Skip to main content
Journal of Assisted Reproduction and Genetics logoLink to Journal of Assisted Reproduction and Genetics
. 2017 Oct 24;35(2):191–212. doi: 10.1007/s10815-017-1062-8

FDA-approved drugs that are spermatotoxic in animals and the utility of animal testing for human risk prediction

Elizabeth R Rayburn 1, Liang Gao 2, Jiayi Ding 3, Hongxia Ding 4, Jun Shao 3, Haibo Li 2,3,
PMCID: PMC5845034  PMID: 29063992

Abstract

Purpose

This study reviews FDA-approved drugs that negatively impact spermatozoa in animals, as well as how these findings reflect on observations in human male gametes.

Methods

The FDA drug warning labels included in the DailyMed database and the peer-reviewed literature in the PubMed database were searched for information to identify single-ingredient, FDA-approved prescription drugs with spermatotoxic effects.

Results

A total of 235 unique, single-ingredient, FDA-approved drugs reported to be spermatotoxic in animals were identified in the drug labels. Forty-nine of these had documented negative effects on humans in either the drug label or literature, while 31 had no effect or a positive impact on human sperm. For the other 155 drugs that were spermatotoxic in animals, no human data was available.

Conclusion

The current animal models are not very effective for predicting human spermatotoxicity, and there is limited information available about the impact of many drugs on human spermatozoa. New approaches should be designed that more accurately reflect the findings in men, including more studies on human sperm in vitro and studies using other systems (ex vivo tissue culture, xenograft models, in silico studies, etc.). In addition, the present data is often incomplete or reported in a manner that prevents interpretation of their clinical relevance. Changes should be made to the requirements for pre-clinical testing, drug surveillance, and the warning labels of drugs to ensure that the potential risks to human fertility are clearly indicated.

Keywords: Drug warning labels, Infertility, Reproductive toxicology, Spermatotoxicity

Introduction

It has been estimated that approximately 5–10% of men in developed countries are affected by infertility due to abnormalities in spermatozoa (number, morphology, motility, etc.) or semen (volume, composition, viscosity, etc.) [1]. There are numerous causes, including genetics, hormone imbalances, dietary insufficiencies, exposure to environmental or occupational toxicants, or the use/abuse of drugs (recreational, over-the-counter, or prescription). Given that approximately half of the US population has taken a prescription drug in the last month [2], prescription drugs may represent a common source of spermatotoxicity.

The present review focuses on the spermatotoxic effects (from the generation of spermatogonia to fertilization) of prescription drugs in various experimental animals. This review was prompted by a previous publication that examined the impact of FDA-approved drugs on human spermatogenesis [3]. In that study, frequent discrepancies were noted in the drug warning labels between the adverse effects of drugs on human and animal spermatogenesis, as well as some inconsistencies between the published peer-reviewed literature and the DailyMed database (a federal repository of drug warning labels). In addition to providing an overview of the spermatotoxicity of prescription drugs in animals, the review also provides a comparison to the reported human spermatotoxicity and addresses how the current data reflect the risk of these drugs to human sperm. This review also suggests ways to improve the current male reproductive toxicity testing and reporting of results.

Materials and methods

Database search and inclusion/exclusion criteria

The FDA indicates that any finding of toxicity should be reported on the drug labels in the “Warnings,” “Adverse Reactions,” and “Precautions” sections, as well as any other area where such a warning would be appropriate. The DailyMed database (http://dailymed.nlm.nih.gov/dailymed/index.cfm) is a federal database that contains approximately 85% of the US FDA-approved drug labels. This database represents the largest and most comprehensive database of the warning labels for FDA-approved drugs and is updated whenever a label changes.

Stemmed key words were used to search the full-text drug labels included in the DailyMed database (also available at: https://www.fda.gov/ScienceResearch/BioinformaticsTools/ucm289739.htm) as described in a previous study [4]. These stemmed keywords (deferen, epidid, interstitial, Leydig, semen, semin, Sertoli, sperm, and testi) covered numerous terms related to spermatogenesis, pathological changes of the male reproductive organs (such as testicular tumors, testitis, and epididymitis), and direct effects on the spermatozoa. To ensure that the effects were due to the specific drugs, drugs with indirect effects on sperm (i.e., drugs with teratogenic effects impacting the fertility of male offspring) were excluded. Only FDA-approved prescription drugs developed for administration to humans were examined. Over-the-counter (OTC) drugs were excluded because the information available on their labels was generally insufficient and most have not been subjected to rigorous reproductive toxicity testing. The drugs with multiple active ingredients were also excluded to reduce the complexity of the analysis. When there were redundant labels for drugs produced by different companies or drugs with more than one label that had been updated at different times, only the most recent label was analyzed. Other labels for drugs intended for veterinary use, medicinal foods, medical devices, dietary supplements, cosmetics, etc., were also excluded. When the final stemmed keyword search was performed on August 8, 2017, there were 95,873 labels included in the DailyMed database. After restricting the information to single-ingredient drugs as described above, a total of 251 unique prescription drug labels were identified that showed a negative impact on sperm, which were further narrowed down to 235 drugs because some of them were the same drug in different formulations that led to the same adverse effects (i.e., aripiprazole/aripiprazole lauroxil).

Data extraction and analysis

Two authors independently searched the DailyMed database and extracted relevant data. When there were differences in the descriptions extracted between these authors, they discussed the findings with additional authors, and the group’s consensus regarding the findings was used for the analysis.

A PubMed search was also performed to determine whether animal or human toxicity had been reported for the 235 drugs identified to impact animal spermatogenesis based on the DailyMed labels. The PubMed database was searched using the terms ([generic drug name] AND sperm*, fertil*, semen, semin*, or testi*).

Results

The general findings of the database searches

A total of 235 FDA-approved drugs intended for human use and containing a single active ingredient were found to impact spermatogenesis in at least one type of animal. All of these drugs could result in a decrease in the sperm count/concentration or a decrease in sperm quality, viability, or fertilization capacity.

Rats were the model most often reported to be affected by the drugs (specifically reported for 184 of the 235 drugs). This was expected, because rats are the most commonly used animal model for reproductive toxicity testing. Dogs are often used to confirm the findings in rats, and were thus also frequently reported to experience changes in spermatogenesis due to the exposure to prescription drugs. There were many instances where only rats were listed as having changes in spermatogenesis following drug exposure, either because there was no toxicity in other animals or because confirmation testing was not performed. Interestingly, in about half of the cases, spermatotoxicity was only reported for dogs. This suggests that dogs may be particularly sensitive to spermatotoxicants. Only 34 of the 235 drugs were reported to affect primate spermatogenesis. However, it is likely that many of the drugs were not tested in primates given the ethical concerns and high expenses associated with testing in primates. In addition, although spermatotoxicity was reported for 11 other drugs in DailyMed, the type of animal(s) affected was not specified in the drug label.

The FDA-approved drugs reported to adversely affect animal spermatogenesis

Unsurprisingly, antineoplastic agents were the category of drugs most often reported to have effects on animal spermatogenesis. Antineoplastic agents can cause temporary or permanent damage to the spermatozoa, germ cells, or support cells. Hormones and hormone antagonists were also expected to have adverse effects on spermatogenesis, so it is not surprising that they make up the other category of drugs most frequently noted to be spermatotoxic in this study. Both classes of drugs are well-recognized as having a potential negative impact on male fertility, and protocols are in place in most hospitals and clinics to counsel patients about these effects and the means to preserve spermatozoa for future use.

On the other hand, there were numerous other drugs with unexpected spermatotoxicity, where the intended target/mechanism of action is not generally considered to impact sperm. These drugs were from various classes, including adrenergic agents (icatibant, silodosin), analgesics (carbamazepine, morphine), antihypertensive agents (tamsulosin, reserpine), anti-infectious agents (metronidazole, miconazole), cardiovascular agents (bosentan, spironolactone), and immunosuppressants (sirolimus, azathioprine). Such agents are of greater concern because the adverse effects of these agents on fertility are not well-recognized and may not be addressed when the drugs are prescribed.

The types of spermatotoxic effects reported for animals

The most common adverse effect of the drugs was a reduced sperm count (due to reduced spermatogenesis or the death of developed and developing sperm), followed by reduced sperm motility, abnormal sperm morphology, testicular atrophy, and tumor formation (most frequently benign Leydig cell tumors in rodents). Similar observations of reduced sperm counts, reduced motility, and abnormal sperm morphology have been noted in humans for some of these drugs (see Table 1). In contrast to animal studies, where drug-related tumor formation was relatively common, drug-induced testicular tumors seem to be rare in humans. While some agents, such as anabolic steroids, are considered to cause testicular atrophy in men [44], the histological findings are generally not available, except for rare cases of injury or malignancy that require surgery. This makes it difficult to assess the full extent of the changes in humans.

Table 1.

FDA-approved drugs with negative effects on sperm in both animals and humans

Main category Drug name (generic) Effect(s) in animals Type of animal(s) affected Human toxicity in DailyMed? Reported effect(s) in humans (DailyMed) Support in PubMed Reported effect(s)
5-Alpha-reductase inhibitor Dutasteride Leydig cell adenomas, reduced cauda epididymal sperm counts, reduced weights of the epididymis, prostate, and seminal vesicles Rats Yes Decreased sperm count, motility, and semen volume [5] Reversible decrease in the number and motility of sperm
Adrenergic agent Silodosin Decreased sperm viability and counts, changes in the testes and epididymides Rats No [6] Prevents sperm emission, but no direct effects on spermatozoa
Alpha-blocker Tamsulosin hydrochloride Changes in semen content Rats No [7] Decreased sperm count
Analgesic Acetaminophen Increased abnormal sperm, reduced spermatogenesis Mice No [8] Reduced motility, DNA fragmentation
Buprenorphine Leydig cell tumors Rats No [9] Leads to anejaculation
Carbamazepine Benign interstitial cell adenomas in the testes, testicular atrophy, aspermatogenesis Rats No [10] Decreased sperm motility and vitality
Morphine sulfate Changes in hormone levels (incl. testosterone and luteinizing hormone) Rats No [11] Decreased sperm motility
Anticonvulsant Divalproex sodium Reduced spermatogenesis, testicular atrophy Rats and dogs No [12] Decreased counts and motility, increased abnormal morphology, decreased testicular volume
Oxcarbazepine Benign testicular interstitial cell tumors Rats No [13] Increased abnormal morphology, decreased testicular volume
Valproate sodium/valproic acid Reduced spermatogenesis and testicular atrophy Rats and dogs No [12] See divalproex above
Antidepressant Clomipramine hydrochloride Phospholipidosis and testicular changes Not specified Yes Testicular tumors [14] Reduced motility and abnormal morphology
Paroxetine Spermatogenic arrest, atrophic changes in the seminiferous tubules Rats Yes Decreased sperm quality, epididymitis [15] Increases DNA fragmentation
Vasopressinb Decrease in the function and fertilizing ability of spermatozoa Mice No [16] Decreased sperm count and motility
Antifungal Ketoconazole Increased abnormal sperm, decreased sperm mobility Rats No [17] Oligospermia, azoopermia
Antihyperlipidemic agent Rosuvastatin calciumb Spermatidic giant cells Dogs and monkeys No [18] Reversible azoospermia
Anti-infective agent Miconazole Chromosomal aberrations in spermatocytes, morphological abnormalities in sperm Mice No [19] Reduced vitality
Lindane Reduced spermatids Rats No [20, 21] Decreased quality, AR
Quinine sulfate Atrophy or degeneration of the seminiferous tubules, decreased sperm count and motility Mice No [22] Decreased motility, altered semen volume
Antineoplastic agent Busulfanc Decreased spermatogenesis Rats Yes Damaged sperm and testicular tissue, testicular atrophy, azoospermia [23] Decreased spermatogenesis
Carmustinec Testicular degeneration Rats No [24] Germ/Leydig cell failure
Cyclophosphamide Decreased weights, atrophy, changes in spermatogenesis Mice and rats Yes Interference with spermatogenesis, testicular atrophy, azoospermia, oligospermia [25] Azoospermia/oligospermia, decreased motility, abnormal morphology
Daunorubicin hydrochloridec Testicular atrophy, aplasia of spermatocytes, complete aspermatogenesis Dogs No [26] Decreased sperm count, motility and vitality
Doxorubicin hydrochloridea, c Decreased testicular weights and hypospermia, diffuse degeneration of the seminiferous tubules, decreased spermatogenesis Mice No [26, 27] Damages DNA, decreased sperm count, motility and vitality
Etoposidec Testicular atrophy Rats No [28] Decrease in sperm count (possibly reversible)
Fludarabine phosphateb Decreased testicular weights, degeneration and necrosis of spermatogenic epithelium of the testes Mice, rats and dogs Yes Damaged sperm and testicular tissue [29] DNA damage
Hydroxyurea Testicular atrophy, decreased spermatogenesis Rats No [30] Adverse effects on sperm production
Ifosfamide (alphabetize) Testicular atrophy with degeneration of the seminiferous tubular epithelium, decreased spermatogenesis Dogs, rats No [31] Sub-fertility, increased FSH and decreased inhibin-B
Imatinib mesylateb Decreased testicular and epididymal weights and percent motile sperm Rats No [32] Oligozoospermia
Leuprolide acetate Testicular interstitial cell adenomas Rats Yes Suppressed testicular steroidogenesis, testicular atrophy [33] Decreased sperm count
Temozolomideb Testicular atrophy, syncytial cells/immature sperm formation Rats and dogs No [34] Decreased motiity, abnormal morphology
Vincristine sulfatec Testicular degeneration and atrophy, epididymal aspermia Rats No [35] Increased infertility
Antipsychotic Risperidoneb Decreased sperm motility and concentration Beagle dogs No [36] Retrograde ejaculation
Antiulcer agent Cimetidine Benign Leydig cell tumors Rats No [37] Reduced sperm count, increased FSH and prolactin
Antiviral agent Ribavirin Abnormalities in sperm Mice No [38] Decreased density, motility and abnormal morphology
Cardiovascular agent Nitroglycerin Testicular tumors, increased interstitial cell tissue and aspermatogenesis Rats No [39] Reversible decrease in motility
Hormones, hormone substitutes, and hormone antagonists Finasteride Testicular Leydig cell adenomas, decreased weights of seminal vesicles and prostate Mice, rabbits Yes Reversible decrease in ejaculate volume and sperm per ejaculation [5] Reversible decrease in number and motility
Oxandroloneb, c Reduction of spermatogenesis and decreased weights of the testes, prostate, and seminal vesicles Rats Yes Suppressed spermatogenesis, testicular atrophy, epididymitis, oligozoospermia [40] Reversible azoospermia
Testosterone Suppressed spermatogenesis Rats Yes Suppressed spermatogenesis, oligozoospermia [41] Decreased concentration
Hypoglycemic agent Atorvastatin calcium Aplasia and aspermia in the epididymis, decreased sperm motility and concentration, and increased abnormal sperm Rats No [42] At least one abnormal parameter in 35% of men (number, viability, motility, morphology, etc.)
Immunosuppressive agent Sirolimus Atrophy of the testes, epididymides, prostate, and seminiferous tubules and/or reduction in sperm counts Rats Yes Reversible azoospermia [43] Decreased concentration and motility

aPubMed data based on in vitro studies

bPubMed data based on small studies involving 10 or fewer patients, including case reports

cPubMed data based on studies of combination therapy, no data exist for the agent administered alone in humans

Of note, most of the reports of adverse effects did not specify how many (what percentage) of the animals were affected. Therefore, it is unclear whether the observed adverse effect was common or relatively rare. The reporting was similarly vague for human effects, with many labels failing to indicate what percentage of the population was affected by the specific type of toxicity.

PubMed findings of human effects for these drugs

Only about 11% (26/235) of the drug warning labels that indicated that the agent had a negative impact on animal sperm (any aspect of spermatogenesis or toxicity to the sperm themselves) also had information in the drug label about a negative effect in humans. There were reports of adverse effects on human sperm for another 36 of these drugs in the PubMed database, although some of these publications were of in vitro studies, case studies, or studies of drug combinations that included the drug of interest (Table 1). Thus, between the drug labels in DailyMed and the reports included in PubMed, less than 30% of the drugs found to adversely affect animal sperm have been found to adversely affect human sperm.

For about half of the drugs that cited a negative impact on animal sperm, there were no data in DailyMed or PubMed about their impact on human sperm (Table 2). Eleven of the drugs without any publications in PubMed indicated that they had effects in humans in the DailyMed labels, although specific details were generally lacking regarding the number of patients affected. Interestingly, there were 33 drugs indicated to have negative effects in animals that were shown to have no effect or a positive impact on human sperm in the published literature (Table 3).

Table 2.

Drugs without clear information about the impact on men

Drug category Generic name Effect(s) on animal spermatogenesis Animal type(s) affected Human toxicity in DailyMed? Reported effect(s) in humans (DailyMed) Notes based on the PubMed search PubMed reference(s)
Analgesic Capsaicin Reduction in the number and percent of motile sperm, reduced sperm counts Rats No May affect motility by binding RPV1, but currently unproven [45]
Anesthetic Lidocaine hydrochloride monohydratea Decreased homogenization-resistant sperm head count, daily sperm production, and spermatogenic efficiency Rats No Increases the spermicidal effects of other compounds [46]
Antibacterial agent Dapsone Reduced sperm motility Rats Yes Orchitis, infertility No studies identified
Moxifloxacin/moxifloxacin hydrochloride Adverse effects on sperm morphology (head-tail separation) Rats No No studies identified
Telavancin hydrochloride Altered sperm parameters Rats No No studies identified
Cefotetan disodium Reduced testicular weight and seminiferous tubule degeneration Rats No No studies identified
Trovafloxacin mesylate Testicular degeneration Rats and dogs No No studies identified
Anticholinergic agent Glycopyrrolate Diminished seminal secretions Dogs No No studies identified
Anticonvulsant Clobazam Increased abnormal sperm Rats No No studies identified
Antidepressant Fluoxetinea Testicular and epididymal microscopic lesions and decreased sperm concentrations Rats No In vitro spermicidal effects [47]
Fluvoxamine maleatea Decreased sperm count, decreased epididymal weight Rats Yes Hematospermia In vitro spermicidal effects [47]
Goserelin acetate Atrophic histological changes in the testes, epididymis, seminal vesicles and prostate gland, suppressed spermatogenesis Rats No No studies identified
Imipramine pamoatea Atrophy of the seminiferous tubules, spermatogenic arrest Dogs No Decreased viability and motility [48]
Methyldopa/methyldopate hydrochloride Decreased sperm count, sperm motility, number of late spermatids Rats No No studies identified
Nimodipinea Leydig cell adenomas Rats No Blocks the AR [49]
Selegiline Epididymal and testicular hypoplasia, decreased sperm count and concentration Dogs, rats No No studies identified
Trimipramine maleate Degeneration of seminiferous tubules Not specified No No studies identified
Nebivolol hydrochloride Testicular Leydig cell hyperplasia and adenomas, effects on spermatogenesis Mice and rats No No studies identified
Antidote for ethylene glycol Fomepizole Decreased testicular mass Rats No No studies identified
Antiemetic Dronabinola Decreases in spermatogenesis, number of developing germ cells, and number of Leydig cells Rats No Decreased motility and AR [50]
Antiepileptic Felbamate Benign interstitial cell tumors of the testes Rats No A case report suggested that it was safe [51]
Rufinamide Decreased sperm count and motility Rats No No studies identified
Antihypertensive agent Hydralazine hydrochloride Benign interstitial cell tumors Rats No No studies identified
Isradipine Benign Leydig cell tumors and testicular hyperplasia Rats No No studies identified
Eplerenone Decreased weights of seminal vesicles and epididymides Rats No No studies identified
Reserpinea Malignant tumors of the seminal vesicles Mice No Reduced motility and fertilization capacity [52]
Anti-infective agent Metronidazolea Effects on testes and sperm production Rats No Spermicidal [53]
Trimetrexate glucuronate Degeneration of the testes and spermatocytes, spermatogenic arrest Mice and rats No No studies identified
Linezolid Reversible reductions in sperm motility and altered sperm morphology, epithelial cell hypertrophy in epididymis Rats, dogs No No studies identified
Mefloquine hydrochloride Histopathological lesions in the epididymides Rats No No studies identified
Micafungin sodium Vacuolation of epididymal ductal epithelial cells and reduced sperm count, seminiferous tubular atrophy and decreased epididymal sperm Rats, dogs No No studies identified
Luliconazole Decreased sperm counts Rats No No studies identified
Miltefosine Testicular Leydig cell adenoma, testicular atrophy, reduced numbers of viable sperm Rats No No studies identified
Anti-inflammatory agent Suprofen Testicular atrophy/hypoplasia Rats No No studies identified
Celecoxib Epididymal hypospermia, slight dilation of the seminiferous tubules Juvenile rats No No studies identified
Zileuton Benign Leydig cell tumors Rats No No studies identified
Antimigraine agents Eletriptan hydrobromide Testicular interstitial cell adenomas Rats No No studies identified
Antineoplastic agent Abiraterone acetate Testicular atrophy, aspermia/hypospermia, reduced sperm counts and motility, altered morphology Rats and monkeys No Decreased testosterone and decreased weights of androgen-responsive organs in a xenograft fetal testis model [54]
Ado-trastuzumab emtansine Degeneration of seminiferous tubules with hemorrhage in the testes, decreased weights of the epididymides, prostate, and seminal vesicles Rats, monkeys No No studies identified
Afatinib Oligo- or azoospermia, increased apoptosis in the testes and atrophy in the seminal vesicles and prostate Rats No No studies identified
Alemtuzumab Adverse effects on sperm parameters, decreased sperm count and motility Transgenic mice No The target of the antibody (CD52) may have homology with gp20 on the sperm membrane, but the clinical impact is unknown [55]
Altretamine Testicular atrophy, decreased spermatogenesis, atrophy of testes, seminal vesicles and ventral prostate Rats No No studies identified
Asparaginase Decreased sperm count and motility Rats No No studies identified
Axitinib Testicular atrophy, decreased numbers of germinal cells, hypospermia or abnormal sperm forms, reduced sperm density and count Rats No No studies identified
Belinostat Reduced organ weights of the testes/epididymides, delay in testicular maturation Dogs No No studies identified
Bexarotene Testicular degeneration Dogs No No studies identified
Bortezomib Degenerative changes in the testes Rats No No studies identified
Brentuximab vedotin Seminiferous tubule degeneration, Sertoli cell vacuolation, reduced spermatogenesis or aspermia Rats No No studies identified
Cabazitaxel Degeneration of seminal vesicles and seminiferous tubule atrophy, minimal epithelial single cell necrosis in epididymis Rats, dogs No No studies identified
Cabozantinib Decreased sperm counts and reproductive organ weights, testicular degeneration and decreased spermatocytes and spermatids Rats, mice No No studies identified
Chlorpromazinea Chromosomal aberrations in spermatocytes and abnormal sperm Rodents No Decreased motility [56]
Cladribine Testicular degeneration Monkeys No No studies identified
Clofarabine Seminiferous tubule and testicular degeneration, atrophy of interstitial cells Mice, rats and dogs No No studies identified
Cobimetinib Testicular degeneration Dogs No No studies identified
Crizotinib Testicular pachytene spermatocyte degeneration Rats No No studies identified
Dabrafenib Testicular degeneration/depletion Rats and dogs Yes Impaired spermatogenesis, decreased sperm count No studies identified
Decitabine Abnormal histology, decreased sperm number Mice No No studies identified
Docetaxel Testicular atrophy or degeneration Dogs No No studies identified
Enzalutamide Atrophy of the prostate and seminal vesicles Rats No No studies identified
Eribulin mesylate Testicular toxicity (hypocellularity of seminiferous epithelium with hypospermia/aspermia) Rats and dogs No No studies identified
Everolimus Decreased sperm motility, sperm count, and plasma testosterone levels Rats Yes Azoospermia or oligozoospermia (< 1% of patients) Increases testosterone, but clinical impact unclear [57]
Fulvestrant Testicular Leydig cell tumors, loss of spermatozoa from seminiferous tubules, seminiferous tubular atrophy, and degenerative changes in the epididymides Rats No No studies identified
Gemcitabine hydrochloride Hypospermatogenesis Mice No No studies identified
Idarubicin Testicular atrophy, inhibition of spermatogenesis and sperm maturation with few or no mature sperm Dogs No No studies identified
Idelalisib Decreased epididymidal and testicular weights, reduced sperm concentration Rats No No studies identified
Irinotecan hydrochloride Atrophy of male reproductive organs Rodents No No studies identified
Ixabepilone Testicular atrophy or degeneration Dogs No No studies identified
Lenvatinib Testicular hypocellularity of the seminiferous epithelium and desquamated seminiferous epithelial cells in the epididymides Dogs No No studies identified
Nilutamide Benign Leydig cell tumors Rats Yes Testicular atrophy No studies identified
Omacetaxine mepesuccinate Degeneration of the seminiferous tubular epithelium, hypospermia/aspermia in the epididymides Mice No No studies identified
Oxaliplatin Testicular degeneration, hypoplasia, and atrophy Dogs No Reduced inhibin-B (predictive of poor spermatogenesis and infertility) [58]
Palbociclib Decreased organ weight, atrophy or degeneration, hypospermia, intratubular cellular debris, lower sperm motility and density Rats and dogs No No studies identified
Panobinostat Prostate atrophy accompanied by reduced secretory granules, testicular degeneration, oligospermia, and increased epididymal debris Dogs No No studies identified
Pazopanib hydrochloride Reduced sperm production and testicular sperm concentrations, epididymal sperm concentrations and sperm motility, atrophy and degeneration of the testes, aspermia, hypospermia, and cribiform changes in the epididymis Rats No No studies identified
Pemetrexed disodium Reduced fertility, hypospermia, and testicular atrophy Mice No No studies identified
Pentostatin Seminiferous tubular degeneration Dogs No No studies identified
Ponatinib Degeneration of the epithelium of the testes Rats and monkeys No No studies identified
Porfimer sodium Discoloration and hypertrophy of the testes Rats No No studies identified
Regorafenib Tubular atrophy and degeneration in the testes, atrophy in the seminal vesicle, and cellular debris and oligospermia in the epididymides Rats No No studies identified
Romidepsin Testicular degeneration Rats No No studies identified
Sorafenib Testicular atrophy or degeneration; degeneration of the epididymis, prostate, and seminal vesicles; oligospermia Rats, dogs No No studies identified
Topotecan Multinucleated spermatogonial giant cells Dogs No No studies identified
Trabectedin Histopathological signs of hemorrhage and degeneration in the testes Rats No No studies identified
Trifluoperazinea Chromosomal aberrations in spermatocytes and abnormal sperm Rodents No Decreased motility [49]
Valrubicin Testicular degeneration, germ cell depletion, spermatid giant cells and karyomegaly Dogs No No studies identified
Venetoclax Testicular toxicity (germ cell loss) Dogs No No studies identified
Vinorelbine Decreased spermatogenesis and prostate/seminal vesicle secretions Rats Yes Damage to spermatozoa No studies identified
Vismodegib Decreased % motile sperm, hypospermia, germ cell degeneration Rats, dogs No Present in semen, but impact unclear [59]
Ziv-aflibercept Decreased sperm motility, alterations in sperm morphology Monkeys No No studies identified
Anti-osteoporosis agents Risedronate sodium Testicular and epididymal atrophy Rats No No studies identified
Anti-Parkinson agent Apomorphine hydrochloride Leydig cell tumors Rats No No studies identified
Ropinirole hydrochloride Testicular Leydig cell adenomas Rats No No studies identified
Rotigotine Leydig cell tumors, decreased epididymal sperm motility Rats No No studies identified
Pimavanserin tartrate Decreased density and motility of sperm, cytoplasmic vacuolation in the epididymis Rats No No studies identified
Antipsychotic agent Aripiprazole/aripiprazole lauroxil Disturbances in spermatogenesis Rats No No studies identified
Paliperidone/paliperidone palmitate Decreased sperm motility and concentration Beagle dogs No No studies identified
Prochlorperazine edisylate/prochlorperazine maleate Chromosomal aberrations in spermatocytes and abnormal sperm Rodents No No studies identified
Antituberculosis agent/anti-infectious Rifabutin Testicular atrophy Baboons and rats No No studies identified
Antiulcer Dexlansoprazole Testicular interstitial cell adenomas Rats No No studies identified
Lansoprazole Testicular interstitial cell adenomas, proliferative changes in the Leydig cells, including benign neoplasia Rats No No studies identified
Antiviral agent Boceprevir Testicular degeneration Rats No No studies identified
Cidofovir Inhibition of spermatogenesis Rats and monkeys No No studies identified
Daclatasvir Reduced prostate/seminal vesicle weights, minimally increased dysmorphic sperm Rats No No studies identified
Entecavir Seminiferous tubular degeneration Rodents and dogs No No studies identified
Famciclovir Atrophy of the seminiferous tubules, reduction in sperm count, and/or increased incidence of sperm with abnormal morphology or reduced motility Rats, mice and dogs No No studies identified
Ganciclovir Hypospermatogenesis Mice, dogs Yes Testicular hypotrophy, aspermatogenesis (dose-dependent) No studies identified
Penciclovir Atrophy of the seminiferous tubules, increased sperm with abnormal morphology, reduced motility Rats and dogs No No studies identified
Trifluridine Testicular atrophy Mice No No studies identified
Valganciclovir Hypospermatogenesis Mice, rats and dogs Yes Inhibited spermatogenesis No studies identified
Attention deficient disorder agent Atomoxetine hydrochloride Decreased epididymal weight and sperm number Rats No No studies identified
Bone density conservation agent Ibandronate sodium Decreased sperm production and altered sperm morphology Rats No No studies identified
Cardiovascular agent Dexrazoxane Testicular atrophy Rats No No studies identified
Naratriptan hydrochloride Testicular/epididymal atrophy accompanied by spermatozoa depletion Rats No No studies identified
Propafenone hydrochloride Decreased spermatogenesis Rabbits, dogs, and monkeys No No studies identified
Ambrisentan Testicular tubular degeneration, effects on the sperm count and morphology Rats and mice No No studies identified
Bosentan Testicular tubular atrophy Rodents Yes Decreased sperm count No studies identified
Dofetilide Testicular atrophy and epididymal oligospermia Rats, mice and dogs No No studies identified
Spironolactone Testicular interstitial cell tumors Rats No 2/9 men showed decreased sperm density, but overall, no significant difference [60]
Macitentan Testicular tubular atrophy Rats No No studies identified
Central nervous system agent Carisoprodol Reduced testes weight and sperm motility Mice No No studies identified
Eszopiclone Decreased sperm number and motility, increase in morphologically abnormal sperm (mid and high doses) Rats No No studies identified
Fentanyl citratea Decreased percent mobile sperm, sperm concentrations, increased abnormal sperm Rats No Modest inhibition of motility (based on a CASA) [61]
Oxazepam Testicular interstitial cell adenomas Rats No
Caffeine citrate Spermatogenic cell degeneration Rats No Different studies have shown conflicting (potentially dose-related) effects [62, 63]
Contrast medium Gadobenate dimeglumine Abnormal spermatogenic cells Rats No No studies identified
Gadopentetate dimeglumine Decreased testes and epididymis weights Rats No No studies identified
Gadoversetamide Reduction and degeneration of spermatocytes, degeneration of the germinal epithelium of the testes, presence of germ cells in the epididymides, reduced sperm count Rats No No studies identified
Dermatological agent Dimethyl fumarate Leydig cell adenomas, increased nonmotile sperm, testicular atrophy, hypospermia, testicular hyperplasia Mice, rats, and dogs No No studies identified
Tretinoin Decreased sperm count and motility Rats No Other retinoids had no effect on sperm [64]
Dispersing agent Hyaluronidase, ovine Testicular degeneration Not specified No The hyaluronidase level is associated with the fertilization rate, but the clinical impact of systemic or nonreproductive treatment is unclear [65]
Enzyme inhibitor Eliglustat Adverse effects on sperm morphology, testes (germ cell necrosis), and sloughed cells in the epididymis Rats No No studies identified
Tofacitinib Benign Leydig cell tumors Rats No No studies identified
Epithelial growth factor Palifermin Decreased epididymal sperm counts Rats No No studies identified
Histamine antagonist Desloratadine Decreased sperm numbers and motility Rats No No studies identified
Hormones, hormone substitutes, and hormone antagonists Conjugated estrogens, estradiol, estradiol acetate, estradiol cypionate Increased frequency of carcinomas of testis Not specified No Estrogen decreases the acrosome reaction in vitro, but is required for normal sperm production. Therefore, effects are likely concentration-dependent [66, 67]
Estropipate Increased frequency of carcinomas of testis Not specified No No studies identified
Flutamide Testicular interstitial cell adenomas of the testes Rats Yes Interference with testosterone, decreased sperm count Unclear, suggested to have minimal transient effects on sperm [68]
Ospemifene Atrophy of the prostate and seminal vesicles Rats No No studies identified
Histrelin acetate Testicular Leydig cell tumors Rats Yes Testicular atrophy No studies identified
Hypnotic agent Doxepin hydrochloride Increased percentages of abnormal sperm and decreased sperm motility Rats No No studies identified
Ramelteon Benign Leydig cell tumors Rats No No studies identified
Rozerem Leydig cell tumors Rats No No studies identified
Hypoglycemic agent Acarbose Benign Leydig cell tumors Rats No No studies identified
Canagliflozin Testicular Leydig cell tumors, decreased sperm velocity, increased number of abnormal sperm Rats No No studies identified
Chlorpropamide Suppression of spermatogenesis Rats No No studies identified
Fenofibrate/fenofibric acid Benign testicular interstitial cell tumors Rats No No studies identified
Fluvastatin sodium Tubular degeneration and aspermatogenesis in testes, vesiculitis of seminal vesicles Hamsters, rats No No studies identified
Mifepristonea Reduced testicular size Rats No Decreased AR [69]
Immunosuppressive agent Azathioprine sodium Reduced spermatogenesis, sperm viability and sperm count Mice No Conflicting findings, may be age-dependent or related to the disease status [70, 71]
Immunological factor Interferon gamma-1b Decreased spermatogenesis and sperm counts, increased abnormal sperm (very high doses) Mice and monkeys No Effect of local or systemic treatment unclear, but no effect on sperm treated in vitro [72]
Pimecrolimus Decreased testicular and epididymal weights, testicular sperm counts, motile sperm Rats No Other mTOR inhibitors had adverse effects [43]
Teriflunomide Reduced epididymal sperm count Rats No
Thalidomide Testicular pathological and histopathological effects Rabbits Yes Orchitis Present in semen, but impact unclear [73]
Muscle relaxant agent Dantrolene sodium Testicular tumors Rats No No studies identified
Ophthalmic drug Aflibercept Changes in sperm morphology and motility Monkeys No No studies identified
PDE5 inhibitor Avanafil Reduced sperm motility, increased percentage of abnormal sperm (broken sperm with detached heads) Rats No No studies identified
PDE4 inhibitor Roflumilast Tubular atrophy, degeneration in the testes and spermiogenic granuloma in the epididymides Rats No No studies identified
Purgative agent Lubiprostone Interstitial cell adenoma of the testes Rats No No studies identified
Naloxegol oxalate Leydig cell adenomas Rats No No studies identified

aPubMed data based on in vitro studies

Table 3.

Drugs where the animal findings were refuted by PubMed publications

Main indication Drug name (generic) Effect(s) in animals Type of animal(s) affected/tested Human toxicity in DailyMed? Reported effect(s) in humans (DailyMed) Effect(s) in humans reported in PubMed PubMed reference
Adrenergic agent Icatibant acetatea Testicular atrophy/degeneration, reduced sperm counts Rats and dogs No No effect [74]
Antibacterial agent Doxycycline/doxycycline hyclate Reduced motility, velocity and concentration, abnormal morphology Rats No May improve sperm parameters in patients with infections [75]
Clarithromycin Testicular atrophy Rats and monkeys No Improves motility [76]
Antiepileptic Pregabalin Decreased sperm counts and sperm motility, increased sperm abnormalities Rats Yes Epidiymitis No effect [77]
Antigout agent Colchicine/colchicinum Abnormal sperm morphology and reduced sperm counts in males Not specified Yes Azoospermia, oligospermia No effect [78]
Antihyperlipidemic agent Pravastatin sodium Sperm abnormalities Rats No No effect [79]
Antihypertensive agent Metoprolol Reversible adverse effects of spermatogenesis (in some studies) Rats No Minimally inhibits motility [80]
Pindolol Testicular atrophy and/or decreased spermatogenesis Rats No No effect [80]
Prazosina Testicular changes consisting of atrophy and necrosis Rats and dogs No No effect [81]
Anti-infective agent Griseofulvinb Suppression of spermatogenesis Rats No No effect [82]
Minocycline hydrochloride Reduced sperm cells and motile sperm, increased abnormal sperm, including absent heads, misshapen heads and abnormal flagella Rats No Improves semen parameters in men with infections [83]
Tinidazole Testicular degeneration, spermatogenic effects Rats No Improves sperm parameters as part of treatment for H. pylori infections [76]
Anti-inflammatory agent Ketoprofen Abnormal spermatogenesis or inhibition of spermatogenesis Rats and dogs No Treatment may improve male infertility [84]
Oxaprozin Testicular degeneration Beagle dogs No No effect [85]
Antineoplastic agent Bicalutamide Testicular benign interstitial (Leydig) cell tumors Rats No No effect [86]
Dasatinib Reduced size and secretion of seminal vesicles, immature prostate, seminal vesicle, and testis Rats No No effect [87]
Epirubicin hydrochloridec Atrophy of the testes and epididymis, reduced spermatogenesis Mice and rats No No effect [88]
Fluorouracila Kills differentiated spermatogonia and spermatocytes Mice No No effect [89]
Letrozole Degeneration of the seminiferous tubular epithelium Rats No Improves sperm parameters as part of treatment for H. pylori infections [90]
Paclitaxelc Testicular atrophy/degeneration Rodents No No effect [91]
Toremifene citrate Testicular tumors Mice No Improves sperm parameters and hormone levels [92]
Antiviral agent Acyclovir Testicular atrophy and aspermatogenesis Rats and dogs No No effect [93]
Miglustat Decreased spermatogenesis with altered sperm morphology and motility, decreased fertility, and testicular interstitial cell adenomas Rats No No effect [94]
Tenofovir None Rats No Decreased progressive motility [95]
Bone density conservation agent Raloxifene hydrochloride Testicular interstitial cell tumors Mice Improves sperm concentration and morphology and increased testosterone in oligospermic med [96]
Cholinergic agonist Pilocarpine Decreased sperm motility, and morphologic evidence of abnormal sperm Rats No No effect [97]
Dermatological agent Clobetasol propionate Increased weights of the seminal vesicles Rats No Treatment of scrotal dermatitis improved the sperm count and motility [98]
Isotretinoin Depression of spermatogenesis Dogs No No effect [64]
Hypoglycemic agent Simvastatin Seminiferous tubule degeneration (necrosis and loss of spermatogenic epithelium, testicular atrophy, decreased spermatogenesis, spermatocytic degeneration and giant cell Rats No No effect [79]
Dogs
Lovastatin Testicular atrophy, decreased spermatogenesis, spermatocytic degeneration, giant cell formation Dogs No No effect [99]
Opioid antagonist Naltrexonea Increased testicular mesotheliomas Rats No Increased motility (opposite the effects of morphine) [100]
Tadalafil Degeneration and atrophy of the seminiferous tubular epithelium, decrease in spermatogenesis Beagle dogs Yes Decreased sperm concentration Improves concentration and motility [101]

aBased on in vitro findings

bBased on small studies of < 10 patients, including case studies

cBased on studies of combination treatment

Discussion

The status of spermatotoxicity testing and relevance of the current testing

Testing for and reporting spermatotoxicity

The guidelines for industry issued by the FDA for new drug development stipulate that “…reproductive or developmental toxicity, whether in valid reproductive/developmental or other relevant nonclinical studies, should be evaluated to estimate the likelihood or other reproductive or developmental risk for humans”. In addition, the “Females and Males of Reproductive Potential” section requires the inclusion of information when there are human and/or animal data suggesting drug-associated effects on fertility and/or preimplantation loss effects (§201.57(c)(9)(iii))” [102]. The FDA, as well as the US Environmental Protection Agency, Occupational Safety and Health Administration, and other national and international organizations, have made efforts to both streamline testing and make the testing as accurate as possible, specifically taking into consideration the route, dose, frequency, and duration of exposure, as well as species-associated differences. The guidelines also emphasize the need to understand the potential impact of drugs on fertility via both pre-clinical studies in animals and subsequent studies in patients, with an understanding that there are some inherent disadvantages associated with testing in animals [103].

Despite the various considerations made to improve the testing process and provide the best predictions of toxicity, there have been numerous studies that reported a lack of efficacy for toxicity testing, including reproductive toxicity testing [104107]. In a previous study, a total of 65 labels for single-ingredient FDA-approved drugs were found in DailyMed that indicated that the drugs had a negative impact on human spermatogenesis [3]. In agreement with our present findings suggesting that animal studies have limited predictive power for human spermatotoxicity, fewer than half of the drug labels (26 of 65) indicated that they had effects on animal spermatogenesis. There was support in the literature for an additional six drugs affecting animal spermatogenesis, but this means that approximately half of the prescription drugs that negatively affect human spermatogenesis did not have an animal correlate reported in either the drug label or the peer-reviewed literature. This may reflect the inadequacy of the animal model(s) tested, insufficient testing, and/or ineffective reporting of the findings of animal studies.

The relationship between the reported effects in animals and the effects in humans

In the present study, approximately 17% of the single-ingredient, FDA-approved, clinically prescribed drugs included in the DailyMed database (235/1318) had an adverse effect on spermatogenesis in at least one animal. Less than 30% of these drugs have been reported to adversely affect human sperm (in either the drug warning label or the published literature included in PubMed). However, this finding does not indicate that the “false positive” rate of animal toxicity testing is > 70%, because there were no data in either database about the impact of more than half of the drugs on human sperm (see Table 2). Therefore, the “false positive” rate for animal testing of the drugs with reported findings was about 35% (33/93).

Interestingly, of the 33 drugs indicated to have negative effects in animals that were shown to have no effect or a positive impact on human sperm in the literature (Table 3), four had data in their FDA labels (included in the DailyMed database) indicating that they had a negative impact on human sperm. Since the DailyMed database is supposed to contain up-to-date information, it is possible that new data had been generated since the PubMed studies were published demonstrating the negative effect reported in the label.

It is also possible that there were differences in the experimental conditions (different ages of subjects, subjects of different ethnicities, subjects with different background diseases or stages of disease, or different timing of sample collection after administration, etc.), and these could have led to the different results. Because details were not present in the FDA labels included in the DailyMed database, it is unknown why there were conflicting data. These findings illustrate the difficulties associated with reproductive toxicity testing. Moreover, they demonstrate the need for better record-keeping and transparency in terms of the data generated, because it is very difficult to interpret the findings in the drug labels based on the data available.

Difficulties associated with animal testing for spermatotoxicity

The current reproductive toxicity testing should be able to provide useful results for clinical extrapolation. However, as noted above, our current study and previous studies about spermatotoxicity [3, 105, 106] have shown that studies in animals have relatively low predictive value for human toxicity. In some cases, the lack of useful findings is due to the high doses of the compounds used in the toxicity studies. Nevertheless, even if the dose is proper in terms of the allometric scaling for animals based on body surface area (~ 12× the human dose (mg/kg b.w.) for a mouse, ~ 2× for a dog, etc. [108]), the plethora of differences between humans and animals reduces the utility of the findings. For example, the pharmacokinetics and pharmacodynamics of drugs may not be similar between the animal model and humans, there may be organ/tissue-related differences that make it impossible to adequately predict or extrapolate the effects, and there may be other fundamental differences between species that abrogate the utility of testing [106]. This is particularly relevant for spermatotoxicity studies, where there are many important differences between rodents and humans, including the time required for spermatogenesis, the development of Leydig cell tumors, and the relative number of sperm produced [109112]. Additional “uncertainty” factors have been used when extrapolating human toxicity from animal findings for occupational and environmental toxicants (i.e., when determining the benchmark dose or the acceptable daily intake based on the no observed adverse effects level (NOAEL) [113], but these types of studies are not usually employed as part of therapeutic drug testing and would be subject to the same issues described above.

Changes that should be made to the testing and reporting of spermatotoxicity

The most important findings of the present study include the following: only 27% (63/235) of the drugs that had a negative effect in animals reported in the drug label were found to have a documented negative impact on human sperm (in the drug labels or literature). Further, a previous study [3] showed that fewer than half of the drugs with labels indicating that they adversely affected human sperm had similar findings in animals. In addition, for more than half of the drugs found to negatively affect animal spermatogenesis or spermatozoa, the impact in humans is unclear either because no testing or surveillance has been performed, or because the results have not been reported. Even in cases where there was clear reporting of the type of human spermatotoxicity and the number of individuals affected, the data were frequently unclear regarding whether the effects were reversible and how long it took to recover potency. These findings suggest that the current system is not performing well and there is a need for better models, different types of pre-clinical testing, different endpoints, and a better system (and/or different regulations) for reporting data.

It should be noted that most of the general methods used to assess reproductive toxicity were often developed more than 50 years ago, although there have been updates in terms of automation and some of the details (i.e., sub-types of sperm motility) of the analyses. Our data add to the already extensive body of evidence that animal testing (certainly for spermatotoxicity, but also for many other types of toxicity) is not adequate to predict human risk. However, completely retiring the rat is not possible, at least at present. There are currently no other models that provide better predictive value for the various steps of reproduction. Direct testing on human sperm samples is useful and should be implemented whenever possible, but cannot identify changes in hormone levels, reproductive tissues, or germ or support cells. These parameters can only be evaluated in vivo.

While animal testing cannot be avoided at present, there are various steps that should be taken immediately to improve the efficacy and clinical relevance of the testing and reporting of data. First, the general protocol for testing should be reconsidered. The usual one-size-fits-all approach to toxicity testing should be scrapped in favor of integrative studies taking into account the differences in pharmacokinetics/pharmacodynamics of the drug (if known), the intended target population (the age and disease background of the patients may be relevant to the toxicity), the effects of structurally and mechanistically similar drugs, and any information that can be obtained from in silico studies (high-throughput testing of the compound’s predicted structure for interactions with characterized receptors and biomacromolecules). It may also be necessary to change the endpoints of studies. Instead of assessing sperm at a single time point (at a pre-specified time after a single dose, or at the end of a chronic study), it would likely be better to collect sperm at several different time points. Collection of multiple samples (including collection on more than 1 day prior to starting treatment) is particularly important for clinical trials given the high variability in human sperm production [111, 114]. Additional testing to confirm recovery would provide helpful information for counseling patients on the long-term adverse effects.

Perhaps the most important changes would be those made to the reporting of the findings. Although the FDA receives the full results of the pre-clinical toxicity studies and clinical safety studies, including the numbers of animals used for pre-clinical testing (or patients included in a clinical trial), this information is frequently lacking from the drug labels in DailyMed. A clear statement of the number of individuals evaluated and the number or percentage of these affected by each adverse effect would help to interpret the findings. It would also be useful for this information to specify the extent of the adverse impact, such as the exact percentages of sperm with abnormal morphology, rather than a blanket statement indicating that there were morphological changes. This information would not need to appear on the main drug label included with the packaging of each drug, but should be linked online (with the link included in the printed information) so that the full details of the study are readily available from DailyMed.

Another point that is commonly lacking in both the DailyMed labels and the data in journal articles is the reversibility of the spermatotoxicity and time to recovery (if recovery is possible). This information is frequently missing for pre-clinical studies, largely because sperm are usually collected at a single time point, sometimes at necropsy. Although clinical trials are more likely to assess the long-term effects of a drug, the long-term reproductive outcomes are often not evaluated. This is partly because such endpoints are difficult to add to standard surveillance protocols because the collection of semen is more invasive and potentially associated with ethical concerns, compared to physical examinations and blood collection.

Finally, computing power has been rapidly increasing during the past few decades, and research studies are helping to illuminate the molecular basis of various physiological processes. Therefore, efforts should be made to better understand the signaling and cellular interactions involved in normal fertility to permit in silico modeling of possible interference by investigational drugs. This would inform confirmatory studies to be performed in vitro on human sperm or iPSC-derived or other sources of germ cells, could suggest biomarkers to allow for more sensitive detection of effects in vivo (in animals and/or humans), and would predict the toxicity likely to be encountered. As in silico modeling continues to improve, it may become possible to evaluate the interactions of various agents and various pathways simultaneously and to combine such findings with those from in vitro and limited in vivo studies for an integrated approach, more accurately predicting the effects in humans [115, 116].

Until toxicity testing is optimized, it is necessary to consider that there is a potential for human risk whenever there is a positive finding of spermatotoxicity in nonclinical or pre-clinical studies for one or more endpoints. The details of the findings and the level of evidence should be included to help physicians and/or pharmacists inform their patients of the risks of specific treatments to their fertility.

Conclusion

Our present findings illustrate the status of male reproductive toxicity testing and provide a useful reference about the FDA-approved drugs known to affect animal spermatogenesis and how these findings correlate with the human clinical setting. These findings emphasize the need for a better understanding of basic sperm biology to allow for the prediction of spermatotoxicity based on a drug’s effects (both targeted and nonspecific). A greater understanding of the normal variations in the sperm of men of various ages and different health status would also help to understand what should be considered abnormal during clinical trials and long-term surveillance. In addition, since the predictive value of animal testing is so low, there is a need for a more comprehensive evaluation of the effects on human sperm in vitro and continued development of in silico and alternative in vitro studies so that potential changes can be estimated before the initiation of human clinical trials. Such an understanding could be used to determine whether the normal observations carried out during clinical trials should be increased or altered to better detect changes.

There are several limitations to this review that should be noted. First, only single-ingredient, FDA-approved drugs indicated for human use that were included in the DailyMed database were analyzed in this study. Although this database is the largest of its type, there are still a few drugs for which information is missing. In addition, all over-the-counter medications, including vitamins, supplements, and herbal/alternative medications, were excluded because the toxicity information on these agents is typically lacking. However, these excluded agents may be associated with spermatotoxicity, either when taken alone or by potentiating the effects of other drugs. The present investigation also focused on the direct effects of drugs on spermatozoa, which represents only a small aspect of male fertility. In addition, there may have been some reporting or publication bias for the databases. However, whenever possible, only large and well-designed clinical trials were included for the PubMed data, and it is noted whenever small-scale studies or case reports are cited in the tables. For the DailyMed database, the data sources are often unclear, so the accuracy of the data is also unclear.

Despite these limitations, the present data are useful in that they provide a comprehensive overview of the prescription drugs that affect animal spermatogenesis, as well as an illustration of the relatively poor performance of animal testing for predicting spermatotoxicity. Several suggestions have been made which might improve the testing and reporting of the data, hopefully leading to better patient care.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflicts of interest.

Footnotes

Elizabeth R. Rayburn and Liang Gao should be considered joint first authors.

References

  • 1.Brezina PR, Yunus FN, Zhao Y. Effects of pharmaceutical medications on male fertility. J Reprod Infertil. 2012;13:3–11. [PMC free article] [PubMed] [Google Scholar]
  • 2.Centers for Disease Control and Prevention. National Center for Health Statistics. Therapeutic drug use. In: Health, United States, 2015: with special feature on racial and ethnic health disparities. [Internet] Hyattsville, MD. 2016; [Cited August 8, 2017]. Available from: http://www.cdc.gov/nchs/fastats/drug-use-therapeutic.htm. [PubMed]
  • 3.Ding J, Shang X, Zhang Z, Jing H, Shao J, Fei Q, et al. FDA-approved medications that impair human spermatogenesis. Oncotarget. 2017;8:10714–10725. doi: 10.18632/oncotarget.12956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Li H, Shi Q. Drugs and diseases interacting with cigarette smoking in US prescription drug labelling. Clin Pharmacokinet. 2015;54:493–501. doi: 10.1007/s40262-015-0246-6. [DOI] [PubMed] [Google Scholar]
  • 5.Smarr MM, Kannan K, Chen Z, Kim S, Buck Louis GM. Male urinary paracetamol and semen quality. Andrology. 2017; 10.1111/andr.12413. [DOI] [PMC free article] [PubMed]
  • 6.Amory JK, Wang C, Swerdloff RS, Anawalt BD, Matsumoto AM, Bremner WJ, et al. The effect of 5alpha-reductase inhibition with dutasteride and finasteride on semen parameters and serum hormones in healthy men. J Clin Endocrinol Metab. 2007;92:1659–1665. doi: 10.1210/jc.2006-2203. [DOI] [PubMed] [Google Scholar]
  • 7.Kobayashi K, Masumori N, Hisasue S, Kato R, Hashimoto K, Itoh N, et al. Inhibition of seminal emission is the main cause of anejaculation induced by a new highly selective alpha1A-blocker in normal volunteers. J Sex Med. 2008;5:2185–2190. doi: 10.1111/j.1743-6109.2008.00779.x. [DOI] [PubMed] [Google Scholar]
  • 8.Ramdurg S, Ambekar A, Lal R. Sexual dysfunction among male patients receiving buprenorphine and naltrexone maintenance therapy for opioid dependence. J Sex Med. 2012;9:3198–3204. doi: 10.1111/j.1743-6109.2011.02219.x. [DOI] [PubMed] [Google Scholar]
  • 9.Reis RM, de Angelo AG, Sakamoto AC, Ferriani RA, Lara LA. Altered sexual and reproductive functions in epileptic men taking carbamazepine. J Sex Med. 2013;10:493–499. doi: 10.1111/j.1743-6109.2012.02951.x. [DOI] [PubMed] [Google Scholar]
  • 10.Agirregoitia E, Subiran N, Valdivia A, Gil J, Zubero J, Irazusta J. Regulation of human sperm motility by opioid receptors. Andrologia. 2012;44(Suppl 1):578–585. doi: 10.1111/j.1439-0272.2011.01230.x. [DOI] [PubMed] [Google Scholar]
  • 11.Hamed SA, Moussa EM, Tohamy AM, Mohamed KO, Mohamad ME, Sherif TM, et al. Seminal fluid analysis and testicular volume in adults with epilepsy receiving valproate. J Clin Neurosci. 2015;22:508–512. doi: 10.1016/j.jocn.2014.08.029. [DOI] [PubMed] [Google Scholar]
  • 12.Isojärvi JI, Löfgren E, Juntunen KS, Pakarinen AJ, Päivänsalo M, Rautakorpi I, et al. Effect of epilepsy and antiepileptic drugs on male reproductive health. Neurology. 2004;62:247–253. doi: 10.1212/01.wnl.0000098936.46730.64. [DOI] [PubMed] [Google Scholar]
  • 13.Maier U, Koinig G. Andrological findings in young patients under long-term antidepressive therapy with clomipramine. Psychopharmacology. 1994;116:357–359. doi: 10.1007/BF02245340. [DOI] [PubMed] [Google Scholar]
  • 14.Tanrikut C, Feldman AS, Altemus M, Paduch DA, Schlegel PN. Adverse effect of paroxetine on sperm. Fertil Steril. 2010;94:1021–1026. doi: 10.1016/j.fertnstert.2009.04.039. [DOI] [PubMed] [Google Scholar]
  • 15.Puri S, Puri VN. Urinary vasopressin in male infertility. Arch Androl. 1985;14:263–265. doi: 10.3109/01485018508988309. [DOI] [PubMed] [Google Scholar]
  • 16.Pont A, Graybill JR, Craven PC, Galgiani JN, Dismukes WE, Reitz RE, et al. High-dose ketoconazole therapy and adrenal and testicular function in humans. Arch Intern Med. 1984;144:2150–2153. [PubMed] [Google Scholar]
  • 17.Tada Y, Hayashi T, Iwaki Y, Karita M, Taguchi S, Funabiki M, et al. Transient azoospermia following rosuvastatin medication for hypercholesterolemia. Clin Exp Obstet Gynecol. 2015;42:545–546. [PubMed] [Google Scholar]
  • 18.Wang J, Zhao Y, Jiang SB, Xia QH, Wei CX, Wang MW, et al. Assessment of tamsulosin as a potential male contraceptive in healthy volunteers. Urology. 2012;80:614–617. doi: 10.1016/j.urology.2012.06.003. [DOI] [PubMed] [Google Scholar]
  • 19.Gulati A, Tiwary AK, Jain S, Moudgil P, Gupta A. Intrasperm Ca2+ modulation and human ejaculated sperm viability: influence of miconazole, clotrimazole and loperamide. J Pharm Pharmacol. 2006;58:1145–1151. doi: 10.1211/jpp.58.8.0017. [DOI] [PubMed] [Google Scholar]
  • 20.Pant N, Shukla M, Upadhyay AD, Chaturvedi PK, Saxena DK, Gupta YK. Association between environmental exposure to p, p′-DDE and lindane and semen quality. Environ Sci Pollut Res Int. 2014;21:11009–11016. doi: 10.1007/s11356-014-2965-x. [DOI] [PubMed] [Google Scholar]
  • 21.Silvestroni L, Fiorini R, Palleschi S. Partition of the organochlorine insecticide lindane into the human sperm surface induces membrane depolarization and Ca2+ influx. Biochem J. 1997;321:691–698. doi: 10.1042/bj3210691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yeung CH, Cooper TG. Effects of the ion-channel blocker quinine on human sperm volume, kinematics and mucus penetration, and the involvement of potassium channels. Mol Hum Reprod. 2001;7:819–828. doi: 10.1093/molehr/7.9.819. [DOI] [PubMed] [Google Scholar]
  • 23.Anserini P, Chiodi S, Spinelli S, Costa M, Conte N, Copello F, et al. Semen analysis following allogeneic bone marrow transplantation. Additional data for evidence-based counselling. Bone Marrow Transplant. 2002;30:447–451. doi: 10.1038/sj.bmt.1703651. [DOI] [PubMed] [Google Scholar]
  • 24.Chatterjee R, Mills W, Katz M, McGarrigle HH, Goldstone AH. Germ cell failure and Leydig cell insufficiency in post-pubertal males after autologous bone marrow transplantation with BEAM for lymphoma. Bone Marrow Transplant. 1994;13:519–522. [PubMed] [Google Scholar]
  • 25.Fairley KF, Barrie JU, Johnson W. Sterility and testicular atrophy related to cyclophosphamide therapy. Lancet. 1972;1:568–569. doi: 10.1016/s0140-6736(72)90358-3. [DOI] [PubMed] [Google Scholar]
  • 26.Bujan L, Walschaerts M, Brugnon F, Daudin M, Berthaut I, Auger J, et al. Impact of lymphoma treatments on spermatogenesis and sperm deoxyribonucleic acid: a multicenter prospective study from the CECOS network. Fertil Steril. 2014;102:667–74.e3. doi: 10.1016/j.fertnstert.2014.06.008. [DOI] [PubMed] [Google Scholar]
  • 27.Baumgartner A, Schmid TE, Cemeli E, Anderson D. Parallel evaluation of doxorubicin-induced genetic damage in human lymphocytes and sperm using the comet assay and spectral karyotyping. Mutagenesis. 2004;19:313–318. doi: 10.1093/mutage/geh032. [DOI] [PubMed] [Google Scholar]
  • 28.Suzuki K, Yumura Y, Ogawa T, Saito K, Kinoshita Y, Noguchi K. Regeneration of spermatogenesis after testicular cancer chemotherapy. Urol Int. 2013;91:445–450. doi: 10.1159/000351189. [DOI] [PubMed] [Google Scholar]
  • 29.Chatterjee R, Haines GA, Perera DM, Goldstone A, Morris ID. Testicular and sperm DNA damage after treatment with fludarabine for chronic lymphocytic leukaemia. Hum Reprod. 2000;15:762–766. doi: 10.1093/humrep/15.4.762. [DOI] [PubMed] [Google Scholar]
  • 30.National Toxicology Program NTP-CERHR monograph on the potential human reproductive and developmental effects of hydroxyurea. NTP CERHR MON. 2008;21:vii–viii. [PubMed] [Google Scholar]
  • 31.Williams D, Crofton PM, Levitt G. Does ifosfamide affect gonadal function? Pediatr Blood Cancer. 2008;50:347–351. doi: 10.1002/pbc.21323. [DOI] [PubMed] [Google Scholar]
  • 32.Mariani S, Basciani S, Fabbri A, Agati L, Ulisse S, Lubrano C, et al. Severe oligozoospermia in a young man with chronic myeloid leukemia on long-term treatment with imatinib started before puberty. Fertil Steril. 2011;95:1120.e15–1120.e17. doi: 10.1016/j.fertnstert.2010.08.060. [DOI] [PubMed] [Google Scholar]
  • 33.Swerdloff RS, Heber D, Bhasin S, Rajfer J. Effect of GnRH superactive analogs (alone and combined with androgen) on testicular function in man and experimental animals. J Steroid Biochem. 1983;19:491–497. doi: 10.1016/0022-4731(83)90208-x. [DOI] [PubMed] [Google Scholar]
  • 34.Strowd RE, Blackwood R, Brown M, Harmon M, Lovato J, Yalcinkaya T, et al. Impact of temozolomide on gonadal function in patients with primary malignant brain tumors. J Oncol Pharm Pract. 2013;19:321–327. doi: 10.1177/1078155212469243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sieniawski M, Reineke T, Josting A, Nogova L, Behringer K, Halbsguth T, et al. Assessment of male fertility in patients with Hodgkin’s lymphoma treated in the German Hodgkin Study Group (GHSG) clinical trials. Ann Oncol. 2008;19:1795–1801. doi: 10.1093/annonc/mdn376. [DOI] [PubMed] [Google Scholar]
  • 36.Holtmann M, Gerstner S, Schmidt MH. Risperidone-associated ejaculatory and urinary dysfunction in male adolescents. J Child Adolesc Psychopharmacol. 2003;13:107–109. doi: 10.1089/104454603321666252. [DOI] [PubMed] [Google Scholar]
  • 37.Wang C, Lai CL, Lam KC, Yeung KK. Effect of cimetidine on gonadal function in man. Br J Clin Pharmacol. 1982;13:791–794. doi: 10.1111/j.1365-2125.1982.tb01867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hofer H, Donnerer J, Sator K, Staufer K, Scherzer TM, Dejaco C, et al. Seminal fluid ribavirin level and functional semen parameters in patients with chronic hepatitis C on antiviral combination therapy. J Hepatol. 2010;52:812–816. doi: 10.1016/j.jhep.2009.12.039. [DOI] [PubMed] [Google Scholar]
  • 39.Mckinney KA, Boyle P, Thompson W. Effect of glyceryl trinitrate on sperm motility and lipid peroxidation in normozoospermic men. Int J Androl. 1995;18:307–312. doi: 10.1111/j.1365-2605.1995.tb00566.x. [DOI] [PubMed] [Google Scholar]
  • 40.Peña JE, Thornton MH, Jr, Sauer MV. Reversible azoospermia: anabolic steroids may profoundly affect human immunodeficiency virus-seropositive men undergoing assisted reproduction. Obstet Gynecol. 2003;101:1073–1075. doi: 10.1016/s0029-7844(02)02330-x. [DOI] [PubMed] [Google Scholar]
  • 41.Kolettis PN, Purcell ML, Parker W, Poston T, Nangia AK. Medical testosterone: an iatrogenic cause of male infertility and a growing problem. Urology. 2015;85:1068–1072. doi: 10.1016/j.urology.2014.12.052. [DOI] [PubMed] [Google Scholar]
  • 42.Pons-Rejraji H, Brugnon F, Sion B, Maqdasy S, Gouby G, Pereira B, et al. Evaluation of atorvastatin efficacy and toxicity on spermatozoa, accessory glands and gonadal hormones of healthy men: a pilot prospective clinical trial. Reprod Biol Endocrinol. 2014;12:65. doi: 10.1186/1477-7827-12-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zuber J, Anglicheau D, Elie C, Bererhi L, Timsit MO, Mamzer-Bruneel MF, et al. Sirolimus may reduce fertility in male renal transplant recipients. Am J Transplant. 2008;8:1471–1479. doi: 10.1111/j.1600-6143.2008.02267.x. [DOI] [PubMed] [Google Scholar]
  • 44.Rahnema CD, Lipshultz LI, Crosnoe LE, Kovac JR, Kim ED. Anabolic steroid-induced hypogonadism: diagnosis and treatment. Fertil Steril. 2014;101:1271–1279. doi: 10.1016/j.fertnstert.2014.02.002. [DOI] [PubMed] [Google Scholar]
  • 45.De Toni L, Garolla A, Menegazzo M, Magagna S, Di Nisio A, Šabović I, et al. Heat sensing receptor TRPV1 is a mediator of thermotaxis in human spermatozoa. PLoS One. 2016;11:e0167622. doi: 10.1371/journal.pone.0167622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Moudgil P, Gupta A, Sharma A, Gupta S, Tiwary AK. Potentiation of spermicidal activity of 2′,4′-dichlorobenzamil by lidocaine. Indian J Exp Biol. 2002;40:1373–1377. [PubMed] [Google Scholar]
  • 47.Kumar VS, Sharma VL, Tiwari P, Singh D, Maikhuri JP, Gupta G, et al. In vitro—the spermicidal and antitrichomonas activities of SSRI antidepressants. Bioorg Med Chem Lett. 2006;16:2509–2512. doi: 10.1016/j.bmcl.2006.01.078. [DOI] [PubMed] [Google Scholar]
  • 48.Levin RM, Amsterdam JD, Winokur A, Wein AJ. Effects of psychotropic drugs on human sperm motility. Fertil Steril. 1981;36:503–506. [PubMed] [Google Scholar]
  • 49.José O, Hernández-Hernández O, Chirinos M, González-González ME, Larrea F, Almanza A, et al. In vitro-recombinant human ZP3-induced sperm acrosome reaction: evidence for the involvement of T- and L-type voltage-gated calcium channels. Biochem Biophys Res Commun. 2010;395:530–534. doi: 10.1016/j.bbrc.2010.04.059. [DOI] [PubMed] [Google Scholar]
  • 50.Whan LB, West MC, McClure N, Lewis SE. Effects of delta-9-tetrahydrocannabinol, the primary psychoactive cannabinoid in marijuana, on human sperm function in vitro. Fertil Steril. 2006;85:653–660. doi: 10.1016/j.fertnstert.2005.08.027. [DOI] [PubMed] [Google Scholar]
  • 51.Yerby MS, McCoy GB. Male infertility: possible association with valproate exposure. Epilepsia. 1999;40:520–521. doi: 10.1111/j.1528-1157.1999.tb00752.x. [DOI] [PubMed] [Google Scholar]
  • 52.Chan SY, Tang LC. Effect of reserpine on fertilizing capacity of human spermatozoa. Contraception. 1984;30:363–369. doi: 10.1016/s0010-7824(84)80028-1. [DOI] [PubMed] [Google Scholar]
  • 53.El-Gizawy SA, Aglan NI. Formulation and evaluation of metronidazole acid gel for vaginal contraception. J Pharm Pharmacol. 2003;55:903–909. doi: 10.1211/0022357021350. [DOI] [PubMed] [Google Scholar]
  • 54.Spade DJ, Hall SJ, Saffarini CM, Huse SM, McDonnell EV, Boekelheide K. Differential response to abiraterone acetate and di-n-butyl phthalate in an androgen-sensitive human fetal testis xenograft bioassay. Toxicol Sci. 2014;138:148–160. doi: 10.1093/toxsci/kft266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Focarelli R, Francavilla S, Francavilla F, Della Giovampaola C, Santucci A, Rosati F. A sialoglycoprotein, gp20, of the human capacitated sperm surface is a homologue of the leukocyte CD52 antigen: analysis of the effect of anti-CD52 monoclonal antibody (CAMPATH-1) on capacitated spermatozoa. Mol Hum Reprod. 1999;5:46–51. doi: 10.1093/molehr/5.1.46. [DOI] [PubMed] [Google Scholar]
  • 56.Hong CY, Chaput de Saintonge DM, Turner P. Effects of chlorpromazine and other drugs acting on the central nervous system on human sperm motility. Eur J Clin Pharmacol. 1982;22:413–416. doi: 10.1007/BF00542545. [DOI] [PubMed] [Google Scholar]
  • 57.Krämer BK, Neumayer HH, Stahl R, Pietrzyk M, Krüger B, Pfalzer B, et al. RADA2307 Study Group. Graft function, cardiovascular risk factors, and sex hormones in renal transplant recipients on an immunosuppressive regimen of everolimus, reduced dose of cyclosporine, and basiliximab. Transplant Proc. 2005;37:1601–1604. doi: 10.1016/j.transproceed.2004.09.026. [DOI] [PubMed] [Google Scholar]
  • 58.Levi M, Shalgi R, Brenner B, Perl G, Purim O, Amit L, et al. The impact of oxaliplatin on the gonads: from bedside to the bench. Mol Hum Reprod. 2015;21:885–893. doi: 10.1093/molehr/gav055. [DOI] [PubMed] [Google Scholar]
  • 59.Grunewald S, Jank A. New systemic agents in dermatology with respect to fertility, pregnancy, and lactation. J Dtsch Dermatol Ges. 2015;13(4):277-89; quiz 290. 10.1111/ddg.12596. [DOI] [PubMed]
  • 60.Caminos-Torres R, Ma L, Snyder PJ. Gynecomastia and semen abnormalities induced by spironolactone in normal men. J Clin Endocrinol Metab. 1977;45:255–260. doi: 10.1210/jcem-45-2-255. [DOI] [PubMed] [Google Scholar]
  • 61.Xu B, Wang ZP, Wang YJ, Lu PH, Wang LJ, Wang XH. The toxic effect of opioid analgesics on human sperm motility in vitro. Drug Chem Toxicol. 2013;36:205–208. doi: 10.3109/01480545.2012.710621. [DOI] [PubMed] [Google Scholar]
  • 62.Sobreiro BP, Lucon AM, Pasqualotto FF, Hallak J, Athayde KS, Arap S. Semen analysis in fertile patients undergoing vasectomy: reference values and variations according to age, length of sexual abstinence, seasonality, smoking habits and caffeine intake. Sao Paulo Med J. 2005;123:161–166. doi: 10.1590/S1516-31802005000400002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Moussa MM. Caffeine and sperm motility. Fertil Steril. 1983;39:845–848. [PubMed] [Google Scholar]
  • 64.Török L, Kádár L, Kása M. Spermatological investigations in patients treated with etretinate and isotretinoin. Andrologia. 1987;19:629–633. doi: 10.1111/j.1439-0272.1987.tb01915.x. [DOI] [PubMed] [Google Scholar]
  • 65.Abdul-Aziz M, MacLusky NJ, Bhavnani BR, Casper RF. Hyaluronidase activity in human semen: correlation with fertilization in vitro. Fertil Steril. 1995;64:1147–1153. [PubMed] [Google Scholar]
  • 66.Vigil P, Toro A, Godoy A. Physiological action of oestradiol on the acrosome reaction in human spermatozoa. Andrologia. 2008;40:146–151. doi: 10.1111/j.1439-0272.2007.00814.x. [DOI] [PubMed] [Google Scholar]
  • 67.Schlegel PN. Aromatase inhibitors for male infertility. Fertil Steril. 2012;98:1359–1362. doi: 10.1016/j.fertnstert.2012.10.023. [DOI] [PubMed] [Google Scholar]
  • 68.Neumann F, Diallo FA, Hasan SH, Schenck B, Traore I. The influence of pharmaceutical compounds on male fertility. Andrologia. 1976;8:203–235. doi: 10.1111/j.1439-0272.1976.tb02137.x. [DOI] [PubMed] [Google Scholar]
  • 69.Ko JK, Huang VW, Li RH, Yeung WS, Ho PC, Chiu PC. An in vitro study of the effect of mifepristone and ulipristal acetate on human sperm functions. Andrology. 2014;2:868–874. doi: 10.1111/j.2047-2927.2014.00261.x. [DOI] [PubMed] [Google Scholar]
  • 70.Koyun M, Baysal YE, Usta MF, Akman S, Güven AG. Evaluation of reproductive functions in male adolescents following renal transplantation. Pediatr Transplant. 2009;13:697–700. doi: 10.1111/j.1399-3046.2008.01052.x. [DOI] [PubMed] [Google Scholar]
  • 71.Dejaco C, Mittermaier C, Reinisch W, Gasche C, Waldhoer T, Strohmer H, et al. Azathioprine treatment and male fertility in inflammatory bowel disease. Gastroenterology. 2001;121:1048–1053. doi: 10.1053/gast.2001.28692. [DOI] [PubMed] [Google Scholar]
  • 72.Estrada LS, Champion HC, Wang R, Rajasekaran M, Hellstrom WJ, Aggarwal B, et al. Effect of tumour necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) on human sperm motility, viability and motion parameters. Int J Androl. 1997;20:237–242. doi: 10.1046/j.1365-2605.1997.00062.x. [DOI] [PubMed] [Google Scholar]
  • 73.Teo SK, Harden JL, Burke AB, Noormohamed FH, Youle M, Johnson MA, et al. Thalidomide is distributed into human semen after oral dosing. Drug Metab Dispos. 2001;29:1355–1357. [PubMed] [Google Scholar]
  • 74.Miska W, Schill WB. Influence of bradykinin antagonists on the motility of human spermatozoa enhanced by bradykinin. Arch Androl. 1994;33:1–5. doi: 10.3109/01485019408987795. [DOI] [PubMed] [Google Scholar]
  • 75.Vicari E. Effectiveness and limits of antimicrobial treatment on seminal leukocyte concentration and related reactive oxygen species production in patients with male accessory gland infection. Hum Reprod. 2000;15:2536–2544. doi: 10.1093/humrep/15.12.2536. [DOI] [PubMed] [Google Scholar]
  • 76.El-Garem Y, El-Sawy M, Mostafa T. Seminal Helicobacter pylori treatment improves sperm motility in infertile asthenozoospermic men. Urology. 2014;84:1347–1350. doi: 10.1016/j.urology.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 77.Sikka SC, Chen C, Almas M, Dula E, Knapp LE, Hellstrom WJ. Pregabalin does not affect sperm production in healthy volunteers: a randomized, double-blind, placebo-controlled, noninferiority study. Pain Pract. 2015;15:150–158. doi: 10.1111/papr.12171. [DOI] [PubMed] [Google Scholar]
  • 78.Haimov-Kochman R, Ben-Chetrit E. No or variable effect—the effect of colchicine treatment on sperm production and function: a review. Hum Reprod. 1998;13:360–362. doi: 10.1093/humrep/13.2.360. [DOI] [PubMed] [Google Scholar]
  • 79.Dobs AS, Miller S, Neri G, Weiss S, Tate AC, Shapiro DR, et al. Effects of simvastatin and pravastatin on gonadal function in male hypercholesterolemic patients. Metabolism. 2000;49:115–121. doi: 10.1016/s0026-0495(00)90938-7. [DOI] [PubMed] [Google Scholar]
  • 80.Curtis-Prior PB, Gadd AL. Beta-adrenoceptor antagonists and human sperm motility. J Pharm Pharmacol. 1990;42:220–222. doi: 10.1111/j.2042-7158.1990.tb05395.x. [DOI] [PubMed] [Google Scholar]
  • 81.Hellstrom WJ, Wang R, Peterson CA, Varady JC, Gesundheit N, Sikka SC. Effects of alprostadil and prazosin on motility, viability and membrane integrity of human sperm. J Urol. 1998;159:1559–1562. doi: 10.1097/00005392-199805000-00041. [DOI] [PubMed] [Google Scholar]
  • 82.Ko EM, Lowry RB, Martin RH. Analysis of sperm karyotypes in a patient treated with griseofulvin. Arch Androl. 2007;53:157–160. doi: 10.1080/01485010701314024. [DOI] [PubMed] [Google Scholar]
  • 83.Malallah YA, Zissis NP. Effect of minocycline on the sperm count and activity in infertile men with high pus cell count in their seminal fluid. J Chemother. 1992;4:286–289. doi: 10.1080/1120009x.1992.11739179. [DOI] [PubMed] [Google Scholar]
  • 84.Barkay J, Harpaz-Kerpel S, Ben-Ezra S, Gordon S, Zuckerman H. The prostaglandin inhibitor effect of antiinflammatory drugs in the therapy of male infertility. Fertil Steril. 1984;42:406–411. doi: 10.1016/s0015-0282(16)48081-7. [DOI] [PubMed] [Google Scholar]
  • 85.Knuth UA, Kühne J, Crosby J, Bals-Pratsch M, Kelly RW, Nieschlag E. Indomethacin and oxaprozin lower seminal prostaglandin levels but do not influence sperm motion characteristics and serum hormones of young healthy men in a placebo-controlled double-blind trial. J Androl. 1989;10:108–119. doi: 10.1002/j.1939-4640.1989.tb00071.x. [DOI] [PubMed] [Google Scholar]
  • 86.Morgante E, Gradini R, Realacci M, Sale P, D'Eramo G, Perrone GA, et al. Effects of long-term treatment with the anti-androgen bicalutamide on human testis: an ultrastructural and morphometric study. Histopathology. 2001;38:195–201. doi: 10.1046/j.1365-2559.2001.01077.x. [DOI] [PubMed] [Google Scholar]
  • 87.Tauer JT, Ulmer A, Glauche I, Jung R, Suttorp M. Long-term imatinib treatment does not cause testicular toxicity in male adolescents with chronic myeloid leukemia and in a juvenile rat model. Klin Padiatr. 2014;226:169–174. doi: 10.1055/s-0034-1372643. [DOI] [PubMed] [Google Scholar]
  • 88.Hill M, Milan S, Cunningham D, Mansi J, Smith I, Catovsky D, et al. Evaluation of the efficacy of the VEEP regimen in adult Hodgkin’s disease with assessment of gonadal and cardiac toxicity. J Clin Oncol. 1995;13:387–395. doi: 10.1200/JCO.1995.13.2.387. [DOI] [PubMed] [Google Scholar]
  • 89.Hong CY, Chiang BN, Ku J, Wu P. Screening the in vitro sperm-immobilizing effect of some anticancer drugs. Hum Toxicol. 1985;4:461–464. doi: 10.1177/096032718500400412. [DOI] [PubMed] [Google Scholar]
  • 90.Ribeiro MA, Gameiro LF, Scarano WR, Briton-Jones C, Kapoor A, Rosa MB, et al. Aromatase inhibitors in the treatment of oligozoospermic or azoospermic men: a systematic review of randomized controlled trials. JBRA Assist Reprod. 2016;20:82–88. doi: 10.5935/1518-0557.20160019. [DOI] [PubMed] [Google Scholar]
  • 91.Pectasides D, Pectasides E, Papaxoinis G, Skondra M, Gerostathou M, Karageorgopoulou S, et al. Testicular function in poor-risk nonseminomatous germ cell tumors treated with methotrexate, paclitaxel, ifosfamide, and cisplatin combination chemotherapy. J Androl. 2009;30:280–286. doi: 10.2164/jandrol.108.006437. [DOI] [PubMed] [Google Scholar]
  • 92.Farmakiotis D, Farmakis C, Rousso D, Kourtis A, Katsikis I, Panidis D. The beneficial effects of toremifene administration on the hypothalamic-pituitary-testicular axis and sperm parameters in men with idiopathic oligozoospermia. Fertil Steril. 2007;88:847–853. doi: 10.1016/j.fertnstert.2006.12.038. [DOI] [PubMed] [Google Scholar]
  • 93.Douglas JM, Jr, David LG, Remington ML, Paulsen CA, Perrin EB, Goodman P, et al. A double-blind, placebo-controlled trial of the effect of chronically administered oral acyclovir on sperm production in men with frequently recurrent genital herpes. J Infect Dis. 1988;157:588–593. doi: 10.1093/infdis/157.3.588. [DOI] [PubMed] [Google Scholar]
  • 94.Amory JK, Muller CH, Page ST, Leifke E, Pagel ER, Bhandari A, et al. Miglustat has no apparent effect on spermatogenesis in normal men. Hum Reprod. 2007;22:702–707. doi: 10.1093/humrep/del414. [DOI] [PubMed] [Google Scholar]
  • 95.van Leeuwen E, Wit FW, Repping S, Eeftinck Schattenkerk JK, Reiss P, van der Veen F, et al. Effects of antiretroviral therapy on semen quality. AIDS. 2008;22:637–642. doi: 10.1097/QAD.0b013e3282f4de10. [DOI] [PubMed] [Google Scholar]
  • 96.Tsourdi E, Kourtis A, Farmakiotis D, Katsikis I, Salmas M, Panidis D. The effect of selective estrogen receptor modulator administration on the hypothalamic-pituitary-testicular axis in men with idiopathic oligozoospermia. Fertil Steril. 2009;91:1427–1430. doi: 10.1016/j.fertnstert.2008.06.002. [DOI] [PubMed] [Google Scholar]
  • 97.Dwivedi C, Long NJ. Effect of cholinergic agents on human spermatozoa motility. Biochem Med Metab Biol. 1989;42:66–70. doi: 10.1016/0885-4505(89)90042-x. [DOI] [PubMed] [Google Scholar]
  • 98.Hendry WF, Munro DD. “Wash leather scrotum” (scrotal dermatitis): a treatable cause of male infertility. Fertil Steril. 1990;53:379–381. doi: 10.1016/s0015-0282(16)53304-4. [DOI] [PubMed] [Google Scholar]
  • 99.Farnsworth WH, Hoeg JM, Maher M, Brittain EH, Sherins RJ, Brewer HB., Jr Testicular function in type II hyperlipoproteinemic patients treated with lovastatin (mevinolin) or neomycin. J Clin Endocrinol Metab. 1987;65:546–550. doi: 10.1210/jcem-65-3-546. [DOI] [PubMed] [Google Scholar]
  • 100.Agirregoitia E, Valdivia A, Carracedo A, Casis L, Gil J, Subiran N, et al. Expression and localization of delta-, kappa-, and mu-opioid receptors in human spermatozoa and implications for sperm motility. J Clin Endocrinol Metab. 2006;91:4969–4975. doi: 10.1210/jc.2006-0599. [DOI] [PubMed] [Google Scholar]
  • 101.Corvasce A, Albino G, Leonetti T, Buonomo AF, Marucco EC. Once-a-day tadalafil administration improves the spermogram parameters in fertile patients. Arch Ital Urol Androl. 2015;87:210–213. doi: 10.4081/aiua.2015.3.210. [DOI] [PubMed] [Google Scholar]
  • 102.U.S. Food and Drug Administration. Guidance for industry: pregnancy, lactation, and reproductive potential: labeling for human prescription drug and biological products—content and format. 2014, Draft guidance. [Internet] [Cited 8 Aug 2017]. Available from: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM425398.pdf.
  • 103.U.S. Food and Drug Administration. Guidance for industry: reproductive and developmental toxicities—integrating study results to assess concerns. [Internet] September 2011. [Cited 8 Aug 2017]. Available from: www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm079240.pdf.
  • 104.Schapaugh AW, McFadden LG, Zorrilla LM, Geter DR, Stuchal LD, Sunger N, et al. Analysis of EPA’s endocrine screening battery and recommendations for further review. Regul Toxicol Pharmacol. 2015;72:552–561. doi: 10.1016/j.yrtph.2015.05.028. [DOI] [PubMed] [Google Scholar]
  • 105.Mak IW, Evaniew N, Ghert M. Lost in translation: animal models and clinical trials in cancer treatment. Am J Transl Res. 2014;6:114–118. [PMC free article] [PubMed] [Google Scholar]
  • 106.Odom DT, Dowell RD, Jacobsen ES, Gordon W, Danford TW, MacIsaac KD, et al. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nat Genet. 2007;39:730–732. doi: 10.1038/ng2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Shanks N, Greek R, Greek J. Are animal models predictive for humans? Philos Ethics Humanit Med. 2009;4:2. doi: 10.1186/1747-5341-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7:27–31. doi: 10.4103/0976-0105.177703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gray LE, Jr, Wilson V, Noriega N, Lambright C, Furr J, Stoker TE, et al. Use of the laboratory rat as a model in endocrine disruptor screening and testing. ILAR J. 2004;45:425–437. doi: 10.1093/ilar.45.4.425. [DOI] [PubMed] [Google Scholar]
  • 110.Cook JC, Klinefelter GR, Hardisty JF, Sharpe RM, Foster PM. Rodent Leydig cell tumorigenesis: a review of the physiology, pathology, mechanisms, and relevance to humans. Crit Rev Toxicol. 1999;29:169–261. doi: 10.1080/10408449991349203. [DOI] [PubMed] [Google Scholar]
  • 111.Johnson L, Petty CS, Neaves WB. A comparative study of daily sperm production and testicular composition in humans and rats. Biol Reprod. 1980;22:11233–11243. doi: 10.1093/biolreprod/22.5.1233. [DOI] [PubMed] [Google Scholar]
  • 112.Misell LM, Holochwost D, Boban D, Santi N, Shefi S, Hellerstein MK, et al. A stable isotope-mass spectrometric method for measuring human spermatogenesis kinetics in vivo. J Urol. 2006;175:242–246. doi: 10.1016/S0022-5347(05)00053-4. [DOI] [PubMed] [Google Scholar]
  • 113.Gaylor D, Ryan L, Krewski D, Zhu Y. Procedures for calculating benchmark doses for health risk assessment. Regul Toxicol Pharmacol. 1998;28:150–164. doi: 10.1006/rtph.1998.1247. [DOI] [PubMed] [Google Scholar]
  • 114.Neaves WB, Johnson L, Porter JC, Parker CR, Jr, Petty CS. Leydig cell numbers, daily sperm production, and serum gonadotropin levels in aging men. J Clin Endocrinol Metab. 1984;59:756–763. doi: 10.1210/jcem-59-4-756. [DOI] [PubMed] [Google Scholar]
  • 115.Oksel C, Ma CY, Liu JJ, Wilkins T, Wang XZ. Literature review of (Q)SAR modelling of nanomaterial toxicity. Adv Exp Med Biol. 2017;947:103–142. doi: 10.1007/978-3-319-47754-1_5. [DOI] [PubMed] [Google Scholar]
  • 116.Ford KA. Refinement, reduction, and replacement of animal toxicity tests by computational methods. ILAR J. 2016;57:226–233. doi: 10.1093/ilar/ilw031. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Assisted Reproduction and Genetics are provided here courtesy of Springer Science+Business Media, LLC

RESOURCES