Male Fertility Preservation: Current Options and Advances in Research



Fig. 8.1
Testicular tissue cryopreservation. Testicular tissues are transported on ice from the operating room to the andrology lab in a sterile specimen container containing medium. (a) Tissue is kept cool and processed in a sterile environment with sterile tools. (b) Most centers cut the testicular tissue into small pieces (2–9 mm3) and deposit in cryovials with DMSO-based freezing medium. (c) Controlled slow rate using a freezing machine




Table 8.1
Cryopreservation of human testicular tissue and cells











































































































Tissue or cells

Freezing method

Freezing conditions

Endpoints

References

Cells

Controlled slow freezing

10 % HSA, 10 % DMSO, 1 % dextran

Viability, Fc – SSEA4, LHR, VASA

[64]

Tissue

Tissue

Controlled slow freezing

0.7 M DMSO, 0.1 M sucrose

Xenografting, IHC – MAGE- A4, Ki67, 3p-HSD

[73]

Vitrification

Eq. sol. – 7.5 % EG, 7.5 % DMSO, 0.25 M sucrose. Vitr. Sol. 15 % EG, 15 % DMSO, 0.5 M sucrose

Tissue

Controlled slow freezing

0.7 M DMSO, 5 % HSA

IHC – MAGE-A4, TEM, organ culture

[67]

Tissue

Controlled slow freezing

0.7 M DMSO, 0.1 M sucrose, 10 mg/ml HSA

IHC – MAGE-A4 and Ki67

[72]

Vitrification

?

Tissue

Controlled slow freezing

1.5 M EG, 0.1 M sucrose, 10% HSA

Morphology, IHC – KIT

[68]

Tissue

Controlled slow freezing

0–2.5 M DMSO or EG or glycerol with 0.1 % ITS and 20 % FBS

Viability, seminiferous tubule culture

[70]

Tissue

Controlled slow freezing

5 % DMSO, 5 % HSA

IHC – MAGE-A4, vimentin, CD34. TEM. Tissue culture

[59]

Tissue

Controlled slow freezing

(1) 0.7 M or 1.5 M DMSO and 5 % HSA (2) 0.7 M DMSO, 0.1 M sucrose, 10 % HSA

IHC – TUNEL, PCNA, UCHL1. TEM

[71]

Uncontrolled slow freezing

0.7 M or 1.5 M DMSO, 0.15 M sucrose, 10 % HSA

Solid-surface vitrification

Eq. sol. – 1.35 M EG, 1.05 M DMSO. Vitr. Sol. 2.7 M EG, 2.1 M DMSO, 20 % HSA

Direct cover vitrification
 

Cells

Controlled slow freezing

1.28 M DMSO, 25 % FBS

Fc – CD45, THY1, SSEA4

[63]

Tissue

Cells

Controlled slow freezing

2 % HSA, 1.4 M DMSO

Cell recovery, viability

[74]

Uncontrolled slow freezing

2 % HSA, 0.7 M DMSO

Vitrification

Eq. sol. – 2 % HSA, 1.1 M DMSO, 1.34 M EG. Vitr. Sol. – 2 % HSA, 0.67 M sucrose, 2.3 M DMSO, 1.34 M EG

Cells

Controlled slow freezing

4 % FBS, 1.5 M DMSO or EG or glycerol or 1,2-propanediol

Viability

[69]


HSA human serum albumin, DMSO dimethyl sulfoxide, Fc flow cytometry, EG ethylene glycol, IHC immunohistochemistry, TEM transmission electron microscopy, FBS fetal bovine serum

Table 8.1 is reproduced from Valli et al., 2015 [65], with permission of Springer




Testicular Cell-Based Methods to Preserve and Restore Male Fertility



Spermatogonial Stem Cell Transplantation


Spermatogonial stem cell transplantation was first described by Ralph Brinster and colleagues in 1994, who demonstrated that SSCs could be isolated and transplanted to regenerate spermatogenesis in infertile recipient mice [75, 76]. SSC transplantation has now been reported in mice, rats, pigs, goats, bulls, sheep, dogs, and monkeys, and donor-derived progeny has been produced by natural breeding in mice, rats, goats, and sheep [7788]. SSCs from donors of all ages, newborn to adult, are competent to regenerate spermatogenesis [78, 89], and SSCs can be cryopreserved and retain spermatogenic function upon thawing and transplantation [85, 90, 91]. Thus, it appears feasible that a testicular tissue biopsy (containing SSCs) could be obtained from a prepubertal boy prior to gonadotoxic therapy, frozen, thawed at a later date, and transplanted back into his testes to regenerate spermatogenesis. If spermatogenesis from transplanted cells is robust, this approach may restore natural fertility, allowing survivors to achieve pregnancy with their partner by natural intercourse and have biological children.

Radford and colleagues already reported cryopreserving testicular cells for 11 adult non-Hodgkin’s lymphoma patients in 1999 and subsequently reported transplanting autologous frozen and thawed testis cells back into the testes of seven survivors [92, 93]. The fertility outcomes for patients in that study have not been reported, and even if the men fathered children, it would not be possible to ascertain whether the sperm arose from transplanted stem cells or surviving endogenous stem cells. This uncertainty will always plague the interpretation of human SSC transplant studies where it is not ethically possible to genetically mark the transplanted cells because the genetic modification would be transmitted to progeny. Therefore, large epidemiological datasets generated over decades will be required to prove the fertility benefit of SSC transplantation. Nonetheless, this study demonstrates that patients are willing to pursue experimental stem cell-based options even when there is no guarantee of a fertile outcome. There are no published reports of SSC transplantation in humans since Radford’s follow-up report of his non-Hodgkin’s lymphoma patients in 2003 [93].


Translating Spermatogonial Stem Cell Transplantation into the Clinic: Challenges and Opportunities


Considering the progress in several animal models and the fact that testicular tissues have already been cryopreserved for hundreds of human patients worldwide [5460, 92, 93], it seem reasonable to expect that SSC transplantation and/or other stem cell technologies will impact the fertility clinic in the next decade. However, there are several safety and feasibility issues that must be considered.


Spermatogonial Stem Cell Culture


Based on our experiences at the Fertility Preservation Program in Pittsburgh [54] and published reports [56, 59], it is reasonable to expect that 50–500 mg of testicular tissue can be obtained by wedge biopsy or needle biopsy from a single testis of a prepubertal boy. This is a small amount of tissue relative to size of adult human testes that can range from 11 to 26 g in size [94]. It is widely believed that the number of stem cells in biopsies from prepubertal boys will be small and that SSCs will have to be expanded in culture prior to transplant. Conditions for maintaining and expanding rodent SSCs in culture are well established and SSCs maintained in long-term culture remain competent to regenerate spermatogenesis and restore fertility [95100].

If cultured human SSCs function similar to cultured rodent SSCs, it should be feasible to expand a few stem cells obtained from the testis biopsy of a prepubertal boy to a number sufficient to produce robust spermatogenesis upon transplantation back into his testes when he is an adult. Several studies have reported culturing human SSCs [57, 58, 101109], including two studies in which cultures were established from the testes of prepubertal patients [58, 101]. Human SSC cultures have been evaluated by quantitative PCR or immunocytochemistry for spermatogonial markers or xenotransplantation into mouse testes. Strategies to isolate and culture human spermatogonia have been unique to each study, and to date no approach has been independently replicated in another laboratory. Also, the field is frustrated by the lack of a functional assay to test the full spermatogenic potential of cultured human cells.


Malignant Contamination


A testicular biopsy obtained from a cancer patient could harbor malignant cells, especially for patients with leukemia. Kim and colleagues [110] reported that 20 % of boys with acute lymphocytic anemia had malignant cells in their testicular tissue prior to the initiation of oncologic treatment. Jahnukainen and colleagues [111] reported the transmission of leukemia after transplantation of testis cells from terminally ill leukemic rats into the testes of non-leukemic recipients. The same group further demonstrated that transplantation of as few as 20 leukemic cells was sufficient for disease transmission, leading to terminal leukemia within 3 weeks.

Because infertility is not life threatening and fertility treatments are elective, it is essential that risk of cancer recurrence after transplant be reduced to zero. Fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS) strategies to isolate and enrich therapeutic spermatogonia while removing malignant contamination have been explored with mixed results. To date, transplantable human spermatogonia have been recovered in the Ep-CAMlo, THY-1lo, CD49f+, SSEA4+, GPR125+, and CD9+ fractions of human testis cells [94, 102, 105, 112114].

Fujita and coworkers isolated germ cells from the testes of leukemic mice in the forward scatter high and side scatter low fraction (positive selection), which was then further divided into fractions that were CD45/MHC class I antigens (H-2Kb/H-2Db) double positive and CD45/MHC class I double negative cells. All recipient males injected with the CD45+/MHC class I+ cells developed terminal leukemia within 40 days. All mice injected with CD45/MHC class I cells survived for 300 days without onset of leukemia and produced donor-derived offspring [115]. In a subsequent study, the same group reported that seven out of eight human leukemic cell lines expressed the cell surface antigens CD45 and MHC class I [116]. In a rat model of Roser’s T-cell leukemia, Hou and colleagues concluded that single parameter selection using either leukemic (CD4 and MHC Class I) or SSC (Ep-CAM) markers was not sufficient to eliminate malignant contamination [117], but malignant contamination was successfully removed using a combination of leukemia and SSC markers (plus/minus selection) [114, 118]. Using similar positive/negative selection strategies, Hermann and colleagues isolated VASA+ germ cells in the THY-1+/CD45 fraction of leukemia-contaminated prepubertal nonhuman primate testis cells [118], and this fraction did not produce tumors in mice. Dovey and colleagues contaminated human testis cells with MOLT-4 acute lymphoblastic leukemia cells and demonstrated by xenotransplantation that the Ep-CAMlo/HLA-ABC/CD49e fraction was enriched 12-fold for transplantable human SSCs and was devoid of malignant contamination [114]. Collectively, these results are encouraging, but caution is still warranted as Geens and colleagues concluded, using EL-4 lymphoma contaminated mouse and human testis cells, that FACS- and MACS-based methods were insufficient to remove malignant contamination [119].

It will not be possible to perform comprehensive in vivo testing on patient samples because this would limit the amount of sample available for fertility therapy. More sensitive PCR-based methods have been described for detection of minimal residual disease (MRD), and this approach has identified malignant contamination in many ovarian tissue samples that were preserved for leukemia patients, even after negative histology and immunocytochemistry examination [120, 121]. However, in one of those studies, Dolmans and colleagues obtained disparate results from histology, qRT-PCR, and xenografting of ovarian tissues from leukemia patients. Quantitative RT-PCR to detect MRD revealed the possibility of malignant contamination in 9 of the 16 samples that was not detected by histological examination. However, when those ovarian tissues were grafted into recipient mice, only five of the nine samples with positive MRD had evidence of leukemic cells 3 months after transplantation [120]. Were the MRD results in the other four cases nefarious, or were they accurate and the leukemic cells simply failed to survive freezing, thawing, and grafting? In the absence of a definitive and practical test of malignant contamination, alternatives to autologous transplantation are needed for patients with hematogenous cancers, testis cancers, or cancers that metastasize to the testes.


De Novo Testicular Morphogenesis


Testicular cells (including germ cells, Sertoli cells, peritubular myoid cells, and Leydig cells) have the remarkable ability to reorganize to form normal looking seminiferous tubules when grafted under the skin of recipient mice [122126]. Ina Dobrinski and colleagues disaggregated neonatal pig and sheep testis cells, pelleted them by centrifugation, and grafted them under the skin of immune-deficient mice. When grafts were recovered between 16 and 41 weeks after transplant, cells had reorganized to form seminiferous tubules with complete spermatogenesis [125, 126]. In a remarkable extension of this approach, Kita and colleagues [124] mixed fetal or neonatal testis cells from mice or rats with GFP+-cultured mouse germline stem cells and growth factor-reduced Matrigel and grafted under the skin of immune-deficient mice. Seven to 10 weeks after grafting, seminiferous tubules with complete spermatogenesis originating from both intrinsic germ cells and cultured (GFP+) germ cells were observed. Tubules were dissected and GFP+ round spermatids were recovered, injected into mouse oocytes. The resulting embryos were transferred to recipient females and gave rise to ten mouse pups, including four with the GFP transgene. It may be feasible to build a human testis from disaggregated human testis cells, but this has not been reported to our knowledge. The human experiment may be complicated by limited availability of fetal, neonatal, or prepubertal human testis cells. It does not appear that anyone has tried to “build a testis” from disaggregated adult testis cells for any species. One day it may be possible to “build a testis,” in vitro or in vivo, on the scaffold of a decellularized human testis [127].


Testicular Tissue-Based Methods to Preserve and Restore Male Fertility



Testicular Tissue Grafting and Xenografting


Testicular tissue grafting may provide an alternative approach for generating fertilization-competent sperm from small testicular biopsies. In contrast to the SSC transplantation method in which SSCs are removed from their cognate niches and transplanted into recipient seminiferous tubules, grafting involves transplantation of the intact SSC/niche unit in pieces of testicular tissue. Honaramooz and colleagues reported that grafted testicular tissue from newborn mice, rats, pigs, and goats, in which spermatogenesis was not yet established, could mature and produce complete spermatogenesis when xenografted into nude mice [128]. The same group later reported the production of live offspring from sperm obtained from mouse testicular tissue grafts [129]. Fertilization-competent sperm was also produced from xenografts of prepubertal nonhuman primate testicular tissue transplanted into mice [130]. These results suggest that it may be possible to obtain fertilization-competent sperm by xenografting small pieces of testicular tissue from a prepubertal cancer patient under the skin of mice or other animal recipients such as pigs that are already an established source for human food consumption, replacement heart valves [131, 132], and potentially other organs [133]. Xenografting would also circumvent the issue of malignant contamination. However, the xenografting approach raises concerns about xenobiotics because viruses from mice, pigs, and other species can be transmitted to human cells [134, 135]. There is no evidence to date that xenografted human testicular tissue can produce spermatogenesis or sperm in mice [136141]. However, there is reason for optimism because Sato and colleagues observed primary spermatocytes 1 year after xenografting testicular tissue from a 3-month-old boy that clearly did not have spermatocytes at the time of transplantation [140]. Xenografting of human testicular tissue to species other than mice has not been tested to our knowledge.

If malignant contamination of the testicular tissue is not a concern, autologous testicular tissue grafting can be considered. Luetjens and colleagues demonstrated that fresh autologous testicular tissue grafts from prepubertal marmosets could produce complete spermatogenesis when transplanted into the scrotum but not under the skin [142]. Frozen and thawed grafts did not produce complete spermatogenesis in that study, but those grafts were only transplanted under the skin. Therefore, additional experimentation is merited. Testicular tissue grafting will not restore natural fertility but could generate haploid sperm that can be used to fertilize oocytes by ICSI.


Testicular Tissue Organ Culture


Sato and colleagues reported that intact testicular tissues from newborn mice (2.5–3.5 days old) could be maintained in organ culture and mature to produce spermatogenesis, including the production of fertilization-competent haploid germ cells [143, 144]. Testicular tissues from neonatal mice were minced into pieces (1–3 mm3) and placed in culture at the gas/liquid interface on a slab of agarose that was soaked in medium. Haploid round spermatids and sperm were recovered from the tissue after 3–6 weeks in culture and used to fertilize mouse eggs by ICSI. The resulting embryos were transferred to pseudopregnant females and gave rise to healthy offspring that matured to adulthood and were fertile. If testicular tissue organ culture can be translated to humans, it will provide an alternative to autologous SSC transplantation, autologous grafting, and xenografting in cases where there is concern about malignant contamination of the testicular tissue. The same authors were also successful to produce haploid germ cells in organ culture of frozen and thawed testicular tissues, which is particularly relevant to the cancer survivor paradigm. However, the fertilization potential of those sperm was not tested [143].


Induced Pluripotent Stem Cell-Based Methods to Preserve and Restore Male Fertility


Several groups have now reported that it is possible to produce germ cells from pluripotent embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) [145158]. Hayashi and coworkers reported that it is possible to differentiate ESCs or iPSCs into epiblast-like cells (EpiLCs) that then give rise to primordial germ cell-like cells (PGCLCs) when cultured in the presence of BMP4 [145]. The resulting germ cells were transplanted into the seminiferous tubules of infertile recipient mice where they regenerated spermatogenesis and produced haploid gametes that were used to fertilize mouse oocytes by ICSI. The embryos were transferred to recipient females and gave rise to live offspring. However, some of the offspring developed tumors in the neck area and died prematurely, suggesting that further optimization of the culture and differentiation protocols will be required [145]. Two groups recently reported the differentiation of human pluripotent stem cells into putative hPGCLCs exhibiting gene expression patterns similar to bona fide human PGCs [146, 147]. Of course, functional validation by generation of progeny is not possible in studies with human cells.

An important implication of the iPSC to germ cell differentiation technology, if responsibly developed, is that it will no longer be necessary to preserve fertility before the initiation of gonadotoxic treatments. An adult survivor of a childhood cancer who desires to start his family and discovers that he is infertile can theoretically produce sperm and biological offspring from his own skin, blood, or other somatic cell type. This scenario applies not only to childhood cancer survivors but all survivors who did not preserve semen or testicular tissue prior to gonadotoxic therapy. Nonhuman primate and human pluripotent stem cells have also been differentiated to the germ lineage, producing putative transplantable germ cells and even rare cells that appear to be haploid [148155, 157159]. The challenge with the human studies is that it is not possible to test the spermatogenic potential or fertilization potential of putative germ cells, which are the gold standards in animal studies. Thus, the burden of proof required of human studies is much lower than animal studies. Spermatogenic lineage development and testicular anatomy in nonhuman primates is similar to humans [22], and this may serve as a platform for safety and feasibility studies in which putative germ cells can be tested by transplantation, and the resulting gametes can be tested by fertilization [85], embryo transfer, and production of live offspring. Perhaps 1 day it will be possible to build a human testis in vitro or in vivo on a decellularized human testis scaffold, and this will provide the ultimate platform to test the spermatogenic potential of experimentally derived human germ cells.


Conclusions


Many centers worldwide are actively preserving testicular tissue or testicular cells for cancer patients in anticipation that those samples can be used in the future for reproductive purposes. Therefore, it is incumbent on the medical and research communities to responsibly develop the technologies that will allow patients to use their samples to achieve their family building goals. This is important because cancer survivors report that fertility has a significant impact on their quality of life after cure. It seems reasonable to assume that similar quality of life issues are relevant to men who are infertile due to genetic (e.g., Klinefelter), surgical, age-related, accidental, or other causes. The first, best, and proven approach for fertility preservation in males is to freeze sperm that can be obtained in a semen sample or extracted from the testis. With IVF and IVF with ICSI, only a relatively small number of sperm are required to achieve fertilization and pregnancy. Unfortunately, sperm banking is not an option for all patients, including prepubertal boys who are not yet producing sperm.

There are several testicular cell- and tissue-based technologies in the research pipeline that may have application for patients who cannot preserve sperm. All of the technologies described in this chapter are dependent on stem cells (SSCs or iPSCs) with the potential to generate or regenerate autologous spermatogenesis. Spermatogonial stem cell transplantation, de novo testicular morphogenesis, testicular tissue organ culture, testicular tissue grafting/xenografting, and iPSC-derived germ cells have all produced spermatogenesis with sperm that are competent to fertilize oocytes and give rise to viable offspring in mice. Several of these methods have also been translated to larger animal models, including nonhuman primates, indicating a potential for application in the human fertility clinic.

The greatest challenge in the development of stem cell technologies for treatment of human male infertility is the lack of experimental tools for testing the spermatogenic and fertile potential of human cells. This means that human studies cannot be held to the same standard for burden of proof that is required of animal studies. While it is not realistic or possible to demonstrate the fertilization potential of human stem cell-derived gametes, it may be possible to develop systems to test the spermatogenic potential of human cells, such as de novo testicular morphogenesis or engraftment of a decellularized testis. Progress along these lines will provide powerful tools to ensure responsible development and validation of stem cell technologies before they are translated to the male fertility clinic.


Acknowledgments

The authors would like to thank the Scaife Foundation, the Richard King Mellon Foundation, the Magee-Womens Research Institute and Foundation, the Children’s Hospital of Pittsburgh Foundation, and the University of Pittsburgh Departments of Obstetrics, Gynecology & Reproductive Sciences and Urology, which have generously provided funds to support the Fertility Preservation Program in Pittsburgh (http://​www.​mwrif.​org/​220). It is in this context that we have had the opportunity to meet the infertile patients that fuel our passion for fertility research. The Orwig lab is supported by the Magee-Womens Research Institute and Foundation, the Eunice Kennedy Shriver National Institute of Child Health and Human Development grants HD075795 and HD076412, the USA-Israel Binational Science Foundation, and gift funds from Montana State University, Sylvia Bernassoli, and Julie and Michael McMullen.


References




2.

Schover LR. Patient attitudes toward fertility preservation. Pediatr Blood Cancer. 2009;53(2):281–4.PubMed


3.

Lee SJ, Schover LR, Partridge AH, Patrizio P, Wallace WH, Hagerty K, Beck LN, Brennan LV, Oktay K. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol Off J Am Soc Clin Oncol. 2006;24(18):2917–31.


4.

Loren AW, Mangu PB, Beck LN, Brennan L, Magdalinski AJ, Partridge AH, Quinn G, Wallace WH, Oktay K, American Society of Clinical Oncology. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol Off J Am Soc Clin Oncol. 2013;31(19):2500–10.


5.

Ethics Committee of the American Society for Reproductive Medicine. Fertility preservation and reproduction in cancer patients. Fertil Steril. 2005;83(6):1622–28.


6.

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):1214–23.


7.

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.PubMed


8.

Meistrich ML. Male gonadal toxicity. Pediatr Blood Cancer. 2009;53(2):261–6.PubMedPubMedCentral


9.

Levine J, Canada A, Stern CJ. Fertility preservation in adolescents and young adults with cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2010;28(32):4831–41.


10.

Green DM, Kawashima T, Stovall M, Leisenring W, Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison LL. Fertility of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol Off J Am Soc Clin Oncol. 2010;28(2):332–9.


11.

Meistrich ML, Vassilopoulou-Sellin R, Lipshultz LI. Adverse effects of treatment: gonadal dysfunction. In: DeVita VT, Hellman S, Rosenberg SA, editors. Principles and practice of oncology. 7th ed. Philadelphia: Lippincott Williams & Wilkins; 2004. p. 2560–74.


12.

Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Waldron W, Altekruse SF, Kosary CL, Ruhl J, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Chen HS, Feuer EJ, Cronin KA, Edwards BK. SEER cancer statistic review 1975–2008. Bethesda: National Cancer Institute; 2010. [cited 2011], Available from: http://​seer.​cancer.​gov/​csr/​1975_​2008/​.


13.

Wasilewski-Masker K, Seidel KD, Leisenring W, Mertens AC, Shnorhavorian M, Ritenour CW, Stovall M, Green DM, Sklar CA, Armstrong GT, Robison LL, Meacham LR. Male infertility in long-term survivors of pediatric cancer: a report from the childhood cancer survivor study. J Cancer Surviv. 2014;8(3):437–47.PubMedPubMedCentral

Sep 24, 2017 | Posted by in GYNECOLOGY | Comments Off on Male Fertility Preservation: Current Options and Advances in Research

Full access? Get Clinical Tree

Get Clinical Tree app for offline access