Skip to main content
  • Research article
  • Open access
  • Published:

Male cancer patient sperm cryopreservation for fertility preservation: 10-year monocentric experience

Abstract

Background

Sperm cryopreservation, an effective method for preserving male fertility, is very advantageous for men suffering from cancer. Unfortunately, as both physicians and cancer patients are unaware of the possibilities for sperm cryopreservation, the data on evaluation of semen parameters and disposition of cryopreserved samples among Chinese cancer patients are scarce.

Results

Male tumours were classified into six major types, germ cell tumours (26 %), haematological neoplasms (28 %), head and neck cancers (19 %), thoracic tumours (4 %), abdominal tumours (10 %), and others (13 %). Haematological neoplasm was the most prevalent cancer among our cohort of patients who opted for sperm banking, followed by germ cell tumours. Patients with germ cell tumours had the lowest pre-thaw and post-thaw seminal sperm concentrations. We separately compared patients with testicular tumours, lymphoma, and leukaemia, and found that leukaemia patients had the lowest pre-thaw sperm concentrations. Most cancer patients (58 %) chose to keep their specimens stored, while 31 % chose to discard the specimens. Over the years, only 13 patients (4 %) returned to use their spermatozoa by assisted reproductive technology. Of the stored samples, patients with germ cell tumours constituted the highest proportion (29.3 %). Moreover, the percentage of haematological neoplasm patients who had no spermatozoa frozen was the highest (46.2 %).

Conclusions

The present data confirm the deleterious impact of various cancers on semen quality. Leukaemia was associated with the worst semen quality and the highest number of semen samples that could not be frozen. We suggest that sperm quality may have decreased even before anti-neoplastic treatment and that sperm banking before treatment should be strongly recommended for cancer patients. A sperm banking programme before gonadotoxic therapy requires close cooperation between assisted reproduction centres and cancer clinics.

Résumé

Contexte

Lacryoconservation des spermatozoïdes, méthode efficace pour préserver lafertilité masculine, est très avantageuse pour les hommes atteints de cancer.Malheureusement, comme les médecins et les patients atteints de cancersignorent les possibilités de cryoconservation du sperme, les données surl’évaluation des paramètres spermatiques et le devenir des échantillonscryopréservés parmi les hommes chinois atteints de cancers sont rares.

Résultats

Lestumeurs masculines ont été classées en six types principaux, tumeurs germinales(26%), néoplasmes hématologiques (28%), cancers de la tête et du cou (19%),tumeurs thoraciques (4%), les tumeurs abdominales (10%), et les autres (13%).Les néoplasmes hématologiques étaient le cancer le plus répandu parmi notrecohorte de patients qui ont opté pour la mise en banque de leur sperme, suivisdes tumeurs à cellules germinales. Les patients atteints de tumeurs germinalesprésentaient les concentrations de spermatozoïdes pré-congélation etpost-décongélation les plus faibles. Nous avons comparé séparément les patientsatteints de tumeurs testiculaires, de lymphome, et de leucémie, et avonsconstaté que ceux atteins de leucémie présentaient les plus bassesconcentrations de spermatozoïdes pré-congélation. Une majorité de patientsatteints de cancer (58 %) ont choisi de conserver en banque leurs échantillonscongelés, tandis que 31 % ont choisi de jeter leurs échantillons congelés. Aufil des ans, seuls 13 patients (4%) sont revenus pour utiliser leurs spermatozoïdespar Assistance Médicale à la Procréation. Parmi les échantillons mis en banque,ceux des patients atteints de tumeurs germinales constituaient la proportion laplus élevée (29,3 %). Par ailleurs, les patients atteins de cancershématologiques constituaient le pourcentage le plus élevé (46.2%) de patientsqui n’avaient eu aucun spermatozoïde congelé.

Conclusions

Lesdonnées rapportées confirment l’impact délétère des divers cancers sur laqualité du sperme. Les leucémies étaientassociées à la pire qualité du sperme et au plus grand nombre d’échantillons desperme qui ne pouvaient pas être congelés. Nous pensons que la qualité desperme pourrait avoir diminué avant même le traitement antinéoplasique, et quela mise en banque de sperme avant le traitement devrait être fortementrecommandée pour les patients atteints de cancers. Un programme de mise enbanque du sperme avant traitement gonadotoxique nécessite une étroitecoopération entre les centres d’Assistance Médicale à la Procréation et les cliniquesde cancérologie.

Introduction

According to the World Health Organization (WHO), infertility will become the third-most prevalent disease in the 21st century after cancer and cardiovascular diseases, affecting human life and health [1]. Studies have reported that the distress related to infertility is more prevalent among male cancer survivors than among unaffected men [2,3,4]. The increase in the number of cancer survivors has highlighted the need for long-term improvement in their quality of life and the demand for reproductive physicians, especially with respect to cancer-related infertility [5, 6]. Therefore, studies have investigated the potential negative effects of cancer therapies, including chemotherapy and radiotherapy, and malignancy itself on male fertility [7]. Sperm banking for male cancer patients is considered as the most effective method for preserving fertility [8, 9]. Several studies have been reported that testicular cancer and haematological diseases affect male fertility, but many other cancers including head and neck tumor and sarcoma that were little reported about male fertility.

In China, only a few reports have been published on fertility preservation in male cancer patients [10]. One of the studies conducted a retrospective review of sperm cryopreservation for 143 male cancer patients at the Human Sperm Bank of the National Research Institute for Family Planning in Beijing [11].

Given the late start of this programme in China, there is not much awareness about human fertility preservation, highlighting the need to develop a systemic and patient-centric programme for offering cryopreservation services to all male cancer patients. The Human Sperm Bank, West China Second University Hospital, Sichuan University, has been offering sperm banking to patients for 10 years. In comparison with other sperm banks in China, our unit has access to a greater number of subjects and has received 1039 sperm samples for cryopreservation for birth demand. Overall, 339 cancer patients banked their spermatozoa for fertility preservation. To our knowledge, we have accessed most male cancer patients who came to bank their spermatozoa for fertility conservation in southwest China.

This is a 10-year retrospective study of a population of patients referred for sperm cryopreservation before cancer therapy. The primary aim of our study was to evaluate semen parameters and the disposition of cryopreserved samples among different cancer patients in our 10-year experience with sperm banking. We simultaneously focused on reproductive outcomes using cryopreserved semen via assisted reproductive technology (ART).

Materials and methods

Patients

In accordance with the guidelines and processes established by our sperm bank, all cancer patients seeking fertility preservation were counselled by an andrology physician and fully informed about the procedure, including the process entailed, costs, future use and storage duration. After receiving written and oral information, men were required to sign a cryostorage consent form that stated that spermatozoa would be discarded upon loss to follow-up and specified how the spermatozoa were to be used. It was stipulated that the stored gametes were only to be used for married couples. Moreover, no use in posthumous conception was allowed. Sperm cryopreservation was performed according to a standardised protocol in the sperm bank and only on semen samples containing motile spermatozoa. If no motile spermtozoa were detected, the findings were discussed with the patient and the sample was not cryopreserved. The disposition categories were continued storage within our unit, discarded, failure to bank, used, and patient’s death. The type of cancer was determined from the oncologist’s letter or by histological diagnosis. We followed up cancer patients by telephone.

Semen samples

Semen samples were obtained by masturbation in sterile containers, which were placed in an incubator at 37 °C until liquefaction and analysed within 1 h. The following semen parameters were evaluated according to the WHO 1999, 2010 guidelines: sperm concentration, volume, progressive motility (grades a + b), and morphology [12, 13]. Sperm motility and concentration were detected using a Suiplus semen analysis (SSA) automatic detection system, which consists of a phase-contrast microscope (OLYMPUS CX41), a micro camera (Basler acA780—75gc), SSA software system (Beijing SuiJia Software Co., Ltd, China) [14] and a Makler counting chamber (Sefi Medical Instruments, Haifa, Israel).

Sperm cryopreservation was performed as follows. The liquefied semen sample was transferred to a 2.0 mL sterile cryotube with Sperm Cryoprotectant Kit (Anhui Anke Biotechnology (Group) Co.,LTD, China, glycerol-egg yolk free cryopreservative medium, final glycerol concentration of 7.5 % [15, 16]) added dropwise until a 3:1 sample: medium ratio. The final volume of diluted seminal fluid in a tube was 1mL.

The straws were stored at 4 °C for 15 min and then suspended 5 cm above of liquid nitrogen for 10 min before being stored in liquid nitrogen. For semen thawing, straws were placed at room temperature for 1 min and then transferred to a beaker at 37 °C until all ice crystals had melted. Azoospermia was confirmed by centrifugation of the entire semen sample at 3000 ×g for 15 min.

Statistical analysis

The statistical methodology comprised one-way analysis of variance and Kruskal-Wallis tests. In all cases, statistical significance was set at p < 0.05.

Results

Study population

A total of 339 male cancer patients who opted for sperm cryopreservation were recruited from January 2010 to December 2019 (Fig. 1). The type of cancers among these 339 patients included germ cell tumours (26 %), haematological neoplasms (28 %), head and neck cancers (19 %), thoracic tumours (4 %), abdominal tumours (10 %), and others (13 %). Germ cell tumours included testicular tumours and extragonadal germ cell tumours. Haematological neoplasms comprised lymphoma and leukaemia. The “other” group was composed of skin, sarcoma, and peripheral Schwannoma. The cancer incidence rate for men in Sichuan Province is 303.60 per 100,000. The six more common cancers among men (based on incidence) were lung cancer (74.26 per 100,000), liver cancer (48.3 per 100,000), oesophageal cancer (42.62 per 100,000), gastric cancer (37.55 per 100,000), colorectal cancer (28.58 per 100,000), and pancreatic cancer (7.38 per 100,000) [17]. We compared the incidence, mortality, and sperm preservation among patients with top six tumour types in Sichuan using the data from the Sichuan cancer registries. Only colon cancer was in the top six tumour types for sperm preservation (Fig. 2). Therefore, the high incidence of tumours may not be related to high sperm preservation rate, owing to high mortality. The types of tumours that preserve fertility are those affected by reproduction or have a younger incidence. In our unit, we found not only testicular tumours but also extragonadal germ cell tumours among men with the need for fertility preservation. The age of the patients was 26.7 ± 6.8 years, and 31 patients (9.9 %) were adolescents (below 18 years of age).

Fig. 1
figure 1

Flow diagram of male cancer patients who presented to Sichuan Human Sperm Bank from 2010 to 2019 for sperm cryopreservation

Fig. 2
figure 2

The incidence of male, mortality of male and the top six malignant tumors in male inducing sperm preservation in Sichuan

Semen parameters

Most samples were collected in sterile containers by masturbation after an abstinence period of 2–7 days. Some patients had sexual abstinence period of longer than 1 week, owing to emergency treatment such as needing radiation or chemotherapy (Tables 1 and 2). The mean semen volume was 3.4 ± 1.5 mL, and the median number of straws stored was 4 per patient among different cancer groups. Before cryopreservation, the overall median sperm concentration was 47 × 106/mL and the progressive motility was 49 %. Post-thaw analyses after cryopreservation revealed a median concentration of 30 × 106/mL and progressive motility of 28 %. No difference was observed in the change from pre-freeze to post-thaw progressive motility and the recovery rate between various cancer groups. The sperm pre-count, total sperm number, and post-thaw concentration were significantly lower in germ cell tumours than in other five cancers (Table 1). We compared the data from leukaemia, lymphoma, and testicular cancer patients, and found that the patients with leukaemia had the poorest sample quality, consistent with the lower pre-freeze count and pre-freeze total sperm number (Table 2).

Table 1 Semen parameters at banking stratified by cancer types
Table 2 Patients’ age and semen parameters of testicular tumor, leukemia and lymphoma

Outcomes of 339 referrals for elective semen cryopreservation

Of the 339 men requesting sperm cryostorage, 313 had cryostored semen samples. The remaining 26 men were either azoospermic (n = 22, 6.5 %) or had immotile spermatozoa (n = 4, 1.2 %). Disposition of cryopreserved sperm categories included continued storage (58 %), discarded (31 %), death (1 %), and use (4 %) (Table 3). Haematological neoplasms were the major cancers related to failed cryopreservation (46.2 %). Semen samples from patients with germ cell tumours (29.3 %) and haematological neoplasms (26.8 %) were the most abundant samples under current storage (Table 3).

Table 3 Sperm banking outcomes of each type of six cancers in each way of disposition

Outcome of ART with cryopreserved semen

We analysed the details of men opting cryopreserved spermatozoa and their reproductive outcomes. As of 31 December 2019, straws from 13 patients (3.8 %) had been used in 15 ART cycles (one patient received three cycles, including intracytoplasmic sperm injection [ICSI], in vitro fertilisation [IVF], intra-uterine insemination [IUI]). Conceptions were achieved in 66.7 % cases (10 out of 15), with 50 % (5 out of 10) pregnancies resulting in delivery and 20 % into spontaneous abortion (2 out of 10). Overall, 30 % (3 out of 10) were clinical pregnancies. Two couples failed to conceive, and one embryo was cryopreserved. The details of these cases are listed in Table 4.

Table 4 Details of men using cryopreserved sperm and their reproductive outcomes

Discussion

As cancer survival rate improves, more attention is being directed from issues of cancer treatment toward enhancing quality of life of cancer survivors. Cryopreservation of semen samples is a non-invasive procedure and the main treatment option for male cancer patients [18].

In our study, we demonstrated that male cancer patients in Sichuan, China, used sperm banking for fertility preservation. During the 10 years of our study, 339 male cancer patients attempted to preserve fertility; this is the largest reported population opting cryopreservation in China. The cancers were divided into six types, germ cell tumours, haematological neoplasms, head and neck cancers, thoracic tumours, abdominal tumours, and other tumours. We found that men with germ cell tumours (including testicular cancer and extragonadal germ cell tumours) had inferior pre- and post-cryopreserved sperm concentrations as compared to those with other types of cancers. However, upon separate comparison of testicular cancer, leukaemia, and lymphoma patients, we found that leukaemia patients had the lowest sperm concentrations. In this study, 46.2 % patients with leukaemia and lymphoma had no spermatozoa to freeze. Some reports have shown that leukaemia patients have an innate suppression of spermatogenesis through an unknown mechanism [19,20,21]. In practice, a few cancer patients visiting our unit give up sperm preservation owing to the cost. We concluded that the urgent treatment time for further treatment might be the reason for this result. A study showed that sperm total motile count (TMC) < 1 million was the second most prevalent category of sperm quality among leukaemia patients during the first visit for semen collection [22]. It is imperative to communicate with clinicians in a timely manner to ensure that leukaemia patients bank sperms before cancer treatment.

Some studies have reported that sperm count and motility are significantly lower in men with testicular tumours [19, 23,24,25,26]. Shankara-Narayana N et al. found that sperm output positively correlated with total testicular volume [26]. Moreover, another study reported that 50.0 % of patients presented with a decrease in sperm concentration after orchiectomy. Even before surgery, approximately half of patients with testicular germ cell tumours presented poor semen quality [27]. Therefore, there are several factors underlying semen quality decline in patients with testicular germ cell tumours, including orchiectomy, disruption of the blood-testis barrier, and endocrine derangements [28, 29].

Cancer patients who choose to use their cryopreserved sperms for fertility will have a good chance of experiencing fatherhood. Nevertheless, studies have reported that the rate of frozen semen usage varies between 7 and 30 %, and about 50 % of them are successful for in vitro fertilisation and intracytoplasmic sperm injection [30,31,32,33]. Over the course of 10 years, 3.8 % patients used their samples in our facility. The time elapsed between sperm freezing and follow-up is a fundamental factor that determines the utilisation rate. The number of patients requiring this service is sharply increasing year after year along with the number of patients who opt to use their samples in assisted conception [33]. Without this information, it is difficult to evaluate the importance of the results. Even if the observation time was 10 years, it is possible that the last recruited patients had a very short follow-up time for evaluation. The limitations of our study are the inability to follow-up couples who were naturally conceived and the lack of knowledge about the reason for sample discard. In the future, we will follow-up all cancer patients who join our cohort for sperm banking for their reproductive outcomes and investigate the reason for sample discard.

Considering the late development of fertility conservation programmes, there is a large gap between China and Europe or the USA. In 2005, the proportion of haematological and germ cell cancer patients between 15 and 19 years of age who opted cryopreservation was 92 and 90 %, respectively in France [34]. At present, there is no report on oncologists providing suggestions on fertility protection in China, but the sperm preservation of tumour patients in various sperm banks in China reflects the current situation of fertility protection to a certain extent. There were 130 male patients who underwent sperm cryopreservation before proceeding to gonadotoxic treatment at the Chinese University of Hong Kong [35]. A total of 97 Chinese male patients underwent sperm cryopreservation in the Hunan Human Sperm Bank from 2004 to 2015. These retrospective audits revealed 52 men with cancer who successfully banked spermatozoa in Zhejiang Human Sperm Bank between 2005 and 2013, 12 men in Shanghai Human Sperm Bank between 2003 and 2009, and 17 men in Jiangsu Human Sperm Bank between 2007 and 2014 [10]. A total of 145 male cancer patients underwent sperm cryopreservation in Beijing Human Sperm Bank [11]. There were 339 male patients with cancer opting fertility preservation option in our unit.

No practice guidelines advocate fertility preservation for cancer in China. One obstacle to sperm banking is inadequate communication between physicians and patients regarding the risk of post-treatment infertility [36]. Specialists in male reproduction or oncologists should discuss sperm preservation possibilities with cancer patients as early as possible. All oncological therapies, including surgical procedures, chemotherapy, and radiotherapy, pose some risk to fertility [4]. These adults can cryopreserve spermatozoa produced by masturbation. To date, sperm cryopreservation is impossible for prepubertal male patients, especially those who cannot provide semen. If collection of seminal spermatozoa is not possible, the testicular tissue containing spermatogonia stem cells can be obtained by biopsy. Exciting basic scientific study is underway to address unmet needs of fertility preservation that may expand fertility options for men in the future.

Conclusions

In conclusion, our study supports the finding that sperm banking is an effective method of fertility preservation in cancer patients. Fertility preservation is very low among Chinese male patients with cancer. Therefore, reproductive physicians and oncologists are required to discuss fertility preservation techniques before gonadotoxic therapy in all male cancer patients. Sperm cryopreservation is a quick and effective technique for preserving fertility. The development of local clinical guidelines and organisation of conferences to promote fertility preservation should be encouraged.

Availability of data and materials

The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

ART:

Assisted reproductive treatments

WHO:

World health organization

SSCs:

Spermatogonia stem cells

EGTC:

Extragonadal germ cell tumours

ALL:

Acute lymphoblastic leukaemia

AML:

Acute myelogenous leukaemia

CML:

Chronic myelocytic leukaemia

NHL:

Non-hodgkin lymphoma

HL:

Hodgkin lymphoma

NPC:

Nasopharyngeal carcinoma

ICSI:

Intracytoplasmic sperm injection

IVF:

In vitro fertilization

IUI:

Intrauterine insemination

References

  1. Rostami Dovom M, Ramezani Tehrani F, Abedini M, Amirshekari G, Hashemi S, Noroozzadeh M. A population-based study on infertility and its influencing factors in four selected provinces in Iran (2008–2010). Iran J Reprod Med. 2014;12(8):561–6.

    PubMed  PubMed Central  Google Scholar 

  2. Williams DHt, Karpman E, Sander JC, Spiess PE, Pisters LL, Lipshultz LI. Pretreatment semen parameters in men with cancer. J Urol. 2009;181(2):736–40.

    Article  Google Scholar 

  3. Bahadur G, Ozturk O, Muneer A, Wafa R, Ashraf A, Jaman N, et al. Semen quality before and after gonadotoxic treatment. Hum Reprod. 2005;20(3):774–81.

    Article  CAS  Google Scholar 

  4. Barak S. Fertility preservation in male patients with cancer. Best Pract Res Clin Obstet Gynaecol. 2019;55:59–66.

    Article  Google Scholar 

  5. Wallace WH, Anderson RA, Irvine DS. Fertility preservation for young patients with cancer: who is at risk and what can be offered? Lancet Oncol. 2005;6(4):209–18.

    Article  Google Scholar 

  6. Moss JL, Choi AW, Fitzgerald Keeter MK, Brannigan RE. Male adolescent fertility preservation. Fertil Steril. 2016;105(2):267–73.

    Article  Google Scholar 

  7. Katz DJ, Kolon TF, Feldman DR, Mulhall JP. Fertility preservation strategies for male patients with cancer. Nat Rev Urol. 2013;10(8):463–72.

    Article  Google Scholar 

  8. Holoch P, Wald M. Current options for preservation of fertility in the male. Fertil Steril. 2011;96(2):286–90.

    Article  Google Scholar 

  9. Jahnukainen K, Ehmcke J, Hou M, Schlatt S. Testicular function and fertility preservation in male cancer patients. Best Pract Res Clin Endocrinol Metab. 2011;25(2):287–302.

    Article  Google Scholar 

  10. Fu LL, Zhang KS, Gu YQ. [Fertility preservation for male adolescent cancer patients]. Zhonghua Nan Ke Xue. 2017;23(3):262–6.

    PubMed  Google Scholar 

  11. Fu L, Zhou F, An Q, Zhang K, Wang X, Xu J, et al. Sperm Cryopreservation for Male Cancer Patients: More than 10 Years of Experience, in Beijing China. Med Sci Monit. 2019;25:3256–61.

    Article  Google Scholar 

  12. World Health Organization. WHO Laboratory Manual for the Examination of Human Semen and Sperm-Cervical Mucus Interaction. 4th Edition. Cambridge University Press. 1999.

  13. World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen. 5th ed. Geneva: Switzerland: WHO Press; 2010.

  14. Luo D, Zhang M, Su X, Liu L, Zhou X, Zhang X, et al. High fat diet impairs spermatogenesis by regulating glucose and lipid metabolism in Sertoli cells. Life Sci. 2020;257:118028.

    Article  CAS  Google Scholar 

  15. Hammadeh ME, Greiner S, Rosenbaum P, Schmidt W. Comparison between human sperm preservation medium and TEST-yolk buffer on protecting chromatin and morphology integrity of human spermatozoa in fertile and subfertile men after freeze-thawing procedure. J Androl. 2001;22(6):1012–8.

    Article  CAS  Google Scholar 

  16. Mahadevan M, Trounson AO. Effect of cryoprotective media and dilution methods on the preservation of human spermatozoa. Andrologia. 1983;15(4):355–66.

    Article  CAS  Google Scholar 

  17. CHENG Shuwen DT, Jing ZENG, Xinyin XU, Ying DENG, Xin ZHANG. Analysis of cancer incidence and mortality in registration areas of Sichuan,2015. Journal of Applied Oncology. 2020;34(4):304–8.

    Google Scholar 

  18. Oktay K, Harvey BE, Partridge AH, Quinn GP, Reinecke J, Taylor HS, et al. Fertility Preservation in Patients With Cancer: ASCO Clinical Practice Guideline Update. J Clin Oncol. 2018;36(19):1994–2001.

    Article  Google Scholar 

  19. Bonetti TC, Pasqualotto FF, Queiroz P, Iaconelli A Jr, Borges E, Jr. Sperm banking for male cancer patients: social and semen profiles. Int Braz J Urol. 2009;35(2):190-7; discussion 7–8.

  20. Botchan A, Hauser R, Gamzu R, Yogev L, Lessing JB, Paz G, et al. Sperm quality in Hodgkin’s disease versus non-Hodgkin’s lymphoma. Hum Reprod. 1997;12(1):73–6.

    Article  CAS  Google Scholar 

  21. Moody JA, Ahmed K, Yap T, Minhas S, Shabbir M. Fertility managment in testicular cancer: the need to establish a standardized and evidence-based patient-centric pathway. BJU Int. 2019;123(1):160–72.

    Article  Google Scholar 

  22. Jayasena CN, Luo R, Dimakopoulou A, Dearing C, Clarke H, Patel N, et al. Prevalence of abnormal semen analysis and levels of adherence with fertility preservation in men undergoing therapy for newly diagnosed cancer: A retrospective study in 2906 patients. Clin Endocrinol (Oxf). 2018;89(6):798–804.

    Article  CAS  Google Scholar 

  23. 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):7–11.

    Article  Google Scholar 

  24. Menon S, Rives N, Mousset-Simeon N, Sibert L, Vannier JP, Mazurier S, et al. Fertility preservation in adolescent males: experience over 22 years at Rouen University Hospital. Hum Reprod. 2009;24(1):37–44.

    Article  CAS  Google Scholar 

  25. Johnson MD, Cooper AR, Jungheim ES, Lanzendorf SE, Odem RR, Ratts VS. Sperm banking for fertility preservation: a 20-year experience. Eur J Obstet Gynecol Reprod Biol. 2013;170(1):177–82.

    Article  Google Scholar 

  26. Shankara-Narayana N, Di Pierro I, Fennell C, Ly LP, Bacha F, Vrga L, et al. Sperm cryopreservation prior to gonadotoxic treatment: experience of a single academic centre over 4 decades. Hum Reprod. 2019;34(5):795–803.

    Article  CAS  Google Scholar 

  27. Andrade MBR, Bertolla RP, Intasqui P, Antoniassi MP, Tibaldi DS, Belardin LB, et al. Effect of orchiectomy on sperm functional aspects and semen oxidative stress in men with testicular tumours. Andrologia. 2019;51(3):e13205.

    Article  CAS  Google Scholar 

  28. Howell SJ, Shalet SM. Spermatogenesis after cancer treatment: damage and recovery. J Natl Cancer Inst Monogr. 2005(34):12–7.

  29. Hotaling JM, Lopushnyan NA, Davenport M, Christensen H, Pagel ER, Muller CH, et al. Raw and test-thaw semen parameters after cryopreservation among men with newly diagnosed cancer. Fertil Steril. 2013;99(2):464–9.

    Article  Google Scholar 

  30. van Casteren NJ, van Santbrink EJ, van Inzen W, Romijn JC, Dohle GR. Use rate and assisted reproduction technologies outcome of cryopreserved semen from 629 cancer patients. Fertil Steril. 2008;90(6):2245–50.

    Article  Google Scholar 

  31. Neal MS, Nagel K, Duckworth J, Bissessar H, Fischer MA, Portwine C, et al. Effectiveness of sperm banking in adolescents and young adults with cancer: a regional experience. Cancer. 2007;110(5):1125–9.

    Article  Google Scholar 

  32. Kelleher S, Wishart SM, Liu PY, Turner L, Di Pierro I, Conway AJ, et al. Long-term outcomes of elective human sperm cryostorage. Hum Reprod. 2001;16(12):2632–9.

    Article  CAS  Google Scholar 

  33. Dearing C, Breen D, Bradshaw A, Ramsay J, Lindsay K. Trends and usage in a London National Health Service Sperm Bank for cancer patients. Hum Fertil (Camb). 2014;17(4):289–96.

    Article  Google Scholar 

  34. Daudin M, Rives N, Walschaerts M, Drouineaud V, Szerman E, Koscinski I, et al. Sperm cryopreservation in adolescents and young adults with cancer: results of the French national sperm banking network (CECOS). Fertil Steril. 2015;103(2):478–86. e1.

    Article  Google Scholar 

  35. Chung JP, Haines CJ, Kong GW. Sperm cryopreservation for Chinese male cancer patients: a 17-year retrospective analysis in an assisted reproductive unit in Hong Kong. Hong Kong Med J. 2013;19(6):525–30.

    PubMed  Google Scholar 

  36. Achille MA, Rosberger Z, Robitaille R, Lebel S, Gouin JP, Bultz BD, et al. Facilitators and obstacles to sperm banking in young men receiving gonadotoxic chemotherapy for cancer: the perspective of survivors and health care professionals. Hum Reprod. 2006;21(12):3206–16.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by Sichuan Science and Technology Program (No. 2020ZYD007).

Author information

Authors and Affiliations

Authors

Contributions

Xiao Liu wrote the article and prepared the tables and figures. Bo Liu, Xiao Xiao, Li Wang, Xiaofang Zhu and Bizhen Shu verified all data collected. Wenrui Zhao carried out the data analysis. Xiao Liu, Bo Liu, Shasha Liu and Yang Xian freezed semen. Min Jiang and Bin Zhou provide technical support. Fuping Li designed the study and edited the article. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Fuping Li.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Ethics Committee of the West China Second University Hospital, Sichuan University. Informed written consent was obtained from all patients prior to their enrollment in this study.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Liu, B., Liu, S. et al. Male cancer patient sperm cryopreservation for fertility preservation: 10-year monocentric experience. Basic Clin. Androl. 31, 24 (2021). https://doi.org/10.1186/s12610-021-00140-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12610-021-00140-w

Keywords

Mots-clés