Open Access
How to translate text using browser tools
9 April 2022 Male fertility preservation and restoration strategies for patients undergoing gonadotoxic therapies
Kien T.D. Tran, Hanna Valli-Pulaski, Amanda Colvin, Kyle E. Orwig
Author Affiliations +
Abstract

Medical treatments for cancers or other conditions can lead to permanent infertility. Infertility is an insidious disease that impacts not only the ability to have a biological child but also the emotional well-being of the infertile individuals, relationships, finances, and overall health. Therefore, all patients should be educated about the effects of their medical treatments on future fertility and about fertility preservation options. The standard fertility preservation option for adolescent and adult men is sperm cryopreservation. Sperms can be frozen and stored for a long period, thawed at a later date, and used to achieve pregnancy with existing assisted reproductive technologies. However, sperm cryopreservation is not applicable for prepubertal patients who do not yet produce sperm. The only fertility preservation option available to prepubertal boys is testicular tissue cryopreservation. Next-generation technologies are being developed to mature those testicular cells or tissues to produce fertilization-competent sperms. When sperm and testicular tissues are not available for fertility preservation, inducing pluripotent stem cells derived from somatic cells, such as blood or skin, may provide an alternative path to produce sperms through a process call in vitro gametogenesis. This review describes standard and experimental options to preserve male fertility as well as the experimental options to produce functional spermatids or sperms from immature cryopreserved testicular tissues or somatic cells.

Summary Sentence

This review describes standard-of-care and experimental approaches to preserve male fertility and treat male infertility. New stem cell- and tissue-based fertility therapies are on the horizon.

Graphical Abstract

img-z1-16_382.jpg

Introduction

Over the past few decades, childhood cancer survival rates have improved dramatically, up to 88% as recorded in 2018 [1]. Each year in the United States, around 10 000 children between the ages 0 and 14 develop cancers that require them to undergo gonadotoxic treatments, such as chemotherapy and radiation [1]. Patients with nonmalignant conditions (e.g., blood and immune deficiencies and autoimmune disorders) often receive myeloablative conditioning prior to bone marrow transplantation, which is also gonadotoxic [2]. Alkylating chemotherapeutic agents, total body irradiation [3, 4], and gonadal radiation [5] put patients at a significant risk of infertility [58]. Studies show that adult survivors of childhood cancers desire to have children [913]. Therefore, it is important to discuss the reproductive side effects before the initiation of gonadotoxic therapies.

To develop fertility preservation strategies, we first have to understand the process of sperm production, called spermatogenesis. The seminiferous tubules (ST) are the factory of spermatogenesis within the testes (Figure 1). Spermatogonial stem cells (SSCs) are located on the basement membrane of the ST and are the foundation of spermatogenesis. SSCs balance self-renewing and differentiating divisions to maintain continuous sperm production after puberty. When SSCs differentiate, they undergo several transit amplifying mitotic divisions, giving rise sequentially to undifferentiated type A progenitor spermatogonia, differentiating type A spermatogonia, differentiated type B spermatogonia, and spermatocytes. Primary spermatocytes migrate off the seminiferous tubule basement membrane, across the blood–testis barrier to the adluminal compartment of the ST, and complete meiosis to produce secondary spermatocytes and round spermatids. Round spermatids undergo spermiogenesis to produce terminally differentiated spermatozoa that are released into the lumen of the ST. Sertoli cells are the only somatic cell type inside the ST; they are in direct contact with all germ cells in the testis and regulate every step of the spermatogenic lineage development—from stem cells to sperm [14]. On the outer side of the basement membrane, peritubular myoid cells contain abundant actin filaments to support the structural integrity of the tubule [15]. They also possess contractile function to transport spermatozoa and testicular f luid in the tubule as well as secrete growth factors that are important for the regulation of spermatogenesis [1517]. The interstitial space is where Leydig cells, endothelial cells, and macrophages are located. Leydig cells are the source of intratesticular testosterone that is required for spermatogenesis [18]. A clear understanding of testicular structure as well as distinct functions of germ cells and somatic cells can help us develop new fertility restoration therapies.

Prior to gonadotoxic treatments, adult and adolescent males have the option to cryopreserve sperms, which can be used to achieve pregnancy in the future using established assisted reproductive technologies (ARTs), including intrauterine insemination [19], in vitro fertilization (IVF) [20], and intracytoplasmic sperm injection (ICSI) [21]. Unfortunately, sperm cryopreservation is not an option for prepubertal patients who are not yet producing sperm. However, prepubertal testicular tissues do have prospermatogonia or SSCs in their testes that are poised to initiate spermatogenesis at puberty [2224]. Cryopreservation of immature testicular tissues, containing SSCs, may preserve their reproductive potential. We will review testicular cell- and tissue-based methods that are in the research pipeline and may be available in the future to produce sperms from cryopreserved immature testicular tissues. At the end of the article, we will describe a potential alternative, nontesticular source of spermatids or sperms produced from patient-derived induced pluripotent stem cells (iPSCs).

Several cell-based and tissue-based technologies utilizing stem cell potential to restore spermatogenesis are in development. Those technologies include SSC transplantation [2332], de novo testicular morphogenesis [33, 34], autologous testicular tissue grafting/xenografting [3541], testicular tissue culture [4245], and germ cells derived from iPSCs [4650] (Figure 2). Centers in the United States and around the world are cryopreserving testicular tissue for prepubertal boys in anticipation that some or all of these technologies will be available for these patients in the future [22, 5164] (Table 1). Therefore, all patients should be counseled about the effects of their medical treatment on fertility and about options (standard or experimental) to preserve fertility prior to their treatments.

Sperm cryopreservation for male adolescents and young adult cancer patients and worldwide fertility cryopreservation programs

Sperm cryopreservation

Sperm cryopreservation was introduced to the human fertility clinic in the 1950s by Bunge and Sherman as a fertility preservation method for adolescent boys and adult men who face a high risk of infertility due to their medical treatments [65]. Moreover, Szell et al. [66] reported that sperms cryopreserved for 40 years retained fertilization function after thawing to produce a healthy offspring. A study showed that 76% of adolescents and young adults (14–40 years old) expressed the desire to have children in the future [67]. The ability to have biological children is part of the recovery path to normalcy, health, and life fulfillment for childhood cancer survivors [9, 11, 13, 68]. However, a study in the United States reported that only 51% of young cancer patients (14–40 years) were offered sperm cryopreservation, and only 24% of them actually froze sperms prior to initiation of their gonadotoxic therapies [67]. An improvement was seen in a 2005 UK study of 13- to 21-year-old adolescents and young adults, in which they were all offered sperm banking; 67% of them successfully banked their sperms [69]. The most common reason cited for failing to preserve sperms was the lack of information among both patients and medical staff [67, 69]. A study in China showed that more than 70% of cancer patients did not know about the existence of sperm banks, and more than 80% of medical staff were not educated about fertility preservation [70]. Moreover, adolescent patients have lower semen volume and sperm motility compared with adult patients [71]. These differences may or may not hinder the success of ARTs in future fertility restoration. Therefore, education on fertility preservation regarding patients who face a significant infertility risk should be broadly provided to medical staff. This allows medical practitioners to thoroughly inform patients at an early stage of their treatments, such that an appropriate plan for preserving the chance of parenthood can be clearly established.

Worldwide testicular tissue cryopreservation programs

Fertility centers around the world are cryopreserving testicular tissues for prepubertal boys with the anticipation that new reproductive technologies will be available to them in the future [22, 5164, 72, 73]. Table 1 summarizes the efforts of 15 centers that perform testicular tissue or cell cryopreservation for patients who were facing the risk of infertility due to their diseases, medical treatments or other circumstances. Three centers reported samples that included adult patients (older than 18 years old) [22, 52, 58]. The rest included patients with the age range between 0 and 18 years old with oncology as the most common diagnosis [5356, 59, 60, 62, 7478]. Eleven out of 15 centers reported using dimethyl sulfoxide (DMSO) as their choice of cryoprotectant [22, 5456, 60, 62, 7478] and controlled slow freezing as their preferred freezing method [22, 5356, 59, 62, 74, 7678]. Most sites in Table 1 are cryopreserving intact pieces of testicular tissue because this preserves the options for both cell- and tissue-based therapies in the future.

Figure 1.

Spermatogenesis occurs within the ST of the testis that are connected to a common collecting reservoir in the testis space where sperms are deposited before flowing to the epididymis. The intratubular space within the ST includes the basal, adluminal, and lumen compartments. Sertoli cells (yellow cells) are the only somatic cell type that directly interacts with germ cells within the intratubular space. SSCs account for a small proportion of undifferentiated spermatogonia located on the basement membrane within the basal compartment. SSCs are responsible for self-renewing and differentiating divisions to maintain the stem cell pool and continuous sperm production process throughout a man's life. When SSCs differentiate, they undergo several transit amplifying mitotic divisions, giving rise sequentially to differentiating type A spermatogonia (dark purple cells) and differentiated type B spermatogonia (light purple cells). Differentiated type B spermatogonia then give rise to primary spermatocytes (light green cells). During meiosis I, primary spermatocytes lift off the basement membrane and pass through the blood–testis–barrier formed between Sertoli cells to enter the adluminal compartment to produce secondary spermatocytes (dark green cells). Secondary spermatocytes complete meiosis II to form round spermatids (magenta cells). Round spermatids undergo spermiogenesis to form elongating spermatids, elongated spermatids, and finally spermatozoa (gray cells). Spermatozoa are then released into the lumen. Peritubular myoid cells and peritubular macrophages are located on the outside of the basement membrane. The interstitial space is the area between STs where Leydig cells, blood vessels, and interstitial macrophages are located.

img-z3-1_382.jpg

The University of Pittsburgh Medical Center (UPMC) Fertility Preservation Program and coordinated centers have cryopreserved testicular tissues for 517 patients between January 2011 and March 2022 (University of Pittsburgh IRB STUDY19020220; Coordinating Centers IRB STUDY19110083). These include patients at risk of infertility due to medical treatments for cancer, myeloablative conditioning prior to bone marrow transplantation for benign diseases (e.g., sickle cell disease, thalassemia, etc.), hormone modulating treatments for gender dysphoria, and gonadectomy for patients with differences in sexual development (Figure 3). The average age of patients was 7.6 years (standard error of the mean (SEM) = 0.22) and ranged from 3 months to 34 years old. The amount of tissue obtained by biopsy is at the discretion of the surgeons who typically recover as much tissue as possible (to maximize options for downstream use in reproduction) without compromising the function of the remaining testis (usually 10–20% of testicular volume). The average amount of tissue collected was 472.9 mg (SEM = 45.5) and ranged 10.1–9824.8 mg. Our center freezes intact pieces of testicular tissue (9–25 mm3, ∼15 mg) in a medium containing 5% DMSO and 5% serum substitute supplement (84% human serum albumin and 16% human globulins) using the method of controlled slow rate freezing described by Keros and colleagues [74]. We have found that the recovery of transplantable stem cells from cryopreserved testicular tissue pieces is better than the recovery of transplantable stem cells from cryopreserved testicular cell suspensions (data not shown). Moreover, tissue pieces of this size (9–25 mm3) are large enough to suture under the skin for testicular tissue grafting, as previously described [40].

Figure 2.

The schematic diagram of standard and experimental male fertility restoration technologies that have produced offspring in at least one mammalian species (Abbreviations: TESE, testicular sperm extraction; IUI, intrauterine insemination; IVF, in vitro fertilization; ICSI, intracytoplasmic sperm injection; SSC, spermatogonial stem cells; iPSCs, induced pluripotent stem cells; PGCLCs, primordial stem cell-like cells).

img-z4-1_382.jpg

Table 1.

Published reports of testicular tissue cryopreservation.

img-z4-6_382.gif

Figure 3.

Testicular tissue cryopreservation of patients at the UPMC Fertility Preservation Program (March 2011–March 2022) (Abbreviation: CNS, central nervous system; BMT, bone marrow transplantation).

img-z5-1_382.jpg

Counseling young patients and their families about the reproductive side effects of medical treatments and options for fertility preservation can be challenging and requires a multidisciplinary effort. However, the effort is justified because adult survivors of childhood cancers report that fertility is important to them and patients are willing to undergo experimental therapies to preserve the possibility of having a biological child [9, 12, 13, 7982].

Cell-based therapies for male fertility restoration

Spermatogonial stem cell transplantation

Mouse SSC transplantation was first reported by Brinster and colleagues in 1994 [30, 83]. These studies demonstrated that mouse SSCs were able to regenerate complete spermatogenesis when transplanted into the testes of infertile males [30, 83]. The efficiency of the regenerated spermatogenesis depends on the physiology of the recipient testes. Engraftment and initiation of spermatogenesis in neonatal testes is more robust than in adult testes [23]. Ablation of endogenous spermatogenesis by gonadotoxic treatment [25] or genetic mutation [23] reduces competition from endogenous stem cells, thus enhancing donor-derived spermatogenesis.

In addition, functional SSCs can be recovered from frozen and thawed mouse testicular cell suspension after 14 years of storage [84]. Indeed, transplantable spermatogonia have been recovered from frozen and thawed cell suspensions of several species [2330, 83, 8591]. Therefore, it is reasonable that testicular tissue frozen from prepubertal patients prior to gonadotoxic therapies can be thawed in the future to obtain functional SSCs for transplantation to regenerate spermatogenesis. These studies have built a strong foundation for translational medicine, so infertile adult survivors of childhood cancers who have cryopreserved testicular biopsies may have a chance to obtain biological children in the future. Autologous and homologous testicular cell transplantation was originally described over 25 years ago in mice [30, 83]. This is a robust technology that has been replicated in many mammalian species, including rats [26, 27], pigs [85, 86], goats [28, 87], bulls [88], sheep [29], dogs [89], and monkeys [24, 9092] (Table 2). Embryos or offspring born from this technology were reported in mice [23, 25, 30, 93], rats [27], goats [28], and monkeys [24, 92] (Table 2).

Radford and colleagues reported the first autologous testicular cell transplantation in human patients in 1999 [94] and in 2003 [52]. Briefly, testicular cell suspensions were cryopreserved for a total of 12 patients with Hodgkin's disease. Seven of those patients returned to have their frozen and thawed testis cells transplanted back into their testes. The outcomes of those transplants were not reported, but the study provides insights into the motivation of men who were willing to undergo an early-stage experimental procedure for the possibility of having a biologically related child. Homologous species SSC transplantation had only been performed in mice and rats when Radford and colleagues reported the first autologous human testicular cell transplantations in 1999. The technique has now been replicated in numerous mammalian species, demonstrating safety and feasibility that may provide stronger support for translation to the human clinic (Table 2). Human studies must proceed with caution and be performed with regulatory approval in experimental human trials.

Table 2.

Stem cell- and tissue-based fertility therapies currently in the research pipeline.

img-z6-2_382.gif

Continued.

img-z7-2_382.gif

Continued.

img-z8-2_382.gif

Continued.

img-z9-2_382.gif

Continued.

img-z10-2_382.gif

Continued.

img-z11-2_382.gif

Spermatogonial stem cell culture

SSC transplantation is a robust technology that may be ready for translation to the human fertility clinic (e.g., for patients who cryopreserved immature testicular tissues with SSCs when they were young). However, based on our experiences in Pittsburgh and coordinated centers and other published reports [22, 74, 95], the amount of tissue obtained by biopsy from prepubertal patients is small (30–400 mg) and may contain a limited number of SSCs. Therefore, in vitro SSC expansion may be needed prior to transplantation to achieve robust engraftment and regeneration of spermatogenesis.

In rodents, SSCs can be maintained in long-term culture with significant expansion in number, and these SSCs retain their potential to restore spermatogenesis and fertility upon transplantation [96101]. The success of SSC culture in rodents required the development of methods to isolate and enrich SSCs while eliminating testicular somatic cells that could rapidly overwhelm the cultures. In their initial report on mouse SSC culture, Kanatsu-Shinohara and colleagues [99] plated heterogeneous testis cells from newborn mice on gelatin-coated plates. Testicular somatic cells rapidly adhered to the plates while germ cells remained floating and could be sequentially aspirated and replated onto secondary plates to gradually remove somatic cells. In contrast, Hamra and colleagues [96] used a positive selection approach with rat pup testis cells that were plated on laminin. SSCs rapidly adhered to the laminin-coated plates and floating testicular somatic cells could be removed by aspiration. Other studies used fluorescence activated cell sorting (FACS) or magnetic-activated cell sorting (MACS) for the cell surface marker THY1 (CD90) to enrich SSCs and reduce contaminating somatic cells prior to culture [102]. Development of a serum-free, defined medium facilitated the discovery of specific growth factors that were required for SSC maintenance and proliferation in culture. While Kanatsu-Shinohara used glial cell-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), epidermal growth factor (EGF), and leukemia inhibitory factor (LIF) [99], others have shown that GDNF is necessary and sufficient to support rodent SSC expansion in culture. The addition of FGF2 and the soluble GDNF family receptor (GFRα1) can increase the rate of SSC expansion [100, 103]. Finally, feeder cells, such as mouse embryonic fibroblasts are frequently needed to maintain SSCs in culture, although feeder-free culture conditions on substrates such as laminin have been described [104]. SSC transplantation provided a definitive bioassay to confirm the presence and number of functional SSCs in culture [105].

A number of laboratories around the world have reported culturing nonhuman primate and human SSCs (hSSCs) [106128]. Langenstroth and colleagues [128] reported on the isolation, enrichment, and short-term 11-day culture of adult marmoset SSCs. Adult marmoset testis cells were placed in regular tissue culture plates with minimal essential media alpha (MEMα) supplemented with 10% fetal bovine serum (FBS) at 35 °C [128]. No feeder cells or other tissue culture substrates (e.g., laminin) were used and no growth factors were added [128]. Similar to the experience with mouse SSC cultures, they found that when heterogeneous testis cell suspensions were placed in culture, the somatic cells rapidly overwhelmed the culture [128]. Therefore, they established a separation culture system in which floating supernatant cells were aspirated after 24 h and placed in a secondary culture dish [128]. Supernatant cells and attached cells were then cultured separately for 11 days [128]. Then, supernatant cells were collected from the supernatant cultures and attached cells were collected from the attached cultures for colonization potential quantification by primate-to-nude-mouse xenotransplantation [128]. Monkey and hSSCs do not produce complete spermatogenesis after xenotransplantation to recipient mouse testes, but they do recapitulate several functions that are unique to SSCs. They migrate and engraft the basement membrane of recipient mouse ST, proliferate to produce chains and clusters of cells with typical spermatogonial appearance (high nuclear to cytoplasmic ratio and frequently connected by intracytoplasmic bridges), and survive long term [107, 129134]. Using primate-to -nude-mouse xenotransplantation, Langenstroth and colleagues [128] found that the 11-day supernatant culture retained about 60% of the number of colonizing spermatogonia that were originally placed in culture on day 0, and this was 8-fold higher than the colonizing activity from 11-day attached cultures.

More than 20 studies on hSSC culture methods have been published [106126], including three with fetal or prepubertal testicular cells [76, 108, 109]. Many have used differential plating on plastic, lectin, collagen, or gelatin to enrich SSCs and reduce testicular somatic cell contamination. Others have used positive or negative FACS or MACS selection alone or in combination with differential plating. Positive selection markers have included integrin subunit alpha 6 (ITGA6), CD9, G-protein coupled receptor 125 (GPR125), expression of stage-specific embryonic antigen 4 (SSEA4), and epithelial cell adhesion molecule (EpCAM). Negative selection markers include KIT proto-oncogene (cKIT), protein tyrosine phosphatase receptor type C (PTPRC or CD45), and THY1 (reviewed in [135]). It is noteworthy that THY1 is a positive selection marker for mouse spermatogonia but a negative selection marker for transplantable human spermatogonia [102, 119, 135, 136].

Most hSSC culture studies used culture conditions similar to those originally described by Kanatsu-Shinohara and colleagues in mice, including StemPro-34 medium supplemented with various combinations of GDNF, FGF2, EGF and LIF. Some of those studies reported significant expansion of hSSC numbers in culture [76, 107, 137], while others reported a rapid decline in hSSC numbers using the same conditions [118, 120, 122, 125]. These disparate outcomes may be explained by differences in starting cell populations, culture conditions, and approaches that were employed to identify and quantify hSSCs in culture (ranging from counting total cells in culture to quantifying xenotransplantation colonizing events). Therefore, there is no consensus “best method” for culturing hSSCs that has been independently replicated in different laboratories and no consensus on best methods to identify and quantify bona fide hSSCs in culture. Nonetheless, a number of research groups have reported hSSC survival and expansion for periods ranging from 1 week to 6 months [106126]. While the results for hSSC culture are promising, they are challenged by the inability to test the full spermatogenic potential of these cells in xenotransplantation; and autologous or homologous transplantation in humans is currently not feasible.

De novo testicular morphogenesis

De novo testicular morphogenesis is defined as the reconstruction of testicular architecture and spermatogenesis from a heterogenous testicular cell suspension to replicate the in vivo cell-to-cell interactions and functions of germ cells and somatic cells. De novo testicular morphogenesis can be performed entirely in culture or by subcutaneous xenotransplantation into animal recipients (Figure 2, Table 2). This approach can be used to dissect, manipulate, or modify the component parts of a complex tissue to learn the impact on testicular cell development or function. The approach may also be used to eliminate unwanted and unnecessary components, such as contaminating malignant cells, which could disrupt development or be unsafe for therapy. Cells from a single testis can be disaggregated to produce many organoids that can be used for high throughput toxicity testing or novel compound screening. De novo testicular morphogenesis has been described in rodents as well as farm animals, nonhuman primates, and humans [33, 138149].

Methods for de novo testicular morphogenesis, in vitro, were first described in rodents (Figure 2, Table 2). Yokonishi and colleagues [150] pioneered the technique by producing organoids from neonatal mouse testis cells in V-bottomed 96-well plates. After 2 days, organoids were collected and cultured at the gas–liquid interface by placing on agarose gel islands half-soaked in culture medium that was supplemented with knockout serum replacement (KSR). Seminiferous tubule-like structures were apparent by two weeks in culture. Sertoli cells and germ cells were arranged inside ST and Leydig cells were located in the space between the de novo-formed tubules. Spermatocytes and spermatids were observed by 60 days in culture. Zhang and colleagues used a variation of the gas–liquid interface culture system; testicular cell/collagen aggregates were deposited on a nucleopore filter that was then floated on culture medium [151]. Seminiferous tubule-like structures contained spermatogonia and spermatocytes [151]. Sertoli cell differentiation occurred only when KSR was included in the medium. Baert and colleagues [152] seeded neonatal mouse testis cells on an alginate lattice scaffold alone or embedded with testicular interstitial cells. The testicular cells formed organoids within the lattice but did not self-organize into ST [152]. Nonetheless germ cell differentiation to the elongated spermatid stage was reported on both the cell-free and cell-laden scaffold [152]. Donor age is a factor. Alves-Lopes and colleagues [139] reported seminiferous tubule formation from 5- to 8-day-old and 20-day-old rat pup testis cells sandwiched in a three-layer Matrigel culture system but not from 60-day old rat testicular cells. To our knowledge, no offspring have been produced from any mouse in vitro de novo testicular morphogenesis approach.

Huleihel and coworkers [153] cultured heterogeneous testicular cell suspensions from prepubertal rhesus macaques in 3-dimensional (3D) methylcellulose or soft agar culture systems. Cells aggregated to produce colonies, including CAMP-responsive element modulator-positive (CREM)+ and acrosin (ACR) + spermatids, but did not reconstitute ST [153]. Sakib and colleagues [149] reported a microwell aggregation approach to producing 3D testicular organoids from neonatal or prepubertal testicular cell suspensions of mice, pigs, macaques, and humans. In all species, the organoids exhibited an inverted seminiferous tubule architecture with germ cells and Sertoli cells on the outside and other somatic cells on the inside of the tubules [149].

Two studies reported adult and pubertal human testicular organoids formation in human testicular extracellular matrix (htECM) [146, 147]. Pendergraft and colleagues [146] used a hanging-drop method to induce organoid formation from cultured adult human spermatogonia mixed with immortalized human Sertoli and Leydig cells suspended in a hydro-gel of htECM. Baert and colleagues [147] seeded adult or pubertal heterogeneous human testis cell suspensions onto a 3-dimensional htECM scaffold that retained the tubular architecture. Both approaches led to the production of organoids including germ cells and somatic cells but neither approach produced seminiferous tubule-like structures [146, 147]. Furthermore, Baert and colleagues [147] reported that human testis cell organoids formed with or without the benefit of the scaffold. Similar to results in rodents, age appears to be a factor in the formation of seminiferous tubule-like structures. Oliver and colleagues [154] sandwiched fresh human fetal testicular cells in a three-layer Matrigel gradient system. Seminiferous cord-like structures appeared within 7 days with a clear basement membrane separating Sertoli cells on the inside from peritubular myoid cells and Leydig cells on the outside [154]. However, although DEAD-box helicase 4 (DDX4 or VASA) + prospermatogonia were observed in the preculture 8-week-old fetal testicular tissues, they disappeared over 7 and 14 days of culture [154]. In summary, fetal or prepubertal testis cell suspensions (mouse, rat, or human) can self-organize to produce seminiferous cord- or seminiferous tubule-like structures that do not appear when pubertal or adult cells are used [154]. Meiotic or postmeiotic cells were occasionally observed, and this does not appear to depend on the formation of tubules [154]. To our knowledge, the function of haploid cells produced by in vitro de novo testicular morphogenesis approaches has not been tested [154].

The second de novo morphogenesis approach is to inject the heterogenous testicular cell suspension under the skin of a mouse host (Figure 2, Table 2). Pelleted testicular cells from mice [33, 34], rats [33, 138, 155, 156], pigs [33, 141, 142], sheep [143], and monkeys [144] have been transplanted into mouse hosts and self-organized in vivo to form seminiferous tubule-like structures (Table 2). Studies from Kita et al. [33] and Zhang et al. [151] confirmed that cells from different sources could be mixed to test their importance in tubule formation or spermatogenesis and that haploid mouse cells produced in de novo reconstructed tubules were competent to fertilize and produce live offspring. Similar seminiferous tubule-like structures containing spermatogenesis with haploid germ cells have been reported from pig [141, 142] and sheep [143] by transplanting neonatal testis cells under the skin of immune-compromised mice. Spermatogenic efficiency was improved by addition of Matrigel but not by introduction of an enriched germ cell fraction into a heterogeneous pig testis cell suspension [142]. The fertilization potential of pig and sheep spermatids in de novo-formed tubules was not tested in those studies [141143]. Neonatal marmoset monkey testis cells self-organized into seminiferous cord-like structures without lumens after subcutaneous xenotransplantation to mouse hosts [144]. The cords contained Sertoli cells and germ cells, but complete spermatogenesis was not observed and there was no evidence of meiotic or postmeiotic cells [144]. Cords were not formed from juvenile or adult marmoset testis cells [144]. To our knowledge, in vivo de novo testicular morphogenesis has not been attempted with human cells. If human testis cells can produce results similar to those reported above for mice, pigs and sheep, de novo-derived haploid germ cells could theoretically be used for clinical applications. This may raise concerns about xenobiotics that could be circumvented if de novo testicular morphogenesis from humans can be achieved in organoid cultures described above. However, this has not occurred yet.

Testicular tissue-based therapies for male fertility restoration

Testicular tissue grafting

For testicular tissue grafting, SSCs are maintained within intact pieces of tissue, preserving the original architecture of the ST, extracellular matrix, and testicular somatic cells with associated paracrine signaling. Tissue grafting is performed by implanting testicular tissue pieces at an orthotopic (e.g., scrotum) or ectopic (e.g., skin) site in the recipient animals (autologous, allogenic, or xenogeneic recipients) with or without exogenous matrices or hormones [3541, 75, 143, 157188] (Table 2). The recipients are usually castrated to stimulate the gonadotropic hormone secretion to promote maturation of the grafted tissues [3541, 75, 143, 157188]. The overall goal is not to restore fertility since grafted testicular tissues are not connected to the excurrent duct system. The goal is to mature the grafted tissue to produce sperm that can be used to fertilize eggs by ICSI and produce offspring. Honaramooz and colleagues [85] reported that immature testicular tissues from mice, pigs, and goats could be transplanted under the back skin of immune-deficient nude mice and matured to produce sperm. Two groups later reported that sperms from fresh [35] or cryopreserved [37] immature mouse testicular tissue grafts were competent to fertilize mouse eggs and produce offspring. Immature testicular tissue grafting (autologous or allogeneic) or xenografting is a robust technology that has been replicated in mice [3537, 157, 158, 160], hamsters [157], rabbits [37], dogs [165], cats [162164], horses [168, 176], pigs [36, 38, 166, 167, 169, 170], bulls [166, 171175], goats [36], sheep [143, 166], and monkeys [39, 41, 168, 177179] with the production of spermatids or spermatozoa (Table 2). Function of graft-derived sperm has been tested by fertilization, and pre-implantation embryo development up to blastocyst stage and/or production of viable offspring were reported in mice [3537], rabbits [37], pigs [38], goats [36], and rhesus macaques [3941] (Table 2). Only immature (fetal, neonatal, pre-pubertal) testicular tissues were able to survive and undergo complete spermatogenesis; whereas adult tissues gradually lost differentiated germ cells and degenerated [75, 158, 161, 163165, 168, 175, 176] (Table 2).

In addition, the efficiency of inducing complete spermatogenesis varies widely among species. In studies of pigs, goats, sheep, and monkeys, at least 50% of tubules contained complete spermatogenesis post xenografting [36, 40, 166]. However, only less than 10% of tubules contained complete spermatogenesis in horse and bull xenografts [171, 173, 174, 176]. Graft retrieval time also greatly affects spermatogenesis efficiency. In mice, grafted tissues recovered after 120 days yielded about four times more tubules with complete spermatogenesis compared with grafts recovered before 120 days [158]. Another mouse study also showed increasing percentage of tubules containing complete spermatogenesis from ∼20 to ∼55% in grafts retrieved at later timepoints between 4–16 weeks post-grafting [36]. However, the number of surviving spermatogonia in pre-pubertal human testicular tissues xenografted to mice decreased over time and complete spermatogenesis was not observed [158]. Honaramooz et al. [189] reported that the fertilization potential of sperm produced in the pig-to-mouse xenografts decreased over time and suggested that this might be due to lack of excurrent ducts to remove older sperm.

Live birth outcomes from autologous, allogeneic, or xenograft experiments suggest that these approaches may have application for maturing prepubertal testicular tissues that were frozen for patients prior to gonadotoxic therapies (Table 2). Furthermore, xenografting of human tissues to mice or other animal hosts may circumvent the risk of reintroducing malignant cells to patient survivors. Spermatogonia survival from fresh and cryopreserved prepubertal grafts were reported at various timepoints, ranging from days to months [75, 158, 159, 182, 183, 187, 190]. The longest observation time was 9 months with orthotopic xenografts retaining spermatogonia survival better than the ectopic grafts [75, 183]. A few studies have reported the initiation of spermatogenesis up to early spermatocytes from fresh or cryopreserved immature human orthotopic or ectopic xenografts [184186, 188]. However, complete spermatogenesis from xenografting human testicular tissues into mice has not yet been achieved [75, 158, 159, 180188]. Recipient choice may be a factor that should be tested in future studies, including porcine or nonhuman primate species, which are more closely related to humans.

Testicular tissue organ culture

Testicular tissue organ culture (TTOC) is an ex vivo system that utilizes small testicular tissue fragments in culture initiation to preserve the original 3D organization of germ cells and somatic cells. TTOC aims to induce in vitro maturation of germ cell and somatic cell compartments. This approach may help circumvent the risks of malignant contamination associated with autologous cell/tissue transplantation and xenobiotic transmission in xenogeneic transplantation. Several TTOC systems have been described in the past two decades culturing immature testicular tissues of mice, rats, monkeys, and humans (Figure 2, Table 2). In 2006, Livera and colleagues [191] cultured fetal mouse or rat testes on floating filter papers, but could not achieve in vitro maturation of prospermatogonia. Since 2010, several studies have reported spermatid or spermatozoa production from cultures of fresh and/or cryopreserved neonatal mouse testicular tissues in either the gas–liquid interphase systems [43, 44, 192201] or microfluidic systems [42, 199, 202] (Figure 2, Table 2). In the conventional gas–liquid interphase system, 0.75–3 mm3 pieces of tissue are cultured on an agarose gel island (0.25–1.5% w/v) half submerged in medium. Neonatal mouse tissues yielded higher in vitro spermatogenesis efficiency compared with adult testes [194]. When starting with immature mouse testicular tissues, the in vitro spermatogenesis timeline was very approximal to in vivo timeline. Round spermatids emerged by day 21 and spermatozoa were observed by day 35 [4244, 192202]. Spermatids or spermatozoa isolated from dissociated cultured tissues were used for fertilization via ICSI from which healthy offspring were reported [4244]. Reda and colleagues [203] reported round spermatids but no spermatozoa in cultures of neonatal rat testicular tissue using the agarose gel system, and the fertilization potential of the round spermatids was not tested.

The testicular tissues cultured on agarose gel tend to mound up after a few days leading to necrosis in the central part of the tissue mound, and spermatogenesis as well as tissue integrity declined after 35 days in culture [34]. Komeya and colleagues invented several microfluidic systems that confine tissues to a small space to prevent mounding of tubules and ensure that all tubules have direct access to the air interface on one side and the medium interface on the other side for exchange of nutrients and waste with medium that flows past the tissue at controlled rates [42, 199, 202]. The pump-driven microfluidic device could maintain continuous spermatogenesis to produce functional spermatozoa for up to six months in culture with more than 90% of tubules containing haploid cells. Healthy offspring were produced from both round spermatid injection (ROSI) and ICSI from spermatozoa [42]. Similarly, pumpless microfluidic devices maintained tubules with spermatogenesis up to the round spermatid stage for at least four months [199, 202]. However, spermatozoa were not observed with the pumpless microfluidics devices, so some optimization of media flow rates or other conditions may be needed.

There are only a few studies describing human TTOC. Jorgensen and colleges were able to preserve the architecture of human fetal gonads and adult testis tissues for up to 2 weeks using a hanging-drop culture system [204, 205]. They reported that fetal germ cells proliferated for at least 2 weeks in culture, and apoptosis was not increased during this time [205]. Differentiated germ cells in adult testicular tissues did not survive past 7–10 days in culture, and proliferation of germ cells also significantly reduced [204]. Yuan et al. cultured fresh gonadal tissue pieces, obtained from 12–19-week-old male fetuses, on agarose gel islands; they reported the presence of round spermatids in 1-month cultures [45]. These in-vitro-derived round spermatids were extracted and used to fertilize human oocytes via ROSI, from which they obtained human blastocysts with normal karyotype and maternal-paternal genetic materials [45]. Moreover, de Michele and colleagues [206] cultured frozen-and-thawed pre-pubertal human testicular tissue pieces in a transwell system and observed round spermatids (1–2 haploid cells per seminiferous tubule cross-section) from 16 to 139 days in culture.

Some of the key medium supplements, including retinoic acid (RA) and follicle stimulating hormone (FSH), were analyzed more in depth in a few studies. RA plays a crucial role in initiating spermatogenesis, but its effects may be age dependent. RA supplement enhanced in vitro spermatogenesis in neonatal mouse testis tissues [195, 197] but was found to be detrimental to fetal human [45, 207] or mouse [195, 208] testicular tissues in culture. FSH and luteinizing hormone (LH) play important roles in maturation of Sertoli cells and Leydig cells, respectively, which are required for germ cell development. Medrano and colleagues [209] reported that FSH promoted Sertoli cell differentiation ((androgen receptor (AR) expression) in prepubertal human testicular tissue cultures. Indeed, FSH and LH promoted in vitro germ cell survival and differentiation in both mouse [195] and human testis tissues [209]. Medrano et al. [209] also reported that replacing fetal bovine serum with KSR improved Sertoli and Leydig cell differentiation as well as the number of undifferentiated embryonic cell transcription factor 1 (UTF1)+ undifferentiated spermatogonia and synaptonemal complex protein 3 (SYCP3)+ premeiotic spermatogonia. Furthermore, Medrano et al. [209] found that sex determining region (SRY)-box transcription factor 9 (SOX9)+ Sertoli and undifferentiated embryonic cell transcription factor 1 (UTF1)+ undifferentiated spermatogonia numbers were higher in cultures maintained a 34 °C (approximating the temperature in the scrotum) than cultures maintained at 37 °C. In a mouse TTOC study, 34 °C cultures yielded higher spermatogenesis efficiency than 32 °C or 37 °C cultures (40.9% at 32 °C, 82.6% at 34 °C, and 0% at 37 °C) [192]. In 2017, de Michele and colleagues [210] reported that when cryopreserved pre-pubertal human testicular tissues were cultured for 139 days in medium supplemented with 50 IU/L FSH, somatic cell maturation, including Sertoli cells and Leydig cells, was observed. Spermatogonia were still present but significantly declined in number after two weeks in culture, and spermatogonia differentiation was not observed in their study [210]. In 2018, the same group reported that a lower concentration of FSH (5 IU/L) supported Sertoli cell and Leydig cell maturation as well as germ cell differentiation to round spermatids [206]. The number of round spermatids was quite low, (1 spermatid/tubule), which precluded further genetic/epigenetic or functional characterization. Nonetheless, this was the most advanced germ cell stage obtained from cultures of frozen-and-thawed pre-pubertal human testicular tissues. As de Michelle and colleagues [206] still encountered a significant decrease of spermatogonia as soon as 2 weeks after culture initiation, further investigations are guaranteed to enhance the survival and self-renewal of spermatogonia to avoid stem cell exhaustion to differentiation.

Induced pluripotent stem cell-based methods for male fertility restoration

For patients who did not cryopreserve sperm or testicular tissues prior to gonadotoxic treatments, it may be possible in the future to generate germ cells from iPSCs derived from any cell type in the body (e.g., skin fibroblasts, blood lymphocytes). The proof in concept studies were performed using pluripotent mouse embryonic stem cells (ESCs) and mouse iPSCs. The process of in vitro gametogenesis (ESCs or iPSCs → primordial germ cell-like cells (PGCLCs) → spermatogonia and/or sperm) recapitulates germ cell development in vivo. During in vivo development, around day 7–8 in mouse embryonic development or week 2–3 in human embryonic development, primordial germ cells (PGCs) are specified in the epiblast stage embryo and segregate to the yolk sac endoderm [211, 212]. PGCs then proliferate and migrate into the embryo with the hindgut endoderm around mouse embryonic day 10.5 or human embryonic week 4 [211, 212]. On mouse embryonic day 12.5 or human embryonic week 5, PGCs arrive at the genital ridges where they continue proliferation in the indifferent gonads [191, 192]. Sex determination is driven by differentiation of gonadal somatic cells at mouse embryonic day 15.5 or human embryonic week 5–6, forming the embryonic testes or embryonic ovaries [211, 212]. In vivo germ cell development provides a blueprint for how to produce germ cells and gametes in vitro, using the appropriate developmental milestones including progression through meiosis to produce haploid germ cells, evidence of recombination, epigenetic reprogramming, euploid chromosome content and production of healthy euploid progeny [213].

Previous studies presented several approaches to differentiate ESCs and iPSCs, derived from mice, monkeys, or humans, into the germ cell lineage (Figure 4, Table 2). In mouse studies, ESCs or iPSCs are differentiated over two days into epiblast stem cells (EpiSCs) or epiblast-like cells (EpiLCs), respectively [4650]. These EpiSCs/EpiLCs can be further differentiated over 3–4 days into PGCLCs as floating aggregates [4650]. Putative mouse PGCLCs can be FACS sorted based on expression of PR/SET domain 1 (Blimp1)-Venus/Stella-Cyan transgenes or cell surface markers SSEA1 and INTEGRINβ3 for transplantation into mouse pup testes to regenerate spermatogenesis and produce offspring [46, 47]. Alternatively, PGCLCs can be further differentiated in vitro to more advanced germ cell stages, including germline stem cell-like cells (GSCLCs) and/or spermatids that are competent to fertilize and produce offspring [4850] (Figure 4, Table 2). PGCLCs showed global transcription profiles that were similar to mouse PGCs on embryonic day 9.5 [47]. PGCLCs could only regenerate complete spermatogenesis to produce functional spermatozoa when transplanted into infertile neonatal mouse recipients, but not adult recipients. The authors proposed that the neonatal testes provided that necessary developmental cues to support the further development of PGCLCs and that these were lacking in the adult testis [46, 47].

If it is true that PGCLCs can only regenerate spermatogenesis when transplanted into neonatal recipients, the application in the human clinic is not obvious. This limitation was partially addressed by Ishikura and colleagues, who developed a method to further differentiate PGCLCs into GSCLCs in a reconstituted testis (rTestis) by mixing PGCLCs with fetal testicular somatic cells (FTSCs) in a transwell plate [49]. The resulting GSCLCs could be maintained in adherent cultures, similar to mouse SSC cultures [102, 214]. GSCLCs could also be transplanted to regenerate complete spermatogenesis in adult recipients, including sperm that were competent to fertilize and produce offspring [50]. A similar rTestis approach recently described mixing human PGCLCs (hPGCLCs) with fetal mouse gonadal cells (xenogeneic, reconstituted (xr) Testis) that resulted in the appearance of DDX4+, piwi-like RNA-mediated gene silencing 4 (PIWIL4)+ prospermatogonia [215]. Zhou and colleagues [48] reported a transplantation-free approach by mixing PGCLCs with FTSCs to form aggregates and continue to culture for 2 more weeks with RA, bone morphogenetic proteins (BMPs), Activin A, FSH, bovine pituitary extract (BPE), and testosterone (Figure 4). Spermatids were present in these cultured aggregates and isolated for ROSI that led to production of healthy, fertile offspring [48]. These studies may have important implications for the human fertility clinic, but they are early stage and need to be independently replicated in other laboratories and in other species. These approaches are further complicated be the requirement for FTSCs, which would be difficult or impossible to obtain for human application. An alternative approach may be the in vitro derivation of the necessary fetal gonadal somatic cell types from pluripotent stem cells, as described by Hayashi and colleagues [216] for the derivation of fetal ovarian somatic-like cells; or direct differentiation of patient somatic cells (e.g., skin fibroblasts) into FTSCs such as Sertoli cells, as described by Buganim and colleagues [217].

Multiple researchers attempted to differentiate nonhuman primate ESCs and iPSCs into the germ cell lineage in vitro [218220] (Figure 4, Table 2). Cynomolgus monkey ESCs were established and used for in vitro differentiation into embryoid bodies (EBs) [219]. EBs started expressing VASA at day 4 post differentiation, suggesting the presence of prospermatogonia [219]. At day 16, synaptonemal complex protein 3 (SCP3) and DNA meiotic recombinase 1 (DMC1) transcriptions were detected, suggesting the presence of meiotic cells [219]. Yamauchi and colleagues [220] reported that addition of bone morphogenetic protein 4 (BMP4) to differentiation culture of Cynomolgus ESCs promoted earlier gene transcription of synaptonemal complex protein 1 (SYP1) in the EBs. In a separate study, Sosa and colleagues [218] examined the capacity of Rhesus iPSC to differentiate into incipient mesoderm-like cells (iMeLCs) and then rPGCLCs in culture (Figure 4). rPGCLCs were then transplanted into adult mouse or Rhesus testes to determine if the in vivo somatic environment could support germ cell engraftment and spermatogenesis [218]. Transplanted rPGCLCs into the adult gonadal niche of either recipient species differentiated into colonies of MAGE family member A4 (MAGEA4)+/Enolase 2 (ENO2)-/VASA+ germ cells and did not differentiate to produce sperm in adult mouse or monkey recipient testes [218]. These results indicated that rPGCLCs acquired transplantation potential (engraftment, proliferation, and survival for long-term upon transplantation) but not spermatogenic potential [218]. Adult Rhesus testicular environment did not provide proper biological cues for to support spermatogenesis from transplanted rPGCLCs [218]. Like previous mouse studies, transplantation into neonatal Rhesus could provide the necessary environment to support spermatogenesis from transplanted PGCLCs. However, those experiments will be expensive and complex, and the justification is questionable since transplantation to neonates in the human clinic is not likely.

Figure 4.

Illustration of various in vitro gametogenesis approaches using iPSCs of mice, monkeys, and humans. (Abbreviation: iPSCs, induced pluripotent stem cells; iMeLCs, incipient mesoderm-like cells; PGCLCs, primordial stem cell-like cells; rTestis, reconstituted testis; GSCLCs, germline stem cell-like cells; SLCs, spermatid-like cells; xrTestis, xenogeneic reconstituted testis).

img-z16-1_382.jpg

A number of laboratories have applied various differentiation conditions (matrices and medium compositions) to differentiate human ESCs (hESCs) and/or human iPSCs (hiPSCs) into hPGCLCs [221225] (Table 2). Clark and colleagues [226] initially reported that hESCs could spontaneously differentiate into VASA+ germ cells in EBs, but the number of VASA+ cells was low. The same group later showed that BMP4 increased the expression of germ cell-specific markers (VASA, SYCP3) during differentiation of hESCs in EBs compared with EB cultures without BMP4 supplement. BMP7 and BMP8b had modest additive effects [222, 227]. Two groups reported a stepwise differentiation of hiPSCs into iMeLCs as an intermediate stage to enhance the efficiency of hPGCLC derivation [215, 224]. EPCAM and INTEGRINα6 were used as surface markers to identify and isolate hPGCLCs [224]. In human PGC specification, sex determining region (SRY)-box transcription factor 17 (SOX17) regulates BLIMP1 to suppress the expression of endoderm and mesoderm genes as well as acting as a key regulator in hPGCLC specification [225].

In a series of studies, Durruthy and colleagues [228] tested the hypothesis that the in vivo environment of mouse ST could direct the differentiation of human pluripotent stem cells into the germ cell lineage. They found that ESCs and iPSCs reprogrammed with the typical Yamanaka OSKM (Oct4, Sox2, Klf4, and c-Myc) factors produced large masses similar to embryonal carcinoma cells and yolk sac tumors. In contrast when VASA was added to the reprogramming mix (OSKMV) (Oct4, Sox2, Klf4, c-Myc, and Vasa), the resulting iPSCs did not form large masses and left the mouse testis structure intact. The authors observed clusters of deleted in azoospermia-like (DAZL)+/VASA+/UTF1+/GFRa1+ germ cell-like cells in the ST [228230]. Several groups have reported in vitro differentiation of hESCs and hiPSCs into haploid cells, indicated by Acrosin expression and a 1 N (haploid) peak observed by Hoechst or propidium iodide (PI) staining followed by flow cytometry [221223, 231]. Functional testing of human haploid cells is important but challenged by federal and state regulations and the need to secure funding from non-federal sources.

Conclusions

Cancer survivors are concerned about the side effects of medical treatments on their future fertility, and many of them have expressed their desire to have children later in life [232]. Prior to gonadotoxic treatments, adolescent and adult men can cryopreserve semen samples containing sperm that can be used to achieve pregnancies with their partners using existing ARTs (Figure 2). However, this is not an option for prepubertal male patients who are not yet producing mature gametes. Many clinics around the world are cryopreserving testicular tissues for these prepubertal patients (Table 1) in anticipation that next-generation technologies will be fully developed in the future to mature those tissues and produce sperm, in vivo or in vitro (Figure 2 and Table 2). This review has described in detail on the progress of each technology being developed. Every technology has its strengths and drawbacks; therefore, it is reasonable to simultaneously invest efforts into several technologies to accommodate various patient circumstances. Some of these techniques, including autologous testicular cell transplantation and testicular tissue grafting, are mature and may be ready for translation to the human clinic (Table 2). This should be done only with approval of an institutional review board. Other technologies reviewed in this chapter (e.g., de novo testicular morphogenesis, TTOC, and iPSC-derived germ cells) are in earlier stages of development (Figures 2 and 4, Table 2). Louise Brown was born over 40 years ago as the world's first baby who was conceived by IVF [20]. Louise was possible because her parents were able to produce eggs and sperms that were combined outside the body to achieve fertilization. Advanced reproductive technologies are in the research pipeline that may allow men/boys without sperms or women/girls without eggs to have biologically related children.

Authors' contributions

K.T.D.T. prepared the manuscript. K.E.O. reviewed and edited the manuscript. H.V.-P. and A.C. provided patient data of the Fertility Preservation Program in Pittsburgh.

Acknowledgment

The authors 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 ( https://fertilitypreservationpittsburgh.org/). It is in this context that we have had the opportunity to meet the infertile patients that fuels our passion for fertility research.

Conflict of interest

There are no conflicts of interest to disclose for any of the authors.

Data availability

The data underlying this article are available in the article and in its online supplementary material.

References

1.

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin 2018; 68:7–30. Google Scholar

2.

Gratwohl A, Baldomero H, Aljurf M, Pasquini MC, Bouzas LF, Yoshimi A, Szer J, Lipton J, Schwendener A, Gratwohl M, Frauendorfer K, Niederwieser D et al. Hematopoietic stem cell transplantation: a global perspective. JAMA 2010; 303: 1617–1624. Google Scholar

3.

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; 6:209–218. Google Scholar

4.

Wallace WH. Oncofertility and preservation of reproductive capacity in children and young adults. Cancer 2011; 117: 2301–2310. Google Scholar

5.

Lambertini M, Del Mastro L, Pescio MC, Andersen CY, Azim HA, Peccatori FA, Costa M, Revelli A, Salvagno F, Gennari A, Ubaldi FM, La Sala GB et al. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med 2016; 14:1. Google Scholar

6.

Green DM, Kawashima T, Stovall M, Leisenring W, Sklar CA, Mertens AC, Donaldson SS, Byrne J, Robison LL. Fertility of male survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol 2010; 28: 332–339. Google Scholar

7.

Green DM, Nolan VG, Goodman PJ, Whitton JA, Srivastava D, Leisenring WM, Neglia JP, Sklar CA, Kaste SC, Hudson MM, Diller LR, Stovall M et al. The cyclophosphamide equivalent dose as an approach for quantifying alkylating agent exposure: a report from the Childhood Cancer Survivor Study. Pediatr Blood Cancer 2014; 61:53–67. Google Scholar

8.

Gordon W, Siegmund K, Stanisic TH, McKnight B, Harris IT, Carroll PR, Paradelo JC, Meyers FJ, Chapman RA, Meyskens FL. A study of reproductive function in patients with seminoma treated with radiotherapy and orchidectomy: (SWOG-8711). Int J Radiat Oncol Biol Phys 1997; 38:83–94. Google Scholar

9.

Nieman CL, Kinahan KE, Yount SE, Rosenbloom SK, Yost KJ, Hahn EA, Volpe T, Dilley KJ, Zoloth L, Woodruff TK. Fertility preservation and adolescent cancer patients: lessons from adult survivors of childhood cancer and their parents. Cancer Treat Res 2007; 138:201–217. Google Scholar

10.

Zebrack BJ, Casillas J, Nohr L, Adams H, Zeltzer LK. Fertility issues for young adult survivors of childhood cancer. Psychooncology 2004; 13:689–699. Google Scholar

11.

Schover LR. Motivation for parenthood after cancer: a review. J Natl Cancer Inst Monogr 2005; 2005:2–5. Google Scholar

12.

Kinahan KE, Didwania A, Nieman CL. Childhood cancer: fertility and psychosocial implications. Cancer Treat Res 2007; 138: 191–200. Google Scholar

13.

Ellis SJ, Wakefield CE, McLoone JK, Robertson EG, Cohn RJ. Fertility concerns among child and adolescent cancer survivors and their parents: a qualitative analysis. J Psychosoc Oncol 2016; 34:347–362. Google Scholar

14.

Chen SR, Liu YX. Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. Reproduction 2015; 149:R159–R167. Google Scholar

15.

Maekawa M, Kamimura K, Nagano T. Peritubular myoid cells in the testis: their structure and function. Arch Histol Cytol 1996; 59:1–13. Google Scholar

16.

Chen LY, Brown PR, Willis WB, Eddy EM. Peritubular myoid cells participate in male mouse spermatogonial stem cell maintenance. Endocrinology 2014; 155:4964–4974. Google Scholar

17.

Thompson RP, Nilsson EE, Skinner MK. Peritubular myoid cells in testis. In: Skinner MK (ed.), Reference Module in Biomedical Sciences , 2nd ed. Cambridge, MA: Academic Press; 2018: 42–46. Google Scholar

18.

Teerds KJ, Huhtaniemi IT. Morphological and functional maturation of Leydig cells: from rodent models to primates. Hum Reprod Update 2015; 21:310–328. Google Scholar

19.

Agarwal A, Allamaneni SR. Artificial insemination. In: Falcone T, Hurd W (eds.), Clinical Reproductive Medicine and Surgery. Philadelphia: Elsevier; 2007: 539–548. Google Scholar

20.

Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet 1978; 2:366. Google Scholar

21.

Palermo G, Joris H, Devroey P, Van Steirteghem AC. Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte. Lancet 1992; 340:17–18. Google Scholar

22.

Valli-Pulaski H, Peters KA, Gassei K, Steimer SR, Sukhwani M, Hermann BP, Dwomor L, David S, Fayomi AP, Munyoki SK, Chu T, Chaudhry R et al. Testicular tissue cryopreservation: 8 years of experience from a coordinated network of academic centers. Hum Reprod 2019; 34:966–977. Google Scholar

23.

Shinohara T, Orwig KE, Avarbock MR, Brinster RL. Remodeling of the postnatal mouse testis is accompanied by dramatic changes in stem cell number and niche accessibility. Proc Natl Acad Sci U S A 2001; 98:6186–6191. Google Scholar

24.

Hermann BP, Sukhwani M, Winkler F, Pascarella JN, Peters KA, Sheng Y, Valli H, Rodriguez M, Ezzelarab M, Dargo G, Peterson K, Masterson K et al. Spermatogonial stem cell transplantation into rhesus testes regenerates spermatogenesis producing functional sperm. Cell Stem Cell 2012; 11:715–726. Google Scholar

25.

Brinster CJ, Ryu BY, Avarbock MR, Karagenc L, Brinster RL, Orwig KE. Restoration of fertility by germ cell transplantation requires effective recipient preparation. Biol Reprod 2003; 69: 412–420. Google Scholar

26.

Ogawa T, Dobrinski I, Brinster RL. Recipient preparation is critical for spermatogonial transplantation in the rat. Tissue Cell 1999; 31:461–472. Google Scholar

27.

Hamra FK, Gatlin J, Chapman KM, Grellhesl DM, Garcia JV, Hammer RE, Garbers DL. Production of transgenic rats by lentiviral transduction of male germ-line stem cells. Proc Natl Acad Sci U S A 2002; 99:14931–14936. Google Scholar

28.

Honaramooz A, Behboodi E, Megee SO, Overton SA, Galantino-Homer H, Echelard Y, Dobrinski I. Fertility and germline transmission of donor haplotype following germ cell transplantation in immunocompetent goats. Biol Reprod 2003; 69:1260–1264. Google Scholar

29.

Herrid M, Olejnik J, Jackson M, Suchowerska N, Stockwell S, Davey R, Hutton K, Hope S, Hill JR. Irradiation enhances the efficiency of testicular germ cell transplantation in sheep. Biol Reprod 2009; 81:898–905. Google Scholar

30.

Brinster RL, Avarbock MR. Germline transmission of donor haplotype following spermatogonial transplantation. Proc Natl Acad Sci U S A 1994; 91:11303–11307. Google Scholar

31.

Azizi H, Niazi Tabar A, Skutella T. Successful transplantation of spermatogonial stem cells into the seminiferous tubules of busulfan-treated mice. Reprod Health 2021; 18:189. Google Scholar

32.

Morimoto H, Ogonuki N, Kanatsu-Shinohara M, Matoba S, Ogura A, Shinohara T. Spermatogonial stem cell transplantation into nonablated mouse recipient testes. Stem Cell Rep 2021; 16: 1832–1844. Google Scholar

33.

Kita K, Watanabe T, Ohsaka K, Hayashi H, Kubota Y, Nagashima Y, Aoki I, Taniguchi H, Noce T, Inoue K, Miki H, Ogonuki N et al. Production of functional spermatids from mouse germline stem cells in ectopically reconstituted seminiferous tubules. Biol Reprod 2007; 76:211–217. Google Scholar

34.

Zhang M, Zhou H, Zheng C, Xiao J, Zuo E, Liu W, Xie D, Shi Y, Wu C, Wang H, Li D, Li J. The roles of testicular c-kit positive cells in de novo morphogenesis of testis. Sci Rep 2014; 4:5936–5936. Google Scholar

35.

Schlatt S, Honaramooz A, Boiani M, Scholer HR, Dobrinski I. Progeny from sperm obtained after ectopic grafting of neonatal mouse testes. Biol Reprod 2003; 68:2331–2335. Google Scholar

36.

Honaramooz A, Snedaker A, Boiani M, Scholer H, Dobrinski I, Schlatt S. Sperm from neonatal mammalian testes grafted in mice. Nature 2002; 418:778–781. Google Scholar

37.

Shinohara T, Inoue K, Ogonuki N, Kanatsu-Shinohara M, Miki H, Nakata K, Kurome M, Nagashima H, Toyokuni S, Kogishi K, Honjo T, Ogura A. Birth of offspring following transplantation of cryopreserved immature testicular pieces and in-vitro microinsemination. Hum Reprod 2002; 17:3039–3045. Google Scholar

38.

Nakai M, Kaneko H, Somfai T, Maedomari N, Ozawa M, Noguchi J, Ito J, Kashiwazaki N, Kikuchi K. Production of viable piglets for the first time using sperm derived from ectopic testicular xenografts. Reproduction 2010; 139:331–335. Google Scholar

39.

Honaramooz A, Li MW, Penedo MCT, Meyers S, Dobrinski I. Accelerated maturation of primate testis by xenografting into mice. Biol Reprod 2004; 70:1500–1503. Google Scholar

40.

Fayomi AP, Peters K, Sukhwani M, Valli-Pulaski H, Shetty G, Meistrich ML, Houser L, Robertson N, Roberts V, Ramsey C, Hanna C, Hennebold JD et al. Autologous grafting of cryopreserved prepubertal rhesus testis produces sperm and offspring. Science 2019; 363:1314–1319. Google Scholar

41.

Liu Z, Nie Y-H, Zhang C-C, Cai Y-J, Wang Y, Lu H-P, Li YZ, Cheng C, Qiu Z-L, Sun Q. Generation of macaques with sperm derived from juvenile monkey testicular xenografts. Cell Res 2016; 26:139–142. Google Scholar

42.

Komeya M, Kimura H, Nakamura H, Yokonishi T, Sato T, Kojima K, Hayashi K, Katagiri K, Yamanaka H, Sanjo H, Yao M, Kamimura S et al. Long-term ex vivo maintenance of testis tissues producing fertile sperm in a microfluidic device. Sci Rep 2016; 6:21472. Google Scholar

43.

Sato T, Katagiri K, Gohbara A, Inoue K, Ogonuki N, Ogura A, Kubota Y, Ogawa T. In vitro production of functional sperm in cultured neonatal mouse testes. Nature 2011; 471: 504–507. Google Scholar

44.

Yokonishi T, Sato T, Komeya M, Katagiri K, Kubota Y, Nakabayashi K, Hata K, Inoue K, Ogonuki N, Ogura A, Ogawa T. Offspring production with sperm grown in vitro from cryopreserved testis tissues. Nat Commun 2014; 5:4320. Google Scholar

45.

Yuan Y, Li L, Cheng Q, Diao F, Zeng Q, Yang X, Wu Y, Zhang H, Huang M, Chen J, Zhou Q, Zhu Y et al. In vitro testicular organogenesis from human fetal gonads produces fertilization-competent spermatids. Cell Res 2020; 30:244–255. Google Scholar

46.

Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 2011; 146:519–532. Google Scholar

47.

Nakaki F, Hayashi K, Ohta H, Kurimoto K, Yabuta Y, Saitou M. Induction of mouse germ-cell fate by transcription factors in vitro. Nature 2013; 501:222–226. Google Scholar

48.

Zhou Q, Wang M, Yuan Y, Wang X, Fu R, Wan H, Xie M, Liu M, Guo X, Zheng Y, Feng G, Shi Q et al. Complete meiosis from embryonic stem cell-derived germ cells in vitro. Cell Stem Cell 2016; 18:330–340. Google Scholar

49.

Ishikura Y, Yabuta Y, Ohta H, Hayashi K, Nakamura T, Okamoto I, Yamamoto T, Kurimoto K, Shirane K, Sasaki H, Saitou M. In vitro derivation and propagation of spermatogonial stem cell activity from mouse pluripotent stem cells. Cell Rep 2016; 17:2789–2804. Google Scholar

50.

Ishikura Y, Ohta H, Sato T, Murase Y, Yabuta Y, Kojima Y, Yamashiro C, Nakamura T, Yamamoto T, Ogawa T, Saitou M. In vitro reconstitution of the whole male germ-cell development from mouse pluripotent stem cells. Cell Stem Cell 2021; 28:2167–2179.e9. Google Scholar

51.

Bahadur G, Chatterjee R, Ralph D. Testicular tissue cryopreservation in boys. Ethical and legal issues: case report. Hum Reprod 2000; 15:1416–1420. Google Scholar

52.

Radford J. Restoration of fertility after treatment for cancer. Horm Res 2003; 59(Suppl 1):21–23. Google Scholar

53.

Kvist K, Thorup J, Byskov AG, Hoyer PE, Mollgard K, Yding AC. Cryopreservation of intact testicular tissue from boys with cryptorchidism. Hum Reprod 2006; 21:484–491. Google Scholar

54.

Ginsberg JP, Li Y, Carlson CA, Gracia CR, Hobbie WL, Miller VA, Mulhall J, Shnorhavorian M, Brinster RL, Kolon TF. Testicular tissue cryopreservation in prepubertal male children: an analysis of parental decision-making. Pediatr Blood Cancer 2014; 61:1673–1678. Google Scholar

55.

Sadri-Ardekani H, McLean TW, Kogan S, Sirintrapun J, Crowell K, Yousif MQ, Hodges SJ, Petty J, Pranikoff T, Sieren L, Zeller K, Atala A. Experimental testicular tissue banking to generate spermatogenesis in the future: A multidisciplinary team approach. Methods 2016; 99:120–127. Google Scholar

56.

Uijldert M, Meißner A, de Melker AA, van Pelt AMM, van de Wetering MD, van Rijn RR, van Wely M, van der Veen F, Repping S. Development of the testis in pre-pubertal boys with cancer after biopsy for fertility preservation. Hum Reprod 2017; 32: 2366–2372. Google Scholar

57.

Ho WLC, Bourne H, Gook D, Clarke G, Kemertzis M, Stern K, Agresta F, Heloury Y, Clark H, Orme L, Jayasinghe Y, Zacharin MR et al. A short report on current fertility preservation strategies for boys. Clin Endocrinol (Oxf) 2017; 87:279–285. Google Scholar

58.

Heckmann L, Langenstroth-Rower D, Pock T, Wistuba J, Stukenborg JB, Zitzmann M, Kliesch S, Schlatt S, Neuhaus N. A diagnostic germ cell score for immature testicular tissue at risk of germ cell loss. Hum Reprod 2018; 33:636–645. Google Scholar

59.

Hildorf S, Cortes D, Gul M, Dong L, Kristensen SG, Jensen CFS, Clasen-Linde E, Fedder J, Andersen CY, Hoffmann ER, Sonksen J, Fossum M et al. Parental acceptance rate of testicular tissue cryopreservation in danish boys with cryptorchidism. Sex Dev 2019; 13:246–257. Google Scholar

60.

Braye A, Tournaye H, Goossens E. Setting up a cryopreservation programme for immature testicular tissue: lessons learned after more than 15 years of experience. Clin Med Insights Reprod Health 2019; 13:1179558119886342. Google Scholar

61.

Borgstrom B, Fridstrom M, Gustafsson B, Ljungman P, Rodriguez-Wallberg KA. A prospective study on the long-term outcome of prepubertal and pubertal boys undergoing testicular biopsy for fertility preservation prior to hematologic stem cell transplantation. Pediatr Blood Cancer 2020; 67:e28507. Google Scholar

62.

Kanbar M, de Michele F, Giudice MG, Desmet L, Poels J, Wyns C. Long-term follow-up of boys who have undergone a testicular biopsy for fertility preservation. Hum Reprod 2020; 36:26–39. Google Scholar

63.

Rives-Feraille A, Liard A, Bubenheim M, Barbotin AL, Giscard d'Estaing S, Mirallie S, Ancelle A, Roux C, Brugnon F, Greze V, Daudin M, Willson-Plat G et al. Assessment of the architecture and integrity of frozen-thawed testicular tissue from (pre)pubertal boys with cancer. Andrology 2022; 10:279–290. Google Scholar

64.

Braye A, Bohler S, Vloeberghs V, De Boe V, De Schepper J, Gies I, Goossens E. Testicular biopsy for fertility preservation in early-diagnosed Klinefelter patients: patient characteristics and long-term follow-up. Reprod Biomed Online 2022; S1472–6483: 00074–00078. Google Scholar

65.

Bunge RG, Sherman JK. Fertilizing capacity of frozen human spermatozoa. Nature 1953; 172:767–768. Google Scholar

66.

Szell AZ, Bierbaum RC, Hazelrigg WB, Chetkowski RJ. Live births from frozen human semen stored for 40 years. J Assist Reprod Genet 2013; 30:743–744. Google Scholar

67.

Schover LR, Brey K, Lichtin A, Lipshultz LI, Jeha S. Knowledge and experience regarding cancer, infertility, and sperm banking in younger male survivors. J Clin Oncol 2002; 20:1880–1889. Google Scholar

68.

Ernst M, Brahler E, Wild PS, Faber J, Merzenich H, Beutel ME. The desire for children among adult survivors of childhood cancer: psychometric evaluation of a cancer-specific questionnaire and relations with sociodemographic and psychological characteristics. Psychooncology 2020; 29:485–492. Google Scholar

69.

Edge B, Holmes D, Makin G. Sperm banking in adolescent cancer patients. Arch Dis Child 2006; 91:149–152. Google Scholar

70.

Li Y, Zhang J, Zhang H, Liu B, Wang G, Cao M, Fu B, Li H, Jiang Q, Yu L, Xian Y, Su B et al. Importance and safety of autologous sperm cryopreservation for fertility preservation in young male patients with cancer. Medicine (Baltimore) 2020; 99:e19589. Google Scholar

71.

Halpern JA, Thirumavalavan N, Kohn TP, Patel AS, Leong JY, Cervellione RM, Keene DJB, Ibrahim E, Brackett NL, Lamb DJ, Ramasamy R. Distribution of semen parameters among adolescent males undergoing fertility preservation in a multicenter international cohort. Urology 2019; 127:119–123. Google Scholar

72.

Picton HM, Wyns C, Anderson RA, Goossens E, Jahnukainen K, Kliesch S, Mitchell RT, Pennings G, Rives N, Tournaye H, van Pelt AM, Eichenlaub-Ritter U et al. A European perspective on testicular tissue cryopreservation for fertility preservation in prepubertal and adolescent boys. Hum Reprod 2015; 30:2463–2475. Google Scholar

73.

Goossens E, Jahnukainen K, Mitchell RT, van Pelt A, Pennings G, Rives N, Poels J, Wyns C, Lane S, Rodriguez-Wallberg KA, Rives A, Valli-Pulaski H et al. Fertility preservation in boys: recent developments and new insights. Hum Reprod Open 2020; 2020:hoaa016. Google Scholar

74.

Keros V, Hultenby K, Borgstrom B, Fridstrom M, Jahnukainen K, Hovatta O. Methods of cryopreservation of testicular tissue with viable spermatogonia in pre-pubertal boys undergoing gonadotoxic cancer treatment. Hum Reprod 2007; 22:1384–1395. Google Scholar

75.

Van Saen D, Goossens E, Bourgain C, Ferster A, Tournaye H. Meiotic activity in orthotopic xenografts derived from human postpubertal testicular tissue. Hum Reprod 2011; 26:282–293. Google Scholar

76.

Sadri-Ardekani H, Akhondi MA, van der Veen F, Repping S, van Pelt AM. In vitro propagation of human prepubertal spermatogonial stem cells. JAMA 2011; 305:2416–2418. Google Scholar

77.

Curaba M, Poels J, van Langendonckt A, Donnez J, Wyns C. Can prepubertal human testicular tissue be cryopreserved by vitrification? Fertil Steril 2011; 95:2123.e2129–2123.e2112. Google Scholar

78.

Pietzak Iii EJ, Tasian GE, Tasian SK, Brinster RL, Carlson C, Ginsberg JP, Kolon TF. Histology of testicular biopsies obtained for experimental fertility preservation protocol in boys with cancer. J Urol 2015; 194:1420–1424. Google Scholar

79.

Schover LR. Patient attitudes toward fertility preservation. Pediatr Blood Cancer 2009; 53:281–284. Google Scholar

80.

Wyns C, Curaba M, Petit S, Vanabelle B, Laurent P, Wese JF, Donnez J. Management of fertility preservation in prepubertal patients: 5 years' experience at the Catholic University of Lou-vain. Hum Reprod 2011; 26:737–747. Google Scholar

81.

Ginsberg JP. New advances in fertility preservation for pediatric cancer patients. Curr Opin Pediatr 2011; 23:9–13. Google Scholar

82.

Sadri-Ardekani H, Akhondi MM, Vossough P, Maleki H, Sedighnejad S, Kamali K, Ghorbani B, van Wely M, van der Veen F, Repping S. 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: 796–802. Google Scholar

83.

Brinster RL, Zimmermann JW. Spermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci U S A 1994; 91: 11298–11302. Google Scholar

84.

Wu X, Goodyear SM, Abramowitz LK, Bartolomei MS, Tobias JW, Avarbock MR, Brinster RL. Fertile offspring derived from mouse spermatogonial stem cells cryopreserved for more than 14 years. Hum Reprod 2012; 27:1249–1259. Google Scholar

85.

Honaramooz A, Megee SO, Dobrinski I. Germ cell transplantation in pigs. Biol Reprod 2002; 66:21–28. Google Scholar

86.

Mikkola M, Sironen A, Kopp C, Taponen J, Sukura A, Vilkki J, Katila T, Andersson M. Transplantation of normal boar testicular cells resulted in complete focal spermatogenesis in a boar affected by the immotile short-tail sperm defect. Reprod Domest Anim 2006; 41:124–128. Google Scholar

87.

Honaramooz A, Behboodi E, Blash S, Megee SO, Dobrinski I. Germ cell transplantation in goats. Mol Reprod Dev 2003; 64: 422–428. Google Scholar

88.

Izadyar F, Den Ouden K, Stout TA, Stout J, Coret J, Lankveld DP, Spoormakers TJ, Colenbrander B, Oldenbroek JK, Van der Ploeg KD, Woelders H, Kal HB et al. Autologous and homologous transplantation of bovine spermatogonial stem cells. Reproduction 2003; 126:765–774. Google Scholar

89.

Kim Y, Turner D, Nelson J, Dobrinski I, McEntee M, Travis AJ. Production of donor-derived sperm after spermatogonial stem cell transplantation in the dog. Reproduction 2008; 136:823–831. Google Scholar

90.

Jahnukainen K, Ehmcke J, Quader MA, Saiful Huq M, Epperly MW, Hergenrother S, Nurmio M, Schlatt S. Testicular recovery after irradiation differs in prepubertal and pubertal non-human primates, and can be enhanced by autologous germ cell transplantation. Hum Reprod 2011; 26:1945–1954. Google Scholar

91.

Shetty G, Uthamanthil RK, Zhou W, Shao SH, Weng CC, Tailor RC, Hermann BP, Orwig KE, Meistrich ML. Hormone suppression with GnRH antagonist promotes spermatogenic recovery from transplanted spermatogonial stem cells in irradiated cynomolgus monkeys. Andrology 2013; 1:886–898. Google Scholar

92.

Shetty G, Mitchell JM, Meyer JM, Wu Z, Lam TNA, Phan TT, Zhang J, Hill L, Tailor RC, Peters KA, Penedo MC, Hanna C et al. Restoration of functional sperm production in irradiated pubertal rhesus monkeys by spermatogonial stem cell transplantation. Andrology 2020; 8:1428–1441. Google Scholar

93.

Dobrinski I, Avarbock MR, Brinster RL. Germ cell transplantation from large domestic animals into mouse testes. Mol Reprod Dev 2000; 57:270–9. Google Scholar

94.

Radford JA, Shalet SM, Lieberman BA. Fertility after treatment for cancer. BMJ 1999; 319:935–936. Google Scholar

95.

Ginsberg JP, Carlson CA, Lin K, Hobbie WL, Wigo E, Wu X, Brinster RL, Kolon TF. An experimental protocol for fertility preservation in prepubertal boys recently diagnosed with cancer: a report of acceptability and safety. Hum Reprod 2010; 25: 37–41. Google Scholar

96.

Hamra FK, Chapman KM, Nguyen DM, Williams-Stephens AA, Hammer RE, Garbers DL. Self renewal, expansion, and transfection of rat spermatogonial stem cells in culture. Proc Natl Acad Sci U S A 2005; 102:17430–17435. Google Scholar

97.

Richardson TE, Chapman KM, Tenenhaus Dann C, Hammer RE, Hamra FK. Sterile testis complementation with spermatogonial lines restores fertility to DAZL-deficient rats and maximizes donor germline transmission. PLoS One 2009; 4: e6308. Google Scholar

98.

Ryu BY, Kubota H, Avarbock MR, Brinster RL. Conservation of spermatogonial stem cell self-renewal signaling between mouse and rat. Proc Natl Acad Sci U S A 2005; 102:14302–14307. Google Scholar

99.

Kanatsu-Shinohara M, Ogonuki N, Inoue K, Miki H, Ogura A, Toyokuni S, Shinohara T. Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod 2003; 69:612–616. Google Scholar

100.

Kubota H, Avarbock MR, Brinster RL. Growth factors essential for self-renewal and expansion of mouse spermatogonial stem cells. Proc Natl Acad Sci U S A 2004; 101:16489–16494. Google Scholar

101.

Kanatsu-Shinohara M, Muneto T, Lee J, Takenaka M, Chuma S, Nakatsuji N, Horiuchi T, Shinohara T. Long-term culture of male germline stem cells from hamster testes. Biol Reprod 2008; 78:611–617. Google Scholar

102.

Kubota H, Avarbock MR, Brinster RL. Culture conditions and single growth factors affect fate determination of mouse spermatogonial stem cells. Biol Reprod 2004; 71:722–731. Google Scholar

103.

Oatley JM, Avarbock MR, Telaranta AI, Fearon DT, Brinster RL. Identifying genes important for spermatogonial stem cell self-renewal and survival. Proc Natl Acad Sci U S A 2006; 103: 9524–9529. Google Scholar

104.

Kanatsu-Shinohara M, Miki H, Inoue K, Ogonuki N, Toyokuni S, Ogura A, Shinohara T. Long-term culture of mouse male germline stem cells under serum-or feeder-free conditions. Biol Reprod 2005; 72:985–991. Google Scholar

105.

Brinster RL. Germline stem cell transplantation and transgenesis. Science 2002; 296:2174–2176. Google Scholar

106.

Murdock MH, David S, Swinehart IT, Reing JE, Tran K, Gassei K, Orwig KE, Badylak SF. Human testis extracellular matrix enhances human spermatogonial stem cell survival in vitro. Tissue Eng Part A 2018; 25:663–676. Google Scholar

107.

Sadri-Ardekani H, Mizrak SC, van Daalen SK, Korver CM, Roepers-Gajadien HL, Koruji M, Hovingh S, de Reijke TM, de la Rosette JJ, van der Veen F, de Rooij DG, Repping S et al. Propagation of human spermatogonial stem cells in vitro. JAMA 2009; 302:2127–2134. Google Scholar

108.

Wu X, Schmidt JA, Avarbock MR, Tobias JW, Carlson CA, Kolon TF, Ginsberg JP, Brinster RL. Prepubertal human spermatogonia and mouse gonocytes share conserved gene expression of germline stem cell regulatory molecules. Proc Natl Acad Sci U S A 2009; 106:21672–21677. Google Scholar

109.

Chen B, Wang YB, Zhang ZL, Xia WL, Wang HX, Xiang ZQ, Hu K, Han YF, Wang YX, Huang YR, Wang Z. Xeno-free culture of human spermatogonial stem cells supported by human embryonic stem cell-derived fibroblast-like cells. Asian J Androl 2009; 11: 557–565. Google Scholar

110.

Lim JJ, Sung SY, Kim HJ, Song SH, Hong JY, Yoon TK, Kim JK, Kim KS, Lee DR. Long-term proliferation and characterization of human spermatogonial stem cells obtained from obstructive and non-obstructive azoospermia under exogenous feeder-free culture conditions. Cell Prolif 2010; 43:405–417. Google Scholar

111.

He Z, Kokkinaki M, Jiang J, Dobrinski I, Dym M. Isolation, characterization, and culture of human spermatogonia. Biol Reprod 2010; 82:363–372. Google Scholar

112.

Kokkinaki M, Djourabtchi A, Golestaneh N. Long-term culture of human SSEA-4 positive spermatogonial stem cells (SSCs). J Stem Cell Res Ther 2011; 2:2488. Google Scholar

113.

Liu S, Tang Z, Xiong T, Tang W. Isolation and characterization of human spermatogonial stem cells. Reprod Biol Endocrinol 2011; 9:141. Google Scholar

114.

Mirzapour T, Movahedin M, Ibrahim TAT, Koruji M, Haron AW, Nowroozi MR, Rafieian SH. Effects of basic fibroblast growth factor and leukaemia inhibitory factor on proliferation and short-term culture of human spermatogonial stem cells. Andrologia 2012; 44:41–55. Google Scholar

115.

Goharbakhsh L, Mohazzab A, Salehkhou S, Heidari M, Zarnani AH, Parivar K, Akhondi MM. Isolation and culture of human spermatogonial stem cells derived from testis biopsy. Avicenna J Med Biotechnol 2013; 5:54–61. Google Scholar

116.

Piravar Z, Jeddi-Tehrani M, Sadeghi MR, Mohazzab A, Eidi A, Akhondi MM. In vitro culture of human testicular stem cells on feeder-free condition. J Reprod Infertil 2013; 14:17–22. Google Scholar

117.

Akhondi MM, Mohazzab A, Jeddi-Tehrani M, Sadeghi MR, Eidi A, Khodadadi A, Piravar Z. Propagation of human germ stem cells in long-term culture. Iran J Reprod Med 2013; 11:551–558. Google Scholar

118.

Zheng Y, Thomas A, Schmidt CM, Dann CT. Quantitative detection of human spermatogonia for optimization of spermatogonial stem cell culture. Hum Reprod 2014; 29:2497–2511. Google Scholar

119.

Smith JF, Yango P, Altman E, Choudhry S, Poelzl A, Zamah AM, Rosen M, Klatsky PC, Tran ND. Testicular niche required for human spermatogonial stem cell expansion. Stem Cells Transl Med 2014; 3:1043–1054. Google Scholar

120.

Baert Y, Braye A, Struijk RB, van Pelt AM, Goossens E. Cryopreservation of testicular tissue before long-term testicular cell culture does not alter in vitro cell dynamics. Fertil Steril 2015; 104:1244–1252–e1241–1244. Google Scholar

121.

Gat I, Maghen L, Filice M, Wyse B, Zohni K, Jarvi K, Lo KC, Gauthier Fisher A, Librach C. Optimal culture conditions are critical for efficient expansion of human testicular somatic and germ cells in vitro. Fertil Steril 2017; 107:595.e597–595.e605. Google Scholar

122.

Medrano JV, Rombaut C, Simon C, Pellicer A, Goossens E. Human spermatogonial stem cells display limited proliferation in vitro under mouse spermatogonial stem cell culture conditions. Fertil Steril 2016; 106:1539–1549.e8. Google Scholar

123.

Ali S, Ravindranath N, Jia MC, Musto N, Tsujimura T, Kitamura Y, Dym M. Expression of multiple c-kit receptor messenger ribonucleic acid transcripts during postnatal development of the rat testis. Biochem Biophys Res Commun 1996; 218: 104–112. Google Scholar

124.

Nowroozi MR, Ahmadi H, Rafiian S, Mirzapour T, Movahedin M. In vitro colonization of human spermatogonia stem cells: effect of patient's clinical characteristics and testicular histologic findings. Urology 2011; 78:1075–1081. Google Scholar

125.

Chikhovskaya JV, van Daalen SKM, Korver CM, Repping S, van Pelt AMM. Mesenchymal origin of multipotent human testis-derived stem cells in human testicular cell cultures. Mol Hum Reprod 2014; 20:155–167. Google Scholar

126.

Abdul Wahab AY, Md Isa ML, Ramli R. Spermatogonial stem cells protein identification in in vitro culture from non-obstructive azoospermia patient. Malays J Med Sci 2016; 23:40–48. Google Scholar

127.

Eildermann K, Gromoll J, Behr R. Misleading and reliable markers to differentiate between primate testis-derived multipotent stromal cells and spermatogonia in culture. Hum Reprod 2012; 27:1754–1767. Google Scholar

128.

Langenstroth D, Kossack N, Westernströer B, Wistuba J, Behr R, Gromoll J, Schlatt S. Separation of somatic and germ cells is required to establish primate spermatogonial cultures. Hum Reprod 2014; 29:2018–2031. Google Scholar

129.

Nagano M, Patrizio P, Brinster RL. Long-term survival of human spermatogonial stem cells in mouse testes. Fertil Steril 2002; 78: 1225–1233. Google Scholar

130.

Hermann BP, Sukhwani M, Lin CC, Sheng Y, Tomko J, Rodriguez M, Shuttleworth JJ, McFarland D, Hobbs RM, Pandolfi PP, Schatten GP, Orwig KE. Characterization, cryopreservation, and ablation of spermatogonial stem cells in adult rhesus macaques. Stem Cells 2007; 25:2330–2338. Google Scholar

131.

Dovey SL, Valli H, Hermann BP, Sukhwani M, Donohue J, Castro CA, Chu T, Sanfilippo JS, Orwig KE. Eliminating malignant contamination from therapeutic human spermatogonial stem cells. J Clin Invest 2013; 123:1833–1843. Google Scholar

132.

Maki CB, Pacchiarotti J, Ramos T, Pascual M, Pham J, Kinjo J, Anorve S, Izadyar F. Phenotypic and molecular characterization of spermatogonial stem cells in adult primate testes. Hum Reprod 2009; 24:1480–1491. Google Scholar

133.

Valli H, Sukhwani M, Dovey SL, Peters KA, Donohue J, Castro CA, Chu T, Marshall GR, Orwig KE. Fluorescence- and magnetic-activated cell sorting strategies to isolate and enrich human spermatogonial stem cells. Fertil Steril 2014; 102:566–580. Google Scholar

134.

Mirzapour T, Movahedin M, Koruji M, Nowroozi MR. Xenotransplantation assessment: morphometric study of human spermatogonial stem cells in recipient mouse testes. Andrologia 2015; 47:626–633. Google Scholar

135.

David S, Orwig KE. Spermatogonial Stem Cell Culture in Oncofertility. Urol Clin North Am 2020; 47:227–244. Google Scholar

136.

Kubota H, Avarbock MR, Brinster RL. Spermatogonial stem cells share some, but not all, phenotypic and functional characteristics with other stem cells. Proc Natl Acad Sci USA 2003; 100: 6487–6492. Google Scholar

137.

Kokkinaki M, Djourabtchi A, Golestaneh N. Long-term culture of human SSEA-4 positive spermatogonial stem cells (SSCs). Stem Cell Res Ther 2011; S2:003. Google Scholar

138.

Gassei K, Ehmcke J, Wood MA, Walker WH, Schlatt S. Immature rat seminiferous tubules reconstructed in vitro express markers of Sertoli cell maturation after xenografting into nude mouse hosts. Mol Hum Reprod 2010; 16:97–110. Google Scholar

139.

Alves-Lopes JP, Soder O, Stukenborg JB. Testicular organoid generation by a novel in vitro three-layer gradient system. Bio-materials 2017; 130:76–89. Google Scholar

140.

Dufour JM, Rajotte RV, Korbutt GS. Development of an in vivo model to study testicular morphogenesis. J Androl 2002; 23: 635–644. Google Scholar

141.

Honaramooz A, Megee SO, Rathi R, Dobrinski I. Building a testis: formation of functional testis tissue after transplantation of isolated porcine (Sus scrofa) testis cells. Biol Reprod 2007; 76: 43–47. Google Scholar

142.

Dores C, Dobrinski I. De novo morphogenesis of testis tissue: an improved bioassay to investigate the role of VEGF165 during testis formation. Reproduction 2014; 148:109–117. Google Scholar

143.

Arregui L, Rathi R, Megee SO, Honaramooz A, Gomendio M, Roldan ER, Dobrinski I. Xenografting of sheep testis tissue and isolated cells as a model for preservation of genetic material from endangered ungulates. Reproduction 2008; 136:85–93. Google Scholar

144.

Aeckerle N, Dressel R, Behr R. Grafting of Neonatal marmoset monkey testicular single-cell suspensions into immunodeficient mice leads to ex situ testicular cord neomorphogenesis. Cells Tissues Organs 2013; 198:209–220. Google Scholar

145.

Baert Y, Stukenborg JB, Landreh M, De Kock J, Jornvall H, Soder O, Goossens E. Derivation and characterization of a cytocompatible scaffold from human testis. Hum Reprod 2015; 30:256–267. Google Scholar

146.

Pendergraft SS, Sadri-Ardekani H, Atala A, Bishop CE. Three-dimensional testicular organoid: a novel tool for the study of human spermatogenesis and gonadotoxicity in vitro†. Biol Reprod 2017; 96:720–732. Google Scholar

147.

Baert Y, De Kock J, Alves-Lopes JP, Söder O, Stukenborg JB, Goossens E. Primary human testicular cells self-organize into organoids with testicular properties. Stem Cell Rep 2017; 8: 30–38. Google Scholar

148.

Zenzes MT, Engel W. The capacity of testicular cells of the postnatal rat to reorganize into histotypic structures. Differentiation 1981; 20:157–161. Google Scholar

149.

Sakib S, Uchida A, Valenzuela-Leon P, Yu Y, Valli-Pulaski H, Orwig K, Ungrin M, Dobrinski I. Formation of organotypic testicular organoids in microwell culture†. Biol Reprod 2019; 100:1648–1660. Google Scholar

150.

Yokonishi T, Sato T, Katagiri K, Komeya M, Kubota Y, Ogawa T. In vitro reconstruction of mouse seminiferous tubules supporting germ cell differentiation. Biol Reprod 2013; 89:15. Google Scholar

151.

Zhang J, Hatakeyama J, Eto K, Abe S. Reconstruction of a seminiferous tubule-like structure in a 3 dimensional culture system of re-aggregated mouse neonatal testicular cells within a collagen matrix. Gen Comp Endocrinol 2014; 205:121–132. Google Scholar

152.

Baert Y, Dvorakova-Hortova K, Margaryan H, Goossens E. Mouse in vitro spermatogenesis on alginate-based 3D bioprinted scaffolds. Biofabrication 2019; 11:035011. Google Scholar

153.

Huleihel M, Nourashrafeddin S, Plant TM. Application of three-dimensional culture systems to study mammalian spermatogenesis, with an emphasis on the rhesus monkey (Macaca mulatta). Asian J Androl 2015; 17:972–980. Google Scholar

154.

Oliver E, Alves-Lopes JP, Harteveld F, Mitchell RT, Akesson E, Soder O, Stukenborg JB. Self-organising human gonads generated by a Matrigel-based gradient system. BMC Biol 2021; 19:212. Google Scholar

155.

Gassei K, Schlatt S, Ehmcke J. De novo morphogenesis of seminiferous tubules from dissociated immature rat testicular cells in xenografts. J Androl 2006; 27:611–618. Google Scholar

156.

Gassei K, Ehmcke J, Schlatt S. Initiation of testicular tubulogenesis is controlled by neurotrophic tyrosine receptor kinases in a three-dimensional Sertoli cell aggregation assay. Reproduction 2008; 136:459–469. Google Scholar

157.

Schlatt S, Kim SS, Gosden R. Spermatogenesis and steroidogenesis in mouse, hamster and monkey testicular tissue after cryopreservation and heterotopic grafting to castrated hosts. Reproduction 2002; 124:339–346. Google Scholar

158.

Geens M, De Block G, Goossens E, Frederickx V, Van Steirteghem A, Tournaye H. Spermatogonial survival after grafting human testicular tissue to immunodeficient mice. Hum Reprod 2006; 21: 390–396. Google Scholar

159.

Yu J, Cai ZM, Wan HJ, Zhang FT, Ye J, Fang JZ, Gui YT, Ye JX. Development of neonatal mouse and fetal human testicular tissue as ectopic grafts in immunodeficient mice. Asian J Androl 2006; 8:393–403. Google Scholar

160.

Goossens E, Frederickx V, Geens M, De Block G, Tournaye H. Cryosurvival and spermatogenesis after allografting prepubertal mouse tissue: comparison of two cryopreservation protocols. Fertil Steril 2008; 89:725–727. Google Scholar

161.

Luetjens CM, Stukenborg J-B, Nieschlag E, Simoni M, Wistuba J. Complete spermatogenesis in orthotopic but not in ectopic transplants of autologously grafted marmoset testicular tissue. Endocrinology 2008; 149:1736–1747. Google Scholar

162.

Snedaker AK, Honaramooz A, Dobrinski I. A game of cat and mouse: xenografting of testis tissue from domestic kittens results in complete cat spermatogenesis in a mouse host. J Androl 2004; 25:926–930. Google Scholar

163.

Kim Y, Selvaraj V, Pukazhenthi B, Travis AJ. Effect of donor age on success of spermatogenesis in feline testis xenografts. Reprod Fertil Dev 2007; 19:869–876. Google Scholar

164.

Arregui L, Dobrinski I, Roldan ERS. Germ cell survival and differentiation after xenotransplantation of testis tissue from three endangered species: Iberian lynx (Lynx pardinus), Cuvier's gazelle (Gazella cuvieri) and Mohor gazelle (G. dama mhorr). Reprod Fertil Dev 2014; 26:817–826. Google Scholar

165.

Abrishami M, Abbasi S, Honaramooz A. The effect of donor age on progression of spermatogenesis in canine testicular tissue after xenografting into immunodeficient mice. Theriogenology 2010; 73:512–522. Google Scholar

166.

Zeng W, Avelar GF, Rathi R, Franca LR, Dobrinski I. The length of the spermatogenic cycle is conserved in porcine and ovine testis xenografts. J Androl 2006; 27:527–533. Google Scholar

167.

Kaneko H, Kikuchi K, Nakai M, Noguchi J. Endocrine status and development of porcine testicular tissues in host mice. J Reprod Dev 2008; 54:480–485. Google Scholar

168.

Arregui L, Rathi R, Zeng W, Honaramooz A, Gomendio M, Roldan ER, Dobrinski I. Xenografting of adult mammalian testis tissue. Anim Reprod Sci 2008; 106:65–76. Google Scholar

169.

Abbasi S, Honaramooz A. Effects of recipient mouse strain, sex and gonadal status on the outcome of testis tissue xenografting. Reprod Fertil Dev 2010; 22:1279–1286. Google Scholar

170.

Kaneko H, Kikuchi K, Men NT, Nakai M, Noguchi J, Kashiwazaki N, Ito J. Production of sperm from porcine fetal testicular tissue after cryopreservation and grafting into nude mice. Theriogenology 2017; 91:154–162. Google Scholar

171.

Oatley JM, de Avila DM, Reeves JJ, McLean DJ. Spermatogenesis and germ cell transgene expression in xenografted bovine testicular tissue. Biol Reprod 2004; 71:494–501. Google Scholar

172.

Oatley JM, Reeves JJ, McLean DJ. Establishment of spermatogenesis in neonatal bovine testicular tissue following ectopic xenografting varies with donor age. Biol Reprod 2005; 72: 358–364. Google Scholar

173.

Rathi R, Honaramooz A, Zeng W, Schlatt S, Dobrinski I. Germ cell fate and seminiferous tubule development in bovine testis xenografts. Reproduction 2005; 130:923–929. Google Scholar

174.

Huang S, Sartini BL, Parks JE. Spermatogenesis in testis xenografts grafted from pre-pubertal Holstein bulls is reestablished by stem cell or early spermatogonia. Anim Reprod Sci 2008; 103:1–12. Google Scholar

175.

Reddy N, Mahla RS, Thathi R, Suman SK, Jose J, Goel S. Gonadal status of male recipient mice influences germ cell development in immature buffalo testis tissue xenograft. Reproduction 2012; 143:59–69. Google Scholar

176.

Rathi R, Honaramooz A, Zeng W, Turner R, Dobrinski I. Germ cell development in equine testis tissue xenografted into mice. Reproduction 2006; 131:1091–1098. Google Scholar

177.

Rathi R, Zeng W, Megee S, Conley A, Meyers S, Dobrinski I. Maturation of testicular tissue from infant monkeys after xenografting into mice. Endocrinology 2008; 149:5288–5296. Google Scholar

178.

Ehmcke J, Gassei K, Westernstroer B, Schlatt S. Immature rhesus monkey (Macaca mulatta) testis xenografts show increased growth, but not enhanced seminiferous differentiation, under human chorionic gonadotropin treatment of nude mouse recipients. Int J Androl 2011; 34:e459–e467. Google Scholar

179.

Ntemou E, Kadam P, Van Saen D, Wistuba J, Mitchell RT, Schlatt S, Goossens E. Complete spermatogenesis in intratesticular testis tissue xenotransplants from immature non-human primate. Hum Reprod 2019; 34:403–413. Google Scholar

180.

Skakkebaek NE, Jensen G, Povlsen CO, Rygaard J. Heterotransplantation of human foetal testicular and ovarian tissue to the mouse mutant nude. A preliminary study. Acta Obstet Gynecol Scand Suppl 1974; 29:73–75. Google Scholar

181.

Schlatt S, Honaramooz A, Ehmcke J, Goebell PJ, Rubben H, Dhir R, Dobrinski I, Patrizio P. Limited survival of adult human testicular tissue as ectopic xenograft. Hum Reprod 2006; 21: 384–389. Google Scholar

182.

Wyns C, Curaba M, Martinez-Madrid B, Van Langendonckt A, Francois-Xavier W, Donnez J. Spermatogonial survival after cryopreservation and short-term orthotopic immature human cryptorchid testicular tissue grafting to immunodeficient mice. Hum Reprod 2007; 22:1603–1611. Google Scholar

183.

Goossens E, Geens M, De Block G, Tournaye H. Spermatogonial survival in long-term human prepubertal xenografts. Fertil Steril 2008; 90:2019–2022. Google Scholar

184.

Wyns C, Van Langendonckt A, Wese FX, Donnez J, Curaba M. Long-term spermatogonial survival in cryopreserved and xenografted immature human testicular tissue. Hum Reprod 2008; 23:2402–2414. Google Scholar

185.

Sato Y, Nozawa S, Yoshiike M, Arai M, Sasaki C, Iwamoto T. Xenografting of testicular tissue from an infant human donor results in accelerated testicular maturation. Hum Reprod 2010; 25:1113–1122. Google Scholar

186.

Poels J, Van Langendonckt A, Many MC, Wese FX, Wyns C. Vitrification preserves proliferation capacity in human spermatogonia. Hum Reprod 2013; 28:578–589. Google Scholar

187.

Poels J, Abou-Ghannam G, Herman S, Van Langendonckt A, Wese F-X, Wyns C. In search of better spermatogonial preservation by supplementation of cryopreserved human immature testicular tissue xenografts with N-acetylcysteine and testosterone. Front Surg 2014; 1. Google Scholar

188.

Ntemou E, Kadam P, Van Laere S, Van Saen D, Vicini E, Goossens E. Effect of recombinant human vascular endothelial growth factor on testis tissue xenotransplants from prepubertal boys: a three-case study. Reprod Biomed Online 2019; 39:119–133. Google Scholar

189.

Honaramooz A, Cui X, Kim N, Dobrinski I. Porcine embryos produced after intracytoplasmic sperm injection using xenogeneic pig sperm from neonatal testis tissue grafted in mice. Reprod Fertil Dev 2008; 20:802–807. Google Scholar

190.

Hutka M, Kadam P, Van Saen D, Homer NZM, Onofre J, Wallace WHB, Smith LB, Stukenborg J-B, Goossens E, Mitchell RT. Fertility preservation in childhood cancer: endocrine activity in prepubertal human testis xenografts exposed to a pubertal hormone environment. Cancer 2020; 12:2830. Google Scholar

191.

Livera G, Delbes G, Pairault C, Rouiller-Fabre V, Habert R. Organotypic culture, a powerful model for studying rat and mouse fetal testis development. Cell Tissue Res 2006; 324: 507–521. Google Scholar

192.

Gohbara A, Katagiri K, Sato T, Kubota Y, Kagechika H, Araki Y, Araki Y, Ogawa T. In vitro murine spermatogenesis in an organ culture system. Biol Reprod 2010; 83:261–267. Google Scholar

193.

Sato T, Katagiri K, Kubota Y, Ogawa T. In vitro sperm production from mouse spermatogonial stem cell lines using an organ culture method. Nat Protoc 2013; 8:2098–2104. Google Scholar

194.

Sato T, Katagiri K, Kojima K, Komeya M, Yao M, Ogawa T. In vitro spermatogenesis in explanted adult mouse testis tissues. PLoS One 2015; 10:e0130171. Google Scholar

195.

Arkoun B, Dumont L, Milazzo JP, Way A, Bironneau A, Wils J, Mace B, Rives N. Retinol improves in vitro differentiation of pre-pubertal mouse spermatogonial stem cells into sperm during the first wave of spermatogenesis. PLoS One 2015; 10: e0116660. Google Scholar

196.

Dumont L, Arkoun B, Jumeau F, Milazzo JP, Bironneau A, Liot D, Wils J, Rondanino C, Rives N. Assessment of the optimal vitrification protocol for pre-pubertal mice testes leading to successful in vitro production of flagellated spermatozoa. Andrology 2015; 3:611–625. Google Scholar

197.

Dumont L, Oblette A, Rondanino C, Jumeau F, Bironneau A, Liot D, Duchesne V, Wils J, Rives N. Vitamin A prevents round spermatid nuclear damage and promotes the production of motile sperm during in vitro maturation of vitrified pre-pubertal mouse testicular tissue. Mol Hum Reprod 2016; 22:819–832. Google Scholar

198.

Reda A, Albalushi H, Montalvo SC, Nurmio M, Sahin Z, Hou M, Geijsen N, Toppari J, Soder O, Stukenborg JB. Knock-out serum replacement and melatonin effects on germ cell differentiation in murine testicular explant cultures. Ann Biomed Eng 2017; 45: 1783–1794. Google Scholar

199.

Komeya M, Hayashi K, Nakamura H, Yamanaka H, Sanjo H, Kojima K, Sato T, Yao M, Kimura H, Fujii T, Ogawa T. Pump-less microfluidic system driven by hydrostatic pressure induces and maintains mouse spermatogenesis in vitro. Sci Rep 2017; 7:15459. Google Scholar

200.

Pence LM, Schmitt TC, Beger RD, Del Valle PL, Nakamura N. Testicular function in cultured postnatal mouse testis fragments is similar to that of animals during the first wave of spermatogenesis. Birth Defects Res 2019; 111:270–280. Google Scholar

201.

Komeya M, Yamanaka H, Sanjo H, Yao M, Nakamura H, Kimura H, Fujii T, Sato T, Ogawa T. In vitro spermatogenesis in two-dimensionally spread mouse testis tissues. Reprod Med Biol 2019; 18:362–369. Google Scholar

202.

Yamanaka H, Komeya M, Nakamura H, Sanjo H, Sato T, Yao M, Kimura H, Fujii T, Ogawa T. A monolayer microfluidic device supporting mouse spermatogenesis with improved visibility. Biochem Biophys Res Commun 2018; 500: 885–891. Google Scholar

203.

Reda A, Hou M, Winton TR, Chapin RE, Soder O, Stukenborg JB. In vitro differentiation of rat spermatogonia into round spermatids in tissue culture. Mol Hum Reprod 2016; 22:601–612. Google Scholar

204.

Jorgensen A, Young J, Nielsen JE, Joensen UN, Toft BG, Rajpert-De Meyts E, Loveland KL. Hanging drop cultures of human testis and testis cancer samples: a model used to investigate activin treatment effects in a preserved niche. Br J Cancer 2014; 110: 2604–2614. Google Scholar

205.

Jorgensen A, Nielsen JE, Perlman S, Lundvall L, Mitchell RT, Juul A, Rajpert-De ME. Ex vivo culture of human fetal gonads: manipulation of meiosis signalling by retinoic acid treatment disrupts testis development. Hum Reprod 2015; 30:2351–2363. Google Scholar

206.

de Michele F, Poels J, Vermeulen M, Ambroise J, Gruson D, Guiot Y, Wyns C. Haploid germ cells generated in organotypic culture of testicular tissue from prepubertal boys. Front Physiol 2018; 9:1413. Google Scholar

207.

Lambrot R, Coffigny H, Pairault C, Donnadieu AC, Frydman R, Habert R, Rouiller-Fabre V. Use of organ culture to study the human fetal testis development: effect of retinoic acid. J Clin Endocrinol Metab 2006; 91:2696–2703. Google Scholar

208.

Trautmann E, Guerquin MJ, Duquenne C, Lahaye JB, Habert R, Livera G. Retinoic acid prevents germ cell mitotic arrest in mouse fetal testes. Cell Cycle 2008; 7:656–664. Google Scholar

209.

Medrano JV, Vilanova-Perez T, Fornes-Ferrer V, Navarro-Gomezlechon A, Martinez-Triguero ML, Garcia S, Gomez-Chacon J, Povo I, Pellicer A, Andres MM, Novella-Maestre E. Influence of temperature, serum, and gonadotropin supplementation in short- and long-term organotypic culture of human immature testicular tissue. Fertil Steril 2018; 110:1045, e1043–1057. Google Scholar

210.

de Michele F, Poels J, Weerens L, Petit C, Evrard Z, Ambroise J, Gruson D, Wyns C. Preserved seminiferous tubule integrity with spermatogonial survival and induction of Sertoli and Leydig cell maturation after long-term organotypic culture of prepubertal human testicular tissue. Hum Reprod 2017; 32:32–45. Google Scholar

211.

Chen D, Clark AT. Mitochondrial DNA selection in human germ cells. Nat Cell Biol 2018; 20:118–120. Google Scholar

212.

Hayashi K. In vitro reconstitution of germ cell developmentdagger. Biol Reprod 2019; 101:567–578. Google Scholar

213.

Handel MA, Eppig JJ, Schimenti JC. Applying "gold standards" to in-vitro-derived germ cells. Cell 2014; 157:1257–1261. Google Scholar

214.

Kanatsu-Shinohara M, Mori Y, Shinohara T. Enrichment of mouse spermatogonial stem cells based on aldehyde dehydrogenase activity. Biol Reprod 2013; 89:140, 141–110. Google Scholar

215.

Hwang YS, Suzuki S, Seita Y, Ito J, Sakata Y, Aso H, Sato K, Hermann BP, Sasaki K. Reconstitution of prospermatogonial specification in vitro from human induced pluripotent stem cells. Nat Commun 2020; 11:5656. Google Scholar

216.

Yoshino T, Suzuki T, Nagamatsu G, Yabukami H, Ikegaya M, Kishima M, Kita H, Imamura T, Nakashima K, Nishinakamura R, Tachibana M, Inoue M et al. Generation of ovarian follicles from mouse pluripotent stem cells. Science 2021; 373. Google Scholar

217.

Buganim Y, Itskovich E, Hu YC, Cheng AW, Ganz K, Sarkar S, Fu D, Welstead GG, Page DC, Jaenisch R. Direct reprogramming of fibroblasts into embryonic Sertoli-like cells by defined factors. Cell Stem Cell 2012; 11:373–386. Google Scholar

218.

Sosa E, Chen D, Rojas EJ, Hennebold JD, Peters KA, Wu Z, Lam TN, Mitchell JM, Sukhwani M, Tailor RC, Meistrich ML, Orwig KE et al. Differentiation of primate primordial germ cell-like cells following transplantation into the adult gonadal niche. Nat Commun 2018; 9:5339. Google Scholar

219.

Teramura T, Takehara T, Kawata N, Fujinami N, Mitani T, Takenoshita M, Matsumoto K, Saeki K, Iritani A, Sagawa N, Hosoi Y. Primate embryonic stem cells proceed to early gametogenesis in vitro. Cloning Stem Cells 2007; 9:144–156. Google Scholar

220.

Yamauchi K, Hasegawa K, Chuma S, Nakatsuji N, Suemori H. In vitro germ cell differentiation from cynomolgus monkey embryonic stem cells. PLoS One 2009; 4:e5338. Google Scholar

221.

Kee K, Angeles VT, Flores M, Nguyen HN, Reijo Pera RA. Human DAZL, DAZ and BOULE genes modulate primordial germ-cell and haploid gamete formation. Nature 2009; 462: 222–225. Google Scholar

222.

Panula S, Medrano JV, Kee K, Bergstrom R, Nguyen HN, Byers B, Wilson KD, Wu JC, Simon C, Hovatta O, Reijo Pera RA. Human germ cell differentiation from fetal- and adult-derived induced pluripotent stem cells. Hum Mol Genet 2011; 20: 752–762. Google Scholar

223.

Easley CA, Phillips BT, MM MG, Barringer JM, Valli H, Hermann BP, Simerly CR, Rajkovic A, Miki T, Orwig KE, Schatten GP. Direct differentiation of human pluripotent stem cells into haploid spermatogenic cells. Cell Rep 2012; 2:440–446. Google Scholar

224.

Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, Ohta H, Moritoki Y, Iwatani C, Tsuchiya H, Nakamura S, Sekiguchi K et al. Robust in vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell 2015; 17:178–194. Google Scholar

225.

Irie N, Weinberger L, Tang WW, Kobayashi T, Viukov S, Manor YS, Dietmann S, Hanna JH, Surani MA. SOX17 is a critical specifier of human primordial germ cell fate. Cell 2015; 160: 253–268. Google Scholar

226.

Clark AT, Bodnar MS, Fox M, Rodriquez RT, Abeyta MJ, Firpo MT, Pera RA. Spontaneous differentiation of germ cells from human embryonic stem cells in vitro. Hum Mol Genet 2004; 13: 727–739. Google Scholar

227.

Kee K, Gonsalves JM, Clark AT, Pera RA. Bone morphogenetic proteins induce germ cell differentiation from human embryonic stem cells. Stem Cells Dev 2006; 15:831–837. Google Scholar

228.

Durruthy Durruthy J, Ramathal C, Sukhwani M, Fang F, Cui J, Orwig KE, Reijo Pera RA. Fate of induced pluripotent stem cells following transplantation to murine seminiferous tubules. Hum Mol Genet 2014; 23:3071–3084. Google Scholar

229.

Ramathal C, Durruthy-Durruthy J, Sukhwani M, Arakaki JE, Turek PJ, Orwig KE, Reijo Pera RA. Fate of iPSCs derived from azoospermic and fertile men following xenotransplantation to murine seminiferous tubules. Cell Rep 2014; 7:1284–1297. Google Scholar

230.

Ramathal C, Angulo B, Sukhwani M, Cui J, Durruthy-Durruthy J, Fang F, Schanes P, Turek PJ, Orwig KE, Reijo PR. DDX3Y gene rescue of a Y chromosome AZFa deletion restores germ cell formation and transcriptional programs. Sci Rep 2015; 5: 15041. Google Scholar

231.

Eguizabal C, Montserrat N, Vassena R, Barragan M, Garreta E, Garcia-Quevedo L, Vidal F, Giorgetti A, Veiga A, Izpisua Belmonte JC. Complete meiosis from human induced pluripotent stem cells. Stem Cells 2011; 29:1186–1195. Google Scholar

232.

Nahata L, Caltabellotta NM, Yeager ND, Lehmann V, Whiteside SL, O'Brien SH, Quinn GP, Gerhardt CA. Fertility perspectives and priorities among male adolescents and young adults in cancer survivorship. Pediatr Blood Cancer 2018; 65:e27019. Google Scholar

233.

Zambrowicz BP, Zimmermann JW, Harendza CJ, Simpson EM, Page DC, Brinster RL, Palmiter RD. Expression of a mouse Zfy-1/lacZ transgene in the somatic cells of the embryonic gonad and germ cells of the adult testis. Development 1994; 120: 1549–1559. Google Scholar

234.

Kanatsu-Shinohara M, Ogonuki N, Matoba S, Ogura A, Shinohara T. Autologous transplantation of spermatogonial stem cells restores fertility in congenitally infertile mice. Proc Natl Acad Sci 2020; 117:7837–7844. Google Scholar

235.

Clouthier DE, Avarbock MR, Maika SD, Hammer RE, Brinster RL. Rat spermatogenesis in mouse testis. Nature 1996; 381: 418–421. Google Scholar

236.

Ogawa T, Dobrinski I, Avarbock MR, Brinster RL. Xenogeneic spermatogenesis following transplantation of hamster germ cells to mouse testes. Biol Reprod 1999; 60:515–521. Google Scholar

237.

Kim Y, Selvaraj V, Dobrinski I, Lee H, Mcentee MC, Travis AJ. Recipient preparation and mixed germ cell isolation for spermatogonial stem cell transplantation in domestic cats. J Androl 2006; 27:248–256. Google Scholar

238.

Dobrinski I, Avarbock MR, Brinster RL. Transplantation of germ cells from rabbits and dogs into mouse testes. Biol Reprod 1999; 61:1331–1339. Google Scholar

239.

Oatley JM, de Avila DM, McLean DJ, Griswold MD, Reeves JJ. Transplantation of bovine germinal cells into mouse testes. J Anim Sci 2002; 80:1925–1931. Google Scholar

240.

Nagano M, McCarrey JR, Brinster RL. Primate spermatogonial stem cells colonize mouse testes. Biol Reprod 2001; 64: 1409–1416. Google Scholar

241.

Schlatt S, Foppiani L, Rolf C, Weinbauer GF, Nieschlag E. Germ cell transplantation into X-irradiated monkey testes. Hum Reprod 2002; 17:55–62. Google Scholar

242.

Brook PF, Radford JA, Shalet SM, Joyce AD, Gosden RG. Isolation of germ cells from human testicular tissue for low temperature storage and autotransplantation. Fertil Steril 2001; 75:269–274. Google Scholar

243.

Patra T, Pathak D, Gupta MK. Comparison of two culture methods during in vitro spermatogenesis of vitrified-warmed testis tissue: organ culture vs. hanging drop culture. Cryobiology 2021; 100:142–150. Google Scholar

244.

Heckmann L, Langenstroth-Rower D, Wistuba J, Portela JMD, Pelt AMM, Redmann K, Stukenborg JB, Schlatt S, Neuhaus N. The initial maturation status of marmoset testicular tissues has an impact on germ cell maintenance and somatic cell response in tissue fragment culture. Mol Hum Reprod 2020; 26: 374–388. Google Scholar

245.

Sharma S, Klaverkamp RS, Wistuba J, Schlatt S. Limited spermatogenic differentiation of testicular tissue from prepubertal marmosets (Callithrix jacchus) in an in vitro organ culture system. Mol Cell Endocrinol 2021; 539: 111488. Google Scholar

246.

Park TS, Galic Z, Conway AE, Lindgren A, van Handel BJ, Magnusson M, Richter L, Teitell MA, Mikkola HK, Lowry WE, Plath K, Clark AT. Derivation of primordial germ cells from human embryonic and induced pluripotent stem cells is significantly improved by coculture with human fetal gonadal cells. Stem Cells 2009; 27:783–795. Google Scholar
© The Author(s) 2022. Published by Oxford University Press on behalf of Society for the Study of Reproduction. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
Kien T.D. Tran, Hanna Valli-Pulaski, Amanda Colvin, and Kyle E. Orwig "Male fertility preservation and restoration strategies for patients undergoing gonadotoxic therapies," Biology of Reproduction 107(2), 382-405, (9 April 2022). https://doi.org/10.1093/biolre/ioac072
Received: 24 September 2021; Accepted: 4 April 2022; Published: 9 April 2022
KEYWORDS
de novo testicular morphogenesis
fertility preservation
grafting
in vitro gametogenesis
induced pluripotent stem cells
Male infertility
Primordial germ cell-like cells
Back to Top