<<

Laboratory Evidence for Male Infertility 3 Neel Parekh and Ashok Agarwal

Key Points 1678, when van Leeuwenhoek first described spermatozoa • The WHO manual is currently in its fifth edition or “semen animals” in the ejaculate [2]. However, it was not and updated the reference values. The lower refer- until 1951 that the integral role of semen in reproduction ence values provide a broader range of “normal” became evident. MacLeod’s landmark study compared parameters. semen parameters of 1000 men who conceived naturally and • Azoospermia is defined as the complete absence of 800 men who were presumed infertile and paved the way for after examination of a centrifuged pellet on modern semen analyses [3]. MacLeod plotted the semen two separate occasions obtained more than 2 weeks parameters on histograms, and the plots were divided into apart. quartiles for each of the two groups of men. He believed that • Healthy sperm DNA is bound to protamine and men in the lowest quartile would be infertile while men tightly packed to protect from stress and breakage above this threshold were likely fertile. The data was instru- during transport through the female reproductive mental in developing reference ranges for semen parameters tract. used in the first edition of the World Health Organization • The TUNEL assay identifies “nicks” or free ends of (WHO) manual in 1980 [4]. Since then, advances in assisted DNA by incorporating fluorescent nucleotides into reproductive technologies (ART) have shown considerably the site of damage. It is both highly specific and more options for subfertile and infertile men to achieve associated with a high positive predictive value to fecundity. Such changes have necessitated a more compre- quantify SDF. hensive and methodological assessment of infertile men. The • An AZFa or AZFb Y-microdeletion has rarely suc- aim of this chapter will be to discuss modern laboratory cessful sperm retrieval rates. However, sperm exams and techniques available to successfully evaluate and retrieval rates are as high as 80% in patients with an treat male factor infertility. isolated AZFc deletion. 3.2 WHO Guidelines for Assessment of Semen Specimen 3.1 Introduction Since 1980, the WHO has endeavored to standardize how The semen analysis has been the primary biomarker to evalu- laboratories analyze and report semen parameters globally. ate male infertility and is a direct reflection of the male’s The WHO manual is currently in its fifth edition which was fertility and overall health in the preceding 74 days [1]. published in 2010 [5]. The fifth edition aims to provide Semen analyses first began with the advent of microscopy in evidence-based­ reference ranges based off of populational studies. The new reference ranges are significantly lower than the previous manuals (Table 3.1) [5, 6]. Cooper et al. N. Parekh Department of Urology, Cleveland Clinic Foundation, analyzed semen samples from more than 4000 men from 14 Cleveland, OH, USA different countries on four continents, and reference values e-mail: [email protected] were calculated from men who naturally conceived with A. Agarwal (*) their partner within 12 months [7]. After the data was exam- American Center for Reproductive Medicine, Cleveland Clinic, ined, the 95th percentile for semen volume, concentration, Cleveland, OH, USA motility, vitality, and morphology was formulated, and the e-mail: [email protected]

© Springer Nature Switzerland AG 2020 27 S. J. Parekattil et al. (eds.), Male Infertility, https://doi.org/10.1007/978-3-030-32300-4_3 28 N. Parekh and A. Agarwal fifth percentile was set as the lower reference limit. There 3.3 Laboratory Evaluation of Male Factor are, however, several limitations to this study including sig- Infertility nificant population biases. Specifically, the majority of the cohort was from Europe, and only 10% of the study popula- 3.3.1 Basic Semen Analysis tion was from the southern hemisphere. Furthermore, the data is derived solely from a fertile cohort and was not The process of natural conception is intricate, culminating in compared to an infertile one; therefore, a “cutoff” between fusion of a healthy sperm to healthy ovum. Semen analyses infertile and fertile men cannot be determined [8]. As a provide valuable information for clinicians and patients but result of the lower reference values, men who would have is only a surrogate for male fertility and does not guarantee been categorized as having abnormal parameters using the paternity. It is unclear why some men with “normal” semen old reference values are now being classified as having nor- analyses suffer from infertility and those with “abnormal” mal parameters. Murray and colleagues determined that semen analyses may remain fertile [8]. Reproductive poten- roughly 15% of men who had one or more aberrant param- tial is also strongly influenced by female factors in many eters using the fourth edition values were categorized as couples and should be assessed when appropriate. To appro- having all parameters within the normal fifth edition refer- priately assess for male factor infertility, a thorough history ence ranges [9]. The fifth edition manual provides clini- and physical exam should be performed. Ideally, two semen cians a table which reviews where individual semen analyses should be obtained and performed one month apart. parameters lie within the 2.5th to 97.5th percentiles for Clinicians should be able to appropriately counsel male men who naturally conceived with their partner within patients on the protocol for proper semen collection and ulti- 12 months (Table 3.2) [5, 7]. This information can be infor- mately interpret the results [9]. mative for men when reviewing semen analysis parameters in the office. 3.3.1.1 Collection It is generally recommended that a semen specimen be col- lected at least 3 months after a febrile illness or stressful life Table 3.1 Evaluation of fourth and fifth editions of the WHO manual event. The WHO recommends providing a semen specimen semen analysis reference values after 2–7 days abstinence [5]. However, the optimal absti- Fifth edition lower nence interval may be 2–4 days, as studies have shown a rela- Fourth edition reference limit fifth Parameter (units) reference value centile (95% CI) tive improvement in total sperm concentration and motility Semen volume (ml) 2.0 1.5 (1.4–1.7) after 4 days [10]. A longer abstinence interval has been asso- Total sperm number 40 39 (33–46) ciated increased sperm DNA damage [11]. Preferably, the (106/ejaculate) specimen should be obtained in a sterile cup via masturbation Sperm concentration 20 15 (12–16) either at home or in the laboratory. However, sexual inter- (106/ml) course using a special collection condom that is not detrimen- Total motility (%) 50 40 (38–42) Progressive motility 25 32 (31–34) tal to sperm may also be utilized. Lubricants should be (%) avoided as they may alter sperm motility [12]. The semen Sperm morphology Not given but 4 (3–4) specimen should be evaluated within an hour of collection normal forms (%) suggests possibly and stored at room or body temperature until then. The 15 Clinical Laboratory Improvement Amendments (CLIA) has % Vitality (live 50 58 (55–63) spermatozoa) specific guidelines by which laboratories should observe to Peroxidase-positive <1.0 <1.0 ensure quality and accurate semen analysis results [13]. Once leukocytes (106/ml) the semen liquefies, typically within 20–60 minutes, a

Table 3.2 Review of semen analysis percentiles for men whose partners had a time to conception of ≤12 months Percentiles Parameters 2.5th 5th 10th 25th 50th 75th 90th 95th 97.5th Semen volume (ml) 1.2 1.5 2.0 2.7 3.7 4.8 6.0 6.8 7.6 Total sperm (106/ejaculate) 23 39 69 142 255 422 647 802 928 Sperm concentration (106/ml) 9 15 22 41 73 116 169 213 259 Total motility (%) 34 40 45 53 61 69 75 78 81 Progressive motility (%) 28 32 39 47 55 62 69 72 75 Sperm morphology normal forms (%) 3 4 5.5 9 15 24.5 36 44 48 3 Laboratory Evidence for Male Infertility 29

­macroscopic assessment of the semen sample is performed opposed to in drops. This is associated with hypofunction of which includes volume, pH, color, and viscosity. Subsequently, the prostate or seminal vesicles, infection, or high levels of microscopic examination is utilized to calculate the concen- leukocytospermia [15]. Trypsin can be used to treat hyper- tration, motility, morphology, and vitality [14]. viscous semen specimens prior to additional testing.

3.3.1.2 Volume 3.3.1.4 pH [normal > 7.2] Over 70% of the semen volume is provided by the seminal Measurement of pH is primarily determined by the balance vesicles, and current WHO criteria for normal semen volume of alkaline seminal vesicle fluid and the acidic prostatic is >1.5 ml [5]. A low semen volume following an appropriate secretions. The normal range of semen pH typically lies abstinence interval and complete collection may indicate between 7.2 and 8.0. An acidic pH is associated with obstruc- partial retrograde ejaculation, ejaculatory duct obstruction tion or seminal vesicle hypoplasia seen in men with congeni- (EDO), congenital absence of the vas deferens, or severe tal absence of the vas deferens [12, 13]. An alkaline pH > 8.0 hypogonadism. A high semen volume (>4 ml) may be seen may be associated with an underlying infection [8, 12]. in instances of prolonged sexual abstinence. 3.3.1.5 Concentration 3.3.1.3 Liquefaction and Viscosity Normal sperm concentration is defined as >15 million sperm/ Fibrinolysin is a proteolytic that is secreted by the mL and is determined after careful light microscopic examina- prostate and facilitates liquefaction of semen from the coag- tion of the wet preparation [5]. Typically, a minimum of 200 ulum state (~30–60 minutes). A defect in liquefaction may spermatozoa are counted utilizing counting chambers within a be indicative of an EDO or inadequate secretion of fibrinoly- grid pattern for accurate assessment (Fig. 3.1). The concentra- sin by the prostate. Semen viscosity is related to the fluid tion is then calculated and reported per milliliter. Alvarez et al. nature of the specimen and can be determined by utilizing a demonstrated that compared to other sperm parameters (count, 1.5 mm pipette to drop a semen specimen into a cup and morphology, motility, and volume), sperm concentration has examining the length of the thread formed. A thread length the highest intra-observer variation [16]. Oligozoospermia is >2 cm is considered abnormally viscous. There is a concern defined as <15 million sperm/mL, but incomplete collection or that increased viscosity impairs sperm motility and therefore short abstinence interval should be ruled out prior to making fertility, but this is controversial [8]. “Non-liquefaction” and this diagnosis. Azoospermia is defined as the complete absence “hyperviscosity” are two distinct conditions that are com- of sperm after examination of a centrifuged pellet on two sepa- monly interchanged inappropriately. “Non-liquefaction” is a rate occasions obtained more than 2 weeks apart. EDO, ejacu- result of the semen remaining in the coagulum state. latory dysfunction, and abnormal are “Hyperviscosity” is when the semen specimen pours thick as potential causes of azoospermia.

Fig. 3.1 Measurement of sperm concentration and motility by a fixed counting chamber and a phase-contrast microscope. (Reprinted with permission, Cleveland Clinic Center for Medical Art & Photography © 2013–2019. All Rights Reserved) 30 N. Parekh and A. Agarwal

Recently, studies have revealed that the total motile sperm been implicated as potential causes of asthenozoospermia count (volume × concentration × % motility) is more predic- [12, 16]. tive of infertility compared to concentration, motility, and volume alone [17, 18]. Compared to infertile men with nor- 3.3.1.7 Morphology mal semen parameters, those with total motile sperm count Over time, there have been significant shifts in the classifica- (TMSC) <1 million experienced 83% fewer natural pregnan- tion of sperm morphology which is a source of controversy. cies, while those with 10–20 million had 55% fewer preg- Normal morphology reference values have shifted from nancies [18]. Surprisingly, among men with TMSC ≥80.5% in the WHO first edition to≥ 4% normal forms in 0–1 million, 23% were able to achieve a natural pregnancy the WHO fifth edition [5, 21]. Stricter approaches and lack of within 3 years, further highlighting the predictive and diag- standardization in technician training may have contributed nostic limitations of semen analyses [18]. TMSC was also to these changes [21, 22]. To appropriately classify morphol- recently shown to be a better predictor than total sperm count ogy, the sperm should routinely be analyzed in a fixed, air for intracytoplasmic sperm injection (ICSI) outcomes dried, and stained semen smear. Either Shorr, Papanicolaou, ­compared with WHO values [19]. Clinically, TMSC facili- or Diff-Quik smear stains are approved by the WHO. This tates decision-making when counseling couples on appropri- process alone has been implicated in affecting morphologic ate treatment options, including assisted reproductive appearance [23, 24]. Normal sperm are typically comprised technologies. of a smooth oval head with a distinct acrosomal region cov- Computer-assisted semen analysis (CASA) is an auto- ering 40%–70% of the sperm head. Normal morphology mated method of determining sperm parameters which offers dimensions have been defined as a sperm head of 3–5 μm in several advantages over manual semen analyses. Specifically, length and 2–3 μm in width. Furthermore, the sperm should the basic parameters evaluated manually by an operator be devoid of head, midpiece, or tail defects [25]. (concentration, motility, and morphology) can be measured more precisely by CASA. CASA also provides useful kine- • Head defects include micro- or megalocephalic heads, matic information. However, CASA systems suffer from tapered head, missing acrosome (globozoospermia), other challenges such as costly equipment, more compli- heads with irregular forms, and bicephalic or multice- cated procedure, and poorer performance with low sperm phalic heads. concentrations [20]. • Midpiece defects consist of abnormal or no tail insertions. Distended, thin, bent, or elongated midpieces are 3.3.1.6 Motility abnormal. Sperm motility is determined by assessing sperm for signs of • Tail defects include coiling (associated with osmotic movement and is a critical indicator of the functionality of stress), multiple, short, 90° bending, or broken tails. spermatozoa. It should be characterized immediately follow- ing liquefaction to avoid temperature changes and dehydra- Sperm morphology male infertility–laboratory evaluation tion. The WHO considers normal total motility to be >40% is reported as the percentage of atypical forms present in the and is dependent on the ability of the sperm to traverse the ejaculate and classified according to the WHO criteria or the epididymis and successfully mature [5]. A motility of <5%– Kruger’s strict criteria. Teratozoospermia is defined as <4% 10% can be attributed to ultrastructural defects in the repro- normal morphology by the WHO method. Early studies ductive tract. The previously utilized WHO subclassification demonstrated that it is most likely the morphologically nor- of progressive motility into fast and slow has fallen out of mal appearing spermatozoa that are best able to navigate the favor due to high variability and inaccuracy among labora- female reproductive tract and fertilize the egg [26, 27]. More tory technicians. Currently, motility is simply divided into recent data suggests that abnormal morphology should not (1) progressive, space gaining motion; (2) nonprogressive, deter intrauterine insemination (IUI) or require couples to motion in place or in small circles; and (3) nonmotile or proceed immediately to in vitro male infertility–laboratory immotile [6]. evaluation: fertilization/intracytoplasmic sperm injection Asthenozoospermia (sperm motility <40%) can also be a (IVF/ICSI) [28–30]. The American Urological Association result of a poor specimen collection that may have been (AUA) recommends against utilizing isolated abnormalities exposed to a rubber condom, lubricants, or spermicides. in strict morphology to counsel couples on treatment deci- Sporadic clumps of agglutinated sperm are typically not con- sions [31]. cerning. However, clumping of sperm seen >10%–15% of the time is associated with antisperm antibodies (ASAs). 3.3.1.8 Agglutination ASAs may impair sperm motility and elicit an unusual Microscopic examination may identify sperm agglutination “shaking pattern” that can impede transit of sperm through or clumping and is characterized by sperm adhering to one the cervical mucous. Lengthy abstinence intervals, infection, another without other cells or debris. Sperm agglutination partial EDO, ultrastructural defects, and varicocele have all may be indictive of ASA. However, if there is clumping of 3 Laboratory Evidence for Male Infertility 31 sperm, cells, and debris, it is most likely a result of aggrega- ism, or lack of emission may present with low-volume ejacu- tion. Aggregates typically only consist of dead sperm while late on semen analysis. A PEAU is a routine laboratory test agglutination from ASA consists of a proportion of motile used to differentiate retrograde ejaculation from the afore- sperm. Clinicians should be aware that a small amount of mentioned conditions. The AUA Best Practice Statement agglutination may be unremarkable [6]. However, when sig- recommends that clinicians perform PEUA in patients with nificant agglutination is present, additional testing with ASA ejaculate volume <1.0 ml and in the absence of CBAVD or testing and/or semen cultures is recommended [12, 31]. hypogonadism [31]. PEUA is performed by microscopically assessing the pellet at ×400 magnification after approxi- 3.3.1.9 Leukocytospermia mately 10 minutes of centrifugation at a minimum of 300 g. Leukocytospermia is defined as >1 × 106/ml WBCs in the In patients diagnosed with aspermia or azoospermia, identi- semen and is commonly associated with idiopathic male fication of any sperm on PEUA is indictive of retrograde infertility [32]. It is important to avoid classifying immature ejaculation. However, in patients with low volume oligozoo- spermatozoa – spermatids and spermatocytes, which appear spermia, “significant numbers” of sperm should be identified as round cells – as seminal leukocytes. The primary sources on PEUA in order to diagnose retrograde ejaculation. There of leukocytes are from the prostate, seminal vesicles, vas is no expert consensus on “significant numbers” of sperm in deferens, and epididymis. They are unlikely to arise from the PEUA. testis, secondary to the blood–testis barrier [33]. If there are >5 round cells per high-powered field (HPF), further investi- gation is recommended. Immunocytochemistry is the gold 3.5 Reactive Oxygen Species (ROS) standard, but due to cost and difficulty standardizing the Testing monoclonal antibodies, it is not commonly performed by most laboratories. Peroxidase staining or the Endtz test is a There is mounting evidence that elevated ROS levels play an reliable substitute to properly identify leukocytes and distin- independent role in the etiology of male infertility [39, 40]. guish them from immature spermatozoa [34]. ROS are produced by leukocytes and abnormal spermatozoa Although leukocytospermia can be indicative of an infec- and are a natural byproduct of metabolic pathways. Small tious process (male accessory gland infection), recent inves- quantities of ROS are actually required to ensure normal tigation has determined that WBCs can exist in the absence spermatogenesis, successful capacitation, and the acrosome of infection or immune response [35]. Furthermore, leukocy- reaction [40, 41]. An overabundance of ROS levels com- tospermia has been implicated in negatively affecting sperm pared with antioxidants, however, may result in oxidative functionality through the formation of reactive oxygen spe- stress (OS) which has been observed to impair spermatogen- cies (ROS) [35, 36]. Sharma and colleagues have shown that esis as well as sperm kinetics [41]. Mitochondria have been ROS levels are increased, even with leukocytospermia levels identified as an essential component of seminal ROS produc- <0.2 million/mL, suggesting leukocyte levels lower than the tion, which is primarily mediated through the formation of WHO “cutoff” are detrimental [37]. superoxide in the electron transport chain [42]. Oxidative Athayde et al. demonstrated that men with a normal Endtz stress has been shown to negatively affect sperm physiology test had a 24% chance of natural conception, while men with and function through several pathways: increased rates of leukocytospermia levels <1 million/mL lowered natural con- DNA fragmentation [43], decrease in sperm motility [44], ception rates to 16% on 12-month follow-up [38]. and diminished quality of sperm [45]. Elevated seminal ROS Unfortunately, identifying leukocytospermia is not standard levels have been identified in 25%–80% of infertile men, but practice in many andrology laboratories or must be ordered levels are significantly lower in the fertile population, and it separately from the basic semen analysis. Routine urine and remains unclear if this scenario is predominately a correla- semen cultures should be performed if leukocytospermia is tive or a causative relationship with infertility [46, 47]. identified. Current therapies to treat leukocytospermia Clinically, measurement of seminal oxidative stress can include anti-inflammatory medications, antioxidants, and identify men where oxidative stress is associated with infer- antihistamines. Antibiotics are used to treat concurrent clini- tility and identifies a patient who may benefit from antioxi- cal infections. dant supplementation [46]. The ideal method of testing for ROS is controversial, likely due to a lack of standardization among laboratories 3.4 Retrograde or Post-Ejaculatory regarding reference values, equipment, and technique. Urinalysis (PEUA) Currently, there are a variety of direct and indirect ROS test- ing modalities utilized to determine seminal OS. However, Men with retrograde ejaculation (semen is redirected into the the most reliable and accurate modality has been luminol-­ urinary bladder), incomplete collection, EDO, congenital based chemiluminescence assay. This technique accurately bilateral absence of the vas deferens (CBAVD), hypogonad- measures both intracellular and extracellular ROS. Luminol 32 N. Parekh and A. Agarwal

Fig. 3.2 Measurement of ab ORP using (a) MiOXSYS analyzer and the (b) sensor. (Reprinted with permission, Cleveland Clinic Center for Medical Art & Photography © 2013–2019. All Rights Reserved)

is also able to react with various forms of ROS, including ­primary function of the is to release hydrogen peroxide, hydroxyl radical, and superoxide anion. the lytic and to digest the An adequate semen specimen is required to perform this test which ultimately allows for spermatozoa (>1 × 106/ml) and analyzed within 1 h of collection. Luminol fusion with the oocyte. Acrosome reaction testing is not adheres to the free radicals to produce a light signal that the routinely performed in many laboratories, but may be luminometer converts to an electrical signal. The quantity of considered when substantial irregularities of head mor- free radicals produced is measured as relative light units phology exist or the setting of recurrent IVF failure. (RLU)/s 106 sperm. Agarwal and colleagues [48] determined Acrosomal status can be examined through triple staining that in healthy controls with normal semen parameters, the with transmission electron microscopy, optic microscopy, physiologic ROS level is 102.2 RLU/s/106. Men above this , and fluorescently labeled lectins threshold value can be considered to be oxidative stress posi- [51–55]. tive and should be counseled appropriately. The latest advancement in laboratory diagnostics is the Male Infertility Oxidative System (MiOXSYS) (Fig. 3.2). It 3.7 Antisperm Antibody (ASA) Testing allows for an easy and accurate measurement of ROS through the oxidation-reduction potential (ORP). ORP, or redox A number of urologic conditions including testicular cancer, ­balance, measures for the homeostasis between reductants torsion, and traumatic or surgical disruption of the testis can and oxidants in a variety of biologic specimens. Several stud- violate the blood–testis barrier [56]. Breakdown of the ies have validated the reproducibility and reliability of the blood–testis barrier may predispose men to an immune MiOXSYS in measuring the ORP levels in semen specimens response to sperm in the form of ASA. ASA are deemed an of patients evaluated for male infertility [49, 50]. The important factor impairing pregnancy rates and are seen in MiOXSYS has several advantages over standard chemilumi- approximately 10% of infertile men as opposed to 2% of fer- nescence assays which include ease of use, smaller sample tile men [57, 58]. Extensive sperm agglutination is a result of volume (30 μL vs. 400 μL), and more forgiving measurement ASA which can impair the process of fertilization by inhibit- protocol (up to 120 minutes after specimen is produced) [50]. ing sperm penetration of cervical mucous and preventing While ROS measurement is not routinely recommended fusion of the sperm and oocyte. Sperm concentration and on initial evaluation of male infertility, it is a clinically useful motility have also been shown to be lower with the presence tool to further evaluate patients with varicocele, lifestyle fac- of ASA [12, 56]. tors (i.e., smoking), and idiopathic infertility [12, 31]. The mixed antiglobulin reaction (MAR) and immunobead test are qualitative laboratory tests used to detect IgA and IgM sperm antibodies. If ≥50% of motile sperm are bound 3.6 Acrosome Reaction Testing by antibodies, then these tests are considered abnormal [5]. The clinical utility of ASA testing is varied, but in men with The acrosome reaction is an essential step of successful isolated asthenospermia and normal concentration or with fertilization. The acrosome is a modified significant sperm agglutination, ASA testing should be con- that encompasses 40%–70% of the sperm head. The sidered [12, 31]. 3 Laboratory Evidence for Male Infertility 33

3.8 Sperm Viability Testing Table 3.3 Methods of evaluating sperm DNA damage Assay Measures Method Sperm viability testing or a vitality assay is a laboratory test Sperm chromatin Susceptibility of Flow cytometry utilized to facilitate identification of living sperm in the setting structure assay (SCSA) sperm DNA to based denaturation by heat of low motility (<25%). In ejaculated specimens with low or low pH motility or nonmotile surgically derived sperm to be used for Terminal Single- and Fluorescence IVF/ICSI, it is necessary to differentiate between necrozoo- deoxynucleotidyl double-stranded microscopy or spermia (dead sperm) and viable immotile sperm (asthenozoo- transferase dUTP nick DNA breaks flow cytometry spermia). This is done by evaluating the integrity of the sperm end labeling (TUNEL) based Single- gel Single- and Objective and which should be intact in living sperm. The fifth electrophoresis assay double-stranded quantitative, edition WHO reference parameter for vitality is ≥58% [5]. Cell (Comet) DNA breaks Altered fluorescence membrane integrity can be evaluated through the hypoosmotic bases microscopy swelling test (HOS test) and the dye exclusion test. Sperm chromatin Absence of sperm Flow cytometry dispersion (SCD) DNA damage - Halo based The dye exclusion test is dependent on the intact cell membrane of a live sperm resisting absorption of specific dyes. Eosin Y and nigrosin are two of the most commonly utilized dyes. Nigrosin is particularly useful as it will stain 3.9.2 Sperm Chromatin Structure Assay the background dark and provide good contrast for sperm (SCSA) assessment. Eosin Y will stain sperm heads red or dark pink if they are dead. It will stain live sperm heads white or light The SCSA is dependent on the principle that heat or acid pink. The HOS test is dependent on the ability of the live will denature sperm with an abnormal chromatin structure. sperm (intact cell membrane) to swell in hypotonic solu- Specifically, SCSA is a measure of the susceptibility of tions. The HOS test is favored over the dye exclusion test due sperm DNA to acid or heat denaturation utilizing flow to the ability of quickly assessing for necrozoospermia with- cytometry [63]. The metachromatic shift from green to red out damaging viable sperm [31]. Therefore, sperm used for a fluorescence is measured using flow cytometry. The per- HOS test can still be used for subsequent IVF/ICSI [58, 59]. centage of spermatozoa with red fluorescence (red– red + green fluorescence) is expressed as DNA fragmentation index (DFI). The clinical threshold for DFI is <30%. 3.9 Advanced Semen Testing Couples with >30% DFI have lower conception rates with natural intercourse and ART. It is a very sensitive assay and 3.9.1 Sperm DNA Fragmentation (SDF) has been reported as an excellent tool to detect dose– response relationship in men exposed to environmental Evaluation of sperm DNA fragmentation has evolved into an toxicants [64]. SCSA can evaluate over 10,000 cells rapidly essential tool for the overall assessment of male infertility. and has a well-standardized­ protocol, thus minimizing Sperm DNA is bound to protamine and tightly packed to pro- interlaboratory variations. The major drawbacks include tect from stress and breakage during transport through the expensive instrumentation (flow cytometry) and skilled female reproductive tract. The cause of DNA damage is technicians. likely multifactorial (i.e., smoking, drug use, environmental exposure, malignancy, chemotherapy, and varicocele) [60]. Elevated SDF has been identified in roughly 8% of subfertile 3.9.3 Terminal Deoxynucleotidyl Transferase men with normal semen parameters [61]. The integrity of dUTP Nick End Labeling (TUNEL) sperm DNA has been shown to affect couple’s fecundity through its impact on successful fertilization, healthy embryo The TUNEL assay identifies “nicks” or free ends of DNA by development, implantation, and pregnancy [62]. incorporating fluorescent nucleotides into the site of damage Several sperm DNA integrity assays have been introduced [65]. Single- and double-stranded DNA damage can be to evaluate the degree of SDF. In general, the assays were examined by either fluorescence microscopy or flow cytom- developed to facilitate clinicians in implementing early and etry. Advantages of the TUNEL assay include proven reli- effective management plans for specific clinical scenarios. ability, accuracy, and ease of use with low interlaboratory These scenarios include couples with recurrent pregnancy discrepancies [66]. Furthermore, by utilizing a threshold of loss, patients with idiopathic infertility, ART failure, and 16.8% for SDF, the TUNEL assay is both highly specific and varicocelectomy candidates. Commonly utilized methods associated with a high positive predictive value and will aid for evaluating SDF are summarized in Table 3.3. in its clinical utility [67]. 34 N. Parekh and A. Agarwal

3.9.4 Single-Cell Gel Electrophoresis Assay collection of blood for hormone testing is recommended due (Comet) to a normal physiologic decrease in testosterone levels throughout the day. If the testosterone level is low, then a The comet assay provides an objective and quantitative mea- repeat measurement of total and free testosterone should be sure of the degree of DNA damage per sperm [68]. Comet is obtained. In addition, measuring serum prolactin and lutein- able to detect not only single- and double-strand breaks but izing hormone (LH) is also advised. also abasic sites. The sperm are subject to electrophoresis at While normal serum FSH levels do not guarantee active high pH which results in structures resembling comets under spermatogenesis, an elevated or “high normal” FSH level is fluorescence microscopy. The number of DNA breaks is indicative of an abnormality in spermatogenesis such as quantified by the intensity of the comet tail relative to the hypergonadotropic hypogonadism (primary testicular fail- head. An advantage of this assay is that it requires only 5000 ure). Causes of primary testicular failure include Klinefelter’s sperm to evaluate SDF and is particularly useful in cases of syndrome, Noonan syndrome, and poorly functioning testes severe oligozoospermia [69]. (i.e., cryptorchidism, atrophy, or torsion). Obstructive azo- ospermia typically presents with normal testosterone and FSH/LH levels. When serum testosterone levels are low and 3.9.5 Sperm Chromatin Dispersion (SCD) gonadotropins are also either low or “inappropriately nor- mal,” hypogonadotropic hypogonadism (secondary testicu- The SCD test or Halosperm test is unique in that it is a mea- lar failure) may be the cause. Elevated prolactin levels, sure of the absence of SDF. The SCD test produces sperm hemochromatosis, opioid use, pituitary/hypothalamic dam- with a central core and a peripheral halo of dis- age, or genetic conditions (i.e., Kallman’s syndrome) are persed DNA loops. Sperm with nonfragmented DNA will associated with hypogonadotropic hypogonadism. demonstrate the characteristic halo, but sperm with DNA If hypogonadotropic hypogonadism is diagnosed or fragmentation will produce very small or no halo on fluores- symptoms suggest prolactinoma, then obtain a serum prolac- cent microscopy. While the SCD test is easy to use, high tin. Due to significant physiologic variability, serum prolac- interlaboratory variations has impeded widespread standard- tin levels should be repeated if initially abnormal. Slight ized usage [12]. elevations of serum prolactin (<50 ng/ml) can be associated with stress, kidney disease, certain medications, or be idio- pathic in nature. However, if the serum prolactin remains 3.9.6 Endocrine Evaluation abnormally elevated or is extremely high, then magnetic resonance imaging (MRI) should be performed to assess for Approximately 3% of infertile men will have a hormonal a pituitary tumor. imbalance as a basis of their subfertility [70]. The 2011 AUA Obese men have increased activity of the hormone aroma- Best Practice Statement recommends endocrinologic assess- tase, an enzyme found in adipose that converts testosterone ment in men with (1) abnormal semen analysis (sperm con- to estrogen. An overabundance of estrogen is associated with centration < 10 million/ml), (2) diminished sexual function, diminished libido, gynecomastia erectile dysfunction, and or (3) clinical findings of a small firm testis or altered mascu- hypogonadism. Evaluation of estrogen should be performed line features. The hypothalamus secretes gonadotropin-­ in obese infertile men or those with symptoms suggesting releasing hormone (GnRH) which traverses the hypophyseal hyperestrogenism. portal system to the anterior pituitary and stimulates the release of FSH and LH. These gonadotropins exert their effect on the testicle in a pulsatile fashion. Under normal conditions, 3.9.7 Genetic Evaluation LH stimulates the testicular Leydig cells to produce testoster- one. Testosterone provides negative feedback onto the hypo- Genetic testing is a key component of the male infertility thalamus by inhibiting GnRH release. FSH stimulates the evaluation. It provides necessary information to establish testicular Sertoli cells to promote spermatogenesis and secrete causes of infertility, identify clinically significant medical inhibin B and activin. Inhibin B causes negative feedback by comorbid conditions, and assess the likelihood of success of inhibiting the release of FSH from the anterior pituitary, certain fertility treatment options (i.e., microdissection tes- whereas activin stimulates the release of FSH from the ante- ticular sperm extraction or varicocele repair). Furthermore, it rior pituitary. The relationship between these hormones can allows the clinician to counsel partners on the potential risk help determine an underlying cause of subfertility. of transmitting genetic conditions to future progeny. The At a minimum, the initial endocrinologic workup should most common genetic causes of male infertility are cystic include a measurement of serum follicle-stimulating hor- fibrosis transmembrane conductance regulator (CFTR) gene mone (FSH) and morning serum testosterone levels. Morning mutations, Y-chromosome microdeletions, and chromosomal 3 Laboratory Evidence for Male Infertility 35

Table 3.4 Frequency of karyotype abnormalities in infertile men function tests have been developed to provide earlier and Azoospermia 10%–15% more individualized treatment for couples [73, 74]. Ongoing Oligozoospermia 5% investigation in the field of proteomics will continue to aid Normal <1% in the development of more successful treatment options All infertile men 7% and provide more answers for couples experiencing infertility. anomalies. The CFTR gene is located on chromosome 7 and is associated with cystic fibrosis (CF), CBAVD, and unilat- 3.10 Review Criteria eral absences of the vas. Almost all men with CF have a CFTR mutation and CBAVD. However, not all CFTR muta- We extensively searched Google Scholar, PubMed, Medline, tions are currently detected, and it is best to assume that men Clinical Key, and Science Direct for articles focusing on with CBAVD have a CFTR mutation and may be carriers of semen analyses, male infertility, advanced sperm testing, CF. It is thus imperative to assess the female partner for hormonal, and genetic assessment. We began our literature CFTR carrier status prior to proceeding with ART. search in September 2018 and completed it by November Karyotype evaluation and Y-chromosome microdeletions 2018. The following key words were utilized in our search: are frequently utilized genetic tests to evaluate patients with “semen analysis,” “sperm DNA fragmentation,” “oxidative severe oligozoospermia (<5 million/ml) or nonobstructive stress,” “WHO manual,” “TUNEL,” “oxidative-reduction azoospermia (NOA). A numeric or structural chromosomal potential,” “computer-assisted semen analysis.” We reviewed abnormality is identified in roughly 7% of infertile men, and only English language articles. Tables were created with the degree of infertility inversely correlates with the presence assistance from Microsoft Excel. of chromosomal abnormalities (see Table 3.4). Klinefelter’s syndrome is the most common chromosomal abnormality in infertile men and accounts for over 60% of chromosomal References abnormalities identified in workup of infertility. Couples should be counseled regarding the potential for paternal 1. Horst G, Koch A. Moving semen analysis into the future. Lancet transmission of chromosomal abnormalities as it can result Lab Newsl. 2017;1:1–2. 2. Lewenhoeck A. Observationes D. Anthonii Lewenhoeck, in miscarriage, birth defects, and other genetic syndromes. de natis e` semini genitali animalcules. Philos Trans. 1665– Y-chromosome microdeletions are identified in roughly 1678;1753(12):1040. 10%–15% of males with severe oligozoospermia or NOA 3. Cobb M. Hereditary before genetics: a history. Nat Rev Genet. [71, 72]. Typically, microdeletions occur in the AZF regions 2006;7:953. 4. World Health Organization. WHO laboratory manual for the exam- (AZFa, AZFb, and AZFc) on the long arm of the ination of human semen and semen-cervical mucus interaction. 1st Y-chromosome (the site of several genes required for normal ed. Singapore: Press Concern; 1980. spermatogenesis). A deletion within the Y-chromosome does 5. World Health Organization. WHO laboratory manual for the exam- not necessarily result in infertility, but sperm retrieval attempts ination and processing of human semen. 5th ed. Geneva: World Health Organization; 2010. are rarely successful with an AZFa or AZFb deletion. 6. World Health Organization. WHO laboratory manual for the exam- However, sperm retrieval rates are as high as 80% in patients ination of human semen and semen-cervical mucus interaction. 4th with an isolated AZFc deletion. It is unclear if there are any ed. Cambridge: Cambridge University Press; 1999. medical conditions associated with Y-chromosome microde- 7. Cooper TG, Noonan E, von Eckardstein S, et al. World Health Organization reference values for human semen characteristics. letions, but all of the male progeny will inherit the abnormal- Hum Reprod Update. 2010;16:231. ity, and appropriate genetic counseling is recommended. 8. Keep BA. How reliable are results from the semen analysis? Fertil Steril. 2004;82(1):41–4. 9. Agarwal A, Gupta S, Sharma R. Diagnostic tests in the evaluation of male infertility. In: Andrological evaluation of male infertility. 3.9.8 Conclusion 1st ed. Switzerland: Springer; 2016. p. 1–10. 10. Levitas E, Lunenfeld E, Weiss N, et al. Relationship between the The laboratory evaluation of male infertility extends far duration of sexual abstinence and semen quality: analysis of 9489 beyond a basic semen analysis. The clinician is tasked in semen samples. Fertil Steril. 2005;83:1680. 11. Agarwal A, Gupta S, Du Plessis S, Sharma R, Esteves SC, Cirenza identifying and treating reversible conditions that may C, et al. Abstinence time and its impact on basic and advanced improve the male’s fertility or even potentially detect harm- semen parameters. Urology. 2016;94:102–10. ful underlying conditions. The fertility evaluation must be 12. Niederberger CS. Male infertility. In Wein A, Kavoussi LR, Partin complemented by a thorough history and physical examina- AW, Peters CA, editors. Campbell-Walsh urology. 11th ed. Elsevier; 2016. p. 556–79. tion with an understanding of genetic and endocrinological 13. CLIA website: www.cdc.gov/clia/default.aspx. Accessed Nov conditions which cause male infertility. Advances in sperm 2018. 36 N. Parekh and A. Agarwal

14. Esteves SC, Zini A, Aziz N, Alvarez JG, Sabanegh ES Jr, Agarwal 35. Lackner JE, Agarwal A, Mahfouz R, du Plessis SS, Schatzl G. The A. Critical appraisal of World Health Organization’s new reference association between leukocytes and sperm quality is concentration values for human semen characteristics and effect on diagnosis and dependent. Reprod Biol Endocrinol. 2010;8:12. treatment of subfertile men. Urology. 2012;79:16–22. 36. Aitken RJ, Clarkson JS. Cellular basis of defective sperm function 15. Du Plessis SS, Gokul S, Agarwal A. Semen hyperviscosity: causes, and its association with the genesis of reactive oxygen species by consequences, and cures. Front Biosci (Elite Ed). 2013;5:224–31. human spermatozoa. J Reprod Fertil. 1987;81(2):459–69. 16. Alvarez C, Castilla JA, Martinez L, Ramirez JP, Vergara F, Gaforio 37. Sharma RK, Pasqualotto AE, Nelson DR, Thomas AJ Jr, Agarwal JJ. Biological variation of seminal parameters in healthy subjects. A. Relationship between seminal white blood cell counts and Hum Reprod. 2003;18:2082–8. oxidative stress in men treated at an infertility clinic. J Androl. 17. Bonde JP, Ernst E, Jensen TK, Hjollund NH, Kolstad H, Henriksen 2001;22:575–83. TB, et al. Relation between semen quality and fertility: a 38. Athayde KS, Cocuzza M, Agarwal A, Krajcir N, Lucon AM, Srougi population-­based study of 430 first-pregnancy planners. Lancet. M, Hallak J. Development of normal reference values for semi- 1998;352:1172–7. nal reactive oxygen species and their correlation with leukocytes 18. Hamilton JA, Cissen M, Brandes M, et al. Total motile sperm count: and semen parameters in a fertile population. J Androl. 2007;28: a better indicator for the severity of male factor infertility than the 613–20. WHO sperm classification system. Hum Reprod. 2015;30:1110–21. 39. Desai N, Sharma R, Makker K, Sabanegh E, Agarwal A. Physiologic 19. Borges E Jr, Setti AS, Braga DP, Figueira RC, Iaconelli A Jr. and pathologic levels of reactive oxygen species in neat semen of Total motile sperm count has a superior predictive value over the infertile men. Fertil Steril. 2009;92(5):1626–31. WHO 2010 cut-off values for the outcomes of intracytoplasmic 40. Agarwal A, Sharma RK, Nallella KP, Thomas AJ Jr, Alvarez JG, sperm injection cycles. Andrology. 2016; https://doi.org/10.1111/ Sikka SC. Reactive oxygen species as an independent marker of andr.12199. male factor infertility. Fertil Steril. 2006;86(4):878–85. 20. Rothmann SA, Reese AA. Semen analysis. In: Lipshultz LI, 41. Menezo YJ, Hazout A, Panteix G, et al. Antioxidants to reduce Howards SS, Niederberger CS, editors. Infertility in the male. 4th sperm DNA fragmentation: an unexpected adverse effect. Reprod ed. New York: Cambridge University Press; 2009. p. 550–73. Biomed Online. 2007;14:418–21. 21. World Health Organization. WHO laboratory manual for the exam- 42. Gosalvez J, Gonzalez-Martinez M, Lopez-Fernandez C, et al. ination of human semen and semen-cervical mucus interaction. 2nd Shorter abstinence decreases sperm deoxyribonucleic acid frag- ed. Cambridge: Cambridge University Press; 1987. mentation in ejaculate. Fertil Steril. 2011;96:1083–6. 22. Morbeck DE, Leonard PH, Weaver AL, et al. Sperm morphology: 43. Matsuura R, Takeuchi T, Yoshida A. Preparation and incubation classification drift over time and clinical implications. Fertil Steril. conditions affect the DNA integrity of ejaculated human spermato- 2011;96:1350. zoa. Asian J Androl. 2010;12:753–9. 23. Niederberger C. Re: Status of sperm morphology assessment: an 44. Agarwal A, Durairajanayagam D, du Plessis SS. Utility of antioxi- evaluation of methodology and clinical value. J Urol. 2015;194:763. dants during assisted reproductive techniques: an evidence based 24. Kashanian JA, Brannigan RE. Sperm morphology and reproductive review. Reprod Biol Endocrinol. 2014;12:112. outcomes: a perplexing relationship. Fertil Steril. 2014;102:1561. 45. Nabi A, Khalili MA, Halvaei I, et al. Prolonged incubation of 25. Abraham-Peskir JV, Chantler E, Uggerhøj E, et al. Response of processed human spermatozoa will increase DNA fragmentation. midpiece vesicles on human sperm to osmotic stress. Hum Reprod. Andrologia. 2014;46:374–9. 2002;17:375. 46. Deepinder F, Cocuzza M, Agarwal A. Should seminal oxidative 26. Menkveld R, Franken DR, Kruger TF, et al. Sperm selection capac- stress measurement be offered routinely to men presenting for ity of the human zona pellucida. Mol Reprod Dev. 1991;30:346. infertility evaluation? Endocr Pract. 2008;14:484–49. 27. Liu DY, Baker HW. Morphology of spermatozoa bound to the zona 47. Lamirande E, Gagnon C. Impact of reactive oxygen species on pellucida of human oocytes that failed to fertilize in vitro. J Reprod spermatozoa: a balancing act between beneficial and detrimental Fertil. 1992;94:71. effects. Hum Reprod. 1995;10:15–21. 28. French DB, Sabanegh ES Jr, Goldfarb J, et al. Does severe tera- 48. Agarwal A, Ahmad G, Sharma R. Reference values of reactive oxy- tozoospermia affect blastocyst formation, live birth rate, and gen species in seminal ejaculates using chemiluminescence assay. J other clinical outcome parameters in ICSI cycles? Fertil Steril. Assist Reprod Genet. 2015;32(12):1721–9. 2010;93:1097. 49. Agarwal A, Sharma R, Roychoudhury R, Du Plessis S, Sabanegh 29. Keegan BR, Barton S, Sanchez X, et al. Isolated teratozoospermia E. MiOXSYS: a novel method of measuring oxidation reduc- does not affect in vitro fertilization outcome and is not an indication tion potential in semen and seminal plasma. Fertil Steril. for intracytoplasmic sperm injection. Fertil Steril. 2007;88:1583. 2016;106(3):566–73. 30. Hotaling JM, Smith JF, Rosen M, et al. The relationship between 50. Fernandez JL, Muriel L, Rivero MT, Goyanes V, Vazquez R, isolated teratozoospermia and clinical pregnancy after in vitro fer- Alvarez JG. The sperm chromatin dispersion test: a simple method tilization with or without intracytoplasmic sperm injection: a sys- for the determination of sperm DNA fragmentation. J Androl. tematic review and meta-analysis. Fertil Steril. 2011;95:1141. 2003;24:59–66. 31. American Urological Association Education and Research, Inc. 51. Kohn FM, Mack SR, Schill WB, Zaneveld LJ. Detection of human The optimal evaluation of the infertile male: AUA best practice sperm acrosome reaction: comparison between methods using dou- statement, 2010. Available at https://www.auanet.org/common/pdf/ ble staining, Pisum sativum agglutinin, concanavalin A and trans- education/clinical-guidance/Male-Infertility-d.pdf. Accessed Nov mission electron microscopy. Hum Reprod. 1997;12(4):714–21. 2018. 52. Lee MA, Trucco GS, Bechtol KB, Wummer N, Kopf GS, Blasco 32. Branigan EF, Spadoni LR, Muller CH. Identification and treat- L, et al. Capacitation and acrosome reactions in human spermato- ment of leukocytospermia in couples with unexplained infertility. zoa monitored by chlortetracycline fluorescence assay. Fertil Steril. J Reprod Med. 1995;40:625–9. 1987;48(4):649–58. 33. el-Demiry MI, Hargreave TB, Busuttil A, et al. Lymphocyte sub- 53. Talbot P, Chacon RS. A new technique for evaluating normal acro- populations in the male genital tract. Br J Urol. 1985;57:769. some reactions of human sperm. J Cell Biol. 1979;83:2089. 34. Shekarriz M, Thomas AJ Jr, Agarwal A. Incidence and level 54. Wolf DP, Boldt J, Byrd W, Bechtol KB. Acrosomal status evalua- of seminal reactive oxygen species in normal men. Urology. tion in human ejaculated sperm with monoclonal antiboides. Biol 1995;45(1):103–7. Reprod. 1985;32(5):1157–62. 3 Laboratory Evidence for Male Infertility 37

55. Fenichel P, Hsi BL, Farahifar D, Donzeau M, Barrier-Delpech D, 65. Sun JG, Jurisicova A, Casper RF. Detection of deoxyribonucleic Yehy CJ. Evaluation of the human sperm acrosome reaction using acid fragmentation in human sperm: correlation with fertilization monoclonal antibody, GB24, and fluorescence-activated cell sorter. in vitro. Biol Reprod. 1997;56:602–7. J Reprod Fertil. 1989;87(2):699–706. 66. LeSaint C, Vingataramin L, Alix S, et al. Correlation between 56. Cui D, Han G, Shang Y, et al. Antisperm antibodies in infertile two sperm DNA fragmentation tests (TUNEL and SCSA) and men and their effect on semen parameters: a systematic review and evaluation of TUNEL assay inter-lab variabiality. Fertil Steril. meta-analysis. Clin Chim Acta. 2015;444:29. 2016;106:e297. 57. Adeghe JH. Male subfertility due to sperm antibodies: a clinical 67. Sharma R, Ahmad G, Esteves SC, et al. Terminal deoxynucleotidyl overview. Obstet Gynecol Surv. 1993;48(1):1–8. transferase dUTP nick end labeling (TUNEL) assay using bench 58. Guzick DS, Overstreet JW, Factor-Litvak P, Brazil CK, Nakajima top flow cytometer for evaluation of sperm DNA fragmentation in S, Coutifaris C, Carson SA, Cisneros P, Steinkampf MP, Hill JA, fertility laboratories: protocol, reference values, and quality con- et al. Sperm morphology, motility, and concentration in fertile and trol. J Assist Reprod Genet. 2016;33:291–300. infertile men. N Engl J Med. 2001;345:1388–93. 68. Singh NP, McCoy MT, Tice RR, et al. A simple technique for quan- 59. Verheyen G, Joris H, Critis K, et al. Comparison of different titation of low levels of DNA damage in individual cells. Exp Cell hypoosmotic swelling solutions to select viable immotile sperma- Res. 1988;175:184–91. tozoa for potential use in intracytoplasmic sperm injection. Hum 69. Lewis SE, John Aitken R, Conner SJ, et al. The impact of sperm Reprod Update. 1997;3:195. DNA damage in assisted conception and beyond: recent advances in 60. Agarwal A, Said T. Role of sperm chromatin abnormalities and diagnosis and treatment. Reprod Biomed Online. 2013;27:325–37. DNA damage in male infertility. Hum Reprod Update. 70. Sigman M, Jarow JP. Ipsilateral testicular hypotrophy is associated 2003;9:331–45. with decreased sperm counts in infertile men with varicoceles. J 61. Aitken RJ, Irvine DS, Wu FC. Prospective analysis of sperm–oocyte Urol. 1997;158:605–7. fusion and reactive oxygen species generation as a criteria for the 71. DeBraekeleer M, Dao TN. Cytogenetic studies in male infertility: a diagnosis of infertility. Am J Obstet Gynecol. 1991;164:542–51. review. Hum Reprod. 1991;6:245–50. 62. Ward WS. Function of sperm chromatin structural elements in fer- 72. Pryor JL, Kent-First M, Muallem A, et al. Microdeletions in the Y tilization and development. Mol Hum Reprod. 2010;16:30–6. chromosome of infertile men. N Engl J Med. 1997;336:534–9. 63. Evenson DP, Jost LK, Marshall D, et al. Utility of the sperm chro- 73. Esteves SC, Hamada A, Kondray V, Pitchika A, Agarwal A. What matin structure assay as a diagnostic and prognostic tool in the every gynecologist should know about male infertility: an update. human fertility clinic. Hum Reprod. 1999;14:1039–49. Arch Gynecol Obstet. 2012;286:217–29. 64. Evenson DP, Wixon R. Environmental toxicants cause sperm DNA 74. Esteves SC, Miyaoka R, Agarwal A. An update on the clinical fragmentation as detected by the Sperm Chromatin Structure Assay assessment of the infertile male. [corrected]. Clinics (Sao Paulo). (SCSA). Toxicol Appl Pharmacol. 2005;207:532–7. 2011;66:691–700. (Erratum in: Clinics (Sao Paulo). 2012; 67: 203).