Skip to content
2000
image of Global Analysis of Regulatory Frameworks and Drug Safety Standards in the Drug Approval Process

Abstract

The drug approval and review process plays a crucial role in the pharmaceutical industry, aiming to ensure that newly marketed drugs are safe, effective, and of high quality. Regulatory authorities overseeing this process, tailored to geographically distinct needs, include the U.S. FDA, EMA, Japan’s Pharmaceuticals and Medical Devices Agency (PMDA), China’s National Medical Products Administration (NMPA), and India’s Central Drugs Standard Control Organization (CDSCO). This analysis offers insight into the various drug approval processes employed by these authorities and examines the International Council for Harmonisation’s ongoing efforts to establish a global consensus on drug regulation standards. It also compares regulatory pathways and highlights current harmonization initiatives. The focus is on analyzing operational aspects of drug regulation and identifying challenges arising from these regulations. The ultimate goal is to present a clear understanding of the intricacies and dynamics of the global drug approval process. Regulating the drug approval process is essential to ensure that new drugs are safe for public consumption, as the introduction of a new drug often faces numerous hurdles beyond safety and efficacy. The challenges highlighted include variations in regulations between authorities, the complexity of modern therapeutics, and the balance between safety and speed. This paper provides an overview of innovations in drug development, their impact on regulatory pathways, ongoing harmonization efforts, and potential obstacles that may arise during the regulatory process.

Loading

Article metrics loading...

/content/journals/cds/10.2174/0115748863392869250827042742
2025-09-04
2025-11-04
Loading full text...

Full text loading...

References

  1. Brown J.S. Mendelsohn A.B. Nam Y.H. The US food and drug administration sentinel system: A national resource for a learning health system. J. Am. Med. Inform. Assoc. 2022 29 12 2191 2200 10.1093/jamia/ocac153 36094070
    [Google Scholar]
  2. Smeltzer M.P. Reeves S.L. Cooper W.O. Common data model for sickle cell disease surveillance: Considerations and implications. JAMIA Open 2023 6 2 ooad036 10.1093/jamiaopen/ooad036 37252051
    [Google Scholar]
  3. Brown J.S. Maro J.C. Nguyen M. Ball R. Using and improving distributed data networks to generate actionable evidence: The case of real-world outcomes in the Food and Drug Administration’s Sentinel system. J. Am. Med. Inform. Assoc. 2020 27 5 793 797 10.1093/jamia/ocaa028 32279080
    [Google Scholar]
  4. Adimadhyam S. Barreto E.F. Cocoros N.M. Leveraging the capabilities of the FDA’s sentinel system to improve kidney care. J. Am. Soc. Nephrol. 2020 31 11 2506 2516 10.1681/ASN.2020040526 33077615
    [Google Scholar]
  5. Morrato E.H. Lennox L.A. Dearing J.W. The evolve to next-gen ACT network: An evolving open-access, real-world data resource primed for real-world evidence research across the clinical and translational science award consortium. J. Clin. Transl. Sci. 2023 7 1 224 10.1017/cts.2023.617 38028333
    [Google Scholar]
  6. Saesen R. Machado M. Crifo B. Involvement of the european medicines agency in multi-stakeholder regulatory science research projects: Experiences of staff members and project coordinators. Front. Med. 2023 10 1181702 10.3389/fmed.2023.1181702 37324145
    [Google Scholar]
  7. Salcher-Konrad M. Naci H. Davis C. Approval of cancer drugs with uncertain therapeutic value: A comparison of regulatory decisions in europe and the united states. Milbank Q. 2020 98 4 1219 1256 10.1111/1468‑0009.12476 33021339
    [Google Scholar]
  8. Murphy A. Bere N. Vamvakas S. Mavris M. The added value of patient engagement in early dialogue at ema: Scientific advice as a case study. Front. Med. 2022 8 811855 10.3389/fmed.2021.811855 35127766
    [Google Scholar]
  9. Banzi R. Bertele’ V. Demotes-Mainard J. Fostering EMA’s transparency policy. Eur. J. Intern. Med. 2014 25 8 681 684 10.1016/j.ejim.2014.07.012 25200801
    [Google Scholar]
  10. Obstacles to transparency over pharmacovigilance data within the EMA. Prescrire Int. 2015 24 165 278 279 26688911
    [Google Scholar]
  11. Lipsky M.S. Sharp L.K. From idea to market: The drug approval process. J. Am. Board Fam. Pract. 2001 14 5 362 367 11572541
    [Google Scholar]
  12. Spielmans G.I. Kirsch I. Drug approval and drug effectiveness. Annu. Rev. Clin. Psychol. 2014 10 1 741 766 10.1146/annurev‑clinpsy‑050212‑185533 24329178
    [Google Scholar]
  13. Boozalis E. Semenov Y.R. Kwatra S.G. Food and drug administration approval process for dermatology drugs in the United States. J. Dermatolog. Treat. 2018 29 6 536 538 10.1080/09546634.2018.1425361 29304565
    [Google Scholar]
  14. Dalwadi S.M. Hunt A. Bonnen M.D. Ghebre Y.T. Computational approaches for drug repurposing in oncology: Untapped opportunity for high value innovation. Front. Oncol. 2023 13 1198284 10.3389/fonc.2023.1198284 37274281
    [Google Scholar]
  15. Fatemi N. Karimpour M. Bahrami H. Current trends and future prospects of drug repositioning in gastrointestinal oncology. Front. Pharmacol. 2024 14 1329244 10.3389/fphar.2023.1329244 38239190
    [Google Scholar]
  16. Meng Y. Wang Y. Xu J. Drug repositioning based on weighted local information augmented graph neural network. Brief. Bioinform. 2023 25 1 bbad431 10.1093/bib/bbad431 38019732
    [Google Scholar]
  17. Harapanhalli R.S. Food and drug administration requirements for testing and approval of new radiopharmaceuticals. Semin. Nucl. Med. 2010 40 5 364 384 10.1053/j.semnuclmed.2010.05.002 20674596
    [Google Scholar]
  18. Lloyd R. Harris J. Wadhwa S. Chambers W. Food and drug administration approval process for ophthalmic drugs in the US. Curr. Opin. Ophthalmol. 2008 19 3 190 194 10.1097/ICU.0b013e3282f97fa1 18408492
    [Google Scholar]
  19. Liebowitz A. Spielman D.B. Schlosser R.J. Stewart M.G. Gudis D.A. Demographic disparities in the federal drug approval process for allergic rhinitis medications. Laryngoscope 2023 133 4 755 763 10.1002/lary.30129 35394648
    [Google Scholar]
  20. Wang B. Avorn J. Kesselheim A.S. Clinical and regulatory features of drugs not initially approved by the FDA. Clin. Pharmacol. Ther. 2013 94 6 670 677 10.1038/clpt.2013.165 23963252
    [Google Scholar]
  21. Menon J.M.L. Ritskes-Hoitinga M. Pound P. van Oort E. The impact of conducting preclinical systematic reviews on researchers and their research: A mixed method case study. PLoS One 2021 16 12 0260619 10.1371/journal.pone.0260619 34898637
    [Google Scholar]
  22. Kousholt B.S. Præstegaard K.F. Stone J.C. Reporting of 3Rs approaches in preclinical animal experimental studies—a nationwide study. Animals 2023 13 19 3005 10.3390/ani13193005 37835611
    [Google Scholar]
  23. Kousholt B.S. Præstegaard K.F. Stone J.C. Reporting quality in preclinical animal experimental research in 2009 and 2018: A nationwide systematic investigation. PLoS One 2022 17 11 0275962 10.1371/journal.pone.0275962 36327216
    [Google Scholar]
  24. Fosse V. Oldoni E. Bietrix F. Recommendations for robust and reproducible preclinical research in personalised medicine. BMC Med. 2023 21 1 14 10.1186/s12916‑022‑02719‑0 36617553
    [Google Scholar]
  25. Romantsik O. Bank M. Menon J.M.L. Malhotra A. Bruschettini M. Value of preclinical systematic reviews and meta-analyses in pediatric research. Pediatr. Res. 2024 96 3 643 653 10.1038/s41390‑024‑03197‑1 38615075
    [Google Scholar]
  26. Hall J. Dib N. Investigational new drug application (IND). J. Cardiovasc. Transl. Res. 2008 1 3 183 184 10.1007/s12265‑008‑9046‑9 20559913
    [Google Scholar]
  27. Southall N.T. Freedom of information act access to an investigational new drug application. ACS Pharmacol. Transl. Sci. 2019 2 6 497 500 10.1021/acsptsci.9b00056 32259081
    [Google Scholar]
  28. Krishnamurthy N. Grimshaw A.A. Axson S.A. Choe S.H. Miller J.E. Drug repurposing: A systematic review on root causes, barriers and facilitators. BMC Health Serv. Res. 2022 22 1 970 10.1186/s12913‑022‑08272‑z 35906687
    [Google Scholar]
  29. Gawai A.A. Shaikh F. Gadekar M. Deokar N. Kolhe S. Biyani K.R. A Review on: Phase ‘0’ Clinical Trials or Exploratory Investigational New Drug. Turk J Pharm Sci 2017 14 1 84 89 10.4274/tjps.63935 32454598
    [Google Scholar]
  30. Raj J.P. Gogtay N.J. Thatte U.M. A two-year evaluation of the minutes of Investigational New Drug committee meetings. Perspect. Clin. Res. 2021 12 4 199 202 10.4103/picr.PICR_83_19 34760647
    [Google Scholar]
  31. Cho N.S. Wong W.K. Nghiemphu P.L. Cloughesy T.F. Ellingson B.M. The future glioblastoma clinical trials landscape: Early phase 0, window of opportunity, and adaptive phase I–III studies. Curr. Oncol. Rep. 2023 25 9 1047 1055 10.1007/s11912‑023‑01433‑1 37402043
    [Google Scholar]
  32. Manning M.L. Thompson M.D. Saber H. Maher V.E. Crich J.Z. Leighton J.K. An FDA analysis of clinical hold deficiencies affecting investigational new drug applications for oncology products. Regul. Toxicol. Pharmacol. 2020 110 104511 10.1016/j.yrtph.2019.104511 31678263
    [Google Scholar]
  33. Davis S. Role of investigational new drug committees in propelling the drug development ecosystem in India. Perspect. Clin. Res. 2021 12 4 177 178 10.4103/picr.picr_190_21 34760642
    [Google Scholar]
  34. Dalakas M.C. Alexopoulos H. Spaeth P.J. Complement in neurological disorders and emerging complement-targeted therapeutics. Nat. Rev. Neurol. 2020 16 11 601 617 10.1038/s41582‑020‑0400‑0 33005040
    [Google Scholar]
  35. Borbély É. Simon M. Fuchs E. Wiborg O. Czéh B. Helyes Z. Novel drug developmental strategies for treatment‐resistant depression. Br. J. Pharmacol. 2022 179 6 1146 1186 10.1111/bph.15753 34822719
    [Google Scholar]
  36. Salminen W.F. Aloba O. Drew A. Marcinowicz A. Huang M.U.S. FDA 505(b)(2) NDA clinical, CMC and regulatory strategy concepts to expedite drug development. Drug Discov. Today 2023 28 7 103618 10.1016/j.drudis.2023.103618 37196760
    [Google Scholar]
  37. Crismon M.L. Walkow J. Sommi R.W. Drug development for new psychiatric drug therapies. Adv. Neurobiol. 2023 30 131 167 10.1007/978‑3‑031‑21054‑9_5 36928848
    [Google Scholar]
  38. Salminen W.F. Wiles M.E. Stevens R.E. Streamlining nonclinical drug development using the FDA 505(b)(2) new drug application regulatory pathway. Drug Discov. Today 2019 24 1 46 56 10.1016/j.drudis.2018.07.005 30041014
    [Google Scholar]
  39. Arondekar B. Duh M.S. Bhak R.H. Real-world evidence in support of oncology product registration: A systematic review of new drug application and biologics license application approvals from 2015–2020. Clin. Cancer Res. 2022 28 1 27 35 10.1158/1078‑0432.CCR‑21‑2639 34667027
    [Google Scholar]
  40. Purpura C.A. Garry E.M. Honig N. Case A. Rassen J.A. The role of real‐world evidence in fda‐approved new drug and biologics license applications. Clin. Pharmacol. Ther. 2022 111 1 135 144 10.1002/cpt.2474 34726771
    [Google Scholar]
  41. Huang Z. Fu Z. Wang J. Review on drug regulatory science promoting covid-19 vaccine development in china. Engineering 2022 10 127 132 10.1016/j.eng.2022.01.001 35096437
    [Google Scholar]
  42. Shi J. Chen X. Hu H. Ung C.O.L. Benchmarking drug regulatory systems for capacity building: An integrative review of tools, practice and recommendations. Int. J. Health Policy Manag. 2023 12 8100 10.34172/ijhpm.2023.8100 38618782
    [Google Scholar]
  43. Dri D.A. Massella M. Gramaglia D. Marianecci C. Petraglia S. Clinical Trials and Machine Learning: Regulatory Approach Review. Rev. Recent Clin. Trials 2021 16 4 341 350 10.2174/1574887116666210715114203 34269668
    [Google Scholar]
  44. Massella M. Dri D.A. Gramaglia D. Regulatory considerations on the use of machine learning based tools in clinical trials. Health Technol. 2022 12 6 1085 1096 10.1007/s12553‑022‑00708‑0 36373014
    [Google Scholar]
  45. Muensterman E.T. Luo Y. Parker J.M. Breakthrough therapy, PRIME and sakigake: A comparison between neuroscience and oncology in obtaining preferred regulatory status. Ther. Innov. Regul. Sci. 2020 54 3 658 666 10.1007/s43441‑019‑00100‑7 33301147
    [Google Scholar]
  46. Gough S. Post-marketing surveillance: A UK/European perspective. Curr. Med. Res. Opin. 2005 21 4 565 570 10.1185/030079905X41426 15899105
    [Google Scholar]
  47. Alomar M. Tawfiq A.M. Hassan N. Palaian S. Post marketing surveillance of suspected adverse drug reactions through spontaneous reporting: Current status, challenges and the future. Ther. Adv. Drug Saf. 2020 11 2042098620938595 10.1177/2042098620938595 32843958
    [Google Scholar]
  48. Gomase V.S. Pharmacovigilance - Technological advancements, recent developments and innovations. Curr. Drug Saf. 2025 20 4 423 449 10.2174/0115748863356840250112181406 39931995
    [Google Scholar]
  49. Gomes K.L.G. da Silva R.E. da Silva J.B. Bosio C.G.P. Novaes M.R.C.G. Post-marketing authorisation safety and efficacy surveillance of advanced therapy medicinal products in Brazil, the European Union, the United States and Japan. Cytotherapy 2023 25 10 1113 1123 10.1016/j.jcyt.2023.06.005 37436339
    [Google Scholar]
  50. Liang D. Sessa M. Post-marketing safety surveillance of erenumab: New insight from Eudravigilance. Expert Opin. Drug Saf. 2022 21 9 1205 1210 10.1080/14740338.2022.2049231 35236231
    [Google Scholar]
  51. Tsoi B. Masucci L. Campbell K. Drummond M. O’Reilly D. Goeree R. Harmonization of reimbursement and regulatory approval processes: A systematic review of international experiences. Expert Rev. Pharmacoecon. Outcomes Res. 2013 13 4 497 511 10.1586/14737167.2013.814962 23977976
    [Google Scholar]
  52. Niazi S.K. Al-Shaqha W.M. Mirza Z. Proposal of international council for harmonization (ich) guideline for the approval of biosimilars. J. Mark. Access Health Policy 2023 11 1 2147286 10.1080/20016689.2022.2147286 36419638
    [Google Scholar]
  53. Chen M.L. Shah V.P. Crommelin D.J. Harmonization of regulatory approaches for evaluating therapeutic equivalence and interchangeability of multisource drug products: Workshop summary report. Eur. J. Pharm. Sci. 2011 44 4 506 513 10.1016/j.ejps.2011.09.010 21946259
    [Google Scholar]
  54. Ofori-Asenso R. Hallgreen C.E. De Bruin M.L. Improving interactions between health technology assessment bodies and regulatory agencies: A systematic review and cross-sectional survey on processes, progress, outcomes, and challenges. Front. Med. 2020 7 582634 10.3389/fmed.2020.582634 33178721
    [Google Scholar]
  55. Qahtani M.A. Al-Jedai A. Wertheimer A. Factors that influence healthcare professionals’ intentions towards biosimilars. Innov. Pharm. 2024 15 1 10.24926/iip.v15i1.5922
    [Google Scholar]
  56. Broich K. Committee for medicinal products for human use (CHMP) assessment on efficacy of antidepressants. Eur. Neuropsychopharmacol. 2009 19 5 305 308 10.1016/j.euroneuro.2009.01.012 19269795
    [Google Scholar]
  57. Committee for medicinal products for human use (CHMP) guideline on the choice of the non‐inferiority margin. Stat. Med. 2006 25 10 1628 1638 10.1002/sim.2584 16639773
    [Google Scholar]
  58. Brown D. Volkers P. Day S. An introductory note to CHMP guidelines: Choice of the non‐inferiority margin and data monitoring committees. Stat. Med. 2006 25 10 1623 1627 10.1002/sim.2561 16639776
    [Google Scholar]
  59. An introductory note to the CHMP guidelines: Choice of the non‐inferiority margin and data monitoring committees by David Brown, Peter Volkers and Simon Day, Statistics in Medicine 2006; 25:1623–1627. Stat. Med. 2007 26 1 230 233 10.1002/sim.2665 16900566
    [Google Scholar]
  60. Hartford C.G. Petchel K.S. Mickail H. Pharmacovigilance during the pre-approval phases: An evolving pharmaceutical industry model in response to ICH E2E, CIOMS VI, FDA and EMEA/CHMP risk-management guidelines. Drug Saf. 2006 29 8 657 673 10.2165/00002018‑200629080‑00003 16872240
    [Google Scholar]
  61. Hojo T. Regulatory science in practice (pharmaceuticals and medical devices agency). Yakugaku Zasshi 2017 137 4 439 442 10.1248/yakushi.16‑00244‑4
    [Google Scholar]
  62. Fukaya-Shiba A. Shimokawa M. Sasaki H. Wakao R. Pharmaceuticals and medical devices agency’s horizon scanning and the science board: Cooperation toward extracellular vesicle‐based products. Br. J. Clin. Pharmacol. 2022 88 3 1392 1394 10.1111/bcp.15065 34528281
    [Google Scholar]
  63. Kondo E. Torii M. Oba I. Okamoto M. The pharmaceuticals and medical devices agency’s approach to facilitate risk communication and its challenges. Yakugaku Zasshi 2018 138 3 307 314 10.1248/yakushi.17‑00185‑2
    [Google Scholar]
  64. Masuyama K. Pharmaceuticals and medical devices agency (pmda)'s new action for pharmaceutical affairs consultation on research and development (r&d) strategy. Yakugaku Zasshi 2013 133 2 183 186 10.1248/yakushi.12‑00246‑1
    [Google Scholar]
  65. Kijima S. Yoshida S. Ochiai Y. Activity and perspective on quantitative modeling and simulation in Japan: Update from the Pharmaceuticals and Medical Devices Agency. CPT Pharma Syst Pharmacol 2022 11 12 1552 1555 10.1002/psp4.12868 36199197
    [Google Scholar]
  66. Zhong L. Li Y. Xiong L. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021 6 1 201 10.1038/s41392‑021‑00572‑w 34054126
    [Google Scholar]
  67. Cao Y. Yue X. Jia M. Wang J. Neuroinflammation and anti-inflammatory therapy for ischemic stroke. Heliyon 2023 9 7 17986 10.1016/j.heliyon.2023.e17986 37519706
    [Google Scholar]
  68. Shen X. Yin F. The mechanisms and clinical application of Traditional Chinese Medicine Lianhua-Qingwen capsule. Biomed. Pharmacother. 2021 142 111998 10.1016/j.biopha.2021.111998 34385103
    [Google Scholar]
  69. Shi G.M. Huang X.Y. Wu D. Toripalimab combined with lenvatinib and GEMOX is a promising regimen as first-line treatment for advanced intrahepatic cholangiocarcinoma: A single-center, single-arm, phase 2 study. Signal Transduct. Target. Ther. 2023 8 1 106 10.1038/s41392‑023‑01317‑7 36928584
    [Google Scholar]
  70. Zhou M. Chen X. Zhang H. China national medical products administration approval summary: Anlotinib for the treatment of advanced non‐small cell lung cancer after two lines of chemotherapy. Cancer Commun. 2019 39 1 1 10 10.1186/s40880‑019‑0383‑7 31221221
    [Google Scholar]
  71. Thatte U.M. Nishandar T.B. Birajdar A.R. Gogtay N.J. Current status of standardized, quality and ethical oversight of clinical research in the country: An audit of the Central Drugs Standard Control Organization (registration of ethics committees) and national accreditation board for hospital and healthcare providers (accreditation) databases. Perspect. Clin. Res. 2019 10 2 84 90 10.4103/picr.PICR_93_18 31008075
    [Google Scholar]
  72. Thangaraju P. Mahesh K. Venkatesan S. Information on migraine drugs in commonly available Indian drug information sources – Whether we satisfied the community needs? Indian J. Pharmacol. 2022 54 3 216 220 10.4103/ijp.ijp_783_20 35848693
    [Google Scholar]
  73. Venugopal B. Nagar A. Kumar Y. Sinha S. Overview of the national ethics committee registry for biomedical and health research in india: Stepping up to safeguard the ethical aspect of research involving human participants. Indian J. Pharmacol. 2023 55 4 251 256 10.4103/ijp.ijp_216_22 37737078
    [Google Scholar]
  74. Dakhale G.N. Kalikar M.V. Giradkar A.B. Impact of accreditation on registered ethics committees in terms of quality and governance in India: A cross-sectional study. Perspect. Clin. Res. 2024 15 2 80 88 10.4103/picr.picr_153_23 38765550
    [Google Scholar]
  75. Thatte U. Marathe P. Ethics committees in india: Past, present and future. Perspect. Clin. Res. 2017 8 1 22 30 10.4103/2229‑3485.198549 28194334
    [Google Scholar]
  76. Cooper C.J. Khan Mirzaei M. Nilsson A.S. Adapting drug approval pathways for bacteriophage-based therapeutics. Front. Microbiol. 2016 7 1209 10.3389/fmicb.2016.01209 27536293
    [Google Scholar]
  77. Cui L. Watanabe S. Miyanaga K. A comprehensive review on phage therapy and phage-based drug development. Antibiotics 2024 13 9 870 10.3390/antibiotics13090870 39335043
    [Google Scholar]
  78. Franco P. Jain R. Rosenkrands-Lange E. Hey C. Koban M.U. Regulatory pathways supporting expedited drug development and approval in ich member countries. Ther. Innov. Regul. Sci. 2023 57 3 484 514 10.1007/s43441‑022‑00480‑3 36463352
    [Google Scholar]
  79. Michaeli D.T. Michaeli T. Albers S. Boch T. Michaeli J.C. Special FDA designations for drug development: Orphan, fast track, accelerated approval, priority review, and breakthrough therapy. Eur. J. Health Econ. 2024 25 6 979 997 10.1007/s10198‑023‑01639‑x 37962724
    [Google Scholar]
  80. Jokura Y. Yano K. Yamato M. Comparison of the new Japanese legislation for expedited approval of regenerative medicine products with the existing systems in the USA and European Union. J. Tissue Eng. Regen. Med. 2018 12 2 e1056 e1062 10.1002/term.2428 28211195
    [Google Scholar]
  81. Scott E.C. Baines A.C. Gong Y. Trends in the approval of cancer therapies by the FDA in the twenty-first century. Nat. Rev. Drug Discov. 2023 22 8 625 640 10.1038/s41573‑023‑00723‑4 37344568
    [Google Scholar]
  82. Wang Y. Nan X. Duan Y. Wang Q. Liang Z. Yin H. FDA-approved small molecule kinase inhibitors for cancer treatment (2001–2015): Medical indication, structural optimization, and binding mode Part I. Bioorg. Med. Chem. 2024 111 117870 10.1016/j.bmc.2024.117870 39128361
    [Google Scholar]
  83. Harrigan J.A. Jacq X. Martin N.M. Jackson S.P. Deubiquitylating enzymes and drug discovery: Emerging opportunities. Nat. Rev. Drug Discov. 2018 17 1 57 78 10.1038/nrd.2017.152 28959952
    [Google Scholar]
  84. Abouelleil M. Deshpande N. Ali R. Emerging trends in neuromodulation for treatment of drug-resistant epilepsy. Front Pain Res 2022 3 839463 10.3389/fpain.2022.839463 35386582
    [Google Scholar]
  85. Liu Q. Ahadpour M. Rocca M. Huang S.M. Clinical pharmacology regulatory sciences in drug development and precision medicine: Current status and emerging trends. AAPS J. 2021 23 3 54 10.1208/s12248‑021‑00563‑3 33846878
    [Google Scholar]
  86. Roessner V. Eichele H. Stern J.S. European clinical guidelines for Tourette syndrome and other tic disorders—version 2.0. Part III: Pharmacological treatment. Eur. Child Adolesc. Psychiatry 2022 31 3 425 441 10.1007/s00787‑021‑01899‑z 34757514
    [Google Scholar]
  87. Raghu G. Ghazipura M. Fleming T.R. Meaningful endpoints for idiopathic pulmonary fibrosis (IPF) clinical trials: Emphasis on ‘feels, functions, survives’. report of a collaborative discussion in a symposium with direct engagement from representatives of patients, investigators, the national institutes of health, a patient advocacy organization, and a regulatory agency. Am. J. Respir. Crit. Care Med. 2024 209 6 647 669 10.1164/rccm.202312‑2213SO 38174955
    [Google Scholar]
  88. Shen J. Swift B. Mamelok R. Pine S. Sinclair J. Attar M. Design and conduct considerations for first‐in‐human trials. Clin. Transl. Sci. 2019 12 1 6 19 10.1111/cts.12582 30048046
    [Google Scholar]
  89. Vella Bonanno P. Ermisch M. Godman B. Adaptive pathways: Possible next steps for payers in preparation for their potential implementation. Front. Pharmacol. 2017 8 497 10.3389/fphar.2017.00497 28878667
    [Google Scholar]
  90. Kållberg C. Mathiesen L. Gopinathan U. Salvesen Blix H. The role of drug regulatory authorities and health technology assessment agencies in shaping incentives for antibiotic R&D: A qualitative study. J. Pharm. Policy Pract. 2023 16 1 53 10.1186/s40545‑023‑00556‑x 36973761
    [Google Scholar]
  91. Copley-Merriman K. Rare diseases: Addressing the challenges in diagnosis, drug approval, and patient access. Value Health 2018 21 5 491 492 10.1016/j.jval.2018.03.009 29753343
    [Google Scholar]
  92. Puccetti M. Pariano M. Schoubben A. Giovagnoli S. Ricci M. Biologics, theranostics, and personalized medicine in drug delivery systems. Pharmacol. Res. 2024 201 107086 10.1016/j.phrs.2024.107086 38295917
    [Google Scholar]
  93. Gupte T. Nitave T. Gobburu J. Regulatory landscape of accelerated approval pathways for medical devices in the United States and the European Union. Front Med Technol 2025 7 1586070 10.3389/fmedt.2025.1586070 40444060
    [Google Scholar]
  94. Kaur S. Yadav S. Sahu V. Sharma N. Shukla V.K. Biosimilar Regulations: Current Framework and Future Prospects. Curr. Drug Saf. 2025 20 10.2174/0115748863360017250509063745 40417752
    [Google Scholar]
  95. Li J. Wang H. Hua Y. Progress and challenges of the new conditional approval process in china: A pooled analysis from 2018 to 2021. Clin. Ther. 2023 45 11 1111 1118 10.1016/j.clinthera.2023.09.006 37806812
    [Google Scholar]
  96. Gomase V.S. Dhamane S.P. Jagdale S.C. Intellectual property rights effects on india’s pharmaceutical industry. Recent Pat. Biotechnol. 2025 19 10.2174/0118722083344893250411032817 40264308
    [Google Scholar]
  97. Gomase V. Kemkar K. Potnis V. Intellectual property rights: Protection of biotechnological inventions in india. Recent Pat. Biotechnol. 2024 18 2 128 143 10.2174/1872208317666230612145600 38282443
    [Google Scholar]
  98. Runge V.M. Dechelation (Transmetalation). Invest. Radiol. 2018 53 10 571 578 10.1097/RLI.0000000000000507 30130320
    [Google Scholar]
  99. Sudsakorn S. Bahadduri P. Fretland J. Lu C. 2020 FDA drug-drug interaction guidance: A comparison analysis and action plan by pharmaceutical industrial scientists. Curr. Drug Metab. 2020 21 6 403 426 10.2174/1389200221666200620210522 32562522
    [Google Scholar]
  100. Gomase V.S. Visokar D.L. Chopade V. Effective integration of artificial intelligence and blockchain technologies for empowerment of drug discovery and development. Curr. Drug Discov. Technol. 2025 22 10.2174/0115701638361312250402125556 40264319
    [Google Scholar]
  101. Alipour S. Nadimi Parashkouhi S. Mojahedian M. Abbasian H. Assessing drug lag in new drug approvals by the Iran Food and Drug Administration compared to the U.S. FDA, EMA, and PMDA: A 20-year analysis (2001–2021). Medicine 2024 103 25 38142 10.1097/MD.0000000000038142 38905367
    [Google Scholar]
  102. Alqahtani S. Seoane-Vazquez E. Rodriguez-Monguio R. Eguale T. Priority review drugs approved by the FDA and the EMA: Time for international regulatory harmonization of pharmaceuticals? Pharmacoepidemiol. Drug Saf. 2015 24 7 709 715 10.1002/pds.3793 26013294
    [Google Scholar]
  103. Roth J.A. Rahshenas M. Nowacki G. A descriptive analysis of real-world oncology biosimilar use in Japan. Future Oncol. 2024 20 25 1837 1850 10.1080/14796694.2024.2352405 38864611
    [Google Scholar]
/content/journals/cds/10.2174/0115748863392869250827042742
Loading
/content/journals/cds/10.2174/0115748863392869250827042742
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test