1
|
Utigalieva E, Morozov A, Shoshany O, Suvorov A, Taratkin M, Manfredi C, Falcone M, Bezrukov E, Fajkovic H, Russo GI, Enikeev D. A systematic review and meta-analysis of the placebo effect on both semen quality and male infertility. Minerva Urol Nephrol 2024; 76:423-435. [PMID: 39051890 DOI: 10.23736/s2724-6051.24.05559-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Placebo influence on such objective indicators, as sperm quality and infertility, has not been studied previously, but some studies report that placebo may distort even objective outcomes. The aim of current study is to assess the placebo effect on fertility in patients suffering from sperm abnormalities and/or infertility. EVIDENCE ACQUISITION We conducted a search of two databases (Scopus and MEDLINE) and identified placebo-controlled clinical trials which focused on sperm abnormalities and/or male infertility treatment. Primary outcomes included changes in semen parameters (volume, total count, sperm concentration in semen, progressive motility, morphology (normal cells)). Secondary outcomes included DNA fragmentation and change in pregnancy rate. EVIDENCE SYNTHESIS Seventy-seven articles published from 1983 to 2022 were included. Statistically significant changes were observed for the following values: total sperm count, mean change 0.16 (95% CI 0.05, 0.26); P=0.004, I2=75.1%; and progressive motility, mean change 0.13 (95% CI 0.02, 0.24); P=0.026, I2=84.9%. In contrast, placebo did not affect sperm concentration, sperm volume, sperm morphology or DNA fragmentation index. The publication bias for all the values measured with Egger's test and funnel plots was low. CONCLUSIONS The current meta-analysis indicated a statistically significant increase of total sperm count and progressive motility in the placebo group. In contrast, placebo did not affect sperm concentration, sperm volume, sperm morphology and DNA fragmentation index. These findings should be considered while planning or analyzing placebo-controlled clinical trials.
Collapse
Affiliation(s)
- Elvira Utigalieva
- Institute for Clinical Medicine, Sechenov University, Moscow, Russia
| | - Andrey Morozov
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Ohad Shoshany
- Urology Section, Beilinson Hospital, Rabin Medical Center, Petah Tiqva, Israel
| | - Aleksandr Suvorov
- Digital Biodesign and Personalized Healthcare World-Class Research Center, Sechenov University, Moscow, Russia
| | - Mark Taratkin
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Celeste Manfredi
- Department of Woman, Child and General and Specialized Surgery, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Marco Falcone
- Section of Urology U, Molinette Hospital, Turin, Italy
- Section of Neurourology, USU/CTO Hospital, Turin, Italy
- AOU Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Evgeny Bezrukov
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Harun Fajkovic
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
| | | | - Dmitry Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia -
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
- Department of Urology, Rabin Medical Center, Petach Tiqwa, Israel
| |
Collapse
|
2
|
Pecora G, Sciarra F, Gangitano E, Venneri MA. How Food Choices Impact on Male Fertility. Curr Nutr Rep 2023; 12:864-876. [PMID: 37861951 PMCID: PMC10766669 DOI: 10.1007/s13668-023-00503-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 10/21/2023]
Abstract
PURPOSE OF REVIEW Increasing evidence on the significance of nutrition in reproduction is emerging from both animal and human studies, suggesting an association between nutrition and male fertility. Here, we have highlighted the impact of the various food groups on reproductive hormones and on spermatogenesis, and the effects of classical and latest dietary patterns such as Mediterranean diet, Western diet, intermittent fasting, ketogenic diet, and vegan/vegetarian diet on male fertility. RECENT FINDINGS Nutrients are the precursors of molecules involved in various body's reactions; therefore, their balance is essential to ensure the correct regulation of different systems including the endocrine system. Hormones are strongly influenced by the nutritional status of the individual, and their alteration can lead to dysfunctions or diseases like infertility. In addition, nutrients affect sperm production and spermatogenesis, controlling sexual development, and maintaining secondary sexual characteristics and behaviors. The consumption of fruit, vegetables, fish, processed meats, dairy products, sugars, alcohol, and caffeine importantly impact on male fertility. Among dietary patterns, the Mediterranean diet and the Western diet are most strongly associated with the quality of semen. Nutrients, dietary patterns, and hormonal levels have an impact on male infertility. Therefore, understanding how these factors interact with each other is important for strategies to improve male fertility.
Collapse
Affiliation(s)
- Giulia Pecora
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 329, 00161, Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 329, 00161, Rome, Italy
| | - Elena Gangitano
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 329, 00161, Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 329, 00161, Rome, Italy.
| |
Collapse
|
3
|
Methorst C, Faix A, Huyghe E. [Medical treatments for male infertility]. Prog Urol 2023; 33:653-680. [PMID: 38012910 DOI: 10.1016/j.purol.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Treatments to stimulate spermatogenesis and antioxidant food supplements are often offered to infertile patients either before sperm extraction surgery to improve results, or as part of medically assisted reproduction or spontaneous fertility to increase the likelihood of a live birth. METHODS A bibliographic search limited to English-language literature on men published before 5/2023 was carried out, including clinical trials, literature reviews and meta-analyses on spermatogenesis-stimulating molecules and antioxidant treatments. RESULTS Several medical treatments seem capable of improving male fertility: they act mainly by stimulating spermatogenesis through hormones, or by reducing the effects of oxidative stress. With regard to oligoasthenozoospermia, the literature shows that certain hormonal treatments stimulating spermatogenesis are useful. In the case of non-obstructive azoospermia, the value of treatment depends on the patient's FSH and testosterone levels. AOX supplementation appears to improve certain spermogram parameters and have an impact on pregnancy and live birth rates. CONCLUSION This review should help urologists gain a better understanding of the various medical treatments and enable them to define an appropriate therapeutic strategy, tailored to the patient and the couple, in order to obtain the best results.
Collapse
Affiliation(s)
- C Methorst
- Service de médecine de la reproduction, hôpital des 4-villes, Saint-Cloud, France
| | - A Faix
- Clinique Saint-Roch, 560 avenue du Colonel-Pavelet-dit-Villars 34000 Montpellier, France
| | - E Huyghe
- Département d'urologie, hôpital de Rangueil, CHU de Toulouse, Toulouse, France; Service de médecine de la reproduction, hôpital Paule-de-Viguier, CHU de Toulouse, Toulouse, France; UMR DEFE, Inserm 1203, université de Toulouse, université de Montpellier, Montpellier, France.
| |
Collapse
|
4
|
Romeo M, Donno V, Spaggiari G, Granata ARM, Simoni M, La Marca A, Santi D. Gonadotropins in the Management of Couple Infertility: Toward the Rational Use of an Empirical Therapy. Semin Reprod Med 2023; 41:258-266. [PMID: 38158195 DOI: 10.1055/s-0043-1777837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Although epidemiology shows that both men and woman can experience infertility, the female partner usually experiences most of the diagnostic and therapeutic burden. Thus, management of couple infertility is a unique example of gender inequality. The use of exogenous gonadotropins in assisted reproductive technology (ART) to induce multifollicular growth is well consolidated in women, but the same is not done with the same level of confidence and purpose in infertile men. Indeed, the treatment of idiopathic male infertility is based on an empirical approach that involves administration of the follicle-stimulating hormone (FSH) in dosages within the replacement therapy range. This treatment has so far been attempted when the endogenous FSH serum levels are within the reference ranges. According to the most recent evidence, a "substitutive" FSH administration may not be effective enough, while a stimulatory approach could boost spermatogenesis over its basal levels without adverse extragonadal effects. This article aims to describe the rationale behind the empirical application of gonadotropins in couple infertility, highlighting the need for a change in the therapeutic approach, especially for the male partner.
Collapse
Affiliation(s)
- Marilina Romeo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Valeria Donno
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgia Spaggiari
- Department of Medical Specialties, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Department of Medical Specialties, Unit of Andrology and Sexual Medicine, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Antonio R M Granata
- Department of Medical Specialties, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Department of Medical Specialties, Unit of Andrology and Sexual Medicine, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Antonio La Marca
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Department of Medical Specialties, Unit of Andrology and Sexual Medicine, Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| |
Collapse
|
5
|
Santi D, Spaggiari G, Dalla Valentina L, Romeo M, Nuzzo F, Serlenga L, Roli L, De Santis MC, Trenti T, Granata ARM, Simoni M. Sperm Concentration Improvement May Be a Parameter Predicting Efficacy of FSH Therapy of Male Idiopathic Infertility. Cells 2023; 12:2236. [PMID: 37759459 PMCID: PMC10527800 DOI: 10.3390/cells12182236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Testis stimulation with follicle-stimulating hormone (FSH) is one of the empirical treatments proposed for male idiopathic infertility, although reliable markers to predict its efficacy are still lacking. This study aimed to identify parameters able to predict FSH efficacy in terms of pregnancy achievement. A real-world study was conducted, enrolling idiopathic infertile men treated with FSH 150IU three times weekly. Patients were treated until pregnancy achievement or for a maximum of two years and two visits were considered: V0 (baseline) and V1 (end of FSH treatment). Primary endpoints were the V1-V0 percentage change in sperm concentration, total sperm count, and total motile sperm number. In total, 48 pregnancies were recorded (27.7%) among 173 men (age 37.9 ± 6.2 years). All three endpoints increased after FSH administration, and only the V1-V0 percentage of sperm concentration significantly predicted pregnancy (p = 0.007). A V1-V0 sperm concentration of 30.8% predicted pregnancy, and the sperm concentration V1-V0 percentage (Y) required to obtain a pregnancy was predicted according to its baseline values (x): Y = 9.8433x2 - 203.67x + 958.29. A higher number of pregnancies was reached in men with baseline sperm concentration below 7.3 million/mL. Thus, the percentage of sperm concentration increasing after FSH administration could predict the treatment efficacy in terms of pregnancy. At the dosage used, the efficacy was significantly higher in patients with a starting sperm concentration < 7.3 mill/mL. Mathematical analyses identified a function able to predict the sperm concentration increase required to obtain a pregnancy in relation to the baseline sperm number.
Collapse
Affiliation(s)
- Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41122 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| | - Giorgia Spaggiari
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| | - Leonardo Dalla Valentina
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41122 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| | - Marilina Romeo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41122 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| | - Federico Nuzzo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41122 Modena, Italy
| | | | - Laura Roli
- Department of Laboratory Medicine and Pathology, Azienda USL of Modena, 41122 Modena, Italy
| | | | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Azienda USL of Modena, 41122 Modena, Italy
| | - Antonio R. M. Granata
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41122 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
- Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 41122 Modena, Italy
| |
Collapse
|
6
|
Cannarella R, Petralia CMB, Condorelli RA, Aversa A, Calogero AE, La Vignera S. Investigational follicle-stimulating hormone receptor agonists for male infertility therapy. Expert Opin Investig Drugs 2023; 32:813-824. [PMID: 37747064 DOI: 10.1080/13543784.2023.2263364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION According to estimates by the World Health Organization, about 17.5% of the adult population - roughly 1 in 6 globally - experience infertility. The causes of male infertility remain poorly understood and have yet to be fully evaluated. Follicle-stimulating hormone (FSH) represents an available and useful therapeutic strategy for the treatment of idiopathic infertility. AREAS COVERED We provide here an overview of the molecular mechanisms by which FSH stimulates Sertoli cells and the schemes, dosages, and formulations of FSH most prescribed so far and reported in the literature. We also evaluated the possible predictor factors of the response to FSH administration and the indications of the latest guidelines on the use of FSH for the treatment of male infertility. EXPERT OPINION FSH therapy should be considered for infertile male patients with oligoasthenoteratozoospermia and normal serum FSH levels to quantitatively and qualitatively improve sperm parameters and pregnancy and birth rates. The grade of evidence is very low to low, due to the limited number of randomized controlled studies and patients available, the heterogeneity of the studies, and the limited effect size. To overcome these limitations, preclinical and clinical research is needed to evaluate the most effective dose and duration of FSH administration.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Cristina M B Petralia
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
7
|
Male infertility and gonadotropin treatment: What can we learn from real-world data? Best Pract Res Clin Obstet Gynaecol 2023; 86:102310. [PMID: 36682942 DOI: 10.1016/j.bpobgyn.2022.102310] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022]
Abstract
Gonadotropin therapy to treat specific male infertility disorders associated with hypogonadotropic hypogonadism is evidence-based and effective in restoring spermatogenesis and fertility. In contrast, its use to improve fertility in men with idiopathic oligozoospermia or nonobstructive azoospermia remains controversial, despite being widely practiced. The existence of two major inter-related pathways for spermatogenesis, including FSH and intratesticular testosterone, provides a rationale for empiric hormone stimulation therapy in both eugonadal and hypogonadal males with idiopathic oligozoospermia or nonobstructive azoospermia. Real-world data (RWD) on gonadotropin stimulating for these patient subsets, mainly using human chorionic gonadotropin and follicle-stimulating hormone, accumulated gradually, showing a positive therapeutic effect in some patients, translated by increased sperm production, sperm quality, and sperm retrieval rates. Although more evidence is needed, current insights from RWD research indicate that selected male infertility patients might be managed more effectively using gonadotropin therapy, with potential gains for all parties involved.
Collapse
|
8
|
Santi D, Spaggiari G, Granata ARM, Simoni M. Real-world evidence analysis of the follicle-stimulating hormone use in male idiopathic infertility. Best Pract Res Clin Obstet Gynaecol 2022; 85:121-133. [PMID: 35618626 DOI: 10.1016/j.bpobgyn.2022.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/11/2022] [Accepted: 04/20/2022] [Indexed: 12/14/2022]
Abstract
Male idiopathic infertility remains a therapeutic challenge in the couple infertility management. In this setting, an empirical treatment with follicle-stimulating hormone (FSH) is allowed, although not recommended. Twenty-one clinical trials and four meta-analyses highlighted an overall increased pregnancy rate in case of FSH administration, but the indiscriminate FSH prescription is still unsupported by clinical evidence in idiopathic infertility. This context could represent an example in which real-world data (RWD) could add useful information. From a nationwide clinical practice survey performed in Italy, emerged the clinicians' attitude to prescribe FSH in the case of impaired semen with a significant improvement of semen parameters, identifying FSH treatment as a therapeutic card in the real-life management. Although more robust data are still needed to optimize FSH treatment in male idiopathic infertility, RWD should be included in the body of evidence considered in healthcare decision-making.
Collapse
Affiliation(s)
- Daniele Santi
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile of Baggiovara, Via Giardini 1355, 41126 Modena, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy.
| | - Giorgia Spaggiari
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile of Baggiovara, Via Giardini 1355, 41126 Modena, Italy
| | - Antonio R M Granata
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile of Baggiovara, Via Giardini 1355, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile of Baggiovara, Via Giardini 1355, 41126 Modena, Italy; Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| |
Collapse
|
9
|
Pallotti F, Barbonetti A, Rastrelli G, Santi D, Corona G, Lombardo F. The impact of male factors and their correct and early diagnosis in the infertile couple's pathway: 2021 perspectives. J Endocrinol Invest 2022; 45:1807-1822. [PMID: 35349114 PMCID: PMC8961097 DOI: 10.1007/s40618-022-01778-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/24/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE The current clinical practice in reproductive medicine should pose the couple at the centre of the diagnostic-therapeutic management of infertility and requires intense collaboration between the andrologist, the gynaecologist and the embryologist. The andrologist, in particular, to adequately support the infertile couple, must undertake important biological, psychological, economical and ethical task. Thus, this paper aims to provide a comprehensive overview of the multifaceted role of the andrologist in the study of male factor infertility. METHODS A comprehensive Medline, Embase and Cochrane search was performed including publications between 1969 and 2021. RESULTS Available evidence indicates that a careful medical history and physical examination, followed by semen analysis, always represent the basic starting points of the diagnostic work up in male partner of an infertile couple. Regarding treatment, gonadotropins are an effective treatment in case of hypogonadotropic hypogonadism and FSH may be used in men with idiopathic infertility, while evidence supporting other hormonal and nonhormonal treatments is either limited or conflicting. In the future, pharmacogenomics of FSHR and FSHB as well as innovative compounds may be considered to develop new therapeutic strategies in the management of infertility. CONCLUSION To provide a high-level of care, the andrologist must face several critical diagnostical and therapeutical steps. Even though ART may be the final and decisive stage of this decisional network, neglecting to treat the male partner may ultimately increase the risks of negative outcome, as well as costs and psychological burden for the couple itself.
Collapse
Affiliation(s)
- F Pallotti
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - A Barbonetti
- Andrology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - G Rastrelli
- Andrology, Women's Endocrinology and Gender Incongruence Unit, Careggi Hospital-Department of Experimental, Clinical and Biomedical Sciences, University of Florence, Florence, Italy
| | - D Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - G Corona
- Endocrinology Unit, Medical Department, Maggiore-Bellaria Hospital, Azienda-Usl Bologna, 40139, Bologna, Italy
| | - F Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, Viale del Policlinico 155, 00161, Rome, Italy.
| |
Collapse
|
10
|
Zhang X, Chen J, Cui Y, Jin Y, Wang X. FSH
can improve semen parameters in patients with idiopathic oligoasthenoteratozoospermia: A systematic review and meta‐analysis. Andrologia 2022; 54:e14596. [DOI: 10.1111/and.14596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Xuebao Zhang
- Reproductive Medicine Center The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai Shandong China
| | - Jie Chen
- Reproductive Medicine Center The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai Shandong China
| | - Yuanqing Cui
- Reproductive Medicine Center The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai Shandong China
| | - Yinshan Jin
- Reproductive Medicine Center The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai Shandong China
| | - Xiong Wang
- Reproductive Medicine Center The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai Shandong China
| |
Collapse
|
11
|
The Roles of Luteinizing Hormone, Follicle-Stimulating Hormone and Testosterone in Spermatogenesis and Folliculogenesis Revisited. Int J Mol Sci 2021; 22:ijms222312735. [PMID: 34884539 PMCID: PMC8658012 DOI: 10.3390/ijms222312735] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
Spermatogenesis and folliculogenesis involve cell–cell interactions and gene expression orchestrated by luteinizing hormone (LH) and follicle-stimulating hormone (FSH). FSH regulates the proliferation and maturation of germ cells independently and in combination with LH. In humans, the requirement for high intratesticular testosterone (T) concentration in spermatogenesis remains both a dogma and an enigma, as it greatly exceeds the requirement for androgen receptor (AR) activation. Several data have challenged this dogma. Here we report our findings on a man with mutant LH beta subunit (LHβ) that markedly reduced T production to 1–2% of normal., but despite this minimal LH stimulation, T production by scarce mature Leydig cells was sufficient to initiate and maintain complete spermatogenesis. Also, in the LH receptor (LHR) knockout (LuRKO) mice, low-dose T supplementation was able to maintain spermatogenesis. In addition, in antiandrogen-treated LuRKO mice, devoid of T action, the transgenic expression of a constitutively activating follicle stimulating hormone receptor (FSHR) mutant was able to rescue spermatogenesis and fertility. Based on rodent models, it is believed that gonadotropin-dependent follicular growth begins at the antral stage, but models of FSHR inactivation in women contradict this claim. The complete loss of FSHR function results in the complete early blockage of folliculogenesis at the primary stage, with a high density of follicles of the prepubertal type. These results should prompt the reassessment of the role of gonadotropins in spermatogenesis, folliculogenesis and therapeutic applications in human hypogonadism and infertility.
Collapse
|
12
|
Shahid MN, Khan TM, Neoh CF, Lean QY, Bukhsh A, Karuppannan M. Effectiveness of Pharmacological Intervention Among Men with Infertility: A Systematic Review and Network Meta-Analysis. Front Pharmacol 2021; 12:638628. [PMID: 34483894 PMCID: PMC8415454 DOI: 10.3389/fphar.2021.638628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 07/09/2021] [Indexed: 01/18/2023] Open
Abstract
Background. Infertility is an emerging health issue for men. Comparative efficacy of different pharmacological interventions on male infertility is not clear. The aim of this review is to investigate the efficacy of various pharmacological interventions among men with idiopathic male infertility. All randomized control trials evaluating the effectuality of interventions on male infertility were included for network meta-analysis (NMA) from inception to 31 April 2020, systematically performed using STATA through the random effect model. The protocol was registered at PROSPERO (CRD42020152891). Results. The outcomes of interest were semen and hormonal parameters. Treatment effects (p < 0.05) were estimated through WMD at the confidence interval of 95%. Upon applying exclusion criteria, n=28 RCTs were found eligible for NMA. Results from NMA indicated that consumption of supplements increases sperm concentration levels [6.26, 95% CI 3.32, 9.21] in comparison to SERMs [4.97, 95% CI 1.61, 8.32], hormones [4.14, 95% CI 1.83, 6.46], and vitamins [0.15, 95% CI -20.86, 21.15)] with placebo, whereas the use of SERMs increased percentage sperm motility [6.69, 95% CI 2.38, 10.99] in comparison to supplements [6.46, 95% CI 2.57, 10.06], hormones [3.47, 95% CI 0.40, 6.54], and vitamins [-1.24, 95% CI -11.84, 9.43] with placebo. Consumption of hormones increased the sperm morphology [3.71, 95% CI, 1.34, 6.07] in contrast to supplements [2.22, 95% CI 0.12, 4.55], SERMs [2.21, 95% CI -0.78, 5.20], and vitamins [0.51, 95% CI -3.60, 4.62] with placebo. Supplements boosted the total testosterone levels [2.70, 95% CI 1.34, 4.07] in comparison to SERMs [1.83, 95% CI 1.16, 2.50], hormones [0.40, 95% CI -0.49, 1.29], and vitamins [-0.70, 95% CI -6.71, 5.31] with placebo. SERMs increase the serum FSH levels [3.63, 95% CI 1.48, 5.79] better than hormones [1.29, 95% CI -0.79, 3.36], vitamins [0.03, 95% CI -2.69, 2.76], and supplements [-4.45, 95% CI -7.15, -1.76] in comparison with placebo. Conclusion. This review establishes that all interventions had a significantly positive effect on male infertility. Statistically significant increased sperm parameters were noted in combinations of zinc sulfate (220 mg BID), clomiphene citrate (50 mg BID), and testosterone undecanoate and CoQ10; tamoxifen citrate and FSH were shown to improve the hormonal profile in infertile males.
Collapse
Affiliation(s)
- Muhammad Nabeel Shahid
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Malaysia
- Department of Pharmacy Practice, Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Tahir Mehmood Khan
- Department of Pharmacy Practice, Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
- School of Pharmacy, Monash University, Subang Jaya, Malaysia
| | - Chin Fen Neoh
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Malaysia
| | - Qi Ying Lean
- Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Pulau Pinang, Malaysia
- Vector-Borne Diseases Research Group (VERDI), Pharmaceutical and Life Sciences CoRe, Universiti Teknologi MARA (UiTM), Shah Alam, Malaysia
| | - Allah Bukhsh
- Department of Pharmacy Practice, Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
- School of Pharmacy, Monash University, Subang Jaya, Malaysia
| | - Mahmathi Karuppannan
- Department of Pharmacy Practice, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Malaysia
| |
Collapse
|
13
|
Cannarella R, La Vignera S, Condorelli RA, Mongioì LM, Calogero AE. FSH dosage effect on conventional sperm parameters: a meta-analysis of randomized controlled studies. Asian J Androl 2021; 22:309-316. [PMID: 31274479 PMCID: PMC7275804 DOI: 10.4103/aja.aja_42_19] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Follicle-stimulating hormone (FSH) represents a therapeutic option in normogonadotropic patients with idiopathic oligozoospermia. The aim of this review was to evaluate the possible dose- and drug-dependent efficacy of FSH treatment on conventional sperm parameters. We performed a comprehensive systematic review via a meta-analysis of all available randomized controlled trials, in which FSH administration was compared with placebo or no treatment when administered to normogonadotropic patients with idiopathic oligozoospermia. Of the 971 articles that were retrieved, 5 were finally included, including a total of 372 patients and 294 controls. Overall, FSH treatment was effective in ameliorating the sperm concentration, total count, progressive motility, but not normal forms. On the basis of the weekly dosage, the studies were classified into those using low (175–262.5 IU per week), intermediate (350–525 IU per week), and high (700–1050 IU per week) doses. At low doses, FSH improved only sperm motility. At intermediate doses, FSH ameliorated sperm concentration and morphology. Total sperm count and progressive motility showed a trend toward the increase. At high doses, FSH increased sperm concentration, total sperm count, and progressive motility. Sperm morphology showed a trend toward the increase. Finally, both highly purified FSH (hpFSH) and recombinant human FSH (rhFSH) improved sperm concentration, total sperm count, progressive motility, but not morphology. No different efficacy was observed between these two preparations. This meta-analysis provides evidence in favor of high FSH doses. The FSH efficacy was not related to the preparation type (recombinant vs highly purified). Further studies are needed to evaluate the effectiveness of long-standing treatment regimes.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania 95123, Italy
| |
Collapse
|
14
|
Verdi A, Nasr-Esfahani MH, Forouzanfar M, Tavalaee M. The Effect of Recombinant Human Follicle-Stimulating Hormone on Sperm Quality, Chromatin Status and Clinical Outcomes of Infertile Oligozoospermic Men Candidate for Intracytoplasmic Sperm Injection: A Randomized Clinical Trial. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2021; 15:1-7. [PMID: 33497040 PMCID: PMC7838760 DOI: 10.22074/ijfs.2021.6210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/11/2020] [Indexed: 01/18/2023]
Abstract
Background Follicle-stimulating hormone (FSH) plays a crucial role in spermatogenesis; in this study, we assessed
the effect of recombinant human FSH (rhFSH) on sperm parameters, chromatin status and clinical outcomes of infer-
tile oligozoospermic men candidates for intracytoplasmic sperm injection (ICSI). Materials and Methods This interventional randomized clinical trials (IRCT) included 40 infertile oligozoospermic
men undergoing ICSI. These individuals were randomized into two groups: 20 men received rhFSH drug for three
months and the other 20 men who did not receive rhFSH drug were considered the control group. Before and 3 months
after treatment initiation, sperm parameters (using computer-assisted semen analysis) and chromatin status [using
chromomycin A3, aniline blue, and sperm chromatin dispersion (SCD) tests] were assessed in these individuals. Fur-
thermore, hormonal profile was assessed using enzyme-linked immunosorbent assay (ELISA). Clinical outcomes of
ICSI were also compared between the two groups.
Results The rhFSH treated group showed a significant increase in the level of FSH, luteinizing hormone (LH), tes-
tosterone (T) and prolactin (PRL), as well as significant improvements in sperm parameters compared to the control
group. Also, after administration of rhFSH, there was asignificant reduction in the percentage of sperm DNA damage,
protamine deficiency and chromatin immaturity, while such a reduction in these parameters was not observed in the
control group. Moreover, the percentage of embryos with grade Aquality, was significantly higher in the rhFSH group
compared to the control group. The pregnancy rate in the rhFSH group was higher than the control group but the dif-
ference was insignificant. Conclusion Administration of rhFSH improves sperm quality in infertile oligozoospermic men and results in higher
rates of good quality embryos post-ICSI (Registration number: IRCT20170923036334N2).
Collapse
Affiliation(s)
- Atefeh Verdi
- Department of Biology, Fars Science and Research Branch, Islamic Azad University, Fars, Iran.,Department of Biology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.,Isfahan Fertility and Infertility Center, Isfahan, Iran
| | - Mohsen Forouzanfar
- Department of Biology, Marvdasht Branch, Islamic Azad University, Marvdasht, Iran. Electronic Address:
| | - Marziyeh Tavalaee
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
15
|
Liu YP, Qi L, Zhang NN, Shi H, Su YC. Follicle-stimulating hormone may predict sperm retrieval rate and guide surgical approach in patients with non-obstructive azoospermia. Reprod Biol 2020; 20:573-579. [PMID: 33203587 DOI: 10.1016/j.repbio.2020.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/14/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023]
Abstract
Testicular sperm aspiration- (TESA) or micro-dissection testicular sperm extraction- (MD-TESE) combined intracytoplasmic sperm injection (ICSI) was the only option for non-obstructive azoospermia (NOA) patients to have a biological offspring and they had different success rates in sperm retrieval. Our study aimed to find predictor(s) for predicting the sperm retrieval rate (SRR) in NOAs and guide clinicians in choosing different surgical approaches, TESA or MD-TESE for NOAs. 294 NOAs who had undergone TESA or MD-TESE were divided into TESA group and MD-TESE group. Depending on sperm retrieval, each group was divided into two subgroups: successful subgroups and failure subgroups. They respectively were 24 cases and 131 cases, 53 cases and 86 cases. Clinical data, including body mass index (BMI), testicular volume, and serum hormone levels, were analyzed in a retrospective manner. The results showed that follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels and SRR were lower in TESA group as compared to these in MD-TESE group, while testicular volume was higher (P < 0.05). The surgical approach of sperm retrieval significantly affected the SRR (P < 0.05). In TESA subgroups, testicular volume, FSH and LH differed significantly (P < 0.05). In MD-TESE subgroups, the level of FSH and LH differed significantly between both groups (P < 0.05). Using logistics regression, we found a negative correlation (β=-0.083) between FSH and the SRR in TESA group but a positive correlation (β = 0.064) in MD-TESE group (P < 0.05). In conclusion, serum FSH level can predict the SRR of NOAs and guide the clinicians while selecting the suitable surgery approach for NOAs.
Collapse
Affiliation(s)
- Ya-Ping Liu
- Department of Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China; Department of Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Lin Qi
- Department of Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China; Department of Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Nan-Nan Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hao Shi
- Department of Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China; Department of Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Ying-Chun Su
- Department of Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China; Department of Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
16
|
Simoni M, Brigante G, Rochira V, Santi D, Casarini L. Prospects for FSH Treatment of Male Infertility. J Clin Endocrinol Metab 2020; 105:5831300. [PMID: 32374828 DOI: 10.1210/clinem/dgaa243] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Despite the new opportunities provided by assisted reproductive technology (ART), male infertility treatment is far from being optimized. One possibility, based on pathophysiological evidence, is to stimulate spermatogenesis with gonadotropins. EVIDENCE ACQUISITION We conducted a comprehensive systematic PubMed literature review, up to January 2020, of studies evaluating the genetic basis of follicle-stimulating hormone (FSH) action, the role of FSH in spermatogenesis, and the effects of its administration in male infertility. Manuscripts evaluating the role of genetic polymorphisms and FSH administration in women undergoing ART were considered whenever relevant. EVIDENCE SYNTHESIS FSH treatment has been successfully used in hypogonadotropic hypogonadism, but with questionable results in idiopathic male infertility. A limitation of this approach is that treatment plans for male infertility have been borrowed from hypogonadism, without daring to overstimulate, as is done in women undergoing ART. FSH effectiveness depends not only on its serum levels, but also on individual genetic variants able to determine hormonal levels, activity, and receptor response. Single-nucleotide polymorphisms in the follicle-stimulating hormone subunit beta (FSHB) and follicle-stimulating hormone receptor (FSHR) genes have been described, with some of them affecting testicular volume and sperm output. The FSHR p.N680S and the FSHB -211G>T variants could be genetic markers to predict FSH response. CONCLUSIONS FSH may be helpful to increase sperm production in infertile men, even if the evidence to recommend the use of FSH in this setting is weak. Placebo-controlled clinical trials, considering the FSHB-FSHR haplotype, are needed to define the most effective dosage, the best treatment length, and the criteria to select candidate responder patients.
Collapse
Affiliation(s)
- Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l'Equitation (IFCE), Université de Tours, Nouzilly, France
| | - Giulia Brigante
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Vincenzo Rochira
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Livio Casarini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| |
Collapse
|
17
|
Follicle-stimulating Hormone (FSH) Action on Spermatogenesis: A Focus on Physiological and Therapeutic Roles. J Clin Med 2020; 9:jcm9041014. [PMID: 32260182 PMCID: PMC7230878 DOI: 10.3390/jcm9041014] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human reproduction is regulated by the combined action of the follicle-stimulating hormone (FSH) and the luteinizing hormone (LH) on the gonads. Although FSH is largely used in female reproduction, in particular in women attending assisted reproductive techniques to stimulate multi-follicular growth, its efficacy in men with idiopathic infertility is not clearly demonstrated. Indeed, whether FSH administration improves fertility in patients with hypogonadotropic hypogonadism, the therapeutic benefit in men presenting alterations in sperm production despite normal FSH serum levels is still unclear. In the present review, we evaluate the potential pharmacological benefits of FSH administration in clinical practice. METHODS This is a narrative review, describing the FSH physiological role in spermatogenesis and its potential therapeutic action in men. RESULTS The FSH role on male fertility is reviewed starting from the physiological control of spermatogenesis, throughout its mechanism of action in Sertoli cells, the genetic regulation of its action on spermatogenesis, until the therapeutic options available to improve sperm production. CONCLUSION FSH administration in infertile men has potential benefits, although its action should be considered by evaluating its synergic action with testosterone, and well-controlled, powerful trials are required. Prospective studies and new compounds could be developed in the near future.
Collapse
|
18
|
Simoni M, Santi D. FSH treatment of male idiopathic infertility: Time for a paradigm change. Andrology 2020; 8:535-544. [DOI: 10.1111/andr.12746] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/21/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Manuela Simoni
- Unit of Endocrinology Department of Biomedical, Metabolic and Neural Sciences University of Modena and Reggio Emilia Modena Italy
- Unit of Endocrinology Department of Medical Specialties Azienda Ospedaliero‐Universitaria of Modena Modena Italy
- BIOS INRA CNRS IFCE Université de Tours Nouzilly France
| | - Daniele Santi
- Unit of Endocrinology Department of Biomedical, Metabolic and Neural Sciences University of Modena and Reggio Emilia Modena Italy
- Unit of Endocrinology Department of Medical Specialties Azienda Ospedaliero‐Universitaria of Modena Modena Italy
| |
Collapse
|
19
|
Omar MI, Pal RP, Kelly BD, Bruins HM, Yuan Y, Diemer T, Krausz C, Tournaye H, Kopa Z, Jungwirth A, Minhas S. Benefits of Empiric Nutritional and Medical Therapy for Semen Parameters and Pregnancy and Live Birth Rates in Couples with Idiopathic Infertility: A Systematic Review and Meta-analysis. Eur Urol 2019; 75:615-625. [DOI: 10.1016/j.eururo.2018.12.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
|
20
|
Huhtaniemi I. MECHANISMS IN ENDOCRINOLOGY: Hormonal regulation of spermatogenesis: mutant mice challenging old paradigms. Eur J Endocrinol 2018; 179:R143-R150. [PMID: 29959220 DOI: 10.1530/eje-18-0396] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/28/2018] [Indexed: 11/08/2022]
Abstract
The two pituitary gonadotrophins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and in particular LH-stimulated high intratesticular testosterone (ITT) concentration, are considered crucial for spermatogenesis. We have revisited these concepts in genetically modified mice, one being the LH receptor (R)-knockout mouse (LuRKO), the other a transgenic mouse expressing in Sertoli cells a highly constitutively active mutated Fshr (Fshr-CAM). It was found that full spermatogenesis was induced by exogenous testosterone treatment in LuRKO mice at doses that restored ITT concentration to a level corresponding to the normal circulating testosterone level in WT mice, ≈5 nmol/L, which is 1.4% of the normal high ITT concentration. When hypogonadal LuRKO and Fshr-CAM mice were crossed, the double-mutant mice with strong FSH signaling, but minimal testosterone production, showed near-normal spermatogenesis, even when their residual androgen action was blocked with the strong antiandrogen flutamide. In conclusion, our findings challenge two dogmas of the hormonal regulation of male fertility: (1) high ITT concentration is not necessary for spermatogenesis and (2) strong FSH stimulation can maintain spermatogenesis without testosterone. These findings have clinical relevance for the development of hormonal male contraception and for the treatment of idiopathic oligozoospermia.
Collapse
Affiliation(s)
- Ilpo Huhtaniemi
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Campus, London, UK
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
21
|
Barbonetti A, Calogero AE, Balercia G, Garolla A, Krausz C, La Vignera S, Lombardo F, Jannini EA, Maggi M, Lenzi A, Foresta C, Ferlin A. The use of follicle stimulating hormone (FSH) for the treatment of the infertile man: position statement from the Italian Society of Andrology and Sexual Medicine (SIAMS). J Endocrinol Invest 2018; 41:1107-1122. [PMID: 29392544 DOI: 10.1007/s40618-018-0843-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/27/2018] [Indexed: 12/18/2022]
Affiliation(s)
- A Barbonetti
- Casa di Cura San Raffaele Sulmona, Sulmona, AQ, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - G Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Umberto I Hospital, Ancona, Polytechnic University of Marche, Ancona, Italy
| | - A Garolla
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - C Krausz
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - F Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - E A Jannini
- Department of Systems Medicine, Chair of Endocrinology and Medical Sexology (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| | - M Maggi
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - A Lenzi
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "La Sapienza", Rome, Italy
| | - C Foresta
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy
| | - A Ferlin
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padua, Italy.
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| |
Collapse
|
22
|
Sperm DNA fragmentation index as a promising predictive tool for male infertility diagnosis and treatment management – meta-analyses. Reprod Biomed Online 2018; 37:315-326. [DOI: 10.1016/j.rbmo.2018.06.023] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 11/17/2022]
|
23
|
Shiraishi K, Matsuyama H. Gonadotoropin actions on spermatogenesis and hormonal therapies for spermatogenic disorders [Review]. Endocr J 2017; 64:123-131. [PMID: 28100869 DOI: 10.1507/endocrj.ej17-0001] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Microdissection testicular sperm extraction and intracytoplasmic sperm injection have made it possible for men with non-obstructive azoospermia (NOA) to conceive a child. A majority of men cannot produce sperm because spermatogenesis per se is believed to be "irreversibly" disturbed. For these men, it has been thought that any hormonal therapy will be ineffective. Further understandings of endocrinological regulation of spermatogenesis are needed and LH or FSH receptor knock out (KO) mice have revealed the roles of gonadotropin separately. Spermatogenesis has been shown to shift during evolution from FSH to LH dominance because LH receptor KO causes infertility while FSH receptor KO does not. High concentrations of intratesticular testosterone secreted from Leydig cells, ranging from 100- to 1,000-fold higher than in the systemic circulation, has pivotal roles during spermatogenesis. This is especially important during spermiogenesis, a post-meiotic step for progression from round to elongating spermatids. Sertoli cells are the target of FSH and have numerous androgen receptors, indicating that Sertoli cells are regulated by FSH and the paracrine functions of testosterone. In combination with Leydig cell-derived growth factors, particularly epidermal growth factor-like growth factors, Sertoli cells support spermatogenesis, especially at proximal levels of spermatogenesis (e.g., spermatogonial proliferation). Taken together, the current knowledge from human studies indicating that testosterone optimization by clomiphene, hCG and/or aromatase inhibitors and high dose hCG/FSH treatment can, at least in part, improve spermatogenesis in NOA. Accordingly hormonal therapy may open a therapeutic window for sperm production in selected patients.
Collapse
Affiliation(s)
- Koji Shiraishi
- Department of Urology, Yamaguchi University School of Medicine, Ube 755-8505, Japan
| | | |
Collapse
|
24
|
Tadros NN, Sabanegh ES. Empiric medical therapy with hormonal agents for idiopathic male infertility. Indian J Urol 2017; 33:194-198. [PMID: 28717268 PMCID: PMC5508429 DOI: 10.4103/iju.iju_368_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Introduction: Infertility affects approximately 15% of all couples, and male factor contribute to up to 50% of cases. Unfortunately, the cause of male infertility is unknown in about 30% of these cases. Infertility of unknown origin is classified as idiopathic male infertility when abnormal semen parameters are present. Despite not having a definable cause, these men may respond to treatment. This review focuses on the use of empiric hormonal therapies for idiopathic male infertility. Methods: A detailed PubMed/MEDLINE search was conducted to identify all publications pertaining to empiric use of hormonal therapies in the treatment of idiopathic male infertility using the keywords “idiopathic,” “male infertility,” “empiric treatment,” “clomiphene,” “SERM,” “gonadotropin,” “aromatase inhibitor,” and “androgen.” These manuscripts were reviewed to identify treatment modalities and results. Results: Gonadotropins, androgens, aromatase inhibitors, and selective estrogen receptor modulators (SERMs) have all been used with varying results. The studies on these treatments are of variable quality. The most well-studied agents are the SERMs which show a modest increase in semen parameters and pregnancy rates. Aromatase inhibitors are most effective in non-idiopathic patients. Gonadotropin treatment is limited by their inconvenience and relative ineffectiveness in this population. Testosterone suppresses spermatogenesis and should not be used to treat infertility. Conclusion: Gonadotropins, SERMs, and aromatase inhibitors may improve semen parameters and hormone levels in men with idiopathic infertility with the best results from SERMs. Testosterone should never be used to treat infertility. Large multicenter randomized controlled studies are needed to better determine the success of empiric use of hormonal therapy on pregnancy rates.
Collapse
Affiliation(s)
- Nicholas N Tadros
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Edmund S Sabanegh
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
25
|
Ulloa-Aguirre A, Lira-Albarrán S. Clinical Applications of Gonadotropins in the Male. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:121-174. [PMID: 27697201 DOI: 10.1016/bs.pmbts.2016.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotropins, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) play a pivotal role in reproduction. The synthesis and secretion of gonadotropins are regulated by complex interactions among several endocrine, paracrine, and autocrine factors of diverse chemical structure. In men, LH regulates the synthesis of androgens by the Leydig cells, whereas FSH promotes Sertoli cell function and thereby influences spermatogenesis. Gonadotropins are complex molecules composed of two subunits, the α- and β-subunit, that are noncovalently associated. Gonadotropins are decorated with glycans that regulate several functions of the protein including folding, heterodimerization, stability, transport, conformational maturation, efficiency of heterodimer secretion, metabolic fate, interaction with their cognate receptor, and selective activation of signaling pathways. A number of congenital and acquired abnormalities lead to gonadotropin deficiency and hypogonadotropic hypogonadism, a condition amenable to treatment with exogenous gonadotropins. Several natural and recombinant preparations of gonadotropins are currently available for therapeutic purposes. The difference between natural and the currently available recombinant preparations (which are massively produced in Chinese hamster ovary cells for commercial purposes) mainly lies in the abundance of some of the carbohydrates that conform the complex glycans attached to the protein core. Whereas administration of exogenous gonadotropins in patients with isolated congenital hypogonadotropic hypogonadism is a well recognized therapeutic approach, their role in treating men with normogonadotropic idiopathic infertility is still controversial. This chapter concentrates on the main structural and functional features of the gonadotropin hormones and how basic concepts have been translated into the clinical arena to guide therapy for gonadotropin deficit in males.
Collapse
Affiliation(s)
- A Ulloa-Aguirre
- Research Support Network, Universidad Nacional Autónoma de México (UNAM)-National Institutes of Health, Mexico City, Mexico.
| | - S Lira-Albarrán
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
26
|
CURRENT STATE OF THE MALE INFERTILITY PROBLEM:THE REVIEW OF EUROPEAN ASSOCIATION OF UROLOGY CLINICAL GUIDELINES. КЛИНИЧЕСКАЯ ПРАКТИКА 2016. [DOI: 10.17816/clinpract7139-50] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The article highlights the problem of male infertility on the basis of review of the latest guidelines of European Association of urology
Collapse
|
27
|
Shiraishi K, Ishikawa T, Watanabe N, Iwamoto T, Matsuyama H. Salvage hormonal therapy after failed microdissection testicular sperm extraction: A multi-institutional prospective study. Int J Urol 2016; 23:496-500. [DOI: 10.1111/iju.13076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/20/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Koji Shiraishi
- Department of Urology; Yamaguchi University School of Medicine; Ube Yamaguchi Japan
| | | | | | - Teruaki Iwamoto
- Center for Infertility and IVF; International University of Health and Welfare Hospital; Nasushiobara Japan
| | - Hideyasu Matsuyama
- Department of Urology; Yamaguchi University School of Medicine; Ube Yamaguchi Japan
| |
Collapse
|
28
|
Ding YM, Zhang XJ, Li JP, Chen SS, Zhang RT, Tan WL, Shi XJ. Treatment of idiopathic oligozoospermia with recombinant human follicle-stimulating hormone: a prospective, randomized, double-blind, placebo-controlled clinical study in Chinese population. Clin Endocrinol (Oxf) 2015; 83:866-71. [PMID: 25761129 DOI: 10.1111/cen.12770] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 02/04/2015] [Accepted: 03/05/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Follicle-stimulating hormone plays a crucial role in spermatogenesis. The aim of this study was to evaluate the efficacy of treatment with FSH in Chinese infertility population. DESIGN Prospective, randomized, double-blind, placebo-controlled clinical study. PATIENTS A total of 354 men affected by idiopathic oligozoospermia from three medical centres. MEASUREMENTS This study contained three parts: (i) treatment with different doses of rhFSH (50 IU, 100 IU, 200 IU and 300 IU); (ii) the efficacy of rhFSH at different periods (2, 3, 4, 5 months); (iii) FSH treatment in patients with different levels of inhibin B (normal-level group, low-level group and high-level group). Semen parameters were evaluated in all subjects. The patients who had not reached spontaneous pregnancy underwent assisted reproductive techniques. RESULTS Sperm number was significantly increased after treatment with FSH at doses of at least 200 IU, and the improvement was observed beginning at the third month. The significant improvement in both morphology and forward motility was observed beginning at the fifth month. Moreover, 300 IU rhFSH administration for 5 months could significantly improve the spontaneous pregnancy rate (12/40) and ART pregnancy rate (14/28), while the rates for placebo group were two of twenty-nine and five of twenty-seven, respectively. The seminal parameters (total sperm count, sperm concentration, forward motility and morphology) were significantly improved in the normal- and low-level inhibin B groups, but no significant variation was observed in the high-level group at the end of the study. CONCLUSIONS The efficacy of FSH treatment was associated with the dose of FSH and duration of treatment, and FSH therapy was more effective in patients with normal level and low level of inhibin B.
Collapse
Affiliation(s)
- Yin-man Ding
- Department of Urology, People's Hospital of Xuancheng City, Xuancheng, An hui, China
| | - Xin-ji Zhang
- Department of Urology, Shunde First People's Hospital Affiliated to Southern Medical University, Guangdong, China
| | - Jing-Ping Li
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - San-san Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ren-tao Zhang
- Department of Urology, People's Hospital of Xuancheng City, Xuancheng, An hui, China
| | - Wan-long Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Jun Shi
- Department of Urology, People's Hospital of Xuancheng City, Xuancheng, An hui, China
| |
Collapse
|
29
|
Santi D, Granata ARM, Simoni M. FSH treatment of male idiopathic infertility improves pregnancy rate: a meta-analysis. Endocr Connect 2015; 4:R46-58. [PMID: 26113521 PMCID: PMC4512277 DOI: 10.1530/ec-15-0050] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/25/2015] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The aim of this study is to comprehensively evaluate whether FSH administration to the male partner of infertile couples improves pregnancy rate, spontaneously and/or after assisted reproductive techniques (ART). METHODS Meta-analysis of controlled clinical trials in which FSH was administered for male idiopathic infertility, compared with placebo or no treatment. Randomization was not considered as an inclusion criterion. RESULTS We found 15 controlled clinical studies (614 men treated with FSH and 661 treated with placebo or untreated). Concerning the type of FSH, eight studies used recombinant FSH, whereas seven studies used purified FSH. Nine studies evaluated spontaneous pregnancy rate, resulting in an overall odds ratio (OR) of about 4.5 (CI: 2.17-9.33). Eight studies evaluated pregnancy rate after ART, showing a significant OR of 1.60 (CI: 1.08-2.37). Sub-dividing studies according to the FSH preparations (purified/recombinant), pregnancy rate improvement remained significant for each preparation. Eleven studies considered sperm quality after FSH treatment, finding a significant improvement of sperm concentration (2.66×10(6)/ml, CI: 0.47-4.84), but not of concentration of sperm with progressive motility (1.22×10(6)/ml, CI: -0.07 to 2.52). Three trials evaluated testicular volume, showing a non-significant increase in men treated (1.35 ml, CI: -0.44 to 3.14). CONCLUSION The results of controlled clinical trials available in the literature indicate an improvement of pregnancy rate after FSH administration to the male partner of infertile couples, both spontaneously and after ART. However, the heterogeneity of studies, the high risk of bias and the lack of precise criteria to guide FSH administration limit the strength of these results. Future studies should be designed to identify the markers of FSH response which are helpful in the decision-making process. Meanwhile, the use of FSH in the treatment of male infertility should be cautious.
Collapse
Affiliation(s)
- D Santi
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, ItalyUnit of EndocrinologyAzienda USL of Modena, NOCSAE, Via P. Giardini 1355, 41126 Modena, Italy Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, ItalyUnit of EndocrinologyAzienda USL of Modena, NOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| | - A R M Granata
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, ItalyUnit of EndocrinologyAzienda USL of Modena, NOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| | - M Simoni
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, ItalyUnit of EndocrinologyAzienda USL of Modena, NOCSAE, Via P. Giardini 1355, 41126 Modena, Italy Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, ItalyUnit of EndocrinologyAzienda USL of Modena, NOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
30
|
Garg H, Kumar R. Empirical Drug Therapy for Idiopathic Male Infertility: What is the New Evidence? Urology 2015; 86:1065-75. [PMID: 26255035 DOI: 10.1016/j.urology.2015.07.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/25/2015] [Accepted: 07/28/2015] [Indexed: 01/08/2023]
Abstract
Idiopathic male infertility is empirically managed using a number of drugs. We reviewed 64 articles published in the last 10 years on such drug therapy. There was severe heterogeneity in data along with poor definition of outcome parameters. Pregnancy or live birth rate was not reported in many studies. Antiestrogens appear to improve pregnancy rates while there is some data supporting the use of aromatase inhibitors. Antioxidants significantly increase the rate of both live birth and pregnancy but the data are limited. However, valid end-points based on data are limited for the empirical use of drugs in idiopathic male infertility.
Collapse
Affiliation(s)
- Harshit Garg
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajeev Kumar
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
31
|
Kobori Y, Suzuki K, Iwahata T, Shin T, Sato R, Nishio K, Yagi H, Arai G, Soh S, Okada H. Induction of spermatogenesis by rhFSH for azoospermia due to spermatogenic dysfunction with maturation arrest: five case series. Syst Biol Reprod Med 2015; 61:168-70. [DOI: 10.3109/19396368.2015.1024897] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Jung JH, Seo JT. Empirical medical therapy in idiopathic male infertility: Promise or panacea? Clin Exp Reprod Med 2014; 41:108-14. [PMID: 25309854 PMCID: PMC4192450 DOI: 10.5653/cerm.2014.41.3.108] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/01/2014] [Accepted: 09/05/2014] [Indexed: 12/05/2022] Open
Abstract
Male factors account for 20%-50% of cases of infertility and in 25% of cases, the etiology of male infertility is unknown. Effective treatments are well-established for hypogonadotropic hypogonadism, male accessory gland infection, retrograde ejaculation, and positive antisperm antibody. However, the appropriate treatment for idiopathic male infertility is unclear. Empirical medical treatment (EMT) has been used in men with idiopathic infertility and can be divided into two categories based on the mode of action: hormonal treatment and antioxidant supplementation. Hormonal medications consist of gonadotropins, androgens, estrogen receptor blockers, and aromatase inhibitors. Antioxidants such as vitamins, zinc, and carnitines have also been widely used to reduce oxidative stress-induced spermatozoa damage. Although scientifically acceptable evidence of EMT is limited because of the lack of large, randomized, controlled studies, recent systematic reviews with meta-analyses have shown that the administration of gonadotropins, anti-estrogens, and oral antioxidants results in a significant increase in the live birth rate compared with control treatments. Therefore, all physicians who treat infertility should bear in mind that EMT can improve semen parameters and subsequent fertility potential through natural intercourse.
Collapse
Affiliation(s)
- Jae Hung Jung
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ju Tae Seo
- Department of Urology, Cheil General Hospital, Kwandong University College of Medicine, Seoul, Korea
| |
Collapse
|
33
|
Condorelli RA, Calogero AE, Vicari E, Mongioi' L, Burgio G, Cannarella R, Giacone F, Iacoviello L, Morgia G, Favilla V, Cimino S, La Vignera S. Reduced Seminal Concentration of CD45pos Cells after Follicle-Stimulating Hormone Treatment in Selected Patients with Idiopathic Oligoasthenoteratozoospermia. Int J Endocrinol 2014; 2014:372060. [PMID: 24550984 PMCID: PMC3914479 DOI: 10.1155/2014/372060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 11/20/2013] [Accepted: 11/20/2013] [Indexed: 11/17/2022] Open
Abstract
The present study evaluated the conventional sperm parameters and the seminal concentration of CD45pos cells (pan-leukocyte marker) of infertile patients with idiopathic oligoasthenoteratozoospermia (OAT). The patients were arbitrarily divided into three groups treated with recombinant follicle-stimulating hormone FSH: α (Group A = 20 patients), recombinant FSH- β (Group B = 20 patients), and highly purified human FSH (Group C = 14 patients). All treated groups achieved a similar improvement of the main sperm parameters (density, progressive motility, and morphology), but only the increase in the percentage of spermatozoa with normal morphology was significant compared to the baseline in all three examined groups. Moreover, all groups had a significant reduction of the seminal concentration of CD45pos cells and of the percentage of immature germ cells. Before and after the treatment, the concentration of CD45pos cells showed a positive linear correlation with the percentage of immature germ cells and a negative correlation with the percentage of spermatozoa with regular morphology. These results demonstrate that treatment with FSH is effective in patients with idiopathic OAT and that there are no significant differences between the different preparations. The novelty of this study is in the significant reduction of the concentration of CD45pos cells observed after the treatment.
Collapse
Affiliation(s)
- Rosita A. Condorelli
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Aldo E. Calogero
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Enzo Vicari
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Laura Mongioi'
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Giovanni Burgio
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Rossella Cannarella
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Filippo Giacone
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Linda Iacoviello
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| | - Giuseppe Morgia
- Department of Urology, University of Catania, Catania, Italy
| | | | | | - Sandro La Vignera
- Department of Medical and Pediatric Sciences, Section of Endocrinology, Andrology and Internal Medicine, University of Catania, Policlinico “G. Rodolico”, Building 4, Rm 2C18, Via S. Sofia 78, 95123 Catania, Italy
| |
Collapse
|
34
|
Paradisi R, Natali F, Fabbri R, Battaglia C, Seracchioli R, Venturoli S. Evidence for a stimulatory role of high doses of recombinant human follicle-stimulating hormone in the treatment of male-factor infertility. Andrologia 2013; 46:1067-72. [DOI: 10.1111/and.12194] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- R. Paradisi
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| | - F. Natali
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| | - R. Fabbri
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| | - C. Battaglia
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| | - R. Seracchioli
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| | - S. Venturoli
- Department of Obstetrics and Gynecology and Reproductive Biology; University Alma Mater Studiorum of Bologna; Bologna Italy
| |
Collapse
|
35
|
Abstract
BACKGROUND Male factors leading to subfertility account for at least half of all cases of subfertility worldwide. Although some causes of male subfertility are treatable, treatment of idiopathic male factor subfertility remains empirical. Researchers have used gonadotrophins to improve sperm parameters in idiopathic male factor subfertility with the ultimate goal of increasing birth and pregnancy rates, but results have been conflicting. OBJECTIVES To determine the effect of systemic follicle-stimulating hormone (FSH) on live birth and pregnancy rates when administered to men with idiopathic male factor subfertility . SEARCH METHODS We searched the Cochrane Menstrual Disorders and Subfertility Group Specialised Register (14 January 2013), the Cochrane Central Register of Controlled Trials (CENTRAL; The Cochrane Library, Issue 12 of 12, 2012), Ovid MEDLINE In-Process & Other Non-Indexed Citations, Ovid MEDLINE Daily and Ovid MEDLINE (1946 to 14 January 2013), Ovid EMBASE (1980 to week 2 of 2013), Ovid PsycINFO (1806 to week 2 of 2013), trial registers for ongoing and registered trials at ClinicalTrials.gov (19 January 2013), the World Health Organisation International Trials Registry Platform (19 January 2013), The Cochrane Library Database of Abstracts of Reviews of Effects (19 January 2013) and OpenGrey for grey literature from Europe (19 January 2013). Searches were not limited by language. Bibliographies of included and excluded trials and abstracts of major meetings were searched for additional trials. SELECTION CRITERIA Randomised controlled trials (RCTs) in which gonadotrophins were compared with placebo or no treatment for participants with idiopathic male factor subfertility. DATA COLLECTION AND ANALYSIS Two review authors independently selected the trials, assessed risk of bias and extracted data on live birth, pregnancy and adverse effects. We included data on pregnancies that occurred during or after gonadotrophin therapy. Study authors and pharmaceutical companies were asked to provide missing and unpublished data and/or additional information. MAIN RESULTS Six RCTs with 456 participants and variable treatment and follow-up periods were included. From the limited data, the live birth rate per couple randomly assigned (27% vs 0%; Peto odds ratio (OR) 9.31, 95% confidence interval (CI) 1.17 to 73.75, one study, 30 participants, very low-quality evidence) and the spontaneous pregnancy rate per couple randomly assigned (16% vs 7%; Peto OR 4.94, 95% CI 2.13 to 11.44, five studies, 412 participants, I(2) = 0%, moderate-quality evidence) were significantly higher in men receiving gonadotrophin treatment than in men receiving placebo or no treatment. No significant difference between groups was noted when intracytoplasmic sperm injection (ICSI) or intrauterine insemination (IUI) was performed. None of the included studies reported miscarriage rates, and adverse events data were sparse. AUTHORS' CONCLUSIONS Encouraging preliminary data suggest a beneficial effect on live birth and pregnancy of gonadotrophin treatment for men with idiopathic male factor subfertility, but because the numbers of trials and participants are small, evidence is insufficient to allow final conclusions. Large multi-centre trials with adequate numbers of participants are needed.
Collapse
Affiliation(s)
- Abdelhamid M Attia
- Faculty of Medicine, Cairo UniversityObstetrics & Gynaecology18 El‐Ghaith St.CairoEgypt12311
| | - Ahmed M Abou‐Setta
- University of ManitobaCentre for Healthcare InnovationGH‐714 820 Sherbrook StreetWinnipegManitobaCanadaR3A 1R9
| | - Hesham G Al‐Inany
- Faculty of Medicine, Cairo UniversityObstetrics & Gynaecology18 El‐Ghaith St.CairoEgypt12311
| | | |
Collapse
|
36
|
Abstract
Several empirical treatments have been proposed to treat idiopathic infertility in men, including follicle-stimulating hormone (FSH). FSH administration is effective in patients with hypogonadotropic hypogonadism, which suggests it might be useful in patients with oligozoospermia who have normal FSH levels. Indeed, many studies have evaluated the efficacy of FSH administration in these patients, several of which have shown improvements in sperm parameters. By contrast, other studies have not reported any significant effect of FSH administration on conventional sperm parameters, although some of have reported the normalization of spermatozoon ultrastructural morphology, as well as reductions in DNA fragmentation, production of reactive oxygen species and aneuploidy. Contemporary studies suggest that the response to FSH treatment in oligozoospermic patients might, at least partially, reflect polymorphisms of the FSH receptor gene. Thus, FSH administration in oligozoospermic men with normal serum FSH levels might be efficacious only in selected patients. For this reason, additional studies are needed to determine the predictive factors and clinical conditions that can be used to identify patients who could benefit from FSH treatment.
Collapse
|
37
|
Foresta C, Selice R, Ferlin A, Garolla A. Recombinant FSH in the treatment of oligozoospermia. Expert Opin Biol Ther 2010; 9:659-66. [PMID: 19379121 DOI: 10.1517/14712590902932673] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND FSH plays a crucial role in human reproduction. Treatment with gonadotrophins has been shown to be effective in males affected by hypogonadotrophic hypogonadism. The success of this treatment has brought about the utilization of the same therapy in infertile oligozoospermic subjects, aimed at obtaining increased sperm count. This physiological role in spermatogenesis has induced various attempts to treat idiopathic oligozoospermic men with FSH, often inducing the restoration of normal spermatogenesis and spontaneous pregnancy. OBJECTIVE To evaluate clinical efficacy of recombinant FSH in male infertility. METHODS Evaluation of pharmacokinetic, pharmacodynamic properties, efficacy and safety of this hormone preparation, on the basis of the data published in literature. CONCLUSIONS Recombinant FSH is effective, safe and well tolerated. Treatment with this hormone may represent a valid tool for infertile men. However it should be performed on selected patients utilizing some predictive parameters able to identify a priori responder subjects with high probability.
Collapse
Affiliation(s)
- Carlo Foresta
- Department of Histology, Section of Clinical Pathology & Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy.
| | | | | | | |
Collapse
|
38
|
Nieschlag E, Kamischke A. Empirical Therapies for Idiopathic Male Infertility. Andrology 2010. [DOI: 10.1007/978-3-540-78355-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
Warne DW, Decosterd G, Okada H, Yano Y, Koide N, Howles CM. A combined analysis of data to identify predictive factors for spermatogenesis in men with hypogonadotropic hypogonadism treated with recombinant human follicle-stimulating hormone and human chorionic gonadotropin. Fertil Steril 2009; 92:594-604. [DOI: 10.1016/j.fertnstert.2008.07.1720] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 05/27/2008] [Accepted: 07/09/2008] [Indexed: 10/21/2022]
|
40
|
Foresta C, Selice R, Garolla A, Ferlin A. Follicle-stimulating hormone treatment in oligozoospermic patients. Expert Rev Endocrinol Metab 2008; 3:761-770. [PMID: 30764065 DOI: 10.1586/17446651.3.6.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Follicle-stimulating hormone (FSH) plays a crucial role in human reproduction. Already, in the fetal and neonatal developmental stages, FSH activates the proliferation of the Sertoli cells and successively, in the pubertal phase, induces the mitotic activity of the spermatogonia and supports cellular differentiation to the round spermatid stage. This physiological role in spermatogenesis has induced various attempts to treat idiopathic oligozoospermic men with FSH. It is well known that treatment with gonadotrophins is very effective in subjects affected by hypogonadotropic hypogonadism, often leading to the restoration of normal spermatogenesis. The success of this treatment in these men has brought the utilization of the therapy with FSH in infertile oligozoospermic subjects, aimed at obtaining a quantitative increase in sperm count. However, the results obtained so far are still controversial. In this article, the literature is reviewed and the authors' experience on using FSH treatment in oligozoospermic subjects is discussed.
Collapse
Affiliation(s)
- Carlo Foresta
- a University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology & Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy.
| | - Riccardo Selice
- b University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology & Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy
| | - Andrea Garolla
- b University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology & Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy
| | - Alberto Ferlin
- b University of Padova, Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology & Centre for Male Gamete Cryopreservation, Via Gabelli 63, 35121 Padova, Italy
| |
Collapse
|
41
|
Bibliography. Current world literature. Parathyroids, bone and mineral metabolism. Curr Opin Endocrinol Diabetes Obes 2007; 14:494-501. [PMID: 17982358 DOI: 10.1097/med.0b013e3282f315ef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Abstract
BACKGROUND Male factor infertility accounts for 50% of all infertility. The treatment of idiopathic male infertility is empirical. Urinary, purified, and recombinant gonadotrophins have been used to improve sperm parameters in idiopathic male infertility with the goal of increasing pregnancy rates. Research addressing pregnancy rates in partners of men treated with gonadotrophins has had conflicting results and needs to be analysed. OBJECTIVES To determine the effectiveness of gonadotrophin administration in men with idiopathic subfertility in improving spontaneous pregnancy rate and in assisted reproductive technique cycles. SEARCH STRATEGY We searched the Cochrane Menstrual Disorders and Subfertility Group trials register (31 May 2007), the Cochrane Central Register of Controlled Trials (The Cochrane Library, issue 2, 2007), MEDLINE (1966 to May 2007), EMBASE and Biological Abstracts (1980 to Week 21 2007). Searches were not limited by language. The bibliographies of included, excluded trials and abstracts of major meetings were searched for additional trials. Authors and pharmaceutical companies were contacted for missing and unpublished data. SELECTION CRITERIA Truly randomised controlled trials where gonadotrophins were administered for the treatment of idiopathic male subfertility with reporting of pregnancy rates were included in the review. DATA COLLECTION AND ANALYSIS Two reviewers independently assessed trial quality and extracted data. Study authors were contacted for additional information. Adverse effects information was collected from the trials. We analysed data regarding pregnancy occurring within three months after gonadotrophin therapy. MAIN RESULTS Four RCTs with 278 participants were included in the analysis. None of the studies had an adequate sample size and they had variable follow-up periods. None of the studies reported live birth or miscarriage rates. Compared to placebo or no treatment, gonadotrophins showed a significantly higher pregnancy rate per couple randomized within three months of completing therapy ( OR 4.17, 95% CI 1.30 to 7.09). AUTHORS' CONCLUSIONS The number of trials and participants is insufficient to draw final conclusions. A large multicenter study with adequate power is needed.
Collapse
Affiliation(s)
- A M Attia
- Faculty Of Medicine, Cairo University, Obstetrics and Gynaecology, 18 El-Ghaith St., El-Agouza, Cairo, Egypt, 12311.
| | | | | | | |
Collapse
|
43
|
Current World Literature. Curr Opin Obstet Gynecol 2007; 19:289-96. [PMID: 17495648 DOI: 10.1097/gco.0b013e3281fc29db] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|