Next Article in Journal
Direct Comparative Analysis of a Pharmacogenomics Panel with PacBio Hifi® Long-Read and Illumina Short-Read Sequencing
Next Article in Special Issue
Sperm DNA Fragmentation after Cryopreservation and Sperm Selection Has No Implications for Clinical Pregnancies and Live Births after Intrauterine Insemination with Donor Sperm
Previous Article in Journal
Desmoid Tumors in the Active Surveillance Era: Evaluation of Treatment Options and Pain Relief in a Single-Center Retrospective Analysis
Previous Article in Special Issue
Factors Affecting the Potential Efficacy of Intrauterine Platelet-Rich Plasma Infusion on Thin Endometrium in Women with Recurrent Implantation Failure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Semen Cryopreservation to Expand Male Fertility in Cancer Patients: Intracase Evaluation of Semen Quality

1
Sperm Bank, Department of Maternal Infant, Annunziata Hospital of Cosenza, 87100 Cosenza, Italy
2
Department of Translational Medicine, Hemostasis & Thrombosis Centre, University of Ferrara, 44121 Ferrara, Italy
3
University Strategic Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
4
Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
5
CERM Hungaria, Centre for Reproductive Medicine, 00198 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(12), 1654; https://doi.org/10.3390/jpm13121654
Submission received: 1 November 2023 / Revised: 21 November 2023 / Accepted: 25 November 2023 / Published: 27 November 2023
(This article belongs to the Special Issue Current Trends and Future Challenges in Assisted Reproduction)

Abstract

:
To preserve male fertility after diagnosis of any kind of cancer, a prompt assessment of the semen quality and an appropriate semen cryopreservation must be performed before radio-chemotherapy starts. The present work aims to evaluate the semen parameters at diagnosis of different cancer patients before cryopreservation and after thawing. Testicular tumors and lymphomas are among the most common cancers in younger patients, and while chemotherapy significantly increases patients’ survival, it can epigenetically alter the semen fluid, resulting in temporary or permanent infertility. We analyzed data from the database of the Gamete Cryopreservation Center (Annunziata Hospital, CS; Italy) in the period of 2011–2020 from a cohort of 254 cancer patients aged 18–56 years. The evaluation was performed in a blind manner and anonymously recovered; the main parameters referring to semen quality were assessed in accordance with the WHO guidelines and decision limits (6th edition; 2021). The cancer types were as follows: testis cancers (TC; n = 135; 53.1%), hematological cancers (HC; n = 76; 29.9%), and other types of cancer (OC; n = 43; 17%). Comparing TC vs. HC (P1) and vs. OC (P2), TC had the worst semen quality: sperm number/mL (P1 = 0.0014; P2 = 0.004), total motility (P1 = 0.02; P2 = 0.07), progressive motility (P1 = 0.04; P2 = 0.05), viability (P1 = 0.01; P2 = 0.02), and percentage of atypical morphology (P1 = 0.05; P2 = 0.03). After semen thawing, viability and progressive motility recovery lowered, accounting for 46.82% and 16.75%, respectively, in the whole cohort; similarly, in the subgroups ascribed to TC, they showed the lowest recovery. Strong correlation existed between pre- and post-cryopreservation viability and progressive motility in the whole cohort (p < 0.001) and in the TC subgroup (p < 0.05). All cancer subgroups, to significantly different extents, had semen findings below the WHO reference values, suggesting diverse sperm susceptibilities to different cancers and cryodamage. Cancer and associated treatments epigenetically affect patients’ semen quality, meaning cryopreservation should be considered a useful personalized prerogative for any kind of cancer in a timely manner.

Graphical Abstract

1. Introduction

Cryopreservation of spermatozoa is one of the most valuable and useful strategies to preserve male reproductive function in patients undergoing pharmacological treatments such as chemo- or radiotherapies characterized by a variety of negative side effects. Recently, sperm cryopreservation also has become available for other pathologies or cancers not directly involving the urogenital system, such as autoimmune diseases and blood cancers requiring treatments that may globally affect sperm viability and quality [1]. It has been demonstrated that the rate of successful recovery after semen thawing greatly fluctuates among different pathologies, depending in part on the molecular clusters of somatic mutations detected in the patient responsible for a dysregulation of key proteins associated with sperm fertility and motility [2]. Therefore, sperm parameters assessed early before cryopreservation may predict the post-thawing recovery with high precision [3,4]. Moreover, specialists are more likely to counsel younger female patients on the reproductive risks before initiation of chemotherapies, as recently reported in a large study published on JAMA, and globally less than 45% of patients received counseling concerning the fertility lowering associated with treatments, showing a strong sex gap, with males reaching counseling only 32% of the time [5].
Testicular tumors and lymphomas are among the most frequent malignancies in males during their reproductive age, and thanks to an early diagnosis and the improved efficacy of novel target chemotherapies, the survival rate has significantly increased; however, in the youngest patients, the great psychological problem of treatment-associated infertility remains [6,7,8]. Consequently, the best option to preserve male fertility is semen cryopreservation performed before any antineoplastic treatment, even in presence of abnormal semen parameters, since the intracytoplasmic sperm injection (ICSI) technique allows the use of a single sperm cell to successfully fertilize one egg. Damaging effects of cancer on fertility have been reported [9], and most malignancies, especially Hodgkin’s lymphoma (HL), extragonadal germinoma, and testicular tumors, are often associated with severely altered semen parameters [10,11]. In this regard, several studies documented the negative effects that these forms of neoplasm directly have on spermatogenesis, compromising sperm quality even before any anti-neoplastic treatment starts. Serious histological alterations have been reported in these patients at the level of the epithelium of the seminiferous tubules, probably due to indirect actions on the hypothalamic–pituitary–gonad axis or direct epigenetic effects on the testicular compartment [11,12,13,14,15,16,17,18,19]. However, the specific epigenetics and molecular effects that also other types of tumors have on spermatogenesis remain rather controversial topics [7,8,11,12,13,14,15,16,17,18,19,20,21,22].
In the present study, the main semen parameters of different cancer patients were evaluated before any radio-chemotherapy treatment or cryopreservation and after thawing to assess viability and progressive motility recovery stratified by the different solid and hematological cancers.

2. Materials and Methods

2.1. Patient Recruitment

Laboratory findings from the database of the Gamete Cryopreservation Center of Annunziata Hospital (CS; Italy) from 2011–2020 were anonymously and retrospectively retrieved, and a total of 254 cancer patients aged 18–56 were analyzed. Cancers were divided into three subgroups according to the specific neoplasia: testicular cancers (TC; n = 135), hematological cancers (HC; n = 76), and other types of solid cancers (OC; n = 43). Data were collected and processed based on the cancer type, age, and quality of the patients’ seminal fluid at the time of cancer diagnosis. Intra-group analyses and computation were performed and the whole data set was compared to the WHO reference indicators. Sperm motility after thawing was assessed for the whole group and the subgroups.
The present study was performed according to the guidelines of the Declaration of Helsinki, and ethical approval was waived since: (i) data were anonymously retrieved and analyzed, (ii) data were obtained during routine clinical visits, (iii) the results did not have any impact on the standard procedure of semen cryopreservation, (iv) the directive staff of the clinic were informed about the ongoing study. Moreover, all the recruited patients signed informed consent to participate in the study.

2.2. Semen Quality Assessment

The global evaluation of the seminal parameters included: total number of spermatozoa and number/mL, total and progressive motility, viability, and the percentage of normal morphology in compliance with the WHO 2021 guidelines (https://www.who.int/publications/i/item/9789240030787; accessed on 1 October 2023). Correlations between the seminal parameters and the age of patients stratified by the different types of malignances were also investigated. Pre- and post-cryopreservation mean comparisons and correlations were performed in the whole group and subgroups of cases. Semen samples were collected in a sterile receptacle after 3–5 days of sexual abstinence. After 20–30 min of liquefaction at 37 °C, semen samples were examined for sperm concentration, total sperm count, total and progressive motility (%), viability (%), and morphology (% of typical forms) in a Makler chamber (Sefi Medical Instruments, Ltd. Tel Aviv, Israel). Semen fluid examinations were carried out by the same specialist operator.

2.2.1. Motility Assessment

Sperm motility was evaluated using a phase contrast microscope at 400× magnification. The percentage of motile spermatozoa was evaluated according to WHO guidelines (2021 edition). Basically, four categories of sperm were classified: rapid progressive, slow progressive, non-progressive, and immotile. As a rule, at least 200 spermatozoa in at least five microscope fields were counted for each sample.

2.2.2. Morphology Assessment

Sperm morphology was assessed by smearing 5–10 μL of the sample onto clean glass slides, which was then allowed to air-dry for 20 min at room temperature. Glass slides were stained using smears in the Diff-Quick Kit (Baxter Dade diagnostics AG, Dubingen, Switzerland). Sperm morphology was assessed at 1000· microscope magnification under oil-immersion.

2.3. Pre-Freezing and Freezing Procedures

All analyses were carried out on swim-up prepared ejaculate. Fertilization medium (FertiCult ™ Flushing Medium FertiPro; MBT, Italy) was the basic medium for spermatozoa preparation. Briefly, raw ejaculate was diluted 1:1 (v/v) in the medium and then was pelleted at 400 g × 10 min and the supernatant was discarded. This step was followed by extreme caution; without disturbing the pellet, 0.5–1.0 mL of fresh medium was added and incubated (45 min, 37 °C) according to Meseguer et al., 2006 [23]. After swim-up, sperm suspensions were recovered and rapidly frozen. Samples were initially mixed dropwise in equal volumes of glycerol-based cryoprotectant (Sperm Freezing Medium, Medicult), the most widely used permeating cryoprotectant for human sperm; then, they were gently mixed with continuous shaking at room temperature and maintained at 37 °C for 10–15 min to allow a proper mixing between the cells and the medium. Samples were packaged in frozen paillettes (CBS High Security sperm straw; Cryo Bio System; L’Aigle, France), placed in plastic storage tubes (mini-goblet), and inserted in larger storage goblets. The entire procedure was performed in a class-A classified room (at least D) according to international good manufacturing practice guidelines by utilizing a slow freeze control system consisting of progressive cooling at 1.5 °C/min (from 20 °C to −6 °C), and then at −6 °C/min to −80 °C. Finally, the paillettes were quickly transferred into liquid nitrogen (−196 °C) and stored.
Thawing was carried out by keeping the straws for 15 min at 37 °C in a dedicated dry incubator before further processing.

2.4. Statistical Analysis

Statistical analysis was carried out using SPSS (v-22) and GraphPad Prism (v-9.5.0), and all the variables shown were expressed as the mean  ±  SD or percentage. Findings related to sperm quality were analyzed using the Chi-squared test, Student’s t-test, or one-way ANOVA. The Pearson correlation test was utilized in the pre- and post-cryopreservation analyses of the whole group and subgroups, and the age-related dynamics of all the semen parameters were centered and scaled according to the formula (x-value − mean value)/SD [Z = (x − μ)/σ]. All the statistical analyses were two-sided, and p-values < 0.05 were considered statistically significant.

3. Results

3.1. Epidemiological Data and Clinical Characteristics

Among a cohort of 254 patients, the most represented cancer was testicular cancer (n = 135; 53.1%; among these patients, 92 had seminoma, 68.1%), followed by hematological cancers (n = 76; 29.9%; among these patients, 37 had HL, 48.7%) and other types of solid cancers (n = 43; 17%; equally distributed among colon, lung, prostate, and bladder).
Figure 1 shows the overall distribution of the entire cohort, depicting the relative frequency of the different cancers from 2011–2020, showing testicular cancer characterized by the highest number of patients compared to the rest of the cancers.
Figure 2 shows the number of patients stratified according to clusters of age that was significantly overrepresented below 35 years when compared with the remaining clusters of age (p-value intra-group comparison, p = 0.001), essentially reflecting the reproductive targets of younger males of active reproductive age (75.9% vs. 24.1%; p = 0.01).
Approximately 15% of patients used their cryopreserved sperm for ART procedures, 5.5% of patients communicated to the Centre to discard their samples because they did not need cryopreservation anymore, and 6.5% of patients died due to cancer.

3.2. Semen Quality Assessment in Cancer Subgroups

Computation and analyses among semen parameters stratified by the various cancer types ascribed the lowest mean number of spermatozoa to testicular cancer patients when compared with the rest of the patients computed together (TC: 15.3 × 106/mL vs. HC + OC: 27.92 × 106/mL; p = 0.0005), with a large number of TC cases (70.37%) below the decision limit indicated by the internationally accepted WHO criteria. Therefore, at a greater extent than for other cancer subgroups, the value expressed as percentage of patients below the normal reference number of 15 × 106/mL was significantly overrepresented in testis cancer when compared with the remaining subgroups (Table 1).
We then compared sperm quality findings in the whole group and subgroups, considering the WHO reference values (Table 2). Whilst ejaculate volumes were within the WHO parameters for any group of patients, the number of spermatozoa/mL was below the reference for all the groups, with testis cancers characterized by the lowest number (15.3 mL × 106 ± 23.0) when compared with other cancers (p = 0.0014) or hematological cancers (p = 0.004). In addition, spermatozoa progressive motility gave the following results: testicular cancer 5.7% ± 7.9; hematological cancers 8.02% ± 9.4 (HL had slight lower motility: 7.02% vs. 8.97%); other cancers 7.1% ± 10.6. This means that testis cancers had the lowest mean value.
Remarkably, morphological analyses expressed as a percentage of the normal phenotype gave mean values above 4% (strict criterion) for all subgroups as follows: testicular cancer 6.59% ± 7.5, hematological cancers 8.75% ± 8.7 (HL had slightly higher rate: 9.54% vs. 8.0%), and other cancers 9.56% ± 9.8, (p < 0.01 testis cancers vs. the remaining two subgroups computed together). The wide range of values, extending from 0–35% in hematological cancers and other cancers and from 0–29% in testis cancers, accounts for the high values of SD found in all the subgroups.
As expected by our previous single analyses, testis cancer patients showed anomalous mean values almost for all the quality parameters considered, reaching significant severest mean values or percentages when compared with the other subgroups. Interestingly, cancers not directly affecting the urogenital system also showed abnormal values when compared with the international WHO reference standards, suggesting sperm cryopreservation and preventive biobanking are also useful strategies for these categories of patients.

3.3. Comparison and Correlation between Pre- and Post-Cryopreservation Semen Parameters

Viability and progressive motility were compared in the whole cohort and in subgroups for samples with concentrations ≥2.0 × 106/mL at baseline (Figure 3A,B). Both parameters were significantly lower after thawing in the whole cohort (p < 0.001) and in the subgroups (p = 0.01). Globally, the whole cohort of cancer patients had a recovery of 46.8% for viability (HC: 47.7%; OC: 46.8%; TC: 45.9%) and of 17.2% for progressive motility (HC: 18.0%; OC: 16.7%; TC: 15.2%). The recovery for the progressive motility of samples with baseline semen concentrations <2.0 × 106/mL was near 0.0% in all the subgroups.
By computing the percentages of pre- and post-cryopreservation viability and progressive motility, significant direct correlations were found in the whole cohort and in the TC subgroup (Table 3); no correlation was found in HC and OC subgroups, probably because of the lower number of cases.

3.4. Age-Dependent Impact on Semen Quality

To investigate the possible impact of patients’ ages on their semen quality, we correlated sperm parameters with age in the whole group and subgroups. Accordingly, correlation analyses yielded borderline or non-significant results, as summarized in Figure 4 (whole group), which shows trend lines for all the sperm quality parameters and the R2-values. In detail, for the whole group, a borderline correlation between age and sperm count was found (R2 = 0.0204; p = 0.05), together with a non significant inverse correlation between age and ejaculate volume (R2 = 0.0059; p = n.s.), responsible for a possible combined effect on the final sperm concentration trend. The remaining parameters did not show statistical significance, although, as discussed above, the TC subgroup had the lowest trends for each comparison and parameter considered.

4. Discussion

Semen analysis represents a common laboratory basic evaluation of male infertility conditions. Cryopreservation of semen started as clinical practice in the early 1960s and is today performed using different strategies. From the first edition in 1980 to the current sixth edition (https://www.who.int/publications/i/item/9789240030787; accessed on 1 October 2023), there have been important changes and advances in semen examination methods and cryopreservation, improving quality control and assurance [24]. Semen parameter assessment is based on the standards and references listed within the laboratory manual for human semen examination and processing [25,26,27]. Exogenous or endogenous situations can affect sperm quality, and the consequences of cancer and cryopreservation for sperm functions are well known. Spermatozoa can be differently damaged and reflect different freezing–thawing consequences on vital parameters. In this scenario, a personalized semen characterization before cryopreservation stratified by the diverse cancers should be obtained.
The main results observed by the present investigation are that any cancer condition can affect the parameters of semen quality compared to the WHO references, and that this is also reflected in the recovery of functions after sperm thawing regardless of the mean age of patients being comparable among subgroups.
The percentage of patients with sperm counts below the WHO reference value (i.e., ≤15 × 106/mL) was 50% and 56% in HC and OC subgroups, respectively, while 70% of TC patients were below the reference. In detail, TC patients also showed the lowest mean parameters when compared with the other subgroups, particularly for sperm number and total motility, which were about 40% of the respective WHO reference ranges. Conversely, HC and OC retained, respectively, 69% and 75% of the reference concentrations, while comparable mean values were retained for total motility (HC, 51.6%; OC, 45.4%; and TC, 38.1%). Interestingly, progressive motility was severely affected in all the subgroups, which retained low residual motility (HC, 26.7%; OC, 23.6%; and TC, 19.0%) compared to the reference value. Finally, as the mean percentage of viability and morphology is concerned, all the subgroups crossed over the limits of the reference cutoffs.
It has been recently reported that there is a lack of detailed information on motility and viability recovery after semen thawing and whether recovery varies among the different cancers or other pathologies requiring sperm banking [1].
We found that both parameters were significantly lower after thawing in the whole cohort and in the subgroups, globally accounting for 46.8% for viability (HC, 47.7%; OC, 46.8%; and TC, 45.9%) and 17.2% for progressive motility (HC, 18.0%; OC, 16.7%; TC, 15.2%). Additionally, considering a comparable percentage of recovery among the subgroups, particular consideration should be given towards TC patients characterized by halved baseline sperm concentrations compared to HC or OC subgroups, predicting in turn a lower absolute recovery. TC patients are characterized by a direct endogenous exposure of germ cells to the tumor environment enriched by high levels of inflammatory mediators and ROS, and this might greatly affect sperm cells’ global viability due to aberrant epigenomic signals affecting the biological tissue age compared to the classical effects of chronological aging [28,29].
In an explorative attempt, we also investigated whether the considered semen quality findings could be affected by age in the whole group and in the different subgroups. Globally, the ejaculate volume decreased in all subgroups as the age of patients increased, while sperm counts slightly increased, with no significant effects of age observed on the rest of the measured parameters, finally confirming that the TC subgroup had the lowest trends for each comparison and parameter considered.
The main alterations in semen parameters at the time of cancer diagnosis are still being studied by various authors with the aim of improving personalized reproductive chances and preserving male fertility [22,30,31,32]. Various hypotheses have been formulated, and considering testicular tumors, these alterations could be related to the histological type and probably to three main factors: i) biochemical and epigenetic direct damages from the cancer environment to the testicular parenchyma and spermatozoa (also mediated by miRNA and ncRNA); ii) altered endocrine balance; and iii) autoimmunity effects [28,33,34]. As hematological and solid cancer is concerned, the causative alterations in the spermatozoa functions might specifically be due to systemic effects, hypothesized to most likely increase pro-inflammatory circulating cytokines and interleukins in this class of patients [35,36]. Strategies aimed to contrast local oxidative stress and inflammation may positively improve basal- and post-cryopreservation parameters, especially for semen samples with suboptimal features [37,38].
Other non-negligible elements may be related to stressor factors, as a significant psychological component linked to the patient’s awareness of having a cancer disease, which adds to the global semen status already affected by unbalanced individual red-ox homeostasis, particularly at the mitochondria level [39,40,41,42]. As for any cell or tissue involved in complex diseases [43,44,45], in any kind of cancer, different molecular clusters of somatic mutations or inherited predispositions of key genes responsible for the dysregulation of the local redox and metal-homeostasis [46,47] may affect sperm viability and functions, finally disturbing spermatogenesis [2,48,49,50].
Cancer affects the entire body in many negative ways, systemically and locally, and this strongly impairs normal gametogenesis both in males and female. How cancer affects male and female fertility and gametes quality is still partially understood, suggesting the need for multi-OMICs approaches to investigations considering infertility, a complex disease in which males and females show definite features that need to be specifically addressed, and also including genomic and epigenomics investigations [51,52,53,54].
Further multicenter studies including larger case series are needed to better investigate the effects that different neoplastic mutations can exert on sperm cell viability and quality; our results further underline and reinforce the importance of specifically characterizing the basal semen parameters in any cancer to perform early actions and to contrast the modifiable factors in order to preserve and improve fertility scores in these frail patients before any antineoplastic treatments start [31,32,55,56].
Considering that the effects of any malignancy on semen parameters are somewhat debatable, in line with the results reported in the present investigation, it has been established that any type of cancer can significantly impair sperm quality parameters, with effects occasionally still within the normal or borderline range [9].
In addition, one of the most common concerns of patients with cancer, particularly testicular cancer, is the quality of the cryopreserved sperm, which is also affected by individual susceptibility to cryodamage. This may have effects on the types of available assisted reproductive technology (ART), since recent advances in ART have enabled males who were in the past considered infertile to successfully father biological offspring. In this line, the combination of IVF and ICSI has allowed the injection of a single sperm directly into the cytoplasm of one egg. Therefore, unfavorable semen parameters might not affect fertilization or conception rates after appropriate cryopreservation as long as one live sperm can be recovered.
Since the epigenome of spermatozoa can be altered in cancer survivors, primarily after chemo-treatment cycles, and, theoretically, modifications of the sperm epigenome may have possible transgenerational transmission [57,58], it is imperative that oncologists and urologists become more familiar with the several personalized options of fertility preservation and inform patients concerning cancer-associated fertility risks.
Although the data obtained by the present investigation have the limitation of a small number of recruited cases, they are in line with other studies and strengthen the indication that it is beneficial to obtain a personalized semen assessment before treatment and early cryopreservation to avoid lower recovery and potential transgenerational transmission of dangerous epigenomic signatures.

5. Conclusions

Cancer, chemotherapy, and other cancer-related treatments have been associated with impairment or loss of future patient fertility, with possible trans-generational transmission. Accordingly, at the time of diagnosis and before the initiation of chemotherapy, detailed counseling explaining the risks must be undertaken. In a study recruiting approximately seven thousand patients, only 45% of cases in reproductive age had a discussion with specialists concerning the risk of infertility associated with treatment [5], a percentage that is too low according to the ASCO recommendations. Globally, there is the need to identify strategies to increase fertility education, and fertility specialists must be involved in counseling at the time of newly diagnosed tumors. Although oncologists recognize the association of chemotherapy and radiotherapy with fertility lowering, they are not well informed on the available fertile gamete preservation options. A stronger cooperation between oncologists, geneticists, and fertility specialists may be of value as a policy to meet the ASCO recommendations.

Author Contributions

Conceptualization, F.S. and G.P.; methodology, G.P.; formal analysis, D.G., V.T., A.V.S. and G.P.; investigation, D.G., V.T., A.V.S. and G.P.; data curation, G.P.; writing—original draft preparation, D.G., G.P., V.T. and F.S.; writing—review and editing, D.G., G.P., V.T., A.V.S. and F.S. All authors have read and agreed to the published version of the manuscript.

Funding

Fondo Ateneo Ricerca (FAR) and Fondo Incentivazione Ricerca (FIR) from D.G. and V.T.

Institutional Review Board Statement

This study was conducted in accordance with the Declaration of Helsinki, and the ethical approval was waived since: (i) data were anonymously retrieved and analyzed, (ii) data were obtained during routine clinical visits, (iii) the results did not have any impact on the standard procedure of semen cryopreservation, and (iv) the directive staff of the clinic were informed on the ongoing study. Moreover, all the recruited patients signed informed consent to participate in the study.

Informed Consent Statement

Written informed consent was obtained from the patients.

Data Availability Statement

The original contributions presented in the study are included in the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marchiani, S.; Degl’Innocenti, S.; Dabizzi, S.; Tamburrino, L.; Fino, M.G.; Traini, G.; Calamai, C.; Maggi, M.; Vignozzi, L.; Baldi, E.; et al. Semen Cryopreservation for Men Banking for Oligozoospermia, Cancers, and Other Conditions: 24 Years’ Experience of an Italian Bank. J. Clin. Med. 2023, 12, 4657. [Google Scholar] [CrossRef]
  2. Panner Selvam, M.K.; Finelli, R.; Baskaran, S.; Agarwal, A. Dysregulation of Key Proteins Associated with Sperm Motility and Fertility Potential in Cancer Patients. Int. J. Mol. Sci. 2020, 21, 6754. [Google Scholar] [CrossRef] [PubMed]
  3. Degl’Innocenti, S.; Filimberti, E.; Magini, A.; Krausz, C.; Lombardi, G.; Fino, M.G.; Rastrelli, G.; Maggi, M.; Baldi, E. Semen cryopreservation for men banking for oligospermia, cancers, and other pathologies: Prediction of post-thaw outcome using basal semen quality. Fertil. Steril. 2013, 100, 1555–1563. [Google Scholar] [CrossRef] [PubMed]
  4. Tamburrino, L.; Traini, G.; Marcellini, A.; Vignozzi, L.; Baldi, E.; Marchiani, S. Cryopreservation of Human Spermatozoa: Functional, Molecular and Clinical Aspects. Int. J. Mol. Sci. 2023, 24, 4656. [Google Scholar] [CrossRef]
  5. Patel, P.; Kohn, T.P.; Cohen, J.; Shiff, B.; Kohn, J.; Ramasamy, R. Evaluation of Reported Fertility Preservation Counseling Before Chemotherapy Using the Quality Oncology Practice Initiative Survey. JAMA Netw. Open 2020, 3, e2010806. [Google Scholar] [CrossRef]
  6. Amirjannati, N.; Sadeghi, M.; Hosseini Jadda, S.H.; Ranjbar, F.; Kamali, K.; Akhondi, M.A. Evaluation of semen quality in patients with malignancies referred for sperm banking before cancer treatment. Andrologia 2011, 43, 317–320. [Google Scholar] [CrossRef] [PubMed]
  7. Bizet, P.; Saias-Magnan, J.; Jouve, E.; Grillo, J.M.; Karsenty, G.; Metzler-Guillemain, C.; Perrin, J. Sperm cryopreservation before cancer treatment: A 15-year monocentric experience. Reprod. Biomed. Online 2012, 24, 321–330. [Google Scholar] [CrossRef]
  8. Bonetti, T.C.; Pasqualotto, F.F.; Queiroz, P.; Iaconelli, A., Jr.; Borges, E., Jr. Sperm banking for male cancer patients: Social and semen profiles. Int. Braz. J. Urol. 2009, 35, 190–197, discussion 197–198. [Google Scholar] [CrossRef]
  9. Shrem, G.; Azani, L.; Feferkorn, I.; Listovsky, T.; Hussaini, S.; Farber, B.; Dahan, M.H.; Salmon-Divon, M. Effect of Malignancy on Semen Parameters. Life 2022, 12, 922. [Google Scholar] [CrossRef]
  10. Williams, D.H.t.; Karpman, E.; Sander, J.C.; Spiess, P.E.; Pisters, L.L.; Lipshultz, L.I. Pretreatment semen parameters in men with cancer. J. Urol. 2009, 181, 736–740. [Google Scholar] [CrossRef]
  11. Crawshaw, M.A.; Glaser, A.W.; Hale, J.P.; Sloper, P. Young males’ experiences of sperm banking following a cancer diagnosis-a qualitative study. Hum. Fertil. 2008, 11, 238–245. [Google Scholar] [CrossRef]
  12. Lee, S.J.; Schover, L.R.; Partridge, A.H.; Patrizio, P.; Wallace, W.H.; Hagerty, K.; Beck, L.N.; Brennan, L.V.; Oktay, K.; American Society of Clinical Oncology. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J. Clin. Oncol. 2006, 24, 2917–2931. [Google Scholar] [CrossRef] [PubMed]
  13. Neal, M.S.; Nagel, K.; Duckworth, J.; Bissessar, H.; Fischer, M.A.; Portwine, C.; Tozer, R.; Barr, R.D. Effectiveness of sperm banking in adolescents and young adults with cancer: A regional experience. Cancer 2007, 110, 1125–1129. [Google Scholar] [CrossRef]
  14. Tal, R.; Botchan, A.; Hauser, R.; Yogev, L.; Paz, G.; Yavetz, H. Follow-up of sperm concentration and motility in patients with lymphoma. Hum. Reprod. 2000, 15, 1985–1988. [Google Scholar] [CrossRef] [PubMed]
  15. Giwercman, A.; Petersen, P.M. Cancer and male infertility. Baillieres Best. Pract. Res. Clin. Endocrinol. Metab. 2000, 14, 453–471. [Google Scholar] [CrossRef] [PubMed]
  16. Liguori, G.; Trombetta, C.; Bucci, S.; Benvenuto, S.; Amodeo, A.; Ocello, G.; Belgrano, E. Semen quality before and after orchiectomy in men with testicular cancer. Arch. Ital. Urol. Androl. 2008, 80, 99–102. [Google Scholar] [PubMed]
  17. Eisenberg, M.L.; Betts, P.; Herder, D.; Lamb, D.J.; Lipshultz, L.I. Increased risk of cancer among azoospermic men. Fertil. Steril. 2013, 100, 681–685. [Google Scholar] [CrossRef] [PubMed]
  18. Loren, A.W.; Mangu, P.B.; Beck, L.N.; Brennan, L.; Magdalinski, A.J.; Partridge, A.H.; Quinn, G.; Wallace, W.H.; Oktay, K.; American Society of Clinical Oncology. Fertility preservation for patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J. Clin. Oncol. 2013, 31, 2500–2510. [Google Scholar] [CrossRef]
  19. van Casteren, N.J.; Boellaard, W.P.; Romijn, J.C.; Dohle, G.R. Gonadal dysfunction in male cancer patients before cytotoxic treatment. Int. J. Androl. 2010, 33, 73–79. [Google Scholar] [CrossRef]
  20. Agarwal, A.; Ranganathan, P.; Kattal, N.; Pasqualotto, F.; Hallak, J.; Khayal, S.; Mascha, E. Fertility after cancer: A prospective review of assisted reproductive outcome with banked semen specimens. Fertil. Steril. 2004, 81, 342–348. [Google Scholar] [CrossRef]
  21. Dunn, H.G.; McBurney, A.K.; Ingram, S.; Hunter, C.M. Maternal cigarette smoking during pregnancy and the child’s subsequent development: I. Physical growth to the age of 6 1/2 years. Can. J. Public. Health 1976, 67, 499–505. [Google Scholar]
  22. Jacobsen, R.; Bostofte, E.; Engholm, G.; Hansen, J.; Olsen, J.H.; Skakkebaek, N.E.; Moller, H. Risk of testicular cancer in men with abnormal semen characteristics: Cohort study. BMJ 2000, 321, 789–792. [Google Scholar] [CrossRef]
  23. Meseguer, M.; Molina, N.; Garcia-Velasco, J.A.; Remohi, J.; Pellicer, A.; Garrido, N. Sperm cryopreservation in oncological patients: A 14-year follow-up study. Fertil. Steril. 2006, 85, 640–645. [Google Scholar] [CrossRef]
  24. Kandil, H.; Agarwal, A.; Saleh, R.; Boitrelle, F.; Arafa, M.; Vogiatzi, P.; Henkel, R.; Zini, A.; Shah, R. Editorial Commentary on Draft of World Health Organization Sixth Edition Laboratory Manual for the Examination and Processing of Human Semen. World J. Mens. Health 2021, 39, 577–580. [Google Scholar] [CrossRef] [PubMed]
  25. Barratt, C.L.R.; Bjorndahl, L.; De Jonge, C.J.; Lamb, D.J.; Osorio Martini, F.; McLachlan, R.; Oates, R.D.; van der Poel, S.; St John, B.; Sigman, M.; et al. The diagnosis of male infertility: An analysis of the evidence to support the development of global WHO guidance-challenges and future research opportunities. Hum. Reprod. Update 2017, 23, 660–680. [Google Scholar] [CrossRef] [PubMed]
  26. Guzick, D.S.; Overstreet, J.W.; Factor-Litvak, P.; Brazil, C.K.; Nakajima, S.T.; Coutifaris, C.; Carson, S.A.; Cisneros, P.; Steinkampf, M.P.; Hill, J.A.; et al. Sperm morphology, motility, and concentration in fertile and infertile men. N. Engl. J. Med. 2001, 345, 1388–1393. [Google Scholar] [CrossRef] [PubMed]
  27. Schlegel, P.N.; Sigman, M.; Collura, B.; De Jonge, C.J.; Eisenberg, M.L.; Lamb, D.J.; Mulhall, J.P.; Niederberger, C.; Sandlow, J.I.; Sokol, R.Z.; et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline part I. Fertil. Steril. 2021, 115, 54–61. [Google Scholar] [CrossRef]
  28. Nunez-Corona, D.; Contreras-Sanzon, E.; Puente-Rivera, J.; Arreola, R.; Camacho-Nuez, M.; Cruz Santiago, J.; Estrella-Parra, E.A.; Torres-Romero, J.C.; Lopez-Camarillo, C.; Alvarez-Sanchez, M.E. Epigenetic Factors and ncRNAs in Testicular Cancer. Int. J. Mol. Sci. 2023, 24, 2194. [Google Scholar] [CrossRef] [PubMed]
  29. Donatti, L.M.; Martello, C.L.; Andrade, G.M.; Oliveira, N.P.; Frantz, N. Advanced Paternal Age Affects the Sperm DNA Fragmentation Index and May Lead to Lower Good-quality Blastocysts. Reprod. Sci. 2023, 30, 2489–2494. [Google Scholar] [CrossRef]
  30. van Casteren, N.J.; van Santbrink, E.J.; van Inzen, W.; Romijn, J.C.; Dohle, G.R. Use rate and assisted reproduction technologies outcome of cryopreserved semen from 629 cancer patients. Fertil. Steril. 2008, 90, 2245–2250. [Google Scholar] [CrossRef]
  31. Shankara-Narayana, N.; Di Pierro, I.; Fennell, C.; Ly, L.P.; Bacha, F.; Vrga, L.; Savkovic, S.; Turner, L.; Jayadev, V.; Conway, A.J.; et al. Sperm cryopreservation prior to gonadotoxic treatment: Experience of a single academic centre over 4 decades. Hum. Reprod. 2019, 34, 795–803. [Google Scholar] [CrossRef] [PubMed]
  32. Liu, X.; Liu, B.; Liu, S.; Xian, Y.; Zhao, W.; Zhou, B.; Xiao, X.; Wang, L.; Zhu, X.; Shu, B.; et al. Male cancer patient sperm cryopreservation for fertility preservation: 10-year monocentric experience. Basic. Clin. Androl. 2021, 31, 24. [Google Scholar] [CrossRef] [PubMed]
  33. Rajpert-De Meyts, E.; Aksglaede, L.; Bandak, M.; Toppari, J.; Jorgensen, N. Testicular Cancer: Pathogenesis, Diagnosis and Management with Focus on Endocrine Aspects. In Endotext; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  34. Cannarella, R.; Gul, M.; Rambhatla, A.; Agarwal, A. Temporal decline of sperm concentration: Role of endocrine disruptors. Endocrine 2023, 79, 1–16. [Google Scholar] [CrossRef] [PubMed]
  35. Masliukaite, I.; Ntemou, E.; Feijen, E.A.M.; van de Wetering, M.; Meissner, A.; Soufan, A.T.; Repping, S.; Kremer, L.M.C.; Jahnukainen, K.; Goossens, E.; et al. Childhood cancer and hematological disorders negatively affect spermatogonial quantity at diagnosis: A retrospective study of a male fertility preservation cohort. Hum. Reprod. 2023, 38, 359–370. [Google Scholar] [CrossRef] [PubMed]
  36. Cosci, I.; Grande, G.; Di Nisio, A.; Rocca, M.S.; Del Fiore, P.; Benna, C.; Mocellin, S.; Ferlin, A. Cutaneous Melanoma and Hormones: Focus on Sex Differences and the Testis. Int. J. Mol. Sci. 2022, 24, 599. [Google Scholar] [CrossRef] [PubMed]
  37. Kumar, A.; Kumar Ghosh, S.; Katiyar, R.; Gemeda, A.E.; Rautela, R.; Bisla, A.; Srivastava, N.; Kumar Bhure, S.; Devi, H.L.; Chandra, V. Supplementation of Mito TEMPO and acetovanillone in semen extender improves freezability of buffalo spermatozoa. Andrology 2022, 10, 775–788. [Google Scholar] [CrossRef] [PubMed]
  38. Tisato, V.; Zauli, G.; Rimondi, E.; Gianesini, S.; Brunelli, L.; Menegatti, E.; Zamboni, P.; Secchiero, P. Inhibitory effect of natural anti-inflammatory compounds on cytokines released by chronic venous disease patient-derived endothelial cells. Mediators Inflamm. 2013, 2013, 423407. [Google Scholar] [CrossRef]
  39. Szabo, A.; Vancsa, S.; Hegyi, P.; Varadi, A.; Forintos, A.; Filipov, T.; Acs, J.; Acs, N.; Szarvas, T.; Nyirady, P.; et al. Lifestyle-, environmental-, and additional health factors associated with an increased sperm DNA fragmentation: A systematic review and meta-analysis. Reprod. Biol. Endocrinol. 2023, 21, 5. [Google Scholar] [CrossRef]
  40. Nouman, H.; Zanbar, L.P. Support or stressor? The community as a predictor of perceptions of infertility. Soc. Work. Health Care 2020, 59, 650–667. [Google Scholar] [CrossRef]
  41. Thanscheidt, C.L.; Patsch, P.; Rosner, S.; Germeyer, A.; Krause, M.; Kentenich, H.; Siercks, I.; Haberlin, F.; Ehrbar, V.; Tschudin, S.; et al. Psychological Aspects of Infertility-Results from an Actor-Partner Interdependence Analysis. Geburtshilfe Frauenheilkd. 2023, 83, 843–849. [Google Scholar] [CrossRef]
  42. Sharma, P.; Kaushal, N.; Saleth, L.R.; Ghavami, S.; Dhingra, S.; Kaur, P. Oxidative stress-induced apoptosis and autophagy: Balancing the contrary forces in spermatogenesis. Biochim. Biophys. Acta Mol. Basis Dis. 2023, 1869, 166742. [Google Scholar] [CrossRef]
  43. Tisato, V.; Zuliani, G.; Vigliano, M.; Longo, G.; Franchini, E.; Secchiero, P.; Zauli, G.; Paraboschi, E.M.; Vikram Singh, A.; Serino, M.L.; et al. Gene-gene interactions among coding genes of iron-homeostasis proteins and APOE-alleles in cognitive impairment diseases. PLoS ONE 2018, 13, e0193867. [Google Scholar] [CrossRef]
  44. Castiglione, A.; Ciorba, A.; Aimoni, C.; Orioli, E.; Zeri, G.; Vigliano, M.; Gemmati, D. Sudden sensorineural hearing loss and polymorphisms in iron homeostasis genes: New insights from a case-control study. Biomed. Res. Int. 2015, 2015, 834736. [Google Scholar] [CrossRef] [PubMed]
  45. Gemmati, D.; Zeri, G.; Orioli, E.; De Gaetano, F.E.; Salvi, F.; Bartolomei, I.; D’Alfonso, S.; Dall’osso, C.; Leone, M.A.; Singh, A.V.; et al. Polymorphisms in the genes coding for iron binding and transporting proteins are associated with disability, severity, and early progression in multiple sclerosis. BMC Med. Genet. 2012, 13, 70. [Google Scholar] [CrossRef]
  46. Zamboni, P.; Gemmati, D. Clinical implications of gene polymorphisms in venous leg ulcer: A model in tissue injury and reparative process. Thromb. Haemost. 2007, 98, 131–137. [Google Scholar]
  47. Parmeggiani, F.; Costagliola, C.; Gemmati, D.; D’Angelo, S.; Perri, P.; Scapoli, G.L.; Catozzi, L.; Federici, F.; Sebastiani, A.; Incorvaia, C. Predictive role of coagulation-balance gene polymorphisms in the efficacy of photodynamic therapy with verteporfin for classic choroidal neovascularization secondary to age-related macular degeneration. Pharmacogenet Genom. 2007, 17, 1039–1046. [Google Scholar] [CrossRef] [PubMed]
  48. Tanwar, P.S.; Kaneko-Tarui, T.; Zhang, L.; Teixeira, J.M. Altered LKB1/AMPK/TSC1/TSC2/mTOR signaling causes disruption of Sertoli cell polarity and spermatogenesis. Hum. Mol. Genet. 2012, 21, 4394–4405. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, K.Q.; Wei, B.H.; Hao, S.L.; Yang, W.X. The PI3K/AKT signaling pathway: How does it regulate development of Sertoli cells and spermatogenic cells? Histol. Histopathol. 2022, 37, 621–636. [Google Scholar] [CrossRef]
  50. Yang, X.; Chen, Y.; Song, W.; Huang, T.; Wang, Y.; Chen, Z.; Chen, F.; Liu, Y.; Wang, X.; Jiang, Y.; et al. Review of the Role of Ferroptosis in Testicular Function. Nutrients 2022, 14, 5268. [Google Scholar] [CrossRef]
  51. Gemmati, D.; Tisato, V. Chapter 24-Genomic and epigenomic signature at the branch-point among genome, phenome, and sexome in health and disease: A multiomics approach. In Principles of Gender-Specific Medicine, 4th ed.; Legato, M.J., Ed.; Academic Press: Cambridge, MA, USA, 2023; pp. 393–408. [Google Scholar]
  52. Gemmati, D.; Varani, K.; Bramanti, B.; Piva, R.; Bonaccorsi, G.; Trentini, A.; Manfrinato, M.C.; Tisato, V.; Care, A.; Bellini, T. “Bridging the Gap” Everything that Could Have Been Avoided If We Had Applied Gender Medicine, Pharmacogenetics and Personalized Medicine in the Gender-Omics and Sex-Omics Era. Int. J. Mol. Sci. 2019, 21, 296. [Google Scholar] [CrossRef]
  53. Hardy, J.; Pollock, N.; Gingrich, T.; Sweet, P.; Ramesh, A.; Kuong, J.; Basar, A.; Jiang, H.; Hwang, K.; Vukina, J.; et al. Genomic testing for copy number and single nucleotide variants in spermatogenic failure. J. Assist. Reprod. Genet. 2022, 39, 2103–2114. [Google Scholar] [CrossRef] [PubMed]
  54. Beaud, H.; Tremblay, A.R.; Chan, P.T.K.; Delbes, G. Sperm DNA Damage in Cancer Patients. Adv. Exp. Med. Biol. 2019, 1166, 189–203. [Google Scholar] [CrossRef] [PubMed]
  55. Andrade, M.B.R.; Bertolla, R.P.; Intasqui, P.; Antoniassi, M.P.; Tibaldi, D.S.; Belardin, L.B.; Spaine, D.M. Effect of orchiectomy on sperm functional aspects and semen oxidative stress in men with testicular tumours. Andrologia 2019, 51, e13205. [Google Scholar] [CrossRef] [PubMed]
  56. Lackamp, N.; Wilkemeyer, I.; Jelas, I.; Keller, U.; Bullinger, L.; Stintzing, S.; le Coutre, P. Survey of Long-Term Experiences of Sperm Cryopreservation in Oncological and Non-Oncological Patients: Usage and Reproductive Outcomes of a Large Monocentric Cohort. Front. Oncol. 2021, 11, 772809. [Google Scholar] [CrossRef]
  57. Tremblay, A.; Beaud, H.; Delbès, G. Transgenerational impact of chemotherapy: Would the father exposure impact the health of future progeny? Gynecol. Obstet. Fertil. Senol. 2017, 45, 609–618. [Google Scholar] [CrossRef]
  58. Delessard, M.; Saulnier, J.; Rives, A.; Dumont, L.; Rondanino, C.; Rives, N. Exposure to Chemotherapy During Childhood or Adulthood and Consequences on Spermatogenesis and Male Fertility. Int. J. Mol. Sci. 2020, 21, 1454. [Google Scholar] [CrossRef]
Figure 1. Number of cases recorded from 2011–2020 stratified according to different cancer subgroups.
Figure 1. Number of cases recorded from 2011–2020 stratified according to different cancer subgroups.
Jpm 13 01654 g001
Figure 2. Patient age stratification among the different cancer subgroups.
Figure 2. Patient age stratification among the different cancer subgroups.
Jpm 13 01654 g002
Figure 3. Box plots of the distribution of pre- (Pre) and post-cryopreservation (Post) for viability (A) and progressive motility (B) stratified by different cancer subgroups. HC, hematological cancers; OC, other cancers; TC, testicular cancer.
Figure 3. Box plots of the distribution of pre- (Pre) and post-cryopreservation (Post) for viability (A) and progressive motility (B) stratified by different cancer subgroups. HC, hematological cancers; OC, other cancers; TC, testicular cancer.
Jpm 13 01654 g003
Figure 4. Correlation analyses of the age dependent effects on the sperm quality parameters in the whole cohort of patients. Variables were centered and scaled as described in the Section 2.
Figure 4. Correlation analyses of the age dependent effects on the sperm quality parameters in the whole cohort of patients. Variables were centered and scaled as described in the Section 2.
Jpm 13 01654 g004
Table 1. Number and percentage of cases with sperm counts below the WHO criteria.
Table 1. Number and percentage of cases with sperm counts below the WHO criteria.
Type of Cancer≤15 × 106/mL>15 × 106/mLp-Value *
a Testicular cancer (n = 135)70.37% (95)29.63% (40)0.003 (a vs. b)
b Hematol. cancers (n = 76)50.0% (38)50.0% (38)0.06 (a vs. c)
c Other cancers (n = 43)55.8% (24)44.2% (19)0.0028 (a vs. b + c)
* p-value was calculated using Chi-squared, comparing the number in brackets for each subgroup. a, b, and c, indicate Testicular cancer, Hematological cancers, and Other cancers subgroup respectively; a vs. b, a vs. c, and a vs. b + c, indicate the subgroups comparison.
Table 2. Semen quality parameters in the whole group and subgroups of cancers.
Table 2. Semen quality parameters in the whole group and subgroups of cancers.
Whole
Cohort
Hematol.
Cancers
Other
Cancers
Testicular Cancerp-ValueWHO 2021
Age, mean ± SD30.2 ± 7.628.4 ± 8.531.9 ± 9.230.7 ± 6.10.02 a
n.s. b
- -
Ejaculate volume, mL2.2 ± 1.32.03 ± 1.22.48 ± 1.52.2 ± 1.3n.s. a
n.s. b
1.4 (1.3–1.5)
Number/mL, ×10621.2 ± 28.726.9 ± 28.3 29.6 ± 39.915.3 ± 23.00.0014 a
0.004 b
39 (35–40)
Total motility, % 18.2 ± 17.821.7 ± 18.619.1 ± 2016.0 ± 16.30.02 a
0.07 b
42 (40–43)
Progressive motility, %6.6 ± 8.98.02 ± 9.47.1 ± 10.65.7 ± 7.90.04 a
0.05 b
30 (29–31)
Viability, %62.3 ± 18.767.1 ± 16.359.4 ± 18.460.3 ± 19.90.01 a
n.s. b
54 (50–56)
Morphology, %7.73 ± 8.48.75 ± 8.79.56 ± 9.86.59 ± 7.50.05 a
0.03 b
4 (3.9–4)
The p-values were calculated by comparing testis cancers with hematological cancers (a), and other cancer (b) subgroups, respectively.
Table 3. Correlation between viability and progressive motility pre- and post-cryopreservation.
Table 3. Correlation between viability and progressive motility pre- and post-cryopreservation.
Pre-Cryopreservation
Post-Cryopreservation Viability (%)Progressive Motility (%)
Whole cohortViability, %R2 = 0.35; p < 0.001R2 = 0.42; p < 0.001
Progressive motility, %R2 = 0.35; p < 0.001R2 = 0.45; p < 0.001
Hematological cancers (HC)Viability, %R2 = 0.12; p = n.s.R2 = 0.16; p = n.s.
Progressive motility, %R2 = 0.18; p = n.s.R2 = 0.13; p = n.s.
Other cancers (OC)Viability, %R2 = 0.09; p = n.sR2 = 0.06; p = n.s
Progressive motility, %R2 = 0.08; p = n.sR2 = 0.07; p = n.s
Testis cancer (TC)Viability, %R2 = 0.2; p < 0.05R2 = 0.18; p < 0.05
Progressive motility, %R2 = 0.16; p < 0.05R2 = 0.2; p < 0.05
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Peluso, G.; Tisato, V.; Singh, A.V.; Gemmati, D.; Scarpellini, F. Semen Cryopreservation to Expand Male Fertility in Cancer Patients: Intracase Evaluation of Semen Quality. J. Pers. Med. 2023, 13, 1654. https://doi.org/10.3390/jpm13121654

AMA Style

Peluso G, Tisato V, Singh AV, Gemmati D, Scarpellini F. Semen Cryopreservation to Expand Male Fertility in Cancer Patients: Intracase Evaluation of Semen Quality. Journal of Personalized Medicine. 2023; 13(12):1654. https://doi.org/10.3390/jpm13121654

Chicago/Turabian Style

Peluso, Giuseppina, Veronica Tisato, Ajay Vikram Singh, Donato Gemmati, and Fabio Scarpellini. 2023. "Semen Cryopreservation to Expand Male Fertility in Cancer Patients: Intracase Evaluation of Semen Quality" Journal of Personalized Medicine 13, no. 12: 1654. https://doi.org/10.3390/jpm13121654

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop