

IVF with preimplantation genetic diagnosis may be an important consideration for people with SCD whose partners are carriers or also have SCD; it is therefore especially important to remove barriers to FP and reduce cost for these patients.192,193 

Barriers to FP include factors related to providers (lack of comfort with or knowledge about fertility, insufficient time to counsel patients); patients (lack of awareness of risks to fertility or of FP options, concern that their physician may not feel it is appropriate, ethical concerns, parental concerns); and the health care system (cost and insurance coverage; access to reproductive health care).7,28,194-197 

American Society for Clinical Oncology guidelines recommend empowering all providers to discuss these issues.8  Direct outreach to patients can improve engagement in FP: providing educational brochures and a facilitated reproductive specialist referral line resulted in a ninefold increase in patient referrals,198  and expedited urology access increased sperm banking sixfold.199 

Essentials of oncofertility program building and key resources are outlined in Tables 2 and 3.5,200-202  Fertile Hope has designated Centers of Excellence203  that embody these components.202 

In the United States, as of 2019, only 6 states mandate insurance coverage for patients with iatrogenic infertility (Connecticut, Delaware, Illinois, Maryland, New York, and Rhode Island).203  This coverage is generally limited to standard methods, and may have a cap or a substantial copayment.204  Similar to other complications of cancer treatments, such as breast reconstruction, coverage should be mandatory for iatrogenic infertility.205-207  American cancer survivors describe significant financial burden and stress related to paying for their FP procedures, using more than 20 different ways to cover their costs.208  Adolescents and young adults with cancer are particularly vulnerable to financial toxicity209 ; the substantial cost of FP may act synergistically to increase distress and may impact lifelong financial health and decisions about family-building.

In a recent study of European nations, 14 of 27 responding countries indicated that oocyte/embryo cryopreservation is covered by law.210  Information is scarce regarding FP in low- and middle-income countries, but 1 study demonstrated that barriers are similar to those in higher resourced settings.211  Cultural and legal challenges in some parts of the globe also represent significant hurdles.152 

Patients who identify as sexual and gender minorities (SGM) have substantial unmet health care needs.212,213  A qualitative study suggests that lesbian, gay, bisexual, transgender, queer/questioning survivors may be more open to nontraditional forms of family-building and less engaged by existing FP educational materials.212  FP interventions for all SGM individuals contemplating hormonal or surgical treatments are recommended,103,214,215  but uptake of these treatments is low.216  FP for SGM people with cancer is an entirely unexplored field.

There are several techniques in development that may open new avenues for FP. Immature oocytes may be retrieved and matured in vitro with either no or minimal ovarian stimulation.217-219  Oocytes and sperm may be developed from immature stem cells, including oogonial stem cells, spermatogonial stem cells,220  gonadal tissue culture,221  and embryonic or induced pluripotent stem cells.222-225  Use of protective agents, such as mTOR inhibitors,226  during chemotherapy may decrease follicular atresia during treatment. Intraovarian artery infusion of granulocyte colony-stimulating factor mobilized HSC may improve ovarian reserve as measured by AMH or AFC and has resulted in a handful of pregnancies in women who were poor responders to COH227,228 ; further studies in cancer patients are needed.

Improving the precision of risk stratification for POF is paramount. AMH, follicle-stimulating hormone, and AFC have all been proposed as biomarkers of ovarian function. Although AMH may predict time to menopause and chemotherapy-induced amenorrhea,229,230  its ability to predict fertility is limited.231  Prediction models must account for age at treatment, prior treatments, prior parity, concomitant medical conditions, and newer putative markers of ovarian reserve.232  Further research is needed to personalize treatment and surveillance plans.

Informing patients facing cancer or HCT about their reproductive risks and options can reduce distress and improve quality of life. The unique needs of patients with hematologic malignancies require a nuanced approach to FP. We must engage in counseling and shared decision-making before initiation of gonadotoxic therapy. Referral to reproductive specialists after completion of therapy is strongly suggested, especially if the provider and the patient agree that pretreatment intervention is not safe or feasible. Advancing technology, improving patient and provider education, facilitating access to reproductive care, and advocating to legislative bodies to support state or nationally mandated coverage is essential to optimize our patients’ reproductive futures.

Contribution: A.W.L. and S.S. wrote, reviewed, and approved the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Alison Wakoff Loren, Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, 6 S Pavilion, University of Pennsylvania, Philadelphia, PA 19104; e-mail: alison.loren@uphs.upenn.edu.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
.
Cancer statistics, 2018
.
CA Cancer J Clin
.
2018
;
68
(
1
):
7
-
30
.
2.
Majhail
NS
,
Tao
L
,
Bredeson
C
, et al
.
Prevalence of hematopoietic cell transplant survivors in the United States
.
Biol Blood Marrow Transplant
.
2013
;
19
(
10
):
1498
-
1501
.
3.
Hewitt
M
,
Greenfield
S
,
Stovall
E
.
From Cancer Patient to Cancer Survivor: Lost in Transition
.
Washington, DC
:
National Academies Press
;
2005
.
4.
Shapiro
CL
.
Cancer survivorship
.
N Engl J Med
.
2018
;
379
(
25
):
2438
-
2450
.
5.
Practice Committee of American Society for Reproductive Medicine
.
Fertility preservation in patients undergoing gonadotoxic therapy or gonadectomy: a committee opinion
.
Fertil Steril
.
2013
;
100
(
5
):
1214
-
1223
.
6.
Coccia
PF
,
Pappo
AS
,
Altman
J
, et al
.
Adolescent and young adult oncology, version 2.2014
.
J Natl Compr Canc Netw
.
2014
;
12
(
1
):
21
-
32
,
quiz 32
.
7.
Loren
AW
,
Mangu
PB
,
Beck
LN
, et al;
American Society of Clinical Oncology
.
Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update
.
J Clin Oncol
.
2013
;
31
(
19
):
2500
-
2510
.
8.
Oktay
K
,
Harvey
BE
,
Partridge
AH
, et al
.
Fertility preservation in patients with cancer: ASCO Clinical Practice Guideline Update
.
J Clin Oncol
.
2018
;
36
(
19
):
1994
-
2001
.
9.
Joshi
S
,
Savani
BN
,
Chow
EJ
, et al
.
Clinical guide to fertility preservation in hematopoietic cell transplant recipients
.
Bone Marrow Transplant
.
2014
;
49
(
4
):
477
-
484
.
10.
Wasilewski-Masker
K
,
Seidel
KD
,
Leisenring
W
, et al
.
Male infertility in long-term survivors of pediatric cancer: a report from the childhood cancer survivor study
.
J Cancer Surviv
.
2014
;
8
(
3
):
437
-
447
.
11.
Green
DM
,
Kawashima
T
,
Stovall
M
, et al
.
Fertility of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study
.
J Clin Oncol
.
2010
;
28
(
2
):
332
-
339
.
12.
Chow
EJ
,
Stratton
KL
,
Leisenring
WM
, et al
.
Pregnancy after chemotherapy in male and female survivors of childhood cancer treated between 1970 and 1999: a report from the Childhood Cancer Survivor Study cohort
.
Lancet Oncol
.
2016
;
17
(
5
):
567
-
576
.
13.
Carter
A
,
Robison
LL
,
Francisco
L
, et al
.
Prevalence of conception and pregnancy outcomes after hematopoietic cell transplantation: report from the Bone Marrow Transplant Survivor Study
.
Bone Marrow Transplant
.
2006
;
37
(
11
):
1023
-
1029
.
14.
Benedict
C
,
Shuk
E
,
Ford
JS
.
Fertility issues in adolescent and young adult cancer survivors
.
J Adolesc Young Adult Oncol
.
2016
;
5
(
1
):
48
-
57
.
15.
Benedict
C
,
Thom
B
,
Friedman
DN
,
Pottenger
E
,
Raghunathan
N
,
Kelvin
JF
.
Fertility information needs and concerns post-treatment contribute to lowered quality of life among young adult female cancer survivors
.
Support Care Cancer
.
2018
;
26
(
7
):
2209
-
2215
.
16.
Gorman
JR
,
Bailey
S
,
Pierce
JP
,
Su
HI
.
How do you feel about fertility and parenthood? The voices of young female cancer survivors
.
J Cancer Surviv
.
2012
;
6
(
2
):
200
-
209
.
17.
Green
D
,
Galvin
H
,
Horne
B
.
The psycho-social impact of infertility on young male cancer survivors: a qualitative investigation
.
Psychooncology
.
2003
;
12
(
2
):
141
-
152
.
18.
Benedict
C
,
Thom
B
,
Kelvin
JF
.
Young adult female cancer survivors’ decision regret about fertility preservation
.
J Adolesc Young Adult Oncol
.
2015
;
4
(
4
):
213
-
218
.
19.
Canada
AL
,
Schover
LR
.
The psychosocial impact of interrupted childbearing in long-term female cancer survivors
.
Psychooncology
.
2012
;
21
(
2
):
134
-
143
.
20.
Howard-Anderson
J
,
Ganz
PA
,
Bower
JE
,
Stanton
AL
.
Quality of life, fertility concerns, and behavioral health outcomes in younger breast cancer survivors: a systematic review
.
J Natl Cancer Inst
.
2012
;
104
(
5
):
386
-
405
.
21.
Brehaut
JC
,
O’Connor
AM
,
Wood
TJ
, et al
.
Validation of a decision regret scale
.
Med Decis Making
.
2003
;
23
(
4
):
281
-
292
.
22.
Chan
JL
,
Letourneau
J
,
Salem
W
, et al
.
Regret around fertility choices is decreased with pre-treatment counseling in gynecologic cancer patients
.
J Cancer Surviv
.
2017
;
11
(
1
):
58
-
63
.
23.
Duffy
CM
,
Allen
SM
,
Clark
MA
.
Discussions regarding reproductive health for young women with breast cancer undergoing chemotherapy
.
J Clin Oncol
.
2005
;
23
(
4
):
766
-
773
.
24.
Schover
LR
,
Brey
K
,
Lichtin
A
,
Lipshultz
LI
,
Jeha
S
.
Oncologists’ attitudes and practices regarding banking sperm before cancer treatment
.
J Clin Oncol
.
2002
;
20
(
7
):
1890
-
1897
.
25.
Schover
LR
,
Rybicki
LA
,
Martin
BA
,
Bringelsen
KA
.
Having children after cancer. A pilot survey of survivors’ attitudes and experiences
.
Cancer
.
1999
;
86
(
4
):
697
-
709
.
26.
Quinn
GP
,
Vadaparampil
ST
,
Lee
JH
, et al
.
Physician referral for fertility preservation in oncology patients: a national study of practice behaviors
.
J Clin Oncol
.
2009
;
27
(
35
):
5952
-
5957
.
27.
Sadri-Ardekani
H
,
Akhondi
MM
,
Vossough
P
, et al
.
Parental attitudes toward fertility preservation in boys with cancer: context of different risk levels of infertility and success rates of fertility restoration
.
Fertil Steril
.
2013
;
99
(
3
):
796
-
802
.
28.
Wyns
C
,
Collienne
C
,
Shenfield
F
, et al
.
Fertility preservation in the male pediatric population: factors influencing the decision of parents and children
.
Hum Reprod
.
2015
;
30
(
9
):
2022
-
2030
.
29.
Vu
JV
,
Llarena
NC
,
Estevez
SL
,
Moravek
MB
,
Jeruss
JS
.
Oncofertility program implementation increases access to fertility preservation options and assisted reproductive procedures for breast cancer patients
.
J Surg Oncol
.
2017
;
115
(
2
):
116
-
121
.
30.
Bradford
NK
,
Walker
R
,
Henney
R
,
Inglis
P
,
Chan
RJ
.
Improvements in clinical practice for fertility preservation among young cancer patients: results from bundled interventions
.
J Adolesc Young Adult Oncol
.
2018
;
7
(
1
):
37
-
45
.
31.
Shah
MS
,
Letourneau
JM
,
Niemasik
EE
,
Bleil
M
,
McCulloch
CE
,
Rosen
MP
.
The role of in-depth reproductive health counseling in addressing reproductive health concerns in female survivors of nongynecologic cancers
.
J Psychosoc Oncol
.
2016
;
34
(
4
):
305
-
317
.
32.
Köhler
TS
,
Kondapalli
LA
,
Shah
A
,
Chan
S
,
Woodruff
TK
,
Brannigan
RE
.
Results from the survey for preservation of adolescent reproduction (SPARE) study: gender disparity in delivery of fertility preservation message to adolescents with cancer
.
J Assist Reprod Genet
.
2011
;
28
(
3
):
269
-
277
.
33.
Quinn
GP
,
Block
RG
,
Clayman
ML
, et al
.
If you did not document it, it did not happen: rates of documentation of discussion of infertility risk in adolescent and young adult oncology patients’ medical records
.
J Oncol Pract
.
2015
;
11
(
2
):
137
-
144
.
34.
Shnorhavorian
M
,
Harlan
LC
,
Smith
AW
, et al;
AYA HOPE Study Collaborative Group
.
Fertility preservation knowledge, counseling, and actions among adolescent and young adult patients with cancer: a population-based study
.
Cancer
.
2015
;
121
(
19
):
3499
-
3506
.
35.
Deshpande
NA
,
Braun
IM
,
Meyer
FL
.
Impact of fertility preservation counseling and treatment on psychological outcomes among women with cancer: a systematic review
.
Cancer
.
2015
;
121
(
22
):
3938
-
3947
.
36.
Faddy
MJ
,
Gosden
RG
,
Gougeon
A
,
Richardson
SJ
,
Nelson
JF
.
Accelerated disappearance of ovarian follicles in mid-life: implications for forecasting menopause
.
Hum Reprod
.
1992
;
7
(
10
):
1342
-
1346
.
37.
Meirow
D
,
Biederman
H
,
Anderson
RA
,
Wallace
WH
.
Toxicity of chemotherapy and radiation on female reproduction
.
Clin Obstet Gynecol
.
2010
;
53
(
4
):
727
-
739
.
38.
Amann
RP
,
Howards
SS
.
Daily spermatozoal production and epididymal spermatozoal reserves of the human male
.
J Urol
.
1980
;
124
(
2
):
211
-
215
.
39.
Amann
RP
.
The cycle of the seminiferous epithelium in humans: a need to revisit?
J Androl
.
2008
;
29
(
5
):
469
-
487
.
40.
Chung
K
,
Irani
J
,
Knee
G
,
Efymow
B
,
Blasco
L
,
Patrizio
P
.
Sperm cryopreservation for male patients with cancer: an epidemiological analysis at the University of Pennsylvania
.
Eur J Obstet Gynecol Reprod Biol
.
2004
;
113
(
Suppl 1
):
S7
-
S11
.
41.
Rueffer
U
,
Breuer
K
,
Josting
A
, et al
.
Male gonadal dysfunction in patients with Hodgkin’s disease prior to treatment
.
Ann Oncol
.
2001
;
12
(
9
):
1307
-
1311
.
42.
Bar-Shira Maymon
B
,
Yogev
L
,
Marks
A
,
Hauser
R
,
Botchan
A
,
Yavetz
H
.
Sertoli cell inactivation by cytotoxic damage to the human testis after cancer chemotherapy
.
Fertil Steril
.
2004
;
81
(
5
):
1391
-
1394
.
43.
Wallace
WH
,
Anderson
RA
,
Irvine
DS
.
Fertility preservation for young patients with cancer: who is at risk and what can be offered?
Lancet Oncol
.
2005
;
6
(
4
):
209
-
218
.
44.
Smart
E
,
Lopes
F
,
Rice
S
, et al
.
Chemotherapy drugs cyclophosphamide, cisplatin and doxorubicin induce germ cell loss in an in vitro model of the prepubertal testis
.
Sci Rep
.
2018
;
8
(
1
):
1773
.
45.
Allen
CM
,
Lopes
F
,
Mitchell
R
,
Spears
N
.
How does chemotherapy treatment damage the prepubertal testis?
Reproduction
.
2018
;
REP-18-0221.R2
.
46.
Nangia
AK
,
Krieg
SA
,
Kim
SS
.
Clinical guidelines for sperm cryopreservation in cancer patients
.
Fertil Steril
.
2013
;
100
(
5
):
1203
-
1209
.
47.
Tournaye
H
,
Dohle
GR
,
Barratt
CL
.
Fertility preservation in men with cancer
.
Lancet
.
2014
;
384
(
9950
):
1295
-
1301
.
48.
Howell
S
,
Shalet
S
.
Gonadal damage from chemotherapy and radiotherapy
.
Endocrinol Metab Clin North Am
.
1998
;
27
(
4
):
927
-
943
.
49.
Green
DM
,
Liu
W
,
Kutteh
WH
, et al
.
Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study
.
Lancet Oncol
.
2014
;
15
(
11
):
1215
-
1223
.
50.
Skinner
R
,
Mulder
RL
,
Kremer
LC
, et al
.
Recommendations for gonadotoxicity surveillance in male childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group in collaboration with the PanCareSurFup Consortium
.
Lancet Oncol
.
2017
;
18
(
2
):
e75
-
e90
.
51.
Meistrich
ML
.
Effects of chemotherapy and radiotherapy on spermatogenesis in humans
.
Fertil Steril
.
2013
;
100
(
5
):
1180
-
1186
.
52.
Ash
P
.
The influence of radiation on fertility in man
.
Br J Radiol
.
1980
;
53
(
628
):
271
-
278
.
53.
Howell
SJ
,
Shalet
SM
.
Spermatogenesis after cancer treatment: damage and recovery
.
J Natl Cancer Inst Monogr
.
2005
;
2005
(
34
):
12
-
17
.
54.
Sanders
JE
,
Hawley
J
,
Levy
W
, et al
.
Pregnancies following high-dose cyclophosphamide with or without high-dose busulfan or total-body irradiation and bone marrow transplantation
.
Blood
.
1996
;
87
(
7
):
3045
-
3052
.
55.
Rovó
A
,
Aljurf
M
,
Chiodi
S
, et al;
Late Effects Working Party of the EBMT
.
Ongoing graft-versus-host disease is a risk factor for azoospermia after allogeneic hematopoietic stem cell transplantation: a survey of the Late Effects Working Party of the European Group for Blood and Marrow Transplantation
.
Haematologica
.
2013
;
98
(
3
):
339
-
345
.
56.
Lie Fong
S
,
Lugtenburg
PJ
,
Schipper
I
, et al
.
Anti-müllerian hormone as a marker of ovarian function in women after chemotherapy and radiotherapy for haematological malignancies
.
Hum Reprod
.
2008
;
23
(
3
):
674
-
678
.
57.
Su
HI
,
Sammel
MD
,
Green
J
, et al
.
Antimullerian hormone and inhibin B are hormone measures of ovarian function in late reproductive-aged breast cancer survivors
.
Cancer
.
2010
;
116
(
3
):
592
-
599
.
58.
van Beek
RD
,
van den Heuvel-Eibrink
MM
,
Laven
JS
, et al
.
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
(
10
):
3869
-
3874
.
59.
Practice Committee of the American Society for Reproductive Medicine
.
Testing and interpreting measures of ovarian reserve: a committee opinion
.
Fertil Steril
.
2015
;
103
(
3
):
e9
-
e17
.
60.
Sklar
CA
,
Mertens
AC
,
Mitby
P
, et al
.
Premature menopause in survivors of childhood cancer: a report from the childhood cancer survivor study
.
J Natl Cancer Inst
.
2006
;
98
(
13
):
890
-
896
.
61.
Letourneau
JM
,
Ebbel
EE
,
Katz
PP
, et al
.
Acute ovarian failure underestimates age-specific reproductive impairment for young women undergoing chemotherapy for cancer
.
Cancer
.
2012
;
118
(
7
):
1933
-
1939
.
62.
Metzger
ML
,
Meacham
LR
,
Patterson
B
, et al
.
Female reproductive health after childhood, adolescent, and young adult cancers: guidelines for the assessment and management of female reproductive complications
.
J Clin Oncol
.
2013
;
31
(
9
):
1239
-
1247
.
63.
Gracia
CR
,
Sammel
MD
,
Freeman
E
, et al
.
Impact of cancer therapies on ovarian reserve
.
Fertil Steril
.
2012
;
97
(
1
):
134
-
140
.
64.
Byrne
J
,
Fears
TR
,
Gail
MH
, et al
.
Early menopause in long-term survivors of cancer during adolescence
.
Am J Obstet Gynecol
.
1992
;
166
(
3
):
788
-
793
.
65.
Chemaitilly
W
,
Li
Z
,
Krasin
MJ
, et al
.
Premature Ovarian insufficiency in childhood cancer survivors: a report from the St. Jude Lifetime Cohort
.
J Clin Endocrinol Metab
.
2017
;
102
(
7
):
2242
-
2250
.
66.
Chiarelli
AM
,
Marrett
LD
,
Darlington
G
.
Early menopause and infertility in females after treatment for childhood cancer diagnosed in 1964-1988 in Ontario, Canada
.
Am J Epidemiol
.
1999
;
150
(
3
):
245
-
254
.
67.
Overbeek
A
,
van den Berg
MH
,
van Leeuwen
FE
,
Kaspers
GJ
,
Lambalk
CB
,
van Dulmen–den Broeder
E
.
Chemotherapy-related late adverse effects on ovarian function in female survivors of childhood and young adult cancer: a systematic review
.
Cancer Treat Rev
.
2017
;
53
:
10
-
24
.
68.
Petrek
JA
,
Naughton
MJ
,
Case
LD
, et al
.
Incidence, time course, and determinants of menstrual bleeding after breast cancer treatment: a prospective study
.
J Clin Oncol
.
2006
;
24
(
7
):
1045
-
1051
.
69.
van der Kaaij
MA
,
Heutte
N
,
Meijnders
P
, et al
.
Premature ovarian failure and fertility in long-term survivors of Hodgkin’s lymphoma: a European Organisation for Research and Treatment of Cancer Lymphoma Group and Groupe d’Etude des Lymphomes de l’Adulte Cohort Study
.
J Clin Oncol
.
2012
;
30
(
3
):
291
-
299
.
70.
Meirow
D
.
Reproduction post-chemotherapy in young cancer patients
.
Mol Cell Endocrinol
.
2000
;
169
(
1-2
):
123
-
131
.
71.
Meirow
D
,
Lewis
H
,
Nugent
D
,
Epstein
M
.
Subclinical depletion of primordial follicular reserve in mice treated with cyclophosphamide: clinical importance and proposed accurate investigative tool
.
Hum Reprod
.
1999
;
14
(
7
):
1903
-
1907
.
72.
Soleimani
R
,
Heytens
E
,
Darzynkiewicz
Z
,
Oktay
K
.
Mechanisms of chemotherapy-induced human ovarian aging: double strand DNA breaks and microvascular compromise
.
Aging (Albany NY)
.
2011
;
3
(
8
):
782
-
793
.
73.
Meirow
D
,
Dor
J
,
Kaufman
B
, et al
.
Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury
.
Hum Reprod
.
2007
;
22
(
6
):
1626
-
1633
.
74.
Chang
EM
,
Lim
E
,
Yoon
S
, et al
.
Cisplatin induces overactivation of the dormant primordial follicle through PTEN/AKT/FOXO3a pathway which leads to loss of ovarian reserve in mice
.
PLoS One
.
2015
;
10
(
12
):
e0144245
.
75.
Kalich-Philosoph
L
,
Roness
H
,
Carmely
A
, et al
.
Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility
.
Sci Transl Med
.
2013
;
5
(
185
):
185ra62
.
76.
Wallace
WH
,
Thomson
AB
,
Saran
F
,
Kelsey
TW
.
Predicting age of ovarian failure after radiation to a field that includes the ovaries
.
Int J Radiat Oncol Biol Phys
.
2005
;
62
(
3
):
738
-
744
.
77.
Shimoji
S
,
Hashimoto
D
,
Tsujigiwa
H
, et al
.
Graft-versus-host disease targets ovary and causes female infertility in mice
.
Blood
.
2017
;
129
(
9
):
1216
-
1225
.
78.
US Food and Drug Administration
.
Prescribing information for Bevacizumab. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761028s000lbl.pdf. Accessed 25 July 2019
.
79.
Weickhardt
AJ
,
Rothman
MS
,
Salian-Mehta
S
, et al
.
Rapid-onset hypogonadism secondary to crizotinib use in men with metastatic nonsmall cell lung cancer
.
Cancer
.
2012
;
118
(
21
):
5302
-
5309
.
80.
Babb
A
,
Farah
N
,
Lyons
C
, et al
.
Uptake and outcome of assisted reproductive techniques in long-term survivors of SCT
.
Bone Marrow Transplant
.
2012
;
47
(
4
):
568
-
573
.
81.
Kenney
LB
,
Antal
Z
,
Ginsberg
JP
, et al
.
Improving male reproductive health after childhood, adolescent, and young adult cancer: progress and future directions for survivorship research
.
J Clin Oncol
.
2018
;
36
(
21
):
2160
-
2168
.
82.
Schmiegelow
ML
,
Sommer
P
,
Carlsen
E
,
Sønksen
JO
,
Schmiegelow
K
,
Müller
JR
.
Penile vibratory stimulation and electroejaculation before anticancer therapy in two pubertal boys
.
J Pediatr Hematol Oncol
.
1998
;
20
(
5
):
429
-
430
.
83.
Adank
MC
,
van Dorp
W
,
Smit
M
, et al
.
Electroejaculation as a method of fertility preservation in boys diagnosed with cancer: a single-center experience and review of the literature
.
Fertil Steril
.
2014
;
102
(
1
):
199
-
205
.
84.
Berookhim
BM
,
Mulhall
JP
.
Outcomes of operative sperm retrieval strategies for fertility preservation among males scheduled to undergo cancer treatment
.
Fertil Steril
.
2014
;
101
(
3
):
805
-
811
.
85.
Barak
S
.
Fertility preservation in male patients with cancer
.
Best Pract Res Clin Obstet Gynaecol
.
2019
;
55
:
59
-
66
.
86.
Chan
PT
,
Palermo
GD
,
Veeck
LL
,
Rosenwaks
Z
,
Schlegel
PN
.
Testicular sperm extraction combined with intracytoplasmic sperm injection in the treatment of men with persistent azoospermia postchemotherapy
.
Cancer
.
2001
;
92
(
6
):
1632
-
1637
.
87.
Furuhashi
K
,
Ishikawa
T
,
Hashimoto
H
, et al
.
Onco-testicular sperm extraction: testicular sperm extraction in azoospermic and very severely oligozoospermic cancer patients
.
Andrologia
.
2013
;
45
(
2
):
107
-
110
.
88.
Schrader
M
,
Müller
M
,
Sofikitis
N
,
Straub
B
,
Krause
H
,
Miller
K
.
“Onco-tese”: testicular sperm extraction in azoospermic cancer patients before chemotherapy-new guidelines?
Urology
.
2003
;
61
(
2
):
421
-
425
.
89.
Bernie
AM
,
Mata
DA
,
Ramasamy
R
,
Schlegel
PN
.
Comparison of microdissection testicular sperm extraction, conventional testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta-analysis
.
Fertil Steril
.
2015
;
104
(
5
):
1099
-
1103
.
90.
Klami
R
,
Mankonen
H
,
Perheentupa
A
.
Successful microdissection testicular sperm extraction for men with non-obstructive azoospermia
.
Reprod Biol
.
2018
;
18
(
2
):
137
-
142
.
91.
Hsiao
W
,
Stahl
PJ
,
Osterberg
EC
, et al
.
Successful treatment of postchemotherapy azoospermia with microsurgical testicular sperm extraction: the Weill Cornell experience
.
J Clin Oncol
.
2011
;
29
(
12
):
1607
-
1611
.
92.
Jahnukainen
K
,
Hou
M
,
Petersen
C
,
Setchell
B
,
Söder
O
.
Intratesticular transplantation of testicular cells from leukemic rats causes transmission of leukemia
.
Cancer Res
.
2001
;
61
(
2
):
706
-
710
.
93.
Giudice
MG
,
de Michele
F
,
Poels
J
,
Vermeulen
M
,
Wyns
C
.
Update on fertility restoration from prepubertal spermatogonial stem cells: How far are we from clinical practice?
Stem Cell Res (Amst)
.
2017
;
21
:
171
-
177
.
94.
Picton
HM
,
Wyns
C
,
Anderson
RA
, et al;
ESHRE Task Force On Fertility Preservation In Severe Diseases
.
A European perspective on testicular tissue cryopreservation for fertility preservation in prepubertal and adolescent boys
.
Hum Reprod
.
2015
;
30
(
11
):
2463
-
2475
.
95.
Jurewicz
M
,
Hillelsohn
J
,
Mehta
S
,
Gilbert
BR
.
Fertility preservation in pubertal and pre-pubertal boys with cancer
.
Pediatr Endocrinol Rev
.
2018
;
15
(
3
):
234
-
243
.
96.
Thomson
AB
,
Anderson
RA
,
Irvine
DS
,
Kelnar
CJ
,
Sharpe
RM
,
Wallace
WH
.
Investigation of suppression of the hypothalamic-pituitary-gonadal axis to restore spermatogenesis in azoospermic men treated for childhood cancer
.
Hum Reprod
.
2002
;
17
(
7
):
1715
-
1723
.
97.
Danis
RB
,
Pereira
N
,
Elias
RT
.
Random start ovarian stimulation for oocyte or embryo cryopreservation in women desiring fertility preservation prior to gonadotoxic cancer therapy
.
Curr Pharm Biotechnol
.
2017
;
18
(
8
):
609
-
613
.
98.
National Summary Report
:
Society for Assisted Reproductive Technology, https://www.sartcorsonline.com/rptCSR_PublicMultYear.aspx?reportingYear=2017#. Accessed 15 July 2019
.
99.
Dolmans
MM
,
Hollanders de Ouderaen
S
,
Demylle
D
,
Pirard
C
.
Utilization rates and results of long-term embryo cryopreservation before gonadotoxic treatment
.
J Assist Reprod Genet
.
2015
;
32
(
8
):
1233
-
1237
.
100.
Cardozo
ER
,
Thomson
AP
,
Karmon
AE
,
Dickinson
KA
,
Wright
DL
,
Sabatini
ME
.
Ovarian stimulation and in-vitro fertilization outcomes of cancer patients undergoing fertility preservation compared to age matched controls: a 17-year experience
.
J Assist Reprod Genet
.
2015
;
32
(
4
):
587
-
596
.
101.
Rienzi
L
,
Gracia
C
,
Maggiulli
R
, et al
.
Oocyte, embryo and blastocyst cryopreservation in ART: systematic review and meta-analysis comparing slow-freezing versus vitrification to produce evidence for the development of global guidance
.
Hum Reprod Update
.
2017
;
23
(
2
):
139
-
155
.
102.
Practice Committees of American Society for Reproductive MedicineSociety for Assisted Reproductive Technology
.
Mature oocyte cryopreservation: a guideline
.
Fertil Steril
.
2013
;
99
(
1
):
37
-
43
.
103.
Ethics Committee of American Society for Reproductive Medicine
.
Fertility preservation and reproduction in patients facing gonadotoxic therapies: a committee opinion
.
Fertil Steril
.
2013
;
100
(
5
):
1224
-
1231
.
104.
Peccatori
FA
,
Azim
HA
Jr
,
Orecchia
R
, et al;
ESMO Guidelines Working Group
.
Cancer, pregnancy and fertility: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
.
2013
;
24
(
Suppl 6
):
vi160
-
vi170
.
105.
Cobo
A
,
Kuwayama
M
,
Pérez
S
,
Ruiz
A
,
Pellicer
A
,
Remohí
J
.
Comparison of concomitant outcome achieved with fresh and cryopreserved donor oocytes vitrified by the Cryotop method
.
Fertil Steril
.
2008
;
89
(
6
):
1657
-
1664
.
106.
Cobo
A
,
Meseguer
M
,
Remohí
J
,
Pellicer
A
.
Use of cryo-banked oocytes in an ovum donation programme: a prospective, randomized, controlled, clinical trial
.
Hum Reprod
.
2010
;
25
(
9
):
2239
-
2246
.
107.
Rienzi
L
,
Romano
S
,
Albricci
L
, et al
.
Embryo development of fresh ‘versus’ vitrified metaphase II oocytes after ICSI: a prospective randomized sibling-oocyte study
.
Hum Reprod
.
2010
;
25
(
1
):
66
-
73
.
108.
Parmegiani
L
,
Cognigni
GE
,
Bernardi
S
, et al
.
Efficiency of aseptic open vitrification and hermetical cryostorage of human oocytes
.
Reprod Biomed Online
.
2011
;
23
(
4
):
505
-
512
.
109.
Potdar
N
,
Gelbaya
TA
,
Nardo
LG
.
Oocyte vitrification in the 21st century and post-warming fertility outcomes: a systematic review and meta-analysis
.
Reprod Biomed Online
.
2014
;
29
(
2
):
159
-
176
.
110.
Druckenmiller
S
,
Goldman
KN
,
Labella
PA
,
Fino
ME
,
Bazzocchi
A
,
Noyes
N
.
Successful oocyte cryopreservation in reproductive-aged cancer survivors
.
Obstet Gynecol
.
2016
;
127
(
3
):
474
-
480
.
111.
Practice Committee of American Society for Reproductive Medicine
.
Ovarian tissue cryopreservation: a committee opinion
.
Fertil Steril
.
2014
;
101
(
5
):
1237
-
1243
.
112.
Forman
EJ
.
Ovarian tissue cryopreservation: still experimental?
Fertil Steril
.
2018
;
109
(
3
):
443
-
444
.
113.
Donfack
NJ
,
Alves
KA
,
Araújo
VR
, et al
.
Expectations and limitations of ovarian tissue transplantation
.
Zygote
.
2017
;
25
(
4
):
391
-
403
.
114.
Donnez
J
,
Dolmans
MM
.
Ovarian cortex transplantation: 60 reported live births brings the success and worldwide expansion of the technique towards routine clinical practice
.
J Assist Reprod Genet
.
2015
;
32
(
8
):
1167
-
1170
.
115.
Van der Ven
H
,
Liebenthron
J
,
Beckmann
M
, et al;
FertiPROTEKT network
.
Ninety-five orthotopic transplantations in 74 women of ovarian tissue after cytotoxic treatment in a fertility preservation network: tissue activity, pregnancy and delivery rates
.
Hum Reprod
.
2016
;
31
(
9
):
2031
-
2041
.
116.
Jadoul
P
,
Guilmain
A
,
Squifflet
J
, et al
.
Efficacy of ovarian tissue cryopreservation for fertility preservation: lessons learned from 545 cases
.
Hum Reprod
.
2017
;
32
(
5
):
1046
-
1054
.
117.
Kim
S
,
Lee
Y
,
Lee
S
,
Kim
T
.
Ovarian tissue cryopreservation and transplantation in patients with cancer
.
Obstet Gynecol Sci
.
2018
;
61
(
4
):
431
-
442
.
118.
Dolmans
MM
,
Luyckx
V
,
Donnez
J
,
Andersen
CY
,
Greve
T
.
Risk of transferring malignant cells with transplanted frozen-thawed ovarian tissue
.
Fertil Steril
.
2013
;
99
(
6
):
1514
-
1522
.
119.
Rosendahl
M
,
Andersen
MT
,
Ralfkiær
E
,
Kjeldsen
L
,
Andersen
MK
,
Andersen
CY
.
Evidence of residual disease in cryopreserved ovarian cortex from female patients with leukemia
.
Fertil Steril
.
2010
;
94
(
6
):
2186
-
2190
.
120.
Dolmans
MM
,
Marinescu
C
,
Saussoy
P
,
Van Langendonckt
A
,
Amorim
C
,
Donnez
J
.
Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe
.
Blood
.
2010
;
116
(
16
):
2908
-
2914
.
121.
Greve
T
,
Clasen-Linde
E
,
Andersen
MT
, et al
.
Cryopreserved ovarian cortex from patients with leukemia in complete remission contains no apparent viable malignant cells
.
Blood
.
2012
;
120
(
22
):
4311
-
4316
.
122.
Amiot
C
,
Angelot-Delettre
F
,
Zver
T
, et al
.
Minimal residual disease detection of leukemic cells in ovarian cortex by eight-color flow cytometry
.
Hum Reprod
.
2013
;
28
(
8
):
2157
-
2167
.
123.
Zver
T
,
Alvergnas-Vieille
M
,
Garnache-Ottou
F
,
Ferrand
C
,
Roux
C
,
Amiot
C
.
Minimal residual disease detection in cryopreserved ovarian tissue by multicolor flow cytometry in acute myeloid leukemia
.
Haematologica
.
2014
;
99
(
12
):
e249
-
e252
.
124.
Bastings
L
,
Beerendonk
CC
,
Westphal
JR
, et al
.
Autotransplantation of cryopreserved ovarian tissue in cancer survivors and the risk of reintroducing malignancy: a systematic review
.
Hum Reprod Update
.
2013
;
19
(
5
):
483
-
506
.
125.
Shapira
M
,
Raanani
H
,
Barshack
I
, et al
.
First delivery in a leukemia survivor after transplantation of cryopreserved ovarian tissue, evaluated for leukemia cells contamination
.
Fertil Steril
.
2018
;
109
(
1
):
48
-
53
.
126.
Demeestere
I
,
Brice
P
,
Peccatori
FA
, et al
.
No evidence for the benefit of gonadotropin-releasing hormone agonist in preserving ovarian function and fertility in lymphoma survivors treated with chemotherapy: final long-term report of a prospective randomized trial
.
J Clin Oncol
.
2016
;
34
(
22
):
2568
-
2574
.
127.
Phelan
R
,
Mann
E
,
Napurski
C
, et al
.
Ovarian function after hematopoietic cell transplantation: a descriptive study following the use of GnRH agonists for myeloablative conditioning and observation only for reduced-intensity conditioning
.
Bone Marrow Transplant
.
2016
;
51
(
10
):
1369
-
1375
.
128.
Blumenfeld
Z
,
Patel
B
,
Leiba
R
,
Zuckerman
T
.
Gonadotropin-releasing hormone agonist may minimize premature ovarian failure in young women undergoing autologous stem cell transplantation
.
Fertil Steril
.
2012
;
98
(
5
):
1266
-
1270
.
129.
Panasiuk
A
,
Nussey
S
,
Veys
P
, et al
.
Gonadal function and fertility after stem cell transplantation in childhood: comparison of a reduced intensity conditioning regimen containing melphalan with a myeloablative regimen containing busulfan
.
Br J Haematol
.
2015
;
170
(
5
):
719
-
726
.
130.
Scott
BL
,
Pasquini
MC
,
Logan
BR
, et al
.
Myeloablative versus reduced-intensity hematopoietic cell transplantation for acute myeloid leukemia and myelodysplastic syndromes
.
J Clin Oncol
.
2017
;
35
(
11
):
1154
-
1161
.
131.
Partridge
AH
,
Gelber
S
,
Peppercorn
J
, et al
.
Web-based survey of fertility issues in young women with breast cancer
.
J Clin Oncol
.
2004
;
22
(
20
):
4174
-
4183
.
132.
Anazodo
A
,
Ataman-Millhouse
L
,
Jayasinghe
Y
,
Woodruff
TK
.
Oncofertility-an emerging discipline rather than a special consideration
.
Pediatr Blood Cancer
.
2018
;
65
(
11
):
e27297
.
133.
Peate
M
,
Meiser
B
,
Cheah
BC
, et al
.
Making hard choices easier: a prospective, multicentre study to assess the efficacy of a fertility-related decision aid in young women with early-stage breast cancer
.
Br J Cancer
.
2012
;
106
(
6
):
1053
-
1061
.
134.
Woodard
TL
,
Hoffman
AS
,
Covarrubias
LA
, et al
.
The Pathways fertility preservation decision aid website for women with cancer: development and field testing
.
J Cancer Surviv
.
2018
;
12
(
1
):
101
-
114
.
135.
Woodard
TL
,
Hoffman
AS
,
Crocker
LC
, et al
.
Pathways: patient-centred decision counselling for women at risk of cancer-related infertility: a protocol for a comparative effectiveness cluster randomised trial
.
BMJ Open
.
2018
;
8
(
2
):
e019994
.
136.
Loren
AW
.
Fertility issues in patients with hematologic malignancies
.
Hematology (Am Soc Hematol Educ Program)
.
2015
;
2015
(
1
):
138
-
145
.
137.
Skaczkowski
G
,
White
V
,
Thompson
K
, et al
.
Factors influencing the provision of fertility counseling and impact on quality of life in adolescents and young adults with cancer
.
J Psychosoc Oncol
.
2018
;
36
(
4
):
484
-
502
.
138.
Ussher
JM
,
Parton
C
,
Perz
J
.
Need for information, honesty and respect: patient perspectives on health care professionals communication about cancer and fertility
.
Reprod Health
.
2018
;
15
(
1
):
2
.
139.
Lee
SJ
,
Schover
LR
,
Partridge
AH
, et al;
American Society of Clinical Oncology
.
American Society of Clinical Oncology recommendations on fertility preservation in cancer patients
.
J Clin Oncol
.
2006
;
24
(
18
):
2917
-
2931
.
140.
Behringer
K
,
Breuer
K
,
Reineke
T
, et al;
German Hodgkin’s Lymphoma Study Group
.
Secondary amenorrhea after Hodgkin’s lymphoma is influenced by age at treatment, stage of disease, chemotherapy regimen, and the use of oral contraceptives during therapy: a report from the German Hodgkin’s Lymphoma Study Group
.
J Clin Oncol
.
2005
;
23
(
30
):
7555
-
7564
.
141.
Behringer
K
,
Mueller
H
,
Goergen
H
, et al
.
Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin Study Group HD13 to HD15 trials
.
J Clin Oncol
.
2013
;
31
(
2
):
231
-
239
.
142.
Senapati
S
,
Morse
CB
,
Sammel
MD
, et al
.
Fertility preservation in patients with haematological disorders: a retrospective cohort study
.
Reprod Biomed Online
.
2014
;
28
(
1
):
92
-
98
.
143.
Rossi
BV
,
Ashby
RK
,
Srouji
SS
.
Embryo banking between induction and consolidation chemotherapy in women with leukemia
.
Fertil Steril
.
2011
;
96
(
6
):
1412
-
1414
.
144.
Das
M
,
Shehata
F
,
Son
WY
,
Tulandi
T
,
Holzer
H
.
Ovarian reserve and response to IVF and in vitro maturation treatment following chemotherapy
.
Hum Reprod
.
2012
;
27
(
8
):
2509
-
2514
.
145.
Nakajima
Y
,
Kuwabara
H
,
Kishimoto
K
, et al
.
Successful pregnancy and delivery via in vitro fertilization with cryopreserved and thawed embryo transfer in an acute myeloid leukemia patient after allogeneic bone marrow transplantation
.
Int J Hematol
.
2015
;
101
(
4
):
417
-
420
.
146.
Mehm
AR
,
Steensma
DP
,
Srouji
SS
,
Ginsburg
ES
,
Brady
PC
.
Fertility preservation in women with marrow failure syndromes prior to allogeneic stem cell transplantation
.
Am J Hematol
.
2018
;
93
(
8
):
E190
-
E193
.
147.
Zolton
JR
,
Parikh
TP
,
Hickstein
DD
, et al
.
Oocyte cryopreservation for women with GATA2 deficiency
.
J Assist Reprod Genet
.
2018
;
35
(
7
):
1201
-
1207
.
148
Carlier
P
,
Markarian
M
,
Bernard
N
, et al
.
Pregnancy outcome among partners of male patients receiving imatinib, dasatinib or nilotinib in chronic myeloid leukemia: reports collected by the French network pharmacovigilance centers [published correction appears in Arch Gynecol Obstet. 2017;295(4):1059]
.
Arch Gynecol Obstet
.
2017
;
295
(
2
):
269
-
271
.
149.
Palani
R
,
Milojkovic
D
,
Apperley
JF
.
Managing pregnancy in chronic myeloid leukaemia
.
Ann Hematol
.
2015
;
94
(
suppl 2
):
S167
-
S176
.
150.
Cortes
J
,
Rea
D
,
Lipton
JH
.
Treatment-free remission with first- and second-generation tyrosine kinase inhibitors
.
Am J Hematol
.
2018
;
94
(
3
):
346
-
357
.
151.
Creative Family Connections
.
Gestational surrogacy law across the United States. Vol. 2019. https://www.creativefamilyconnections.com/us-surrogacy-law-map/. Accessed 15 July 2019
.
152.
Rashedi
AS
,
de Roo
SF
,
Ataman
LM
, et al
.
Survey of fertility preservation options available to patients with cancer around the globe
.
J Glob Oncol
.
2018
;
4
:
1
-
16
.
153.
Hughes
TP
,
Ross
DM
.
Moving treatment-free remission into mainstream clinical practice in CML
.
Blood
.
2016
;
128
(
1
):
17
-
23
.
154.
Balsat
M
,
Etienne
M
,
Elhamri
M
,
Hayette
S
,
Salles
G
,
Thomas
X
.
Successful pregnancies in patients with BCR-ABL-positive leukemias treated with interferon-alpha therapy during the tyrosine kinase inhibitors era
.
Eur J Haematol
.
2018
;
101
(
6
):
774
-
780
.
155.
Lasica
M
,
Willcox
A
,
Burbury
K
, et al
.
The effect of tyrosine kinase inhibitor interruption and interferon use on pregnancy outcomes and long-term disease control in chronic myeloid leukemia
.
Leuk Lymphoma
.
2019
;
60
(
7
):
1796
-
1802
.
156.
Luskin
MR
.
Chronic myeloid leukemia and pregnancy: patient and partner perspectives
.
Expert Rev Hematol
.
2018
;
11
(
8
):
597
-
599
.
157.
P
P
,
Samal
R
,
Ghose
S
;
P P. Chronic myeloid leukaemia in pregnancy: call for guidelines
.
J Obstet Gynaecol
.
2019
;
39
(
4
):
582
-
583
.
159.
Pasquini
MC
,
Voltarelli
J
,
Atkins
HL
, et al
.
Transplantation for autoimmune diseases in North and South America: a report of the Center for International Blood and Marrow Transplant Research
.
Biol Blood Marrow Transplant
.
2012
;
18
(
10
):
1471
-
1478
.
160.
Snowden
JA
,
Badoglio
M
,
Labopin
M
, et al;
EBMT (JACIE)
.
Evolution, trends, outcomes, and economics of hematopoietic stem cell transplantation in severe autoimmune diseases
.
Blood Adv
.
2017
;
1
(
27
):
2742
-
2755
.
161.
Snowden
JA
,
Saccardi
R
,
Allez
M
, et al;
Paediatric Diseases Working Party (PDWP)
.
Haematopoietic SCT in severe autoimmune diseases: updated guidelines of the European Group for Blood and Marrow Transplantation
.
Bone Marrow Transplant
.
2012
;
47
(
6
):
770
-
790
.
162.
Muraro
PA
,
Martin
R
,
Mancardi
GL
,
Nicholas
R
,
Sormani
MP
,
Saccardi
R
.
Autologous haematopoietic stem cell transplantation for treatment of multiple sclerosis
.
Nat Rev Neurol
.
2017
;
13
(
7
):
391
-
405
.
163.
Muraro
PA
,
Pasquini
M
,
Atkins
HL
, et al;
Multiple Sclerosis–Autologous Hematopoietic Stem Cell Transplantation (MS-AHSCT) Long-term Outcomes Study Group
.
Long-term outcomes after autologous hematopoietic stem cell transplantation for multiple sclerosis
.
JAMA Neurol
.
2017
;
74
(
4
):
459
-
469
.
164.
Shouval
R
,
Furie
N
,
Raanani
P
,
Nagler
A
,
Gafter-Gvili
A
.
Autologous hematopoietic stem cell transplantation for systemic sclerosis: a systematic review and meta-analysis
.
Biol Blood Marrow Transplant
.
2018
;
24
(
5
):
937
-
944
.
165.
Sullivan
KM
,
Majhail
NS
,
Bredeson
C
, et al
.
Systemic sclerosis as an indication for autologous hematopoietic cell transplantation: Position Statement from the American Society for Blood and Marrow Transplantation
.
Biol Blood Marrow Transplant
.
2018
;
24
(
10
):
1961
-
1964
.
166.
Maciejewska
M
,
Snarski
E
,
Wiktor-Jędrzejczak
W
.
A preliminary online study on menstruation recovery in women after autologous hematopoietic stem cell transplant for autoimmune diseases
.
Exp Clin Transplant
.
2016
;
14
(
6
):
665
-
669
.
167.
Massenkeil
G
,
Alexander
T
,
Rosen
O
, et al
.
Long-term follow-up of fertility and pregnancy in autoimmune diseases after autologous haematopoietic stem cell transplantation
.
Rheumatol Int
.
2016
;
36
(
11
):
1563
-
1568
.
168.
Smith-Whitley
K
.
Reproductive issues in sickle cell disease
.
Blood
.
2014
;
124
(
24
):
3538
-
3543
.
169.
Ghafuri
DL
,
Stimpson
SJ
,
Day
ME
,
James
A
,
DeBaun
MR
,
Sharma
D
.
Fertility challenges for women with sickle cell disease
.
Expert Rev Hematol
.
2017
;
10
(
10
):
891
-
901
.
170.
Lukusa
AK
,
Vermylen
C
,
Vanabelle
B
, et al
.
Bone marrow transplantation or hydroxyurea for sickle cell anemia: long-term effects on semen variables and hormone profiles
.
Pediatr Hematol Oncol
.
2009
;
26
(
4
):
186
-
194
.
171.
Taddesse
A
,
Woldie
IL
,
Khana
P
, et al
.
Hypogonadism in patients with sickle cell disease: central or peripheral?
Acta Haematol
.
2012
;
128
(
2
):
65
-
68
.
172.
Huang
AW
,
Muneyyirci-Delale
O
.
Reproductive endocrine issues in men with sickle cell anemia
.
Andrology
.
2017
;
5
(
4
):
679
-
690
.
173.
Osegbe
DN
,
Akinyanju
O
,
Amaku
EO
.
Fertility in males with sickle cell disease
.
Lancet
.
1981
;
2
(
8241
):
275
-
276
.
174.
Berthaut
I
,
Guignedoux
G
,
Kirsch-Noir
F
, et al
.
Influence of sickle cell disease and treatment with hydroxyurea on sperm parameters and fertility of human males
.
Haematologica
.
2008
;
93
(
7
):
988
-
993
.
175.
DeBaun
MR
.
Hydroxyurea therapy contributes to infertility in adult men with sickle cell disease: a review
.
Expert Rev Hematol
.
2014
;
7
(
6
):
767
-
773
.
176.
Sahoo
LK
,
Kullu
BK
,
Patel
S
, et al
.
Study of seminal fluid parameters and fertility of male sickle cell disease patients and potential impact of hydroxyurea treatment
.
J Assoc Physicians India
.
2017
;
65
(
6
):
22
-
25
.
177.
Soliman
AT
,
Yasin
M
,
El-Awwa
A
,
Abdelrahman
MO
,
De Sanctis
V
.
Does blood transfusion affect pituitary gonadal axis and sperm parameters in young males with sickle cell disease?
Indian J Endocrinol Metab
.
2013
;
17
(
6
):
962
-
968
.
178.
Ferster
A
,
De Valck
C
,
Azzi
N
,
Fondu
P
,
Toppet
M
,
Sariban
E
.
Bone marrow transplantation for severe sickle cell anaemia
.
Br J Haematol
.
1992
;
80
(
1
):
102
-
105
.
179.
Panepinto
JA
,
Walters
MC
,
Carreras
J
, et al;
Non-Malignant Marrow Disorders Working Committee, Center for International Blood and Marrow Transplant Research
.
Matched-related donor transplantation for sickle cell disease: report from the Center for International Blood and Transplant Research
.
Br J Haematol
.
2007
;
137
(
5
):
479
-
485
.
180.
Walters
MC
,
Hardy
K
,
Edwards
S
, et al;
Multicenter Study of Bone Marrow Transplantation for Sickle Cell Disease
.
Pulmonary, gonadal, and central nervous system status after bone marrow transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2010
;
16
(
2
):
263
-
272
.
181.
Arnold
SD
,
Jin
Z
,
Sands
S
,
Bhatia
M
,
Kung
AL
,
Satwani
P
.
Allogeneic hematopoietic cell transplantation for children with sickle cell disease is beneficial and cost-effective: a single-center analysis
.
Biol Blood Marrow Transplant
.
2015
;
21
(
7
):
1258
-
1265
.
182.
Bhatia
M
,
Kolva
E
,
Cimini
L
, et al
.
Health-related quality of life after allogeneic hematopoietic stem cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2015
;
21
(
4
):
666
-
672
.
183.
Shenoy
S
,
Angelucci
E
,
Arnold
SD
, et al
.
Current results and future research priorities in late effects after hematopoietic stem cell transplantation for children with sickle cell disease and thalassemia: A Consensus Statement from the Second Pediatric Blood and Marrow Transplant Consortium International Conference on Late Effects after Pediatric Hematopoietic Stem Cell Transplantation
.
Biol Blood Marrow Transplant
.
2017
;
23
(
4
):
552
-
561
.
184.
Walters
MC
,
Storb
R
,
Patience
M
, et al
.
Impact of bone marrow transplantation for symptomatic sickle cell disease: an interim report. Multicenter investigation of bone marrow transplantation for sickle cell disease
.
Blood
.
2000
;
95
(
6
):
1918
-
1924
.
185.
Dallas
MH
,
Triplett
B
,
Shook
DR
, et al
.
Long-term outcome and evaluation of organ function in pediatric patients undergoing haploidentical and matched related hematopoietic cell transplantation for sickle cell disease
.
Biol Blood Marrow Transplant
.
2013
;
19
(
5
):
820
-
830
.
186.
Madden
LM
,
Hayashi
RJ
,
Chan
KW
, et al
.
Long-term follow-up after reduced-intensity conditioning and stem cell transplantation for childhood nonmalignant disorders
.
Biol Blood Marrow Transplant
.
2016
;
22
(
8
):
1467
-
1472
.
187.
Chakrabarti
S
,
Bareford
D
.
A survey on patient perception of reduced-intensity transplantation in adults with sickle cell disease
.
Bone Marrow Transplant
.
2007
;
39
(
8
):
447
-
451
.
188.
Practice Committee of the American Society for Reproductive Medicine. Committee of the American Society for Reproductive Medicine
.
Prevention and treatment of moderate and severe ovarian hyperstimulation syndrome: a guideline
.
Fertil Steril
.
2016
;
106
(
7
):
1634
-
1647
.
189.
Pecker
LH
,
Maher
JY
,
Law
JY
,
Beach
MC
,
Lanzkron
S
,
Christianson
MS
.
Risks associated with fertility preservation for women with sickle cell anemia
.
Fertil Steril
.
2018
;
110
(
4
):
720
-
731
.
190.
Lavery
SA
,
Islam
R
,
Hunt
J
,
Carby
A
,
Anderson
RA
.
The medical and ethical challenges of fertility preservation in teenage girls: a case series of sickle cell anaemia patients prior to bone marrow transplant
.
Hum Reprod
.
2016
;
31
(
7
):
1501
-
1507
.
191.
Demeestere
I
,
Simon
P
,
Dedeken
L
, et al
.
Live birth after autograft of ovarian tissue cryopreserved during childhood
.
Hum Reprod
.
2015
;
30
(
9
):
2107
-
2109
.
192.
Mishkin
AD
,
Mapara
MY
,
Reshef
R
.
Iatrogenic infertility after curative stem cell transplantation in patients with sickle cell disease
.
Ann Intern Med
.
2018
;
168
(
12
):
881
-
882
.
193.
Pecker
LH
,
Naik
RP
.
The current state of sickle-cell trait: implications for reproductive and genetic counseling
.
Blood
.
2018
;
blood-2018-06-848705
.
194.
Clayman
ML
,
Harper
MM
,
Quinn
GP
,
Reinecke
J
,
Shah
S
.
Oncofertility resources at NCI-designated comprehensive cancer centers
.
J Natl Compr Canc Netw
.
2013
;
11
(
12
):
1504
-
1509
.
195.
Jones
G
,
Hughes
J
,
Mahmoodi
N
,
Smith
E
,
Skull
J
,
Ledger
W
.
What factors hinder the decision-making process for women with cancer and contemplating fertility preservation treatment?
Hum Reprod Update
.
2017
;
23
(
4
):
433
-
457
.
196.
Vindrola-Padros
C
,
Dyer
KE
,
Cyrus
J
,
Lubker
IM
.
Healthcare professionals’ views on discussing fertility preservation with young cancer patients: a mixed method systematic review of the literature
.
Psychooncology
.
2017
;
26
(
1
):
4
-
14
.
197.
Frederick
NN
,
Campbell
K
,
Kenney
LB
,
Moss
K
,
Speckhart
A
,
Bober
SL
.
Barriers and facilitators to sexual and reproductive health communication between pediatric oncology clinicians and adolescent and young adult patients: the clinician perspective
.
Pediatr Blood Cancer
.
2018
;
65
(
8
):
e27087
.
198.
Quinn
GP
,
Vadaparampil
ST
,
Gwede
CK
,
Reinecke
JD
,
Mason
TM
,
Silva
C
.
Developing a referral system for fertility preservation among patients with newly diagnosed cancer
.
J Natl Compr Canc Netw
.
2011
;
9
(
11
):
1219
-
1225
.
199.
Lopategui
DM
,
Ibrahim
E
,
Aballa
TC
, et al
.
Effect of a formal oncofertility program on fertility preservation rates-first year experience
.
Transl Androl Urol
.
2018
;
7
(
S3 Suppl 3
):
S271
-
S275
.
200.
Levine
JM
,
Kelvin
JF
,
Quinn
GP
,
Gracia
CR
.
Infertility in reproductive-age female cancer survivors
.
Cancer
.
2015
;
121
(
10
):
1532
-
1539
.
201.
Su
HI
,
Ray
L
,
Chang
RJ
.
Setting Up an Oncofertility Program
. In:
Gracia
C
,
Woodruff
TK
, eds.
Oncofertility Medical Practice: Clinical Issues and Implementation
.
New York
:
Springer
;
2012
:
163
-
173
202.
Reinecke
JD
,
Kelvin
JF
,
Arvey
SR
, et al
.
Implementing a systematic approach to meeting patients’ cancer and fertility needs: a review of the Fertile Hope Centers Of Excellence program
.
J Oncol Pract
.
2012
;
8
(
5
):
303
-
308
.
203.
Alliance for Fertility Preservation
. .
204.
Resolve: The National Infertility Association
. .
205.
Campo-Engelstein
L
.
Consistency in insurance coverage for iatrogenic conditions resulting from cancer treatment including fertility preservation
.
J Clin Oncol
.
2010
;
28
(
8
):
1284
-
1286
.
206.
Walter
JR
,
Xu
S
,
Woodruff
TK
.
A call for fertility preservation coverage for breast cancer patients: the cost of consistency
.
J Natl Cancer Inst
.
2017
;
109
(
5
).
207.
Cardozo
ER
,
Huber
WJ
,
Stuckey
AR
,
Alvero
RJ
.
Mandating coverage for fertility preservation - a step in the right direction
.
N Engl J Med
.
2017
;
377
(
17
):
1607
-
1609
.
208.
Inhorn
MC
,
Birenbaum-Carmeli
D
,
Westphal
LM
, et al
.
Medical egg freezing: how cost and lack of insurance cover impact women and their families
.
Reprod Biomed Soc Online
.
2018
;
5
:
82
-
92
.
209.
Thom
B
,
Benedict
C
,
Friedman
DN
,
Kelvin
JF
.
The intersection of financial toxicity and family building in young adult cancer survivors
.
Cancer
.
2018
;
124
(
16
):
3284
-
3289
.
210.
ESHRE Working Group on Oocyte CryoPreservation in Europe
,
Shenfield
F
,
de Mouzon
J
, et al
.
Oocyte and ovarian tissue cryopreservation in European countries: statutory background, practice, storage and use
.
Hum Reprod Open
.
2017
;
2017
(
1
):
hox003
.
211.
Salama
M
,
Ataman
L
,
Taha
T
, et al
.
Building oncofertility core competency in developing countries: experience from Egypt, Tunisia, Brazil, Peru, and Panama
.
J Glob Oncol
.
2018
;
4
:
1
-
11
.
212.
M Russell
A
,
Galvin
KM
,
Harper
MM
,
Clayman
ML
.
A comparison of heterosexual and LGBTQ cancer survivors’ outlooks on relationships, family building, possible infertility, and patient-doctor fertility risk communication [published correction in J Cancer Surviv. 2016;10(5):943]
.
J Cancer Surviv
.
2016
;
10
(
5
):
935
-
942
.
213.
Quinn
GP
,
Sanchez
JA
,
Sutton
SK
, et al
.
Cancer and lesbian, gay, bisexual, transgender/transsexual, and queer/questioning (LGBTQ) populations
.
CA Cancer J Clin
.
2015
;
65
(
5
):
384
-
400
.
214.
Deutsch
MB
,
Feldman
JL
.
Updated recommendations from the world professional association for transgender health standards of care
.
Am Fam Physician
.
2013
;
87
(
2
):
89
-
93
.
215.
Hembree
WC
,
Cohen-Kettenis
P
,
Delemarre-van de Waal
HA
, et al;
Endocrine Society
.
Endocrine treatment of transsexual persons: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab
.
2009
;
94
(
9
):
3132
-
3154
.
216
Nahata
L
,
Tishelman
AC
,
Caltabellotta
NM
,
Quinn
GP
.
Low fertility preservation utilization among transgender youth
.
J Adolesc Health
.
2017
;
61
(
1
):
40
-
44
.
217
Chian
RC
,
Buckett
WM
,
Tan
SL
.
In-vitro maturation of human oocytes
.
Reprod Biomed Online
.
2004
;
8
(
2
):
148
-
166
.
218.
Chian
RC
,
Uzelac
PS
,
Nargund
G
.
In vitro maturation of human immature oocytes for fertility preservation
.
Fertil Steril
.
2013
;
99
(
5
):
1173
-
1181
.
219.
Hudson
JN
,
Stanley
NB
,
Nahata
L
,
Bowman-Curci
M
,
Quinn
GP
.
New promising strategies in oncofertility
.
Expert Rev Qual Life Cancer Care
.
2017
;
2
(
2
):
67
-
78
.
220.
de Michele
F
,
Vermeulen
M
,
Wyns
C
.
Fertility restoration with spermatogonial stem cells
.
Curr Opin Endocrinol Diabetes Obes
.
2017
;
24
(
6
):
424
-
431
.
221.
de Michele
F
,
Poels
J
,
Vermeulen
M
, et al
.
Haploid germ cells generated in organotypic culture of testicular tissue from prepubertal boys
.
Front Physiol
.
2018
;
9
:
1413
.
222.
Dovey
SL
,
Valli
H
,
Hermann
BP
, et al
.
Eliminating malignant contamination from therapeutic human spermatogonial stem cells
.
J Clin Invest
.
2013
;
123
(
4
):
1833
-
1843
.
223.
Bhartiya
D
,
Anand
S
,
Patel
H
,
Parte
S
.
Making gametes from alternate sources of stem cells: past, present and future
.
Reprod Biol Endocrinol
.
2017
;
15
(
1
):
89
.
224.
Sarma
UC
,
Findlay
JK
,
Hutt
KJ
.
Oocytes from stem cells
.
Best Pract Res Clin Obstet Gynaecol
.
2019
;
55
:
14
-
22
.
225.
Wang
JJ
,
Ge
W
,
Liu
JC
, et al
.
Complete in vitro oogenesis: retrospects and prospects
.
Cell Death Differ
.
2017
;
24
(
11
):
1845
-
1852
.
226.
Goldman
KN
,
Chenette
D
,
Arju
R
, et al
.
mTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy
.
Proc Natl Acad Sci USA
.
2017
;
114
(
12
):
3186
-
3191
.
227.
Herraiz
S
,
Buigues
A
,
Diaz-Garcia
C
, et al
.
Fertility rescue and ovarian follicle growth promotion by bone marrow stem cell infusion
.
Fertil Steril.
2018
;
109
(
5
):
908
-
918
.
228.
Herraiz
S
,
Romeu
M
,
Buigues
A
, et al
.
Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders
.
Fertil Steril.
2018
;
110
(
3
):
496
-
505
.
229.
Gohari
MR
,
Ramezani Tehrani
F
,
Chenouri
S
,
Solaymani-Dodaran
M
,
Azizi
F
.
Individualized predictions of time to menopause using multiple measurements of antimüllerian hormone
.
Menopause
.
2016
;
23
(
8
):
839
-
845
.
230.
Ruddy
KJ
,
O’Neill
A
,
Miller
KD
, et al
.
Biomarker prediction of chemotherapy-related amenorrhea in premenopausal women with breast cancer participating in E5103
.
Breast Cancer Res Treat
.
2014
;
144
(
3
):
591
-
597
.
231.
Steiner
AZ
,
Pritchard
D
,
Stanczyk
FZ
, et al
.
Association between biomarkers of ovarian reserve and infertility among older women of reproductive age
.
JAMA
.
2017
;
318
(
14
):
1367
-
1376
.
232.
Lee
DH
,
Pei
CZ
,
Song
JY
, et al
.
Identification of serum biomarkers for premature ovarian failure
.
Biochim Biophys Acta Proteins Proteomics
.
2019
;
1867
(
3
):
219
-
226
.
Sign in via your Institution