Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Abstract
Abstract Fertivision 2017
Case Report
Clinical Practice Guideline
Commentary
Edirorial
Editor's view point
Editorial
Editorial View Point
Fertivision 2015 - Abstracts
Guest Editorial
IFS pages
Letter to the Editor
Original Article
Original Research
PCOS Guideline
Point of View
Review Article
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Abstract
Abstract Fertivision 2017
Case Report
Clinical Practice Guideline
Commentary
Edirorial
Editor's view point
Editorial
Editorial View Point
Fertivision 2015 - Abstracts
Guest Editorial
IFS pages
Letter to the Editor
Original Article
Original Research
PCOS Guideline
Point of View
Review Article
View/Download PDF

Translate this page into:

Review Article
2024
:11;
4
doi:
10.25259/fsr_46_23

Semen Cryopreservation in Oncofertility

Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India

*Corresponding author: Miss. Shruthivishali Muthukumar, Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India. m.shruthivishali@manipal.edu

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Muthukumar S, Tholeti P. Semen Cryopreservation in Oncofertility. Fertil Sci Res. 2024;11:4. doi: 10.25259/fsr_46_23

Abstract

Oncofertility, an interdisciplinary field, aims at improving the uality of life of cancer patients prone to iatrogenic infertility by offering fertility preservation options for a potentially fertile future. Semen cryopreservation (SC) is a well-established and effective way of preserving fertility in adolescent and young adult cancer-diagnosed males as they face potential fertility loss due to cancer and its treatments. The pathophysiology of certain cancers, such as testicular cancer or Hodgkin’s lymphoma, has been shown to have an adverse effect on semen uality. Ablative therapies like chemotherapy and radiotherapy can result in compromised sperm parameters depending on the dose and the type of the drug or radiation. Hence, it is recommended to encourage cancer-diagnosed males to store sperm prior to gonadotoxic therapy to restore potential fertility in the future. Despite the feasibility of SC, this option remains underutilised due to several limitations.

Keywords

Oncofertility
semen cryopreservation
fertility preservation
young adult males

INTRODUCTION

Oncofertility is an emerging multi-disciplinary field that encompasses various fertility preservation strategies for cancer patients at risk of infertility from cancer and its treatments.[1] Fertility preservation (FP) aims at preserving the gametes or gonadal tissue of the cancer-affected individual prior to ablation therapy, thereby facilitating potential parenthood in the future. In adolescent and young adult (AYA) males, semen cryopreservation (SC), which is a well-established and effective way of preserving fertility, is the gold standard method of FP, and the therapeutic applications of frozen sperm are rapidly expanding.[2] With the increasing population of AYA male cancer survivors facing infertility in adulthood, as a conseuence of cancer and its treatment, sperm cryopreservation before treatment has received more prominence. It is recommended that all AYA males be offered sperm cryopreservation prior to cancer therapy, irrespective of the toxicity risk.[3] Despite its ease of utility, sperm banking is underutilised in oncofertility due to several challenges. This current review aims to highlight the effect of cancer pathophysiology and its associated therapies on semen uality, the paramount importance of sperm banking in cancer-diagnosed males, laboratory aspects, recommendations, and guidelines, as well as the ethical aspects involved.

Impact of cancer on spermatogenesis and sperm uality

Men with cancer often experience impaired gonadal function and present poor sperm uality.[4] The pathophysiology of cancer can be attributed to metabolic changes, endocrine alterations and systemic conseuences of the disease.[5] In germ-cell cancers, it has been reported that the direct effect of the tumour, hormonal secretions and immune-mediated factors can interfere with spermatogenesis and sperm functions. Tumours can impinge or infiltrate neurologic structures, leading to ejaculatory disorders, or even affect other parts of the male reproductive system, such as the epididymis, vas deferens and ejaculatory duct, thereby impairing fertility potential.[6] In haematological cancers, altered general functions, hyperthermia or testicular infiltration could be additional mechanisms responsible for reduced sperm production.[7] Among various cancers, Hodgkin’s lymphoma (HL) and testicular cancer (TC) are the most prevalent cancers affecting men of reproductive age, with an incidence of 6% and 13–15%, respectively.[8] Studies have shown that patients with TC, HL and other genitourinary malignancies more often present altered semen profile, particularly reduced semen volume, sperm concentration, and motility when compared to men with other cancers.[9,10] These variations could be due to the systemic and direct effects of the malignancy, such as inflammation, cytokine production or fever. This could suggest that the mechanisms underlying the decline in sperm uality may be associated with the type and origin of the malignancy.[11]

Conseuences of cancer treatment on male fertility

With a focus on cancer cure, over time, cancer treatments have evolved, but the common modalities remain chemotherapy and radiotherapy, which are effective in combating malignancies despite their associated gonadotoxicity resulting in male infertility.[12,13]

Chemotherapy

Chemotherapeutic agents have been shown to cause germinal epithelial damage, resulting in oligo- or azoospermia, as well as Sertoli and Leydig cell damage affecting the hormonal pathways.[14] The impact of chemotherapy on gonadal function is determined by variables such as the type of drugs, combination regimens and the dose administered.[15] Each class of cytotoxic drugs has diverse effects on the germ cell stages and depletion rate depending on their modes of action. As a result, the duration and permanence of induced azoospermia are determined by the dosage of the drug, the freuency and the cumulative impact of the other agents administered. Alkylating drugs such as cyclophosphamide, chlorambucil, procarbazine and busulphan can pose a high risk to fertility. For instance, a single dose of 300 mg/kg cyclophosphamide causes azoospermia in over 80% of males within 50–60 days of treatment, which can lead to irreversible infertility.[16] Cisplatin, at a threshold level of 600 mg/m2, results in temporary azoospermia with a recovery of spermatogenesis in about 50% of patients in 2 years, whereas vinblastine, at 50 g/m2, has an intermediate risk of infertility, resulting in temporary oligozoospermia, but is usually administered in conjunction with other sterilising agents.[15,17]

Radiation therapy

The testis is a highly radiosensitive organ as it contains the proliferating spermatogonial stem cells (SSC). The testicular function can be impaired by direct testicular, pelvic, hypothalamic–pituitary axis irradiation or scattered radiation.[18] As radiation directly targets the DNA of proliferating cells, the SSC gets affected as a conseuence, but the differentiated spermatocytes, spermatids and spermatozoa are less vulnerable to radiation toxicity. Direct testicular irradiation can result in azoospermia (>2.0 Gy), permanent oligozoospermia (0.8–2.0 Gy) or transient oligozoospermia (<0.8 Gy). A dose of 8 Gy can induce azoospermia in nearly all men, incurring permanent damage to spermatogenesis.[19] The severity and duration of gonadal damage are influenced by a number of variables, such as dose, fraction size and the specific target cell population.[20]

Time for spermatogenic recovery after therapeutic insult

Testicular dysfunction and germinal epithelial damage can result from cytotoxic treatment, SSC uality and their differentiation ability. Gonadotoxic effects and recovery time depend on initial semen uality, delivery method, treatment regimens, as well as the spermatogenesis phase.[21] For instance, recolonisation of surviving SSC can be detected by 6 months after exposure to a dose of 0.2 Gy, or up to 9–18 months following a dose of 1 Gy, and sometimes greater than 4 years after exposure to 10 Gy. Combination regimens of CP with busulphan or thiotepa have shown sperm recovery after 3 years, and in 50 % of cases, sperm were observed in the ejaculate after 7 years.[22] Combination treatment with radiotherapy and chemotherapy can induce further gonadotoxic damage to the testis.

Genetic normalcy of sperm after cancer therapy

Cancer survivors are more likely to have sperm with aneuploidy, DNA damage, aberrant chromatin structure and epigenetic modifications, even after 2 years of initial treatment in addition to alterations to conventional semen parameters.[23] Sperm DNA aberration is a serious clinical concern because male cancer survivors might use gametes with potentially damaged genomes for assisted reproduction technology (ART) cycles, which could result in adverse reproductive outcomes.[24] A study has shown a reduced pregnancy rate (20%) in ART cycles, which used sperm exposed to cancer treatments compared to the use of sperm that was cryopreserved before cancer therapy, which reported a higher pregnancy rate (51.4%).[25] The altered sperm epigenome of treated males could lead to a transgenerational transmission mechanism affecting the progeny.[26] Sperm chromatin damage resulting in genome instability is one of the most important reproductive side effects owing to the malignancy and/or its treatment[27] which cannot be reversed. Therefore, it’s crucial for these individuals to opt for FP before cancer treatment, as the chances of genetic damage to the sperm increase significantly even 1 day post-treatment with alkylating agents.[28]

Semen cryopreservation: The gold standard

SC is a well-established, non-invasive FP strategy for adolescents and adult males.[29] SC, in conjugation with ART, has revolutionised the FP and restoration options in oncofertility thereby improving the uality of life of cancer survivors.[30] The birth of twins through IVF (in vitro fertilisation) using semen that had been frozen for about 40 years has been chronicled, reassuring that even with long-term cryopreservation, sperm retain the fertilisation ability, resulting in healthy offspring.[31] Some patients may be unable to ejaculate on demand due to various factors such as prevailing illness, age, local pain, psychological issues, cultural factors, or religious concerns. For such patients, penile vibratory stimulation or electroejaculation (EJ) can be performed to obtain the semen sample. Even for patients with time constraints, EJ is more suitable. SC is not just limited to adults; peripubertal boys at around the age of 12 years usually reach spermarche, at which point they acuire secondary sexual characteristics. Depending on their physical capacity and emotional maturity to provide a sample by masturbation, SC can be offered.[32]

SC pre- and post-therapy: Laboratory aspects

Semen banking can be offered to all AYA male cancer patients at the time of diagnosis after thorough counselling for FP. SC involves semen collection, analysis, preparation, and freezing with the addition of cryoprotectants and storage in liuid nitrogen. Among the various cryopreservation techniues, rapid freezing (RF) is the uickest and most effective techniue that has shown superior post-thaw motility and cryo-survival than slow freezing.[33] RF involves drop-wise mixing of sample and cryoprotectant in a 1:1 ratio; followed by incubation at 4°C for 10 minutes. After that, the straws are placed in the vapour phase of liuid nitrogen (−80°C) for 15 minutes before being immersed in liuid nitrogen.[34] The protocols for sperm banking have been established globally and any modifications to the freezing protocol would be dependent on the type of commercially available sperm freezing medium.[35]

Typically, 3–5 days of ejaculatory abstinence is followed for routine therapeutic sperm banking. However, such compliance may delay the initiation of the treatment in cancer patients. A study on sperm collected for FP revealed that the post-thaw uality of sperm produced after 24-48 hours of abstinence was comparable to that of longer abstinence.[36] This consideration will allow for a more freuent collection schedule, enabling patients to store multiple samples. The number of samples to be stored is determined by the semen uality, time-to-treatment, health status of the patient and fertility restoration strategy. For patients with mild or severe oligoasthenozoospermia, it is crucial to store multiple samples or aliuots. Nevertheless, in such cases of limitedly available samples, ICSI (intracytoplasmic sperm injection) could potentially enable them to father a child. As it is evident that cancer therapies result in sperm DNA damage and sperm concentration depletion, SC should ideally be performed prior to initiation of the therapy. However, when a patient desires to store sperm after cancer treatment, experts recommend a 1-year waiting period after the completion of the last cycle of chemo- or radiotherapy.[37]

Guidelines for male FP

Due to the negative impact of cancer and/or its treatment on fertility, international organisations such as the American Society of Clinical Oncology (ASCO), ASRM and ESHRE recommend that healthcare professionals should discuss sperm banking with adolescents and young adult males before their cancer treatment.[3841] Although sperm uality may be reduced even before starting therapy, and there may be a need to start chemotherapy immediately, leaving patients with limited time to provide optimal number of ejaculates for FP; these concerns should not dissuade patients from sperm banking.[42] Individualised oncofertility counselling should be provided with a focus on the patient’s interest and understanding capacity, their age, as well as the planned treatment regimen. Wherever possible, patients should be provided with written information or access to online resources during oncofertility counselling.[43]

The indications for oncofertility include childhood malignancy, treatment with high-risk chemotherapy drugs or total body irradiation, or conditioning before haematopoietic stem cell transplantation.[44] However, as there is no absolute threshold of anticancer therapy exposure that determines gonadal failure and infertility, every patient should be regarded as potentially at risk of developing treatment-related gonadotoxicity.[43]

Barriers to FP and ethical considerations

Despite the professional international guidelines and recommendations for FP in cancer patients, timely referrals and the number and patients opting for FP remain low owing to several barriers.[45] The barriers to FP referrals include limited knowledge and training among health care providers, prioritising cancer treatment, additional costs for the patients, emotional impact, limited time to treatment and a lack of familial support. Another aspect is the complexity of FP discussions in the face of life-threatening situations such as a cancer diagnosis.[46] However, it is very important to obtain informed consent from patients or parents of adolescent boys before the procedure.[47]

The barriers at the level of the institution include a lack of trained professionals, insufficient resources or a lack of infrastructure.[45,48] The disposition of cryopreserved samples is another critical consideration, especially for pre- or peripubertal boys who might reuire long-term storage. Other barriers such as underutilisation of stored samples, decision-making for disposal and a lack of follow-up can also pose a significant ethical burden on the FP units.[49] By bridging care gaps and addressing access barriers, providing oncofertility education, resources, support and timely access to fertility-related care, FP can be easily implemented as part of standard cancer care.[50]

CONCLUSION

Cancer, related chemotherapy and radiation therapy can elicit a negative impact on sperm production, its uality and sexual functions in cancer-affected males. As these individuals are susceptible to fertility loss, FP, prior to treatment is crucial. SC is considered the gold standard strategy for FP in adolescents and young adult males. However, even with well-established storage protocols and international guidelines, FP is not widely implemented in oncofertility due to several limitations, such as lack of awareness among the patients or physicians, limited time to therapy and financial burden.

Ethical approval

Institutional Review Board approval is not reuired.

Declaration of patient consent

Patient’s consent not reuired as patients identity is not disclosed or compromised.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

Use of artificial intelligence (AI)-assisted technology for manuscript preparation

The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript and no images were manipulated using AI.

REFERENCE

  1. , . Oncofertility and the Male Cancer Patient. Curr Treat Options Oncol. 2012;13:146-60.
    [CrossRef] [PubMed] [Google Scholar]
  2. , , , , , . Fertility Preservation in Males with Cancer: 16-year Monocentric Experience of Sperm Banking and Post-Thaw Reproductive Outcomes. Ther Adv Med Oncol. 2016;8:412-20.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  3. , , , , , . What should be Done in Terms of Fertility Preservation for Patients with Cancer? The French 2021 Guidelines. Eur J Cancer. 2022;173:146-66.
    [CrossRef] [PubMed] [Google Scholar]
  4. . Male Infertility in Cancer Patients: Review of the Literature. Int J Urol. 2010;17:327-31.
    [CrossRef] [PubMed] [Google Scholar]
  5. , . Disruption of Spermatogenesis by the Cancer Disease Process. J Natl Cancer Inst Monogr 2005:9-12.
    [CrossRef] [PubMed] [Google Scholar]
  6. , , . Fertility Preservation in Men: A Contemporary Overview and a Look Toward Emerging Technologies. Fertil Steril. 2021;115:1126-39.
    [CrossRef] [PubMed] [Google Scholar]
  7. , , . Semen Quality of 4480 Young Cancer and Systemic Disease Patients: Baseline Data and Clinical Considerations. Basic Clin Androl. 2016;18:26:3.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , , , . Improved Survival for Adolescents and Young Adults with Hodgkin Lymphoma and Continued High Survival for Children in the Netherlands: A Population-based Study during 1990–2015. Br J Haematol. 2020;189:1093-106.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  9. , , . Genitourinary Cancer Patients have Worse Baseline Semen Parameters than Healthy Sperm Bankers. Andrology. 2019;7:449-453.
    [CrossRef] [PubMed] [Google Scholar]
  10. , , , , , . Do Malignant Diseases Affect Semen Quality? Sperm Parameters of Men with Cancers. Andrologia. 2016;48:333-40.
    [CrossRef] [PubMed] [Google Scholar]
  11. , , . Cancer Chemotherapy. 2023 Feb 27. In: Botchan A, Hauser R, Gamzu R, Yogev L, Lessing JB, Paz G, Yavetz H. Sperm Quality in Hodgkin’s Disease Versus Non-Hodgkin’s Lymphoma. Hum Reprod. 1997;12:73-6.
    [CrossRef] [PubMed] [Google Scholar]
  12. StatPearls. Treasure Island (FL): StatPearls Publishing; .
  13. . Male Gonadal Toxicity. Pediatr Blood Cancer. 2009;53:261-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  14. , , , , , . Exposure to Chemotherapy during Childhood or Adulthood and Conseuences on Spermatogenesis and Male Fertility. Int J Mol Sci. 2020;21:1454.
    [CrossRef] [PubMed] [Google Scholar]
  15. , . Spermatogenesis after Cancer Treatment: Damage and Recovery. J Natl Cancer Inst Monogr 2005:12-7.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , , , , . Successful Treatment of Postchemotherapy Azoospermia with Microsurgical Testicular Sperm Extraction: The Weill Cornell Experience. J Clin Oncol. 2011;29:1607-11.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , , , , . American Society of Clinical Oncology. Fertility Preservation for Patients with Cancer: American Society of Clinical Oncology Clinical Practise Guideline Update. J Clin Oncol. 2013;31:2500-10.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  18. , , , , , . Radiation Effects on Male Fertility. Andrology. 2019;7:2-7.
    [CrossRef] [PubMed] [Google Scholar]
  19. , , . Effects of Chemotherapy and Radiotherapy on Spermatogenesis: The Role of Testicular Immunology. Int J Mol Sci. 2019;20:957.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  20. . Effects of Chemotherapy and Radiotherapy on Spermatogenesis in Humans. Fertil Steril. 2013;100:1180-6.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  21. , , , , , . Effects of Chemotherapeutic Agents on Male Germ Cells and Possible Ameliorating Impact of Antioxidants. Biomed Pharmacother. 2021;142:112040.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , . Impact of Cancer on Cryopreserved Sperm Quality and Fertility: A Cohort Study. Health Sci Rep. 2022;5:e726.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  23. , , , . Sperm DNA Damage in Cancer Patients. Adv Exp Med Biol. 2019;1166:189-203.
    [PubMed] [Google Scholar]
  24. , , , . Fatherhood and Sperm DNA Damage in Testicular Cancer Patients. Front Endocrinol (Lausanne). 2018;9:506.
    [PubMed] [Google Scholar]
  25. , , , , , . Sperm and Testicular Tissue Cryopreservation and Assisted Reproductive Technology Outcomes in Male Cancer Patients: A 15-year Experience. J Cancer Res Clin Oncol. 2023;149:5321-30.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  26. , , , , , . Differential DNA Methylation Regions in Adult Human Sperm Following Adolescent Chemotherapy: Potential for Epigenetic Inheritance. PLoS One. 2017;12:e0170085.
    [CrossRef] [PubMed] [Google Scholar]
  27. . Sperm DNA Damage and Cancer Treatment. Int J Androl. 2002;25:255-61.
    [PubMed] [Google Scholar]
  28. . Risks of Genetic Damage in Offspring Conceived using Spermatozoa Produced during Chemotherapy or Radiotherapy. Andrology. 2020;8:545-58.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , , . Male Fertility Preservation and Restoration Strategies for Patients Undergoing Gonadotoxic Therapies†. Biol Reprod. 2022;107:382-405.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  30. , , , . Fertility Preservation Strategies for Male Patients with Cancer. Nat Rev Urol. 2013;10:463-472.
    [CrossRef] [PubMed] [Google Scholar]
  31. , , , . Live Births from Frozen Human Semen Stored for 40 Years. J Assist Reprod Genet. 2013;30:743-4.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , , , , . Fertility Preservation for Young Adults, Adolescents, and Children with Cancer. Ups J Med Sci. 2020;125:112-20.
    [CrossRef] [PubMed] [Google Scholar]
  33. , , . Rapid Freezing Versus Slow Programmable Freezing of Human Spermatozoa. Fertil Steril. 2010;93:1921-8.
    [CrossRef] [PubMed] [Google Scholar]
  34. , , , . Human Sperm Cryopreservation: Update on Techniues, Effect on DNA Integrity, and Implications for ART. Adv Urol. 2012;2012:854837.
    [PubMed] [Google Scholar]
  35. , , , , , . Cryopreservation Media Differentially Affect Sperm Motility, Morphology and DNA Integrity. Andrology. 2018;6:836-45.
    [CrossRef] [PubMed] [Google Scholar]
  36. , , , . Optimum Abstinence Time for Cryopreservation of Semen in Cancer Patients. J Urol. 1995;154:86-8.
    [PubMed] [Google Scholar]
  37. , . Fertility and Reproduction After Chemotherapy of Adult Cancer Patients: Malignant Lymphoma and Testicular Cancer. Ann Oncol. 2004;15:iv259-65.
    [CrossRef] [PubMed] [Google Scholar]
  38. , , , , , . Fertility Preservation in Patients with Cancer: ASCO Clinical Practise Guideline Update. J Clin Oncol. 2018;36:1994-2001.
    [CrossRef] [PubMed] [Google Scholar]
  39. , , , , , . American Society of Clinical Oncology Recommendations on Fertility Preservation in Cancer Patients. J Clin Oncol. 2006;24:2917-31. Epub 2006 May 1. Erratum in: J Clin Oncol 2006;24:5790
    [CrossRef] [PubMed] [Google Scholar]
  40. . Fertility Preservation and Reproduction in Patients Facing Gonadotoxic Therapies: A Committee Opinion. Fertil Steril. 2013;100:1224-31.
    [CrossRef] [PubMed] [Google Scholar]
  41. Clinical Management Planning for Fertility Preservation in Female Cancer Patient. Stockholm: European Society of Human Reproduction and Embryology (ESHRE); .
    [CrossRef] [PubMed]
  42. , , . Clinical Guidelines for Sperm Cryopreservation in Cancer Patients. Fertil Steril. 2013;100:1203-9.
    [CrossRef] [PubMed] [Google Scholar]
  43. , , , , , . Electronic Address: Clinicalguidelines@esmo.org. Fertility Preservation and Post-treatment Pregnancies in Post-pubertal Cancer Patients: ESMO Clinical Practise Guidelines†. Ann Oncol. 2020;31:1664-78.
    [CrossRef] [PubMed] [Google Scholar]
  44. , , , , , . Cancer Treatment-related Infertility: A Critical Review of the Evidence. JNCI Cancer Spectr. 2019;3:pkz008.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  45. , , , , , . Addressing Barriers to Fertility Preservation for Cancer Patients: The Role of Oncofertility Patient Navigation. J Oncol Navig Surviv. 2021;12:332-48.
    [PubMed] [PubMed Central] [Google Scholar]
  46. , , . Fertility Preservation Discussions with Male Adolescents with Cancer and their Parents: “Ultimately, it’s his Decision”. JAMA Pediatr. 2018;172:799-800.
    [CrossRef] [PubMed] [Google Scholar]
  47. , , , . Fertility Preservation for Prepubertal Boys: Lessons Learned from the Past and Update on Remaining Challenges Towards Clinical Translation. Hum Reprod Update. 2021;27:433-59.
    [CrossRef] [PubMed] [Google Scholar]
  48. , , , , , . Infrastructure of Fertility Preservation Services for Pediatric Cancer Patients: A Report from the Children’s Oncology Group. JCO Oncol Pract. 2022;18:e325-e333.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  49. , , , , , . Use and Effectiveness of Sperm Cryopreservation for Adolescents and Young Adults: A 37-year Bicentric Experience. J Adolesc Young Adult Oncol. 2021;10:78-84.
    [CrossRef] [PubMed] [Google Scholar]
  50. , , , , , . How can we Improve Oncofertility Care for Patients? A Systematic Scoping Review of Current International Practise and Models of Care. Hum Reprod Update. 2019;25:159-79.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
Show Sections