World J Mens Health. 2024;42:e42. Forthcoming. English.
Published online Apr 04, 2024.
Copyright © 2024 Korean Society for Sexual Medicine and Andrology
Original Article

Global Practice Patterns and Variations in the Medical and Surgical Management of Non-Obstructive Azoospermia: Results of a World-Wide Survey, Guidelines and Expert Recommendations

Amarnath Rambhatla,1 Rupin Shah,2 Imad Ziouziou,3 Priyank Kothari,4 Gianmaria Salvio,5 Murat Gul,6 Taha Hamoda,7,8 Parviz Kavoussi,9 Widi Atmoko,10 Tuncay Toprak,11 Ponco Birowo,10 Edmund Ko,12 Mohamed Arafa,13,14,15 Ramy Abou Ghayda,16 Vilvapathy Senguttuvan Karthikeyan,17 Giorgio Ivan Russo,18 Germar-Michael Pinggera,19 Eric Chung,20 Ahmed M. Harraz,21,22,23 Marlon Martinez,24 Nguyen Ho Vinh Phuoc,25 Nicholas Tadros,26 Ramadan Saleh,27 Missy Savira,10 Giovanni M. Colpi,28 Wael Zohdy,14 Edoardo Pescatori,29 Hyun Jun Park,30,31 Shinichiro Fukuhara,32 Akira Tsujimura,33 Cesar Rojas-Cruz,34 Angelo Marino,35 Siu King Mak,36 Edouard Amar,37 Wael Ibrahim,38 Puneet Sindhwani,39 Naif Alhathal,40 Gian Maria Busetto,41 Manaf Al Hashimi,42,43 Ahmed El-Sakka,44 Asci Ramazan,45 Fotios Dimitriadis,46 Massimiliano Timpano,47 Davor Jezek,48 Baris Altay,49 Daniel Suslik Zylbersztejn,50 Michael YC Wong,51 Du Geon Moon,52 Christine Wyns,53 Safar Gamidov,54 Hamed Akhavizadegan,55 Alessandro Franceschelli,56 Kaan Aydos,57 Nguyen Quang,58,59 Shedeed Ashour,14 Adel Al Dayel,60 Mohammed S. Al-Marhoon,61 Sava Micic,62 Saleh Binsaleh,63 Alayman Hussein,8 Haitham Elbardisi,13,15 Taymour Mostafa,14 Jonathan Ramsay,64 Athanasios Zachariou,65 Islam Fathy Soliman Abdelrahman,14,66 Osvaldo Rajmil,67 Arif Kalkanli,68 Juan Manuel Corral Molina,69 Kadir Bocu,70 Gede Wirya Kusuma Duarsa,71 Gökhan Çeker,72 Ege Can Serefoglu,73 Fahmi Bahar,74 Nazim Gherabi,75 Shinnosuke Kuroda,76 Abderrazak Bouzouita,77 Ahmet Gudeloglu,78 Erman Ceyhan,79 Mohamed Saeed Mohamed Hasan,80 Muhammad Ujudud Musa,81 Ahmad Motawi,14 Chak-Lam Cho,82 Hisanori Taniguchi,83 Christopher Chee Kong Ho,84 Jesus Fernando Solorzano Vazquez,85 Shingai Mutambirwa,86 Nur Dokuzeylul Gungor,87 Marion Bendayan,88,89 Carlo Giulioni,90 Aykut Baser,91 Marco Falcone,47 Luca Boeri,92 Gideon Blecher,93,94 Alireza Kheradmand,95 Tamilselvi Sethupathy,96 Ricky Adriansjah,97 Nima Narimani,98 Charalampos Konstantinidis,99 Tuan Thanh Nguyen,100,101,102 Andrian Japari,103 Parisa Dolati,104 Keerti Singh,105,106 Cevahir Ozer,79 Selcuk Sarikaya,107 Nadia Sheibak,108,109 Ndagijimana Jean Bosco,110 Mehmet Serkan Özkent,111 Sang Thanh Le,8,103 Ioannis Sokolakis,112 Darren Katz,113,114,115 Ryan Smith,116 Manh Nguyen Truong,104,117 Tan V. Le,25,118 Zhongwei Huang,119 Muslim Dogan Deger,120 Umut Arslan,11 Gokhan Calik,121 Giorgio Franco,122 Ayman Rashed,123 Oguzhan Kahraman,79 Sotiris Andreadakis,124 Rosadi Putra,125 Giancarlo Balercia,5 Kareim Khalafalla,20,126 Rossella Cannarella,18 Anh Đặng Tuấn,127 Amr El Meliegy,14 Birute Zilaitiene,128 Marlene Lizbeth Zamora Ramirez,129 Filippo Giacone,130 Aldo E. Calogero,18 Konstantinos Makarounis,131 Sunil Jindal,132 Bac Nguyen Hoai,133 Ravi Banthia,134 Marcelo Rodriguez Peña,135 Dharani Moorthy,136 Aram Adamyan,137 Deniz Kulaksiz,138 Hussein Kandil,139 Nikolaos Sofikitis,65 Ciro Salzano,140 Andreas Jungwirth,141 Surendra Reddy Banka,142 Tiago Cesar Mierzwa,143 Tahsin Turunç,144 Divyanu Jain,145 Armen Avoyan,138 Pietro Salacone,146 Ateş Kadıoğlu,147 Chirag Gupta,148 Haocheng Lin,149 Iman Shamohammadi,150 Nasser Mogharabian,151 Trenton Barrett,152 Yavuz Onur Danacıoğlu,153 Andrea Crafa,18 Salima Daoud,154 Vineet Malhotra,155 Abdulmalik Almardawi,156 Osama Mohamed Selim,14 Mohamad Moussa,157,158 Saeid Haghdani,159 Mesut Berkan Duran,160 Yannic Kunz,19 Mirko Preto,47 Elena Eugeni,161,162 Thang Nguyen,138 Ahmed Rashad Elshahid,123 Seso Sulijaya Suyono,163 Dyandra Parikesit,164 Essam Nada,27 Eduardo Gutiérrez Orozco,165 Florence Boitrelle,88,89 Nguyen Thi Minh Trang,102 Mounir Jamali,166 Raju Nair,167 Mikhail Ruzaev,168 Franco Gadda,92 Charalampos Thomas,99 Raphael Henrique Ferreira,169 Umit Gul,170 Serena Maruccia,171 Ajay Kanbur,172,173 Ella Kinzikeeva,174 Saad Mohammed Abumelha,156 Raghavender Kosgi,175 Fatih Gokalp,176 Mohammad Ayodhia Soebadi,177 Gustavo Marquesine Paul,178 Hesamoddin Sajadi,179 Deepak Gupte,180 Rafael F. Ambar,143 Emrullah Sogutdelen,181 Karun Singla,182 Ari Basukarno,183 Shannon Hee Kyung Kim,184 Mohammad Ali Sadighi Gilani,179 Koichi Nagao,185 Sakti Ronggowardhana Brodjonegoro,186 Andri Rezano,187,188 Mohamed Elkhouly,189 Rossella Mazzilli,190 Hasan M. A. Farsi,7 Hung Nguyen Ba,191 Hamed Alali,184 Dimitrios Kafetzis,192 Tran Quang Tien Long,193 Sami Alsaid,14 Hoang Bao Ngoc Cuong,194 Knigavko Oleksandr,195 Akhmad Mustafa,97 Herik Acosta,196 Hrishikesh Pai,197 Bahadır Şahin,198 Eko Arianto,199 Colin Teo,200 Sanjay Prakash Jayaprakash,17 Rinaldo Indra Rachman,10 Mustafa Gurkan Yenice,153 Omar Sefrioui,201 Shivam Priyadarshi,202 Marko Tanic,203 Noor Kareem Alfatlaw,204 Fikri Rizaldi,187 Ranjit B. Vishwakarma,2 George Kanakis,205 Dinesh Thomas Cherian,206 Joe Lee,207 Raisa Galstyan,208 Hakan Keskin,80 Janan Wurzacher,19 Doddy Hami Seno,209 Bambang S Noegroho,97 Ria Margiana,199,210,211,212 Qaisar Javed,213 Fabrizio Castiglioni,214 Raman Tanwar,215 Ana Puigvert,87,216 Coşkun Kaya,217 Medianto Purnomo,218 Chadi Yazbeck,219 Azwar Amir,220 Edson Borges,221 Marina Bellavia,28 Isaac Ardianson Deswanto,10 Vinod KV,222 Giovanni Liguori,223 Dang Hoang Minh,59 Kashif Siddiqi,224 Fulvio Colombo,29 Armand Zini,225 Niket Patel,226 Selahittin Çayan,227 Ula Al-kawaz,228 Maged Ragab,229 Guadalupe Hernández Hebrard,230 Jean de la Rosette,121 Ozan Efesoy,229 Ivan Hoffmann,231,232 Thiago Afonso Teixeira,233,234,235 Barış Saylam,227 Daniela Delgadillo,85 Ashok Agarwal,236 and Global Andrology Forum
    • 1Department of Urology, Henry Ford Health System, Vattikuti Urology Institute, Detroit, MI, USA.
    • 2Division of Andrology, Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India.
    • 3Department of Urology, College of Medicine and Pharmacy, Ibn Zohr University, Agadir, Morocco.
    • 4Department of Urology, Topiwala National Medical College, B.Y.L Nair Charitable Hospital, Mumbai, India.
    • 5Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy.
    • 6Department of Urology, Selçuk University School of Medicine, Konya, Turkey.
    • 7Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia.
    • 8Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt.
    • 9Department of Reproductive Urology, Austin Fertility & Reproductive Medicine/Westlake IVF, Austin, TX, USA.
    • 10Department of Urology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
    • 11Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey.
    • 12Department of Urology, Loma Linda University Health, Loma Linda, CA, USA.
    • 13Department of Urology, Hamad Medical Corporation, Doha, Qatar.
    • 14Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt.
    • 15Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar.
    • 16Urology Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, USA.
    • 17Andrology Unit, Department of Urology, Apollo Hospitals, Chennai, India.
    • 18Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
    • 19Department of Urology, University Hospital Innsbruck, Innsbruck, Austria.
    • 20Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia.
    • 21Department of Urology, Mansoura University Urology and Nephrology Center, Mansoura, Egypt.
    • 22Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait.
    • 23Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait.
    • 24Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines.
    • 25Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam.
    • 26Division of Urology, Southern Illinois University School of Medicine, Springfield, IL, USA.
    • 27Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt.
    • 28Andrology and IVF Center, Next Fertility Procrea, Lugano, Switzerland.
    • 29Andrology and Reproductive Medicine Unit, Next Fertility GynePro, Bologna, Italy.
    • 30Department of Urology, Pusan National University School of Medicine, Busan, Korea.
    • 31Medical Research Institute of Pusan National University Hospital, Busan, Korea.
    • 32Department of Urology, Graduate School of Medicine, Osaka University, Osaka, Japan.
    • 33Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan.
    • 34Department of Urology, University Hospital of Rostock, Rostock, Germany.
    • 35Reproductive Medicine Unit, ANDROS Day Surgery Clinic, Palermo, Italy.
    • 36Department of Surgery, Union Hospital Reproductive Medicine Centre (Tsim Sha Tsui), Kowloon, China.
    • 37Department of Urology, American Hospital of Paris, Paris, France.
    • 38Department of Obstetrics Gynecology and Reproductive Medicine, Fertility Care Center in Cairo, Cairo, Egypt.
    • 39Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
    • 40Department of Urology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
    • 41Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy.
    • 42Department of Urology, Burjeel Hospital, Abu Dhabi, UAE.
    • 43Department of Urology, Khalifa University College of Medicine and Health Science, Abu Dhabi, UAE.
    • 44Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
    • 4545Department of Urology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey.
    • 461st Urology Department, Aristotle University of Thessaloniki, Thessaloniki, Greece.
    • 47Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy.
    • 48Department for Transfusion Medicine and Transplantation Biology, Reproductive Tissue Bank, University Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia.
    • 49Department of Urology, Ege University Medical School, Bornova, Turkey.
    • 50Department of Surgery, Discipline of Urology, Fleury Group and Hospital Israelita Albert Einstein, São Paulo, Brazil.
    • 51Department of Andrology, International Urology, Fertility and Gynecology Centre, Mount Elizabeth Hospital, Singapore.
    • 52Department of Urology, Korea University Guro Hospital, Seoul, Korea.
    • 53Department of Gynaecology-Andrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium.
    • 54V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, Ministry of Health of Russia, Moscow, Russia.
    • 55Department of Urology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran.
    • 56Andrology Unit, University Hospital S. Orsola, Bologna, Italy.
    • 57Department of Urology, Ankara University, Ankara, Turkey.
    • 58Center for Andrology and Sexual Medicine, Viet Duc University Hospital, Hanoi, Vietnam.
    • 59Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam.
    • 60Men's Health Clinic Dammam, Dammam, Saudi Arabia.
    • 61Division of Urology, Department of Surgery, Sultan Qaboos University, Muscat, Oman.
    • 62Department of Andrology, Uromedica Polyclinic, Belgrade, Serbia.
    • 63Division of Urology, Deparment of Surgery, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.
    • 64Department of Andrology, Hammersmith Hospital, London, UK.
    • 65Department of Urology, Ioannina University School of Medicine, Ioannina, Greece.
    • 66Department of Andrology, Armed Forces College of Medicine, Cairo, Egypt.
    • 67Department of Andrology, Fundació Puigvert, Barcelona, Spain.
    • 68Department of Urology, Taksim Education and Research Hospital, Istanbul, Turkey.
    • 69Department of Urology, Hospital Clínico de Barcelona, Barcelona, Spain.
    • 70Urology Department, Niğde Omer Halis Demir University, Faculty of Medicine, Sirnak, Turkey.
    • 71Department of Urology, Udayana University, Denpasar, Indonesia.
    • 72Department of Urology, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey.
    • 73Department of Urology, Biruni University School of Medicine, Istanbul, Turkey.
    • 74Andrology Section, Siloam Sriwijaya Hospital, Palembang, Indonesia.
    • 75Department of Medicine, University of Algiers 1, Algiers, Algeria.
    • 76Department of Urology, Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
    • 77Department of Anatomy, Faculty of Medicine, Faculty Tunis Manar, Tunis, Tunisia.
    • 78Department of Urology, Hacettepe University Faculty of Medicine, Ankara, Turkey.
    • 79Department of Urology, Faculty of Medicine, Baskent University, Ankara, Turkey.
    • 80Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt.
    • 81Urology Unit, Department of Surgery, Federal Medical Center, Katsina State, Nigeria.
    • 82Department of Surgery, S. H. Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong.
    • 83Department of Urology and Andrology, Kansai Medical University, Osaka, Japan.
    • 84Department of Surgery, School of Medicine, Taylor's University, Subang Jaya, Selangor, Malaysia.
    • 85IVF Department, CITMER Reproductive Medicine, Mexico City, Mexico.
    • 86Department of Urology, Dr. George Mukhari Academic Hospital, Sefako Makgatho Health Science University, Medunsa, South Africa.
    • 87Department of Obstetrics and Gynecology, Reproductive Endocrinology and IVF Unit, School of Medicine, Bahcesehir University, Istanbul, Turkey.
    • 88Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France.
    • 89Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France.
    • 90Department of Urology, Polytechnic University of Marche, Ancona, Italy.
    • 91Department of Urology, Faculty of Medicine, Bandırma Onyedi Eylül University, Balıkesir, Turkey.
    • 92Department of Urology, IRCCS Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.
    • 93Department of Surgery, School of Clinical Sciences, Monash University, Melbourne, Australia.
    • 94Department of Urology, The Alfred Hospital, Melbourne, Australia.
    • 95Department of Urology, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
    • 96Department of Obstetrics and Gynaecology, CK Medical Centre Hospital, Erode, India.
    • 97Department of Urology, Hasan Sadikin General Hospital, Faculty of Medicine of Padjadjaran University, Bandung, Indonesia.
    • 98Department of Urology, School of Medicine, Hasheminejad Kidney Center, Iran University of Medical Science, Tehran, Iran.
    • 99Urology and Neuro-Urology Unit, National Rehabilitation Center, Athens, Greece.
    • 100Department of Urology, University of California, Irvine, CA, USA.
    • 101Department of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam.
    • 102Department of Urology, Cho Ray Hospital, Ho Chi Minh City, Vietnam.
    • 103Department of Urology, Fertility Clinic, Telogorejo Hospital, Central Java, Indonesia.
    • 104Department of Animal Science, Faculty of Agriculture, University of Shiraz, Shiraz, Iran.
    • 105Department of Preclinical and Health Sciences, Faculty of Medical Sciences, The University of West Indies, Bridgetown, Barbados.
    • 106Windsor Medical Centre, Bridgetown, Barbados.
    • 107Department of Urology, Gulhane Research and Training Hospital, University of Health Sciences, Ankara, Turkey.
    • 108Department of Anatomical Sciences, Reproductive Sciences and Technology Research Center, Iran University of Medical Sciences, Tehran, Iran.
    • 109Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran.
    • 110Department of Dermatology, Venereology & Andrology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt.
    • 111Department of Urology, Konya City Hospital, Konya, Turkey.
    • 112Department of Urology, Martha-Maria Hospital Nuremberg, Nuremberg, Germany.
    • 113Men’s Health Melbourne, Victoria, Australia.
    • 114Department of Surgery, Western Precinct, University of Melbourne, Victoria, Australia.
    • 115Department of Urology, Western Health, Victoria, Australia.
    • 116Department of Urology, University of Virginia School of Medicine, Charlottesville, VA, USA.
    • 117Fertility Centre, Hanh Phuc International Hospital, Binh Duong, Vietnam.
    • 118Department of Urology and Andrology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam.
    • 119Department of Obstetrics and Gynaecology, National University Health Systems, Singapore.
    • 120Department of Urology, Edirne Sultan 1st Murat State Hospital, Edirne, Turkey.
    • 121Department of Urology, Istanbul Medipol University, Istanbul, Turkey.
    • 122Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy.
    • 123123Department of Urology, Faculty of Medicine, 6th of October University, Giza, Egypt.
    • 124Private Practice, Thessaloniki, Greece.
    • 125Department of Urology, RSUD Ciawi Regional General Hospital, West Java, Indonesia.
    • 126Department of Urology, University of Illinois, Chicago, IL, USA.
    • 127Tam Anh IVF Center, Tam Anh General Hospital, Hanoi, Vietnam.
    • 128Institute of Endocrinology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
    • 129Reproductive Medicine Unit, Español Hospital, Mexico City, Mexico.
    • 130HERA Center, Unit of Reproductive Medicine, Sant'Agata Li Battiati, Catania, Italy.
    • 131Department of Urology and Andrology, Locus Medicus, Athens, Greece.
    • 132Department of Andrology & Reproductive Medicine, Jindal Hospital & Fertility Center, Meerut, India.
    • 133Department of Andrology and Sexual Medicine, Hanoi Medical University Hospital, Hanoi, Vietnam.
    • 134Department of Urology, Western General Hospital, Edinburgh, UK.
    • 135Institute of Gynecology and Fertility (IFER), University of Buenos Aires, Buenos Aires, Argentina.
    • 136IVF Department, Swarupa Fertility & IVF Centre, Vijayawada, India.
    • 137Department of Urology, Astghik Medical Center, Yerevan, Armenia.
    • 138Department of Obstetrics and Gynecology, Kanuni Training and Research Hospital, University of Health Sciences, Trabzon, Turkey.
    • 139Fakih IVF Fertility Center, Abu Dhabi, UAE.
    • 140PO San Giovanni Bosco, ASL Napoli 1 Centro, Napoli, Italy.
    • 141St. Barabara Private Clinic, Bad Vigaun, Austria.
    • 142Department of Andrology, Androcare Institute of Andrology and Men's Health, Hyderabad, India.
    • 143Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil.
    • 144Urology Clinic, Iskenderun Gelisim Hospital, Iskenderun, Turkey.
    • 145Department of Obstetrics and Gynecology, Jaipur Golden Hospital, New Delhi, India.
    • 146Andrology and Pathophysiology of Reproduction Unit, Santa Maria Goretti Hospital, Latina, Italy.
    • 147Section of Andrology, Department of Urology, Istanbul University Faculty of Medicine, Istanbul, Turkey.
    • 148Department of Urology, Jaipur National University, Jaipur, India.
    • 149Department of Urology, Peking University Third Hospital, Peking University, Beijing, China.
    • 150Department of Urology, Shiraz University of Medical Sciences, Shiraz, Iran.
    • 151Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
    • 152Perth Urology Clinic, Perth, WA, Australia.
    • 153Department of Urology, University of Health Science, Istanbul Bakırköy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey.
    • 154Laboratory of Histo-Embryology and Reproductive Biology, Faculty of Medicine of Sfax, University of Sfax, Sfax, Tunisia.
    • 155Department of Urology and Andrology, VNA Hospital, New Delhi, India.
    • 156Department of Urology, Prince Sultan Millitary Medical City, Riyadh, Saudi Arabia.
    • 157Department of Urology, Lebanese University, Beirut, Lebanon.
    • 158Department of Urology, Al Zahraa Hospital, UMC, Beirut, Lebanon.
    • 159Andrology Research Center, Yazd Reproductive Science Institute, Isfahan Fertility and Infertility Center, Isfahan, Iran.
    • 160Department of Urology, Pamukkale University School of Medicine, Denizli, Turkey.
    • 161Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
    • 162Department of Medicine and Medical Specialties, Division of Medical Andrology and Endocrinology of Reproduction, University of Terni, Terni, Italy.
    • 163Gladiool IVF, Magelang, Indonesia.
    • 164Department of Urology, Faculty of Medicine, Universitas Indonesia Hospital, Depok, Indonesia.
    • 165IVF Department, CITMER Reproductive Medicine, Nuevo Leon, Mexico.
    • 166Department of Urology, Military Teaching Hospital, Rabat, Morocco.
    • 167Department of Reproductive Medicine, Mitera Hospital, Kottayam, India.
    • 168Urology Clinic, Moscow, Russia.
    • 169Department of Urology, Santa Casa de Ribeirão Preto, São Paulo, Brazil.
    • 170Private EPC Hospital, Adana, Turkey.
    • 171Department of Urology, ASST Santi Paolo e Carlo, San Paolo Hospital, Milano, Italy.
    • 172Department of Andrology, Kanbur Clinic, Thane, India.
    • 173Department of Urosurgery, Jupiter Hospital, Thane, India.
    • 174Andrology Unit, Zucchi Clinical Institutes, Monza, Italy.
    • 175Department of Andrology and Men’s Health, Apollo Hospitals, Hyderabad, India.
    • 176Department of Urology, Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Turkey.
    • 177Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
    • 178Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
    • 179Department of Urology, Bombay Hospital and Medical Research Center, Mumbai, India.
    • 180Department of Urology, Bolu Abant Izzet Baysal University, Bolu, Turkey.
    • 181Urology Department, Clarity Health, Chandigarh, India.
    • 182Department of Urology, Dr. Dradjat Hospital, Serang, Indonesia.
    • 183IVF Australia, Sydney, Australia.
    • 184Department of Urology, Macquarie University Faculty of Medicine and Health Sciences, Sydney, Australia.
    • 185Department of Urology, Toho University Faculty of Medicine, Tokyo, Japan.
    • 186Division of Urology, Department of Surgery, Prof. Dr. Sardjito Hospital, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia.
    • 187Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
    • 188Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang, Indonesia.
    • 189IVF Department, Bourn Hall Fertility Center, Dubai, UAE.
    • 190Unit of Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.
    • 191Andrology Unit, ART Center, Vinmec Times City International Hospital, Hanoi, Vietnam.
    • 192Orchid IVF Clinic, Dubai, UAE.
    • 193Department of Obstetrics and Gynecology, Hanoi Obstetrics and Gynecology Hospital, Hanoi, Vietnam.
    • 194Department of Surgery, Hai Phong University of Medicine and Pharmacy, Hai Phong, Vietnam
    • 195Department of Urology, Nephrology and Andrology Kharkiv National Medical University, Kharkiv, Ukraine.
    • 196Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan.
    • 197IVF Department, Bloom IVF Group, Mombai, India.
    • 198Department of Urology, School of Medicine, Marmara University, Istanbul, Turkey.
    • 199Department of Urology, Prof R.D. Kandou Hospital, Manado, Indonesia.
    • 200Department of Urology, Gleneagles Hospital, Singapore.
    • 201African Fertility Center, Casablanca, Morocco.
    • 202Department of Urology, Sawai Man Singh Medical College and Hospital, Jaipur, Rajasthan, India.
    • 203Department of Urology, General Hospital, Cuprija, Serbia.
    • 204Fertility Center of Al-Najaf, Al-Sadr Medical City, Babylon Health Directorate, Iraqi Ministry of Health, Baghdad, Iraq.
    • 205Department of Endocrinology, Diabetes and Metabolism, Athens Naval & VA Hospital, Athens, Greece.
    • 206Urology Department, Aster Medcity, Kochi, India.
    • 207Department of Urology, National University Hospital, Singapore.
    • 208Department of Urology, Yerevan State Medical University, Yerevan, Armenia.
    • 209Division of Urology, Department of Surgery, Persahabatan General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
    • 210Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
    • 211Master’s Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
    • 212Urology Unit, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
    • 213Department of Urology, Al-Ahlia Hospital, Abu Dhabi, UAE.
    • 214Andrology Unit, ART Center - San Carlo Clinic, Milan, Italy.
    • 215Urology Center, Jyoti Hospital, Gurugram, India.
    • 216Institute of Andrology and Sexual Medicine (IANDROMS), Barcelona, Spain.
    • 217Department of Urology, Health Science University Eskisehir City HPRH, Eskisehir, Turkey.
    • 218Department of Urology, Brawijaya University, Malang, Indonesia.
    • 219Department of Obstetrics Gynecology and Reproductive Medicine, Reprogynes Medical Institute, Paris, France.
    • 220Department of Urology, Dr Wahidin Sudirohusodo Hospital, Makassar, Indonesia.
    • 221IVF Department, Fertility Assisted Fertilization Center, São Paulo, Brazil.
    • 222Department of Urology, Cure & SK Hospital, Trivandrum, India.
    • 223Department of Urology, University of Trieste, Trieste, Italy.
    • 224Department of Urology, Cleveland Clinic, Abu Dhabi, UAE.
    • 225Department of Surgery, McGill University, Montreal, QC, Canada.
    • 226Akanksha Hospital and Research Institute, Anand, Gujarat, India.
    • 227Department of Urology, University of Mersin School of Medicine, Mersin, Turkey.
    • 228High Institute for Infertility Diagnosis and Assisted Reproductive Technologies, Al-Nahrain University, Baghdad, Iraq.
    • 229Department of Andrology, Tanta University, Tanta, Egypt.
    • 230IVF Department, CITMER Reproductive Medicine, Puebla, Mexico.
    • 231Department of Reproductive Medicine and Andrology, University Clinic Halle (Saale), Halle, Germany.
    • 232Reproductive Center Dr. Hoffmann, Berlin, Germany.
    • 233Division of Urology, University Hospital, Federal University of Amapa, Macapá, Brazil.
    • 234Men’s Health Study Group, Institute for Advanced Studies, University of São Paulo, São Paulo, Brazil.
    • 235Androscience-Science and Innovation Center and High Complexity Clinical and Research Andrology Laboratory, São Paulo, Brazil.
    • 236Cleveland Clinic Foundation, Cleveland, OH, USA.
Received November 22, 2023; Revised December 05, 2023; Accepted December 07, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Purpose

Non-obstructive azoospermia (NOA) is a common, but complex problem, with multiple therapeutic options and a lack of clear guidelines. Hence, there is considerable controversy and marked variation in the management of NOA. This survey evaluates contemporary global practices related to medical and surgical management for patients with NOA.

Materials and Methods

A 56-question online survey covering various aspects of the evaluation and management of NOA was sent to specialists around the globe. This paper analyzes the results of the second half of the survey dealing with the management of NOA. Results have been compared to current guidelines, and expert recommendations have been provided using a Delphi process.

Results

Participants from 49 countries submitted 336 valid responses. Hormonal therapy for 3 to 6 months was suggested before surgical sperm retrieval (SSR) by 29.6% and 23.6% of participants for normogonadotropic hypogonadism and hypergonadotropic hypogonadism respectively. The SSR rate was reported as 50.0% by 26.0% to 50.0% of participants. Interestingly, 46.0% reported successful SSR in <10% of men with Klinefelter syndrome and 41.3% routinely recommended preimplantation genetic testing. Varicocele repair prior to SSR is recommended by 57.7%. Half of the respondents (57.4%) reported using ultrasound to identify the most vascularized areas in the testis for SSR. One-third proceed directly to microdissection testicular sperm extraction (mTESE) in every case of NOA while others use a staged approach. After a failed conventional TESE, 23.8% wait for 3 months, while 33.1% wait for 6 months before proceeding to mTESE. The cut-off of follicle-stimulating hormone for positive SSR was reported to be 12–19 IU/mL by 22.5% of participants and 20–40 IU/mL by 27.8%, while 31.8% reported no upper limit.

Conclusions

This is the largest survey to date on the real-world medical and surgical management of NOA by reproductive experts. It demonstrates a diverse practice pattern and highlights the need for evidence-based international consensus guidelines.

Keywords
Azoospermia; Hypogonadism; Infertility, male; Semen; Sperm retrieval

INTRODUCTION

Azoospermia refers to the complete absence of sperm in the semen and the centrifuged pellets. Approximately 1% of the general male population and 5%–10% of infertile men are azoospermic [1, 2]. Azoospermia can be classified as obstructive (OA) or non-obstructive azoospermia (NOA) and each has different etiologies and management [3]. NOA is due to impairment in testicular sperm production and constitutes 60% of azoospermia cases [4]. Currently, there is a lack of society guidelines, especially for the management of NOA. There is still controversy on the role of hormonal therapy, antioxidants, and varicocele repair (VR) prior to sperm retrieval. Moreover, there are questions about the best approach to surgical sperm retrieval (SSR), though microdissection testicular sperm extraction (mTESE) appears to be the most appropriate with the best chances of sperm retrieval. Further, it remains unclear whether it is better to perform mTESE simultaneously with oocyte retrieval or whether mTESE with cryopreservation followed by intracytoplasmic sperm injection (ICSI) at a later date gives equal results when considering different clinical scenarios. Additionally, factors such as cost, and accessibility also need to be considered.

The current study aims to assess contemporary expert global practices related to the medical and surgical management of NOA and to compare it with the available international practice guidelines. Also, an “expert recommendation” is provided to clarify the current best practices of NOA management based on global practices, society guidelines, and available evidence from the literature.

MATERIALS AND METHODS

The global survey was approved by the Internal Review Board (IR-02-23-110). The checklist for Reporting Results of Internet E-Survey (CHERRIES) was used to guide the construction, dissemination, and analysis of the questionnaire [5]. The checklist is provided in the Supplement File 1.

1. Target population

The survey was conducted among physicians who treat NOA patients in their daily practice. The group included andrologists, urologists, and reproductive specialists. Clinicians with no experience in dealing with these patients were excluded.

2. Questionnaire creation and structure

Members of the Global Andrology Forum (GAF) were invited to submit multiple-choice questions (MCQs) on various aspects of NOA evaluation and treatment [6]. These MCQs were reviewed and integrated into a questionnaire by a group of ten experts who were experienced reproductive urologists actively engaged in treating patients with NOA. The option “not applicable” was added for any respondents who do not encounter the scenario in clinical practice. The final questionnaire comprised 56 questions divided into 4 sections: demographic data (Q1–8), medical management (Q9–15), surgical therapy (Q16–40), and future horizons (Q41–43) with the invitation letter on the first page including the aims of the survey and consent to participate (Supplement File 2). While completing the survey, respondents were able to scan, review, and edit answers before final submission.

3. Questionnaire dissemination

The questionnaire was created using Google Forms and was available online from July 21st, 2022, to September 9th, 2022. The survey was published on the website of the GAF and all members who fit the inclusion criteria were invited to complete the survey. On the final page, the respondents were able to recommend other experts who met the inclusion criteria. The survey questionnaire was also distributed to the members of the different organizations listed under the Acknowledgements.

4. Data collection and analysis

The survey data were extracted from the Google Sheet associated with the Google Form. A total of 367 responses were submitted. Three hundred thirty-six responses were analyzed after the exclusion of invalid responses. Responses were excluded if respondents were clinicians not treating NOA (25 responses) or if respondents only completed the demographics section of the survey and no other sections (6 responses). Some questions allowed more than one response which increased the number of responses analyzed for that particular question. The question responses were calculated based on the number and percentage of each answer. For questions with the option (select all that apply), the analysis was based on the total number of responses. We determined percentages for each answer selection and graphically presented the data.

5. Society guidelines

There are limited available guidelines on the evaluation and management of men with NOA. Different societies such as the American Urological Association/American Society for Reproductive Medicine (AUA/ASRM), European Association of Urology (EAU), and European Academy of Andrology (EAA) have released guidelines pertaining to male infertility.

6. GAF expert recommendations

GAF expert recommendations were formulated using a Delphi process (Supplement File 3). Two senior authors (RS, AR) formulated the statements based on the survey results, available professional society guidelines, and the latest literature on the subject. These were then forwarded to 43 GAF clinicians with at least 15 years of experience in treating NOA. They rated the recommendations on a scale of 1 to 10, 1 being “completely disagree” and 10 being “completely agree.” When they disagreed with the statement, they were asked to provide a new changed recommendation. The final recommendation was accepted once 70% of the respondents scored 7 or above.

RESULTS AND DISCUSSION

We grouped the results of our survey by question topic. For each group of questions, we discuss guideline recommendations from the major male infertility guidelines (AUA/ASRM, EAU, EAA) (Table 1) and provide GAF expert recommendations based on the Delphi process (all recommendations were accepted in the first round).

Table 1
Comparison of the different guidelines on the medical and surgical management of NOA

1. Demographics

A total of 367 participants submitted their responses. Of these, 31 responses were excluded due to incomplete data (n=6) or submission by an ineligible respondent (n=25). A total of 336 participants were included in the study. Highest responses were from Italy (n=36, 10.7%), India (n=35, 10.4%), and Türkiye (n=35, 10.4%). Respondents and countries are presented in Fig. 1.

Fig. 1
Geographical distribution of respondents. The number of respondents is shown in brackets after the name of each country. The map is color-coded according to the number of respondents in each country.

1) Respondent demographics

Most respondents were attending physicians, with training in general urology or completed andrology fellowships, which were equally divided between academic and private practice (Table 2). Almost half of them (48.5%) had 11 or more years of experience (Fig. 2) with more than 30% of them evaluating 11–20 new infertile men per week (Fig. 3).

Fig. 2
Years of practice of the respondents.

Fig. 3
The number of new couples evaluated per week by the respondents.

Table 2
Demographic characteristics of the respondents

2. Medical management of NOA

1) Hormonal therapy in men with NOA

(1) Survey results

Hormonal therapy was suggested prior to sperm retrieval for NOA patients with hypogonadotropic hypogonadism, normogonadotropic hypogonadism, and hypergonadotropic hypogonadism by 74.6%, 29.6%, and 23.6% of participants respectively (Fig. 4). Almost 24.6% of participants offer hormonal therapy for NOA patients after the first failure of sperm retrieval while 9.9% offer it in all NOA patients undergoing an initial attempt at SSR.

Fig. 4
Characteristics of NOA patients who are offered hormonal therapy. NOA: non-obstructive azoospermia.

When hormone therapy was recommended before sperm retrieval in NOA patients (excluding hypogonadotropic hypogonadism), 30.6% of respondents recommended it for a period for 6 months or less (Fig. 5).

Fig. 5
Duration of hormonal therapy before sperm retrieval in NOA patient. NOA: non-obstructive azoospermia.

The effect of hormonal therapy before sperm retrieval was evaluated by testosterone to estradiol ratio (T/E2), testosterone (T), follicle-stimulating hormone (FSH), luteinizing hormone (LH) levels, by 41.5%, 52.5%, 56.9%, and 34.3% of participants, respectively (Fig. 6).

Fig. 6
Evaluation of the effect of hormonal therapy before sperm retrieval. LH: luteinizing hormone, FSH: follicle-stimulating hormone, T: testosterone, T/E2: testosterone to estradiol ratio.

Regarding the choice of hormone therapy prior to sperm retrieval in hypergonadotropic NOA, a marked diversity of choices was seen ranging from various oral and injectable drugs, singly or in combination (Fig. 7). A similar diversity of choices, with a preference for clomiphene or aromatase inhibitors, were reported for men with NOA and normal gonadotropins (Fig. 8).

Fig. 7
Hormonal therapy before sperm retrieval in hypergonadotropic NOA patient. NOA: non-obstructive azoospermia, HCG: human chorionic gonadotropin, FSH: follicle-stimulating hormone, HMG: human menopausal gonadotropin.

Fig. 8
Hormonal therapy before sperm retrieval in eugonadotropic NOA patient. NOA: non-obstructive azoospermia, HCG: human chorionic gonadotropin, FSH: follicle-stimulating hormone, HMG: human menopausal gonadotropin.

(2) Guidelines

The EAU guidelines conclude that routine hormonal therapy should not be recommended before cTESE/mTESE [7]. The AUA/ASRM guidelines recommend that patients with NOA should be advised that there is limited data to support hormone therapy prior to SSR [8].

(3) Discussion

The heterogeneity of answers in the survey with over 70% of respondents using hormonal therapy (Fig. 4, 5, 6) even when the guidelines don’t support its use may be explained by the lack of robust evidence with regard to the effectiveness of hormonal therapy in patients with NOA. A recent meta-analysis failed to demonstrate an improvement in SSR rate after pretreatment with hormonal therapy in men with NOA and hypergonadotropic hypogonadism, while some improvement in SSR rates was demonstrated in eugonadal men with NOA, but the authors stated that the literature had a moderate to severe risk of bias [9]. A prospective study reported that pure FSH treatment improved the success of TESE for eugonadal NOA patients with normal FSH levels and the best outcome was when the histopathological analysis revealed focal spermatogenesis or hypospermatogenesis [10]. Foresta et al [11] reported similar findings with improvement in patients with hypospermatogenesis.

(4) Expert recommendation

The quality of evidence supporting hormonal therapy before SSR in NOA patients is low. However, it may improve the SSR rates in some NOA patients. Given the limited and poor-quality evidence, it is not routinely recommended but may be considered after adequate counseling.

2) Exogenous testosterone and NOA

(1) Survey results

With regard to the management of NOA due to exogenous testosterone, there was considerable variation in the treatment strategies reported by the respondents with about one-third (32.5%) opting to discontinue exogenous testosterone and allow natural recovery, while the rest choosing to discontinue testosterone, concomitantly starting stimulation with either clomiphene or human chorionic gonadotropin (hCG) or clomiphene citrate plus hCG or hCG and HMG/FSH (Fig. 9).

Fig. 9
Advice for patient with NOA due to exogenous testosterone. NOA: non-obstructive azoospermia, HCG: human chorionic gonadotropin, FSH: follicle-stimulating hormone, HMG: human menopausal gonadotropin.

When asked about the use of exogenous testosterone in men with NOA and hypogonadism before undergoing TESE, surprisingly, 4.8% of respondents would still prescribe exogenous testosterone and another 9.6% would prescribe it sometimes. The majority of respondents (72.2%) would prescribe hCG/LH, selective estrogen receptor modulators (SERMs), or aromatase inhibitors (Fig. 10).

Fig. 10
Proportion of respondents who prescribe exogenous testosterone therapy when a man with NOA has a low testosterone level and is yet to undergo sperm retrieval. NOA: non-obstructive azoospermia, HCG: human chorionic gonadotropin, LH: luteinizing hormone, SERM: selective estrogen receptor modulators

(2) Guidelines

Neither the EAA nor the AUA/ASRM guidelines make recommendations for the management of men with NOA induced by exogenous testosterone. However, the EAU guidelines recommend withdrawal of exogenous testosterone as the initial treatment [7]. If there are no sperm in the semen by 6 to 12 months then hCG alone or in combination with FSH or clomiphene citrate can be prescribed [12].

However, there is poor quality of data supporting treatment algorithms in this scenario. The EAU guidelines recommend avoiding testosterone therapy for male infertility (strong level of recommendation) [7]. Similarly, a clinical principal recommendation from the AUA/ASRM guidelines states that “For the male interested in current or future fertility, testosterone monotherapy should not be prescribed.” [8].

(3) Discussion

Exogenous testosterone has been increasingly used worldwide, especially in the form of anabolic steroid use with approximately 3 million users in the USA alone and with estimates as high as 25% of young men who work out in gyms [13, 14]. Exogenous testosterone results in negative feedback on the hypothalamic-pituitary-gonadal axis leading to suppression of FSH and LH secretion resulting in a decrease in intratesticular androgen levels, thereby inhibiting spermatogenesis and resulting in NOA [15]. This negative effect on testicular function is expected to recover within 6 months of exogenous testosterone cessation. However, not all men will respond similarly. Only one-third of the patients will return to their baseline pre-treatment sperm density [16] and there remains a theoretical risk of permanent azoospermia in some men especially if they had poor pre-treatment spermatogenesis at baseline. There is no international consensus on the treatment regimen for NOA cases secondary to exogenous testosterone therapy. This is evident by the lack of recommendations from international societies. This is also reflected by the results of our survey, where there is no overwhelming agreement on one type of therapy. For infertile men with NOA who are planning to undergo SSR, exogenous testosterone therapy is contraindicated due to the pathophysiological effect of such treatment. Surprisingly, in our survey up to 15% of respondents would still use exogenous testosterone therapy in infertile men with NOA. A similar result was also reported by an earlier study surveying 387 urologists, where 25% of the respondents reported that they would treat infertile males with exogenous testosterone therapy while the couple actively pursues pregnancy [17].

(4) Expert recommendation

Exogenous testosterone should not be used for men with NOA who are still interested in testicular sperm retrieval and future fertility. Instead, SERMs, aromatase inhibitors, or hCG can be used to raise testosterone without compromising spermatogenesis.

3. Surgical management

1) Surgical therapy related to mTESE

(1) Survey result

Out of the 332 respondents, 14.5% indicated that they perform mTESE in <10% of NOA cases while 14.8% perform mTESE in 75% to 100% of cases (Fig. 11). The average SSR rate in men with NOA was reported to be 50.0% by 26%–50% of participants while 19% of the participants reported successful SSR in <10% of cases (Fig. 12). Approximately 2/3 of the respondents (68.1%) reported that most couples opted for mTESE to obtain their sperm, while 9% of the couples chose to have both options of own and donor sperm in the same cycle (Fig. 13).

Fig. 11
Frequency performing mTESE for NOA cases. mTESE: microdissection testicular sperm extraction, NOA: non-obstructive azoospermia.

Fig. 12
Overall surgical sperm retrieval rate in men with NOA. mTESE: microdissection testicular sperm extraction, NOA: non-obstructive azoospermia.

Fig. 13
General acceptability of mTESE in your practice. mTESE: microdissection testicular sperm extraction, OA: obstructive azoospermia, NOA: non-obstructive azoospermia

(2) Discussion

The proportion of surgeons performing mTESE is relatively low. Approximately one third of the respondents performed mTESE in <50% of their patients; however, a few surgeons perform mTESE in the majority of their patients with NOA. The average SSR rates are around 50% but some reported it to be as low as <10%. The wide range of SSR rates reported by the survey participants are probably due to the heterogenous nature of patients with NOA (age, location, testicular size, hormonal levels, etiology), and variations in surgeon experience and technique. The SSR rates will also be affected by the experience of the embryologists involved in finding sperm in mTESE specimens. Results of this survey also indicate that while most couples prefer to have their own biological children, some are open to donor sperm.

(3) Expert recommendation

mTESE is the most efficient procedure for sperm retrieval in men with NOA who wish to have biological children. The experience of the surgeon and the time spent by the embryologists can impact the success of mTESE.

2) Sperm retrieval in Klinefelter syndrome (KS) patients

(1) Survey result

Out of the 326 participants, 46.0% reported successful SSR in <10% of KS cases and another 30.4% reported success in only 10%–25% of cases, while 4.0% reported that they had successful SSR in >50% of these cases (Fig. 14).

Fig. 14
Sperm retrieval rate in patients with Klinefelter syndrome.

(2) Guidelines

Sperm production is variable in patients with KS and in those who are azoopsermic. TESE or mTESE can help recover sperm in up to 50% of cases [7, 8]. The EAA male infertility guideline does not specifically discuss sperm retrieval in patients with KS.

(3) Discussion

While the guidelines and published studies claim SSR rates of 40%–60% in men with KS, the majority of respondents (76.4%) reported SSR rates of less than 25%, with the majority (46.0%) claiming that they found sperm in <10% of their KS patients. Though it is tempting to attribute the higher success reported in published papers to superior technique and experience, the results of such a large number of respondents cannot be ignored. Perhaps, there are unrecognized geographical differences in testicular histopathology of KS. A comparison of results between centers of the same region, and between different regions, may throw light on this significant discrepancy and allow for better and more realistic patient counseling. The 2020 AUA/ASRM and EAU guidelines report that the rate of spermatozoa found upon mTESE in up to 50%–60% of 47, XXY men [7, 8] while other studies have reported SSR in 20%–60% of men with KS [18]. A study comparing rates of sperm retrieval between KS patients and other NOA patients with normal karyotype using skewed regression analysis showed a success rate of 28.4% for the KS group compared to 22.0% for the NOA with normal karyotype group. A statistically significant difference existed with higher testosterone levels in the successful SSR group [19]. The retrieval rates in KS patients are variable and testosterone levels could be an indicator for predicting success. Further studies are needed to provide more comprehensive data on this subject.

(4) Expert recommendation

The SSR rates reported in the literature in patients with KS undergoing mTESE range between 20%–60%. mTESE in patients with KS can be performed after explaining realistic success rates to patients. High volume mTESE centers may have better success in SSR in patients with KS.

3) Sperm retrieval: special considerations

(1) Survey result

The majority (80.2%) of the respondents have their laboratory team working in the operating room (37.7%) or in a nearby room in the same facility (42.5%) (Fig. 15). Only 32.9% of the respondents stated that their embryologist spent at least one hour looking for sperm before declaring the absence of sperm in the retrieved samples while the others reported that the time spent was less than 30 minutes (29.8%) or between 30 and 60 minutes (22.6%) (Fig. 16). In 59.8% of the centers performing mTESE, fresh sperm is preferred over cryopreserved sperm (Fig. 17).

Fig. 15
Working place of laboratory team during surgical sperm retrieval.

Fig. 16
Time needed by embryologist to look for sperm before declaring absence of sperm in retrieved samples at the time of mTESE. mTESE: microdissection testicular sperm extraction.

Fig. 17
Preference of sperm during mTESE for ICSI. mTESE: microdissection testicular sperm extraction, ICSI: intracytoplasmic sperm injection, IVF: in vitro fertilization.

(2) Discussion

There is no consensus guideline on this topic. Most centers have their laboratory team near the operating room or inside the same facility to identify sperm in the retrieved samples. The standard time for searching for sperm is around 60 minutes and may last up to 120 minutes before declaring it negative for sperm [8]. The results of this survey indicate that fresh sperm are preferred over cryopreserved sperm in most centers [20].

(3) Expert recommendation

It is preferable for the laboratory team to be in reasonable proximity to the operating theater to facilitate the transfer of mTESE specimens. The samples should be subject to meticulous scrutiny and examination by the embryologist for at least 60 minutes in an attempt to identify sperm. Both fresh and cryopreserved sperm can be used depending upon the expertise of the center and the embryologist.

4) Testicular biopsy for histopathology during surgical sperm retrieval

(1) Survey result

Nearly 40% of the 335 respondents would routinely perform a bilateral testicular biopsy for histopathology at the time of SSR, while 17.9% of the participants would perform it unilaterally routinely. A quarter of the participants (25.4%) would perform it in selected patients, and 10.7% of the clinicians would not perform a testicular biopsy for histopathology at the time of SSR under any circumstance (Fig. 18).

Fig. 18
Testicular biopsy for histopathology at the time of surgical sperm retrieval.

(2) Guidelines

Formal diagnostic biopsy is not recommended in this clinical setting by the EAU guidelines because patients with spermatogenic failure (e.g., Sertoli cell-only syndrome [SCOS]) may harbor focal areas of spermatogenesis. Neither the EAA, AUA/ASRM guidelines mention any specific role of testicular histopathology in defining the prognosis and success of sperm retrieval rates of TESE in NOA patients.

(3) Discussion

Sperm retrieval in patients with NOA is successful in 29.5% to 90.0% of cases. There is some evidence that shows a positive correlation between the histology found at testicular biopsy and the likelihood of finding mature sperm cells during testicular sperm retrieval. Historically, the presence of hypospermatogenesis at testicular biopsy showed a positive correlation in predicting successful sperm retrieval up to 93.3% after either single or multiple conventional TESEs or mTESE compared with early maturation arrest (13.3%), late maturation arrest pattern (66.7%) or SCOS (18.1%) [21]. However, it is impractical to obtain a testicular biopsy prior to the sperm retrieval procedure.

Biopsy is best done during the SSR procedure to look for the histopathological findings. This approach is considered to be more cost-effective, and the histopathological finding can help guide the clinician in a scenario when repeat testicular sperm retrieval is necessary [22]. In our survey, most of the respondents perform testicular biopsy for histopathology at the time of SSR. Although most guidelines do not recommend taking a biopsy to determine prognosis before the procedure, they do not offer any recommendation regarding taking a biopsy during the procedure. In this regard, sending a testicular biopsy for histopathology during SSR may be reasonable to determine the subsequent prognosis if no sperm is identified in the sample. Additionally, testicular biopsy for histopathological testing was also performed in men with risk factors for testicular malignancy such as cryptorchidism or intratesticular microlithiasis when no sperm are identified [23]. It is important to detect intratubular germ cell neoplasia in situ (GCNIS) which represents a high risk of development of testicular cancer [23]. There is a high percentage of testicular nodules and malignancies among azoospermic males with complete SCOS [24]. Vice versa, bilateral testicular tumor, testicular hypotrophy, higher tumor stage, GCNIS, smaller testes, SCOS, and history of undescended testicles can impact the spermatogenesis process and result in azoospermia [25].

(4) Expert recommendation

Sending a testicular biopsy during SSR may be considered as it can establish a histological diagnosis and prognosis for a subsequent sperm retrieval (if required) and may identify GCNIS. However, patients should be informed of the small added risk of a diagnostic testicular biopsy and the chance that this biopsy will contain spermatozoa despite a negative SSR.

5) Surgical therapy in men with genetic abnormalities

(1) Survey result

Y chromosome azoospermia factor c (AZFc) microdeletion (70.5%) and 47, XXY karyotype (67.3%) were the most chosen answers to indicate the need for SSR in patients with NOA. A significant percentage of respondents would offer SSR for men with AZFa (19.9%) and AZFb (23.2%) microdeletion, while only 8.3% of the participants would directly recommend donor sperm or adoption in the cases of genetic disorders (Fig. 19).

Fig. 19
Genetic conditions to perform surgical sperm retrieval in patients with NOA. mTESE: microdissection testicular sperm extraction, cTESE: conventional testicular sperm extraction, NOA: non-obstructive azoospermia, AZF: azoospermia factor.

(2) Guidelines

In the EAU guidelines, it is recommended that TESE, regardless of technique, should not be attempted in patients with complete deletions that include the AZFa and AZFb regions, since they indicate a very poor chance of successful sperm retrieval. Although non-specific recommendations are made, the AUA/ASRM guidelines also acknowledge that sperm have not been retrieved by TESE in men with complete AZFa and/or AZFb microdeletions, so surgical intervention is not indicated. Similarly, EAA guidelines also recommend against attempting sperm retrieval in cases of complete deletion of AZFa region, whereas in azoospermic carriers of deletions in AZFbc regions with proximal breakpoint in the P4 palindrome, a mTESE may be attempted. EAA guidelines also state that either standard or mTESE can be done in patients with KS.

(3) Discussion

Approximately 27.3% to 35.0% of men with NOA have genetic abnormalities [26, 27]. According to a recent study by Gao et al [28] in 2022, men with certain NOA etiologies such as KS, Y chromosome microdeletion, cryptorchidism, and mumps orchitis have a higher or lower rate of successful SSR than idiopathic NOA [28]. A clinical factor that has been associated with a better prognosis in azoospermic men (higher probability of successful sperm retrieval) is a history of cryptorchidism with prior orchidopexy. One study revealed a SSR rate of 21.4% to 71.4% for men with KS [28, 29]. For men with a Y chromosome microdeletion, sperm retrieval rates vary drastically depending on the site of the microdeletion.

Based on a systematic review of 32 studies, ranges as wide as 13% to 100% have been reported as the sperm retrieval rate among patients with AZFc microdeletion, with a mean of 47% [30]. Some men with AZFc microdeletions may even have low sperm counts in their ejaculate [30, 31]. On contrary, there have been no instances of sperm retrieval from men with complete AZFa or AZFb microdeletions [31]. Rare sperm retrieval in men with AZFb microdeletion has been reported if partial deletion is present [32]. Standard tests for these microdeletions may not specify if a partial or complete deletion is present. Another study showed unfavorable sperm retrieval rates, despite the use of mTESE, in NOA patients with chromosomal anomalies excluding those patients with KS [33]. The result of the survey showed that 70.5% and 67.3% of respondents recommend SSR in patients with NOA with an AZFc microdeletion and KS (47, XXY) respectively, which is consistent with the literature that reports a better sperm retrieval rate for patients in these categories. Interestingly, 19.9% and 23.2% of participants still perform sperm retrieval attempts in AZFa and AZFb microdeletion patients despite the current literature revealing only rare cases of successful sperm retrieval in men with deletions of these regions of the Y chromosome.

Several studies described the possibility of sperm cells identified in patients with partial deletion of AZFa and AZFb [34, 35]. Two cases of a non-classical, aberrant pattern of AZFb microdeletion, detected using extra Sequence-Tagged Sites markers in or around the AZFb region with some residual sperm production, have been reported [32]. In a study of 1,030 infertile men in Japan, the SSR rate of patients with gr/gr deletion (18.7%) was lower than those without gr/gr deletion, although not statistically significant. Further studies are needed to elucidate the effect of gr/gr deletion on SSR [36].

(4) Expert recommendation

The genetic status of a male has a significant impact on the success rate of SSR. There is a reasonable chance of finding sperm in men with KS and Y chromosome AZFc microdeletion. A complete deletion of AZFa and AZFb correlates with severe spermatogenesis impairment and sperm retrieval is not advised in these conditions. Sperm retrieval may be rarely successful in incomplete, aberrant, or non-classical AZFa and AZFb microdeletions. It is essential to offer proper counseling regarding the likelihood of sperm retrieval, the transmission of the AZF deletion to the male offspring, and the option for alternatives such as donor sperm and adoption.

6) Varicocele and NOA

(1) Survey result

In men with NOA and a large varicocele, 57.7% of the surgeons stated that they recommend microsurgical VR in most cases (Fig. 20). Factors favoring VR were smaller ipsilateral testis (35.1%), younger partners (34.2%), and FSH levels <10 IU/L (29.1%) (Fig. 21). In the presence of a genetic abnormality (such as AZFc deletion or 47, XXY), 36.8% of surgeons chose VR followed by sperm retrieval while an equal number (36.8%) of respondents chose to perform sperm retrieval only and would not do VR (Fig. 22). Most respondents chose to recommend VR (81.5%) for subclinical varicocele with NOA (Fig. 23). The proportion of sperm recovery in the ejaculate after VR was reported to be <10% by 38.1% and 10%–25% by 25.8% of respondents (Fig. 24).

Fig. 20
Microsurgical varicocelectomy in case of large varicocele associated with NOA. NOA: non-obstructive azoospermia.

Fig. 21
Conditions to recommend surgery for a varicocele associated with NOA. NOA: non-obstructive azoospermia, FSH: follicle-stimulating hormone.

Fig. 22
Treatment option for patient with genetic abnormality as well as clinical varicocele and NOA. AZF: azoospermia factor, NOA: non-obstructive azoospermia.

Fig. 23
Surgery for subclinical varicocele in NOA patient without other pathology present. NOA: non-obstructive azoospermia.

Fig. 24
Proportion of sperm recovery in the ejaculate of NOA men following varicocele repair. NOA: non-obstructive azoospermia.

(2) Guidelines

The AUA/ASRM guideline (statement 27) states, “couples should be informed of the absence of definitive evidence supporting VR prior to ART” [8]. The EAU guideline (10.4.3.3.3) states that VR in men with NOA can result in the appearance of sperm in the ejaculate (20.8% to 55.0%) and results in improved SSR (odd ratio: 2.65; 95% confidence interval: 1.69–4.14) [37]. However, it cautions that the evidence is based on observational studies and advises that “the risks and benefits of VR must be discussed fully with the patient with NOA and a clinically significant varicocele”. The current AUA/ASRM guidelines (statement 26) and EAU (10.4.3.3.2) guidelines are unequivocal in recommending against the repair of a subclinical varicocele [8, 37].

(3) Discussion

The management of varicocele in men with NOA is controversial which is reflected in the divergent practice patterns reported by the survey respondents in this study. A high proportion (57.7%) of the survey participants stated that they recommend microsurgical VR in infertile men presenting with NOA and a clinically palpable varicocele. Smaller size of the ipsilateral testis, normal FSH levels in serum, and younger age of female partners were considered by the surgeons as factors that favor the decision of VR in these patients. In a contemporary study beginning four months after VR surgery, 12/28 men (42.9%) had sperm in their ejaculates, with a mean sperm concentration of 1.2±3.6×106/mL at 24 months of follow-up. They reported two pregnancies following assisted reproductive technology (ART) treatment; however, there were no spontaneous pregnancies [38]. A recent prospective noncontrolled study, reported the recovery of motile sperm in the ejaculate of 10 of 31 (32.3%) men with NOA and clinically palpable varicoceles following subinguinal microsurgical varicocelectomy [39]. In the same study, there were greater chances of sperm recovery in cases of azoospermia with hypospermatogenesis and late spermatocyte arrest. Kim [38] further demonstrated the recovery of motile sperm in 43% of 28 men with NOA after microsurgical inguinal VR. In their series, 55% of men with hypospermatogenesis and 50% with late maturation arrest at the spermatid stage achieved recovery of sperm in ejaculate, whereas none of the patients with SCOS or early maturation arrest at the spermatocyte stage showed sperm recovery. Likewise, Esteves and Glina reported recovery of sperm in the ejaculate of 47% of men after VR [40]. Only men with hypospermatogenesis or maturation arrest patterns demonstrated improvement after surgery while all patients with SCOS continued to be azoospermic. Some studies have shown higher sperm retrieval during mTESE following prior correction of varicocele [41, 42]. Additionally, the results of a meta-analysis indicated a strong trend towards increased live birth rates following VR prior to ICSI (odd ratio=2.208, p=0.052) [43]. However, the quality of evidence on the outcome of VR in men with clinical varicocele and NOA is still poor, and a clinician should consider correcting a clinically significant varicocele only after optimal patient counselling that the quality of evidence is poor, and the outcome of varicocele correction may be the appearance of a few sperm in the ejaculate which may take up to 12 months and would still need ICSI. Surprisingly, the majority of respondents (81.5%) stated that they would perform VR in men with subclinical varicocele and NOA, though this is completely contrary to the guidelines and evidence in the literature, perhaps as an attempt to grasp any possibility in a desperate situation.

(4) Expert recommendation

Evidence supporting VR in men with NOA is limited. The decision to perform VR in cases of NOA is a shared decision between the physician and the couple after a detailed discussion of the risks and benefits. The decision may be guided by parameters such as testicular volume, FSH level, female partner’s age, testicular histology if available, and overall fertility status. VR for subclinical varicoceles is not recommended.

7) Techniques to optimize sperm retrieval.

(1) Survey result

Only a minority of the participants (5.4%) use ultrasound to locate the most vascularized areas of the testicular parenchyma for testicular biopsy in cases of NOA (Fig. 25). Additionally, most respondents (88.1%) reported not utilizing any innovative techniques during mTESE (Fig. 26).

Fig. 25
Imaging targeted testicular biopsies in NOA. NOA: non-obstructive azoospermia.

Fig. 26
Innovations to increase sperm retrieval rate during mTESE. mTESE: microdissection testicular sperm extraction.

Only about 5% routinely used fine needle aspiration (FNA) mapping prior to sperm retrieval, and 52.5% of respondents felt that it was not useful, with 20.4% of these stating that it may even be harmful in case of subsequent sperm retrieval (Fig. 27).

Fig. 27
Fine needle aspiration (FNA) mapping prior to sperm retrieval procedure.

(2) Guidelines

According to the AUA/ASRM guidelines, mTESE has a success rate that is twice as high as other techniques, including FNA.

(3) Discussion

Currently, there is no evidence to support the use of imaging during testicular biopsy nor the use of innovative techniques during mTESE to improve success rates [7]. However, the authors specified that shared decision-making between the physician and patient should determine which procedure to choose.

(4) GAF Expert recommendation

The evidence supporting the utility of diagnostic testicular FNA mapping prior to SSR is limited and the procedure may cause testicular damage. Hence, it is not routinely recommended. There is currently no evidence supporting the use of imaging techniques to improve the success of sperm retrieval.

8) Comparison of sperm retrieval techniques (TESA vs cTESE vs mTESE)

(1) Survey result

Of the 330 participants who answered a question regarding the use of cTESE versus mTESE for sperm retrieval in infertile men with NOA, 36 (10.9%) reported routinely beginning with a testicular sperm aspiration (TESA) in men with NOA. If the initial TESA failed to identify any usable sperm, then the participants perform cTESE or mTESE at a second session. Thirty-eight (11.5%) reported routinely performing cTESE first, followed by mTESE at the same session if the cTESE attempt fails to find sperm. One third of respondents (107, 32.4%) stated that they always proceed directly to mTESE (Fig. 28). A small testicular volume was an indication to directly perform mTESE for almost half the respondents (48.3%) (Fig. 29). Other conditions that led to primary mTESE included patients with very high FSH levels (38.5%), history of testicular insult or injury (19.0%), and KS (38.8%) (Fig. 29).

Fig. 28
Planning TESA vs cTESE vs mTESE in men with NOA. TESA: testicular sperm aspiration, cTESE: conventional testicular sperm extraction, mTESE: microdissection testicular sperm extraction, NOA: non-obstructive azoospermia.

Fig. 29
Clinical situations where mTESE is significantly superior to cTESE. mTESE: microdissection testicular sperm extraction, cTESE: conventional testicular sperm extraction, NOA: non-obstructive azoospermia, FSH: follicle-stimulating hormone.

(2) Guidelines

Most guidelines and expert opinions recommend mTESE as the initial choice when available, as it has been found to be superior to both cTESE [44] and TESA [45]. The AUA/ASRM guidelines support this recommendation, stating that mTESE should be initially considered for men with NOA undergoing sperm retrieval.

(3) Discussion

Surgical techniques such as TESA, cTESE, and mTESE are used to extract sperm in patients with NOA for ICSI purposes [44, 46]. The order of these techniques may vary depending on practice, providers, and settings. Multiple studies have shown that mTESE has a higher success rate compared to cTESE, which in turn has a higher success rate than TESA [46]. However, there is lack of evidence if a less invasive TESA should be attempted before cTESE or mTESE. FSH and testicular volume were suggested as predictive factors for successful TESE and TESA [47].

(4) GAF expert recommendation

mTESE is considered the preferred method for sperm extraction due to its overall higher SSR rate compared to other procedures such as TESA and cTESE. For some testicular histological patterns, cTESE and TESA may have acceptable sperm retrieval rates. Hence, it may be acceptable, in some cases, to perform a cTESE as the first step of a mTESE.

9) Timing of repeat surgical sperm retrieval

(1) Survey results

Out of 332 respondents, 79 (23.8%) reported waiting for 3 months and 110 (33.1%) reported waiting for 6 months before proceeding to mTESE after a failed cTESE in men with NOA. Waiting for longer periods was less common (19 respondents; 5.7%) (Fig. 30). In patients with successful mTESE, approximately one-third (119, 35.7%) waited for 6 months before repeating a second mTESE, while 18.9% waited for 3 months (Fig. 31). For patients with a failed initial mTESE, 44 (13.2%) reported repeating mTESE in 3 months, and 90 (27.7%) reported repeating it in 6 months. Interestingly, 85 respondents (25.5%) stated that they do not repeat mTESE following a failed first attempt (Fig. 32).

Fig. 30
Waiting period to perform mTESE after a failed cTESE in men with NOA. mTESE: microdissection testicular sperm extraction, cTESE: conventional testicular sperm extraction, NOA: non-obstructive azoospermia.

Fig. 31
Period of waiting to perform mTESE after successful mTESE in men with NOA. mTESE: microdissection testicular sperm extraction, NOA: non-obstructive azoospermia.

Fig. 32
Period of waiting to perform mTESE after failed mTESE. mTESE: microdissection testicular sperm extraction.

(2) Guidelines

AUA/ASRM, EAU, and EAA guidelines do not address an optimal timing for repeat TESE.

(3) Discussion

There is currently insufficient evidence to determine the ideal time for repeating a TESE, regardless of whether the initial yield is positive or negative. However, several studies have reported that the time interval between the first and second biopsies can significantly impact the success rate of the second sperm extraction procedure [48, 49, 50]. The literature commonly recommends waiting for a duration of 6 months between the initial and subsequent TESE procedures [51].

(4) GAF expert recommendation

A repeat mTESE can still be successful regardless of the outcome of the initial sperm retrieval procedure, although the chance of successful SSR during a repeat operation is higher if the original surgery found sperm. The recommended duration between the two procedures is 6 months. Waiting for 6 months after the first procedure may allow for the testicles to recover their function from the previous surgery.

10) mTESE miscellaneous questions

(1) Survey results

In men with symmetrical testes, approximately two-thirds (69.6%) of respondents would proceed with the contralateral testis with a 5% to 20% of finding sperm, while 8.1% had >20% expectation to find sperm in the opposite testis (Fig. 33). The cut off FSH value for positive sperm retrieval following mTESE was reported to be 12–19 IU/mL by 22.5% of participants, and 20–40 IU/mL by 27.8%, while 31.8% of respondents declared that there were no upper limits (Fig. 34). In men with KS (47, XXY) undergoing in vitro fertilization (IVF)/ICSI, 41.3% recommended preimplantation genetic testing (PGT) of embryos while 21.7% reported that it was not a routine practice (Fig. 35).

Fig. 33
Procedure for man with NOA and bilaterally symmetrical testes if no sperm found on one side during mTESE. NOA: non-obstructive azoospermia, mTESE: microdissection testicular sperm extraction.

Fig. 34
The highest FSH level to surgically obtain sperm in NOA. FSH: follicle-stimulating hormone, NOA: non-obstructive azoospermia.

Fig. 35
Preimplantation genetic testing of embryos in couples doing IVF/ICSI when sperm have been retrieved with mTESE in men with 47XXY. IVF: in vitro fertilization, ICSI: intracytoplasmic sperm injection, mTESE: microdissection testicular sperm extraction.

(2) Guidelines

The AUA/ASRM, EAU, and EAA guidelines do not offer definitive statements regarding proceeding with TESE in the contralateral testes with an initial negative result and PGT for KS patients. The guidelines do suggest that there is no cut-off value for FSH to proceed with mTESE.

(3) Discussion

In men with NOA with bilateral symmetrical testes, most surgeons prefer to perform mTESE on the second side if the initial search of one testis is negative [52]. Though there is no strict cut-off for FSH, most often the participants stated that mTESE could be successful when serum levels of FSH are less than 40 IU/mL [7, 46, 53]. While research has suggested that the risk of genetic abnormalities in the offspring of KS men is lower than previously believed [54], many fertility practitioners still consider PGT as a precautionary measure for safety reasons. Whenever feasible, it is advisable to undergo PGT to decrease the likelihood of transmitting the 47, XXY chromosome abnormality to offspring [55].

(4) GAF expert recommendation

With a negative mTESE on one side, mTESE can be attempted on the opposite side in bilateral symmetrical testes with a 10% chance of finding sperm on the second side. Though there is no defined cut-off value for FSH, higher success rates have been reported with FSH <40 IU/mL. In embryos formed from 47, XXY men, PGT can be offered whenever feasible, however, studies suggest that the majority of embryos from men with KS have no chromosomal abnormalities.

4. Future horizons

1) Treatment before sperm retrieval procedures

(1) Survey results

Stem cell therapy and platelet-rich plasma (PRP) are used by 5 (1.05%) and 3 (0.75%) responders, respectively. Most of the respondents (208, 62.5%) do not utilize any of the proposed techniques. The remaining respondents stated, “not applicable to my practice” (92, 27.6%) or “other” (19, 6.6%), the latter including acupuncture, antioxidants, and hormone therapy (Fig. 36).

Fig. 36
Treatment before sperm retrieval procedure in NOA patients. NOA: non-obstructive azoospermia.

(2) Guidelines

AUA/ASRM, EAU, and EAA guidelines include stem cells and PRP as experimental therapies. The EAA guideline does not discuss these.

(3) Discussion

The use of spermatogonial stem cell (SSC)-based therapy for fertility preservation is gaining attention as a promising alternative to overcome infertility caused by gonadotoxic treatments [56]. Cryopreserving SSC from pre-pubertal testicular tissue obtained after surgery is essential, although alternative adult tissue can be used provided that spermatogonial function is not compromised. There may also be a possibility for genome editing prior to transplantation in the future. Testicular (xeno) grafting has been shown to lead to successful spermatogenesis in testicular tissues of many mammalian species, including mice, pigs, and monkeys [57, 58, 59], with reported success rate of live offspring as 87% by the use of ICSI [57, 58, 60]. Despite the promising but challenging results, current international guidelines do not support such treatment and thus we are far from specific recommendations. The literature regarding the application of PRP for azoospermia is scarce. An experimental study from Khadivi et al [61] conducted on different two-dimensional (2D) and three-dimensional (3D) culture systems of SSCs demonstrated the occurrence of proliferating SSCs. The number and diameter of colonies in the PRP-2D group showed a considerable increase (p<0.01) as compared to the control group. In the PRP-scaffold group, a significant increase (p<0.01) was seen only in the number of colonies related to the control group.

(4) GAF expert recommendation

Currently, there is not enough evidence to recommend the routine use of stem cells and PRP in the treatment of NOA. These are evolving and promising therapies but currently their use should be restricted to experimental settings.

2) Use of testicular MRI prior to sperm retrieval

(1) Survey result

The majority of the respondents (285, 89.6%) consider MRI useful in clinical practice, whereas 43 (13.1%) consider it as an experimental instrument (Fig. 37).

Fig. 37
Use of testicular MRI prior to sperm retrieval procedures in patients with the majority of the respondents (285, 86.9%) consider MRI useful in clinical practice, whereas 43 (13.1%) consider it as an experimental instrument. MRI: magnetic resonance imaging, NOA: non-obstructive azoospermia.

(2) Guidelines

AUA/ASRM, EAU, and EAA guidelines do not address this.

(3) Discussion

Previous preliminary studies have shown that diffusion-weighted imaging can be useful in evaluating patients with NOA [62, 63]. Additionally, an increased apparent diffusion coefficient (ADC) has been observed in patients with histological patterns of maturation arrest and SCOS [64]. Tsili et al [65] reported that NOA with hypospermatogenesis had a lower 25th percentile of ADC compared to NOA with severe hypospermatogenesis and that the median ADC was the most significant metric (p=0.007) for predicting the presence of sperm.

(4) GAF expert recommendation

More evidence is needed before its use can be routinely recommended for identifying areas of spermatogenesis or potentially favorable patients for SSR.

3) Advancements over the next 10 years

(1) Survey result

We also provided a question on what the respondents thought the biggest advancement in the treatment of NOA over the next 10 years would be. Among the possibilities for future advancements, most of the participants answered gene therapy (189, 57.3%) or stem cells (223, 67.6%). Other advancements were PRP (48, 14.5%), advances in imaging studies (110, 33.3%), artificial sperm (62, 18.8%), 3D printing of testes or sperm (48, 14.5%), and artificial intelligence (88, 26.7%). A total of 11 subjects answered “other”, including whole exome sequencing, in vitro spermatogenesis, and genetic testing (Fig. 38).

Fig. 38
The biggest advancement in the treatment of NOA over the next 10 years. NOA: non-obstructive azoospermia.

(2) Guidelines

AUA/ASRM and EAU guidelines include stem cells and PRP as experimental therapies. The EAA guideline does not discuss future therapies.

(3) Discussion

Gene therapy represents the most innovative technology able to change clinical practice in the near future in patients with NOA. In particular, CRISPR/Cas9 is a technology for RNA-directed modification of target sequences by Cas proteins, consisting mainly of CRISPR clusters, leading sequences (leaders), repeating sequence regions (tracers), and a set of conserved CRISPR-associated genes (Cas genes) [66, 67]. In this context, gene therapy can be used to treat male infertility caused by genetic defects.

(4) GAF recommendation

Gene therapy with CRISPR/Cas9 may be able to cure NOA in selected patients. This is possible in theory, but future studies are needed to demonstrate its applicability.

5. Limitations

There are several limitations to the current survey. Firstly, the survey was distributed through a global group of experts with significant experience in NOA management, rather than to all male infertility or reproductive specialists, which may limit the generalizability of the findings. Furthermore, it was not possible to determine the total response rate due to the various ways in which the questionnaire was disseminated, including emails, direct communication, and professional society websites. As a result, the total number of invitations is unknown. In terms of demographics, there is a preponderance of responses from certain countries such as Italy, Turkey, and India, while other large countries such as Russia and Canada are noticeably under-represented. Finally, subgroup analysis based on specialization, practice setting, or years of practice was also limited due to the high heterogeneity of variables. The highlights of our global survey are summarized in a SWOT (strengths, weaknesses, opportunities, and threats). Using the SWOT method, the main advantages and limitations of our study are concisely displayed. We also provided opportunities for improvement in the future to treat men with NOA and posed threats which can limit advancements in NOA (Fig. 39).

Fig. 39
The strength weakness, opportunities, and threat (SWOT) analysis.

CONCLUSIONS

This global survey provides a valuable and comprehensive perspective on global practices related to the medical and surgical management of NOA. It represents the first global survey for NOA and addresses important issues for clinicians. The results demonstrate a diverse range of practices in the medical and surgical management of NOA and underscore the need for evidence-based international consensus guidelines to ensure the highest standard of care for all patients.

The management of NOA is complex and success is defined by the ability to achieve the appearance of few sperms in the ejaculate via medical or surgical management or via successful SSR. The present guidelines are not very clear in many aspects about the management of NOA. This survey underlines the heterogeneity in the current worldwide practices among clinicians treating NOA. Some of the practices are clearly not in line with society recommendations. There is great variation in the results of various SSR techniques among the respondents which needs to be evaluated critically to find out whether a bias is responsible for these variations or there is a geographical impact on the SSRs. There is an urgent need for evidence-based international consensus guidelines for treatment as well as reporting. Well-planned meetings and research would be a step in the right direction.

Supplementary Materials

Supplementary materials can be found via https://doi.org/10.5534/wjmh.230339.

Supplement File 1

Click here to view.(157K, pdf)

Supplement File 2

Click here to view.(715K, pdf)

Supplement File 3

Click here to view.(460K, pdf)

Notes

Conflict of Interest:The authors have nothing to disclose.

Funding:None.

Author Contribution:

  • Conceptualization: AA, RS.

  • Statistical analysis: AH.

  • Supervision: AS, RS.

  • Writing – original draft: RS, AS, WA, MM, IZ, PK, NT, NHVO, PV, AH, GS, MG, TH, TT, PB, EK, MA, RAG, VSK, RS, GIR, GMP, EC.

  • Writing – review & editing: all authors.

  • All authors have read and agreed to the published version of the manuscript.

Acknowledgements

We are thankful to Dr. Damayanthi Durairajanayagam (Malaysia) for her help with the scientific editing of our article and Ms. Daniela Delgadillo (Mexico) for her assistance with creating the figures and manuscript submission.

The authors are thankful to the following societies for promoting the online survey through the efforts of their members.

  • 1. AK Andrologie und Sexuelle Funktionsstörungen as part of the Österreichische Gesellschaft für Urologie und Andrologie (Germar-Michael Pinggera, MD, Austria).

  • 2. Algerian Association of Urology (Nazim Gherabi, MD, Algeria).

  • 3. Andrology Working Group, Society of Urologic Surgery in Turkey (Gökhan Çeker, MD, Turkey; Oğuzhan Kahraman, MD, Turkey; Erman Ceyhan, MD, Turkey).

  • 4. Egyptian Society for Sexual Medicine & Surgery (Ahmed El-Sakka, MD, Egypt).

  • 5. Egyptian Society of Andrology (Taymour Mostafa, MD, Egypt).

  • 6. Egyptian Urological Association (Ayman Rashed, MD, PhD and Alayman Hussein MD, PhD)

  • 7. Indonesian Urological Association (InaUA) and InaUA Section of Andrological Urology (Ponco Birowo, MD, PhD, Indonesia; Gede Wirya Kusuma Duarsa, MD, PhD, Indonesia; Ricky Adriansjah, MD, Indonesia; Widi Atmoko, MD, Indonesia).

  • 8. Italian Society of Andrology and Sexual Medicine (Aldo E. Calogero, MD, Italy).

  • 9. Italian Society of Human Reproduction (Carlo Trotta, MD, Italy; Giovanni M. Colpi, MD, Italy; Lucia Rocco, PhD, Italy).

  • 10. Italian Society of Urology (Gian Maria Busetto, MD, PhD, Italy).

  • 11. L ebanese Society of Urology (Mohamad Moussa, MD, Lebanon).

  • 12. Malaysian Society of Andrology and the Study of the Aging Male (Christopher Ho, MD, Malaysia; Kay Seong, NGOO, MD, Malaysia).

  • 13. Malaysian Urological Association (Teng Aik Ong, MD, Malaysia).

  • 14. Mediterranean Society for Reproductive Medicine (Hassan Sallam, MD, PhD, Egypt).

  • 15. Middle East Society for Sexual Medicine (Amr El Meliegy, MD, Egypt).

  • 16. Moroccan Association of Andrology (AMAN) (Imad Ziouziou, MD, Morrocco).

  • 17. Oman Urology Society (OUS) (Mohammed S. Al-Marhoon, MD, Oman).

  • 18. Romanian Association for Sexual Medicine (Catalina Zenoaga-Barbarosie, MSc, Romania).

  • 19. Saudi Andrology Group (Naif Alhathal, MD, Saudi Arabia).

  • 20. Society for Men's Health Singapore (King Chien; Joe Lee, MD, Singapore).

  • 21. Society of Egyptian Fellows and Members of the Royal College of Obstetricians and Gynecologists (Hassan Sallam MD, PhD, Egypt).

  • 22. Society of Urological Surgery in Turkey (SUST) (Murat Gul, MD, Turkey).

  • 23. Turkish Society of Andrology (TSA) (Ateş Kadıoğlu, MD, Turkey).

  • 24. Turkish Association of Urology (Arif Kalkanli, MD, Turkey; Ateş Kadıoğlu, MD, Turkey).

  • 25. Vietnamese Society for Sexual Medicine (Quang Nguyen, MD, PhD; Ho Vinh Phuoc Nguyen, MD; Tan V. Le, MD; Quang Tien Long Tran, MD).

References

    1. Jarow JP, Espeland MA, Lipshultz LI. Evaluation of the azoospermic patient. J Urol 1989;142:62–65.
    1. Mazzilli F, Rossi T, Delfino M, Sarandrea N, Dondero F. Azoospermia: incidence, and biochemical evaluation of seminal plasma by the differential pH method. Panminerva Med 2000;42:27–31.
    1. Wosnitzer M, Goldstein M, Hardy MP. Review of azoospermia. Spermatogenesis 2014;4:e28218
    1. Gudeloglu A, Parekattil SJ. Update in the evaluation of the azoospermic male. Clinics (Sao Paulo) 2013;68 Suppl 1:27–34.
    1. Eysenbach G. Improving the quality of Web surveys: the Checklist for Reporting Results of Internet E-Surveys (CHERRIES). J Med Internet Res 2004;6:e34
      Erratum in: doi: 10.2196/jmir.2042.
    1. Agarwal A, Saleh R, Boitrelle F, Cannarella R, Hamoda TAA, Durairajanayagam D, et al. The Global Andrology Forum (GAF): a world-wide, innovative, online initiative to bridge the gaps in research and clinical practice of male infertility and sexual health. World J Mens Health 2022;40:537–542.
    1. Salonia A, Bettocchi C, Capogrosso P, Carvalho J, Corona G, Hatzichristodoulou G, et al. EAU guidelines on sexual and reproductive health [Internet]. European Association of Urology; c2022 [cited 2023 Oct 17].
    1. Schlegel PN, Sigman M, Collura B, De Jonge CJ, Eisenberg ML, Lamb DJ, et al. Diagnosis and treatment of infertility in men: AUA/ASRM guideline PART II. J Urol 2021;205:44–51.
    1. Tharakan T, Corona G, Foran D, Salonia A, Sofikitis N, Giwercman A, et al. Does hormonal therapy improve sperm retrieval rates in men with non-obstructive azoospermia: a systematic review and meta-analysis. Hum Reprod Update 2022;28:609–628.
    1. Aydos K, Unlü C, Demirel LC, Evirgen O, Tolunay O. The effect of pure FSH administration in non-obstructive azoospermic men on testicular sperm retrieval. Eur J Obstet Gynecol Reprod Biol 2003;108:54–58.
    1. Foresta C, Bettella A, Ferlin A, Garolla A, Rossato M. Evidence for a stimulatory role of follicle-stimulating hormone on the spermatogonial population in adult males. Fertil Steril 1998;69:636–642.
    1. El Osta R, Almont T, Diligent C, Hubert N, Eschwège P, Hubert J. Anabolic steroids abuse and male infertility. Basic Clin Androl 2016;26:2
    1. Pope HG Jr, Wood RI, Rogol A, Nyberg F, Bowers L, Bhasin S. Adverse health consequences of performance-enhancing drugs: an Endocrine Society scientific statement. Endocr Rev 2014;35:341–375.
    1. Parkinson AB, Evans NA. Anabolic androgenic steroids: a survey of 500 users. Med Sci Sports Exerc 2006;38:644–651.
    1. McLachlan RI, O'Donnell L, Meachem SJ, Stanton PG, de K, Pratis K, et al. Hormonal regulation of spermatogenesis in primates and man: insights for development of the male hormonal contraceptive. J Androl 2002;23:149–162.
    1. World Health Organization Task Force on Methods for the Regulation of Male Fertility. Contraceptive efficacy of testosterone-induced azoospermia and oligozoospermia in normal men. Fertil Steril 1996;65:821–829.
      Erratum in: Fertil Steril 1996;65:1267.
    1. Ko EY, Siddiqi K, Brannigan RE, Sabanegh ES Jr. Empirical medical therapy for idiopathic male infertility: a survey of the American Urological Association. J Urol 2012;187:973–978.
    1. Caroppo E, Colpi GM. Update on the management of non-obstructive azoospermia: current evidence and unmet needs. J Clin Med 2021;11:62
    1. Chehrazi M, Rahimiforoushani A, Sabbaghian M, Nourijelyani K, Sadighi Gilani MA, Hoseini M, et al. Sperm retrieval in patients with Klinefelter syndrome: a skewed regression model analysis. Int J Fertil Steril 2017;11:117–122.
    1. Asanad K, Matthew Coward R, Mehta A, Smith JF, Vij SC, Nusbaum DJ, et al. factors influencing the decision for fresh vs cryopreserved microdissection testicular sperm extraction for non-obstructive azoospermia. Urology 2021;157:131–137.
    1. Ku MH, Huang IS, Lin AT, Chen KK, Huang WJ. The predictive value of parameters of clinical presentations for sperm yield in patients with nonobstructive azoospermia receiving microdissection testicular sperm extraction. Urol Sci 2017;28:243–247.
    1. Abdel Raheem A, Garaffa G, Rushwan N, De Luca F, Zacharakis E, Abdel Raheem T, et al. Testicular histopathology as a predictor of a positive sperm retrieval in men with non-obstructive azoospermia. BJU Int 2013;111:492–499.
    1. Dohle GR, Elzanaty S, van Casteren NJ. Testicular biopsy: clinical practice and interpretation. Asian J Androl 2012;14:88–93.
    1. Mancini M, Carmignani L, Gazzano G, Sagone P, Gadda F, Bosari S, et al. High prevalence of testicular cancer in azoospermic men without spermatogenesis. Hum Reprod 2007;22:1042–1046.
    1. Pang KH, Osman NI, Muneer A, Alnajjar HM. The relationship between testicular tumour characteristics and azoospermia: a systematic review. Int J Impot Res 2022;34:543–551.
    1. Pylyp LY, Spinenko LO, Verhoglyad NV, Zukin VD. Chromosomal abnormalities in patients with oligozoospermia and non-obstructive azoospermia. J Assist Reprod Genet 2013;30:729–732.
    1. Xie C, Chen X, Liu Y, Wu Z, Ping P. Multicenter study of genetic abnormalities associated with severe oligospermia and non-obstructive azoospermia. J Int Med Res 2018;46:107–114.
    1. Gao S, Yang X, Xiao X, Yin S, Guan Y, Chen J, et al. Outcomes and affecting factors for ICSI and microTESE treatments in nonobstructive azoospermia patients with different etiologies: a retrospective analysis. Front Endocrinol (Lausanne) 2022;13:1006208
    1. Boeri L, Palmisano F, Preto M, Sibona M, Capogrosso P, Franceschelli A, et al. Sperm retrieval rates in non-mosaic Klinefelter patients undergoing testicular sperm extraction: what expectations do we have in the real-life setting? Andrology 2020;8:680–687.
    1. Yuen W, Golin AP, Flannigan R, Schlegel PN. Histology and sperm retrieval among men with Y chromosome microdeletions. Transl Androl Urol 2021;10:1442–1456.
    1. Park SH, Lee HS, Choe JH, Lee JS, Seo JT. Success rate of microsurgical multiple testicular sperm extraction and sperm presence in the ejaculate in Korean men with y chromosome microdeletions. Korean J Urol 2013;54:536–540.
    1. Stouffs K, Vloeberghs V, Gheldof A, Tournaye H, Seneca S. Are AZFb deletions always incompatible with sperm production? Andrology 2017;5:691–694.
    1. Takeda T, Iwatsuki S, Hamakawa T, Mizuno K, Kamiya H, Umemoto Y, et al. Chromosomal anomalies and sperm retrieval outcomes of patients with non-obstructive azoospermia: a case series. Andrology 2017;5:473–476.
    1. Kleiman SE, Almog R, Yogev L, Hauser R, Lehavi O, Paz G, et al. Screening for partial AZFa microdeletions in the Y chromosome of infertile men: is it of clinical relevance? Fertil Steril 2012;98:43–47.
    1. Kleiman SE, Yogev L, Lehavi O, Hauser R, Botchan A, Paz G, et al. The likelihood of finding mature sperm cells in men with AZFb or AZFb-c deletions: six new cases and a review of the literature (1994-2010). Fertil Steril 2011;95:2005–2012. 2012.e1–2012.e4.
    1. Iijima M, Shigehara K, Igarashi H, Kyono K, Suzuki Y, Tsuji Y, et al. Y chromosome microdeletion screening using a new molecular diagnostic method in 1030 Japanese males with infertility. Asian J Androl 2020;22:368–371.
    1. Minhas S, Bettocchi C, Boeri L, Capogrosso P, Carvalho J, Cilesiz NC, et al. European Association of Urology guidelines on male sexual and reproductive health: 2021 update on male infertility. Eur Urol 2021;80:603–620.
    1. Kim ED, Leibman BB, Grinblat DM, Lipshultz LI. Varicocele repair improves semen parameters in azoospermic men with spermatogenic failure. J Urol 1999;162(3 Pt 1):737–740.
    1. Abdel-Meguid TA. Predictors of sperm recovery and azoospermia relapse in men with nonobstructive azoospermia after varicocele repair. J Urol 2012;187:222–226.
    1. Esteves SC, Glina S. Recovery of spermatogenesis after microsurgical subinguinal varicocele repair in azoospermic men based on testicular histology. Int Braz J Urol 2005;31:541–548.
    1. Inci K, Hascicek M, Kara O, Dikmen AV, Gürgan T, Ergen A. Sperm retrieval and intracytoplasmic sperm injection in men with nonobstructive azoospermia, and treated and untreated varicocele. J Urol 2009;182:1500–1505.
    1. Haydardedeoglu B, Turunc T, Kilicdag EB, Gul U, Bagis T. The effect of prior varicocelectomy in patients with nonobstructive azoospermia on intracytoplasmic sperm injection outcomes: a retrospective pilot study. Urology 2010;75:83–86.
    1. Kirby EW, Wiener LE, Rajanahally S, Crowell K, Coward RM. Undergoing varicocele repair before assisted reproduction improves pregnancy rate and live birth rate in azoospermic and oligospermic men with a varicocele: a systematic review and meta-analysis. Fertil Steril 2016;106:1338–1343.
    1. Colpi GM, Colpi EM, Piediferro G, Giacchetta D, Gazzano G, Castiglioni FM, et al. Microsurgical TESE versus conventional TESE for ICSI in non-obstructive azoospermia: a randomized controlled study. Reprod Biomed Online 2009;18:315–319.
    1. El-Haggar S, Mostafa T, Abdel Nasser T, Hany R, Abdel Hadi A. Fine needle aspiration vs. mTESE in non-obstructive azoospermia. Int J Androl 2008;31:595–601.
    1. Bernie AM, Mata DA, Ramasamy R, Schlegel PN. Comparison of microdissection testicular sperm extraction, conventional testicular sperm extraction, and testicular sperm aspiration for nonobstructive azoospermia: a systematic review and meta-analysis. Fertil Steril 2015;104:1099–1103.e1-3.
    1. Tsujimura A, Matsumiya K, Miyagawa Y, Takao T, Fujita K, Koga M, et al. Prediction of successful outcome of microdissection testicular sperm extraction in men with idiopathic nonobstructive azoospermia. J Urol 2004;172(5 Pt 1):1944–1947.
    1. Ghalayini IF, Alazab R, Halalsheh O, Al-Mohtaseb AH, Al-Ghazo MA. Repeated microdissection testicular sperm extraction in patients with non-obstructive azoospermia: outcome and predictive factors. Arab J Urol 2022;20:137–143.
    1. Ramasamy R, Ricci JA, Leung RA, Schlegel PN. Successful repeat microdissection testicular sperm extraction in men with nonobstructive azoospermia. J Urol 2011;185:1027–1031.
    1. Vernaeve V, Verheyen G, Goossens A, Van Steirteghem A, Devroey P, Tournaye H. How successful is repeat testicular sperm extraction in patients with azoospermia? Hum Reprod 2006;21:1551–1554.
    1. Schlegel PN, Su LM. Physiological consequences of testicular sperm extraction. Hum Reprod 1997;12:1688–1692.
    1. Alkandari MH, Bouhadana D, Zini A. Is a contralateral testicular exploration required at microdissection testicular sperm extraction for men with nonobstructive azoospermia, cryptozoospermia or severe oligozoospermia? Andrologia 2021;53:e14208
    1. Ramasamy R, Lin K, Gosden LV, Rosenwaks Z, Palermo GD, Schlegel PN. High serum FSH levels in men with nonobstructive azoospermia does not affect success of microdissection testicular sperm extraction. Fertil Steril 2009;92:590–593.
    1. Tong J, Zhao XM, Wan AR, Zhang T. PGT or ICSI? The impression of NGS-based PGT outcomes in nonmosaic Klinefelter syndrome. Asian J Androl 2021;23:621–626.
    1. Chen W, Bai MZ, Yang Y, Sun D, Wu S, Sun J, et al. ART strategies in Klinefelter syndrome. J Assist Reprod Genet 2020;37:2053–2079.
    1. Sanou I, van Maaren J, Eliveld J, Lei Q, Meißner A, de Melker AA, et al. Spermatogonial stem cell-based therapies: taking preclinical research to the next level. Front Endocrinol (Lausanne) 2022;13:850219
    1. Kaneko H, Kikuchi K, Men NT, Nakai M, Noguchi J, Kashiwazaki N, et al. Production of sperm from porcine fetal testicular tissue after cryopreservation and grafting into nude mice. Theriogenology 2017;91:154–162.
    1. Kaneko H, Kikuchi K, Nakai M, Somfai T, Noguchi J, Tanihara F, et al. Generation of live piglets for the first time using sperm retrieved from immature testicular tissue cryopreserved and grafted into nude mice. PLoS One 2013;8:e70989
    1. Honaramooz A, Li MW, Penedo MC, Meyers S, Dobrinski I. Accelerated maturation of primate testis by xenografting into mice. Biol Reprod 2004;70:1500–1503.
    1. Shinohara T, Inoue K, Ogonuki N, Kanatsu-Shinohara M, Miki H, Nakata K, et al. Birth of offspring following transplantation of cryopreserved immature testicular pieces and in-vitro microinsemination. Hum Reprod 2002;17:3039–3045.
    1. Khadivi F, Koruji M, Akbari M, Jabari A, Talebi A, Ashouri Movassagh S, et al. Application of platelet-rich plasma (PRP) improves self-renewal of human spermatogonial stem cells in two-dimensional and three-dimensional culture systems. Acta Histochem 2020;122:151627
    1. Han BH, Park SB, Seo JT, Chun YK. Usefulness of testicular volume, apparent diffusion coefficient, and normalized apparent diffusion coefficient in the MRI evaluation of infertile men with azoospermia. AJR Am J Roentgenol 2018;210:543–548.
    1. Wang H, Guan J, Lin J, Zhang Z, Li S, Guo Y, et al. Diffusion-weighted and magnetization transfer imaging in testicular spermatogenic function evaluation: preliminary results. J Magn Reson Imaging 2018;47:186–190.
    1. Ntorkou A, Tsili AC, Goussia A, Astrakas LG, Maliakas V, Sofikitis N, et al. Testicular apparent diffusion coefficient and magnetization transfer ratio: can these MRI parameters be used to predict successful sperm retrieval in nonobstructive azoospermia? AJR Am J Roentgenol 2019;213:610–618.
    1. Tsili AC, Astrakas LG, Goussia AC, Sofikitis N, Argyropoulou MI. Volumetric apparent diffusion coefficient histogram analysis of the testes in nonobstructive azoospermia: a noninvasive fingerprint of impaired spermatogenesis? Eur Radiol 2022;32:7522–7531.
    1. Liu G, Lin Q, Jin S, Gao C. The CRISPR-Cas toolbox and gene editing technologies. Mol Cell 2022;82:333–347.
    1. Wang HQ, Wang T, Gao F, Ren WZ. Application of CRISPR/Cas technology in spermatogenesis research and male infertility treatment. Genes (Basel) 2022;13:1000

Metrics
Share
Figures

1 / 39

Tables

1 / 2

ORCID IDs
PERMALINK