1
|
Feng Y. Letter to the editor regarding: "Understanding delays to parathyroidectomy: A mixed-methods approach". Surgery 2025; 178:108851. [PMID: 39414469 DOI: 10.1016/j.surg.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024]
Affiliation(s)
- Yingying Feng
- Blood Purification Center, Shaoxing Second Hospital, Shaoxing, Zhejiang, China.
| |
Collapse
|
2
|
Barghout SH, Meti N, Chotai S, Kim CJH, Patel D, Brown MC, Hueniken K, Zhan LJ, Raptis S, Al-Agha F, Deutschman C, Grant B, Pienkowski M, Moriarty P, de Almeida J, Goldstein DP, Bratman SV, Shepherd FA, Tsao MS, Freedman AN, Xu W, Liu G. Adaptive Universal Principles for Real-world Observational Studies (AUPROS): an approach to designing real-world observational studies for clinical, epidemiologic, and precision oncology research. Br J Cancer 2025; 132:139-153. [PMID: 39572762 DOI: 10.1038/s41416-024-02899-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 01/22/2025] Open
Abstract
The field of precision oncology has witnessed several advances that stimulated the development of new clinical trial designs and the emergence of real-world data (RWD) as an important resource for evidence generation in healthcare decision-making. Here, we highlight our experience with an innovative approach to a set of Adaptive, Universal Principles for Real-world Observational Studies (AUPROS). To demonstrate the utility of these principles, we used a mixed-methods approach to assess three studies that follow AUPROS at Princess Margaret Cancer Centre: (1) Molecular Epidemiology of ThorAcic Lesions (METAL), (2) Translational Head And NecK Study (THANKS), and (3) CAnadian CAncers With Rare Molecular Alterations (CARMA; NCT04151342). We performed resource assessments, stakeholder-directed surveys and discussions, analysis of funding, research output, collaborations, and a Strengths-Weaknesses-Opportunities-Threats (SWOT) analysis. Based on these analyses, AUPROS is an approach that is applicable to a wide range of observational study designs. The universality of AUPROS allows for multi-purpose analyses of various RWD, and the adaptive nature creates opportunities for multi-source funding and collaborations. Following AUPROS can offer cost and logistical benefits and may lead to increased research productivity. Several challenges were identified pertinent to ethics approvals, sustainability, complex coordination, and data quality that require local adaptation of these principles.
Collapse
Affiliation(s)
- Samir H Barghout
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nicholas Meti
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Simren Chotai
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Royal College of Surgeons, Dublin, Ireland
| | - Christina J H Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of General Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Devalben Patel
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - M Catherine Brown
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Katrina Hueniken
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Luna J Zhan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Stavroula Raptis
- Applied Health Research Centre, Unity Health, Toronto, ON, Canada
| | - Faisal Al-Agha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Benjamin Grant
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Martha Pienkowski
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - John de Almeida
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Otolaryngology-Head and Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David P Goldstein
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Otolaryngology-Head and Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Frances A Shepherd
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ming S Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrew N Freedman
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Wei Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Blacklock KLB, Donnelly K, Lu Y, Pozo JD, Glendinning L, Polton G, Selmic L, Tanis JB, Killick D, Parys M, Morris JS, Breathnach I, Zago S, Gould SM, Shaw DJ, Tivers MS, Malucelli D, Marques A, Purzycka K, Cantatore M, Mathers ME, Stares M, Meynert A, Patton EE. Oronasal mucosal melanoma is defined by two transcriptional subtypes in humans and dogs with implications for diagnosis and therapy. J Pathol 2025. [PMID: 39828982 DOI: 10.1002/path.6377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/31/2024] [Indexed: 01/22/2025]
Abstract
Mucosal melanoma is a rare melanoma subtype associated with a poor prognosis and limited existing therapeutic interventions, in part due to a lack of actionable targets and translational animal models for preclinical trials. Comprehensive data on this tumour type are scarce, and existing data often overlooks the importance of the anatomical site of origin. We evaluated human and canine oronasal mucosal melanoma (OMM) to determine whether the common canine disease could inform the rare human equivalent. Using a human and canine primary OMM cohort of treatment-naive archival tissue, alongside clinicopathological data, we obtained transcriptomic, immunohistochemical, and microbiome data from both species. We defined the transcriptomic landscape in both species and linked our findings to immunohistochemical, microbiome, and clinical data. Human and dog OMM stratified into two distinctive transcriptional groups, which we defined using a species-independent 41-gene signature. These two subgroups are termed CTLA4-high and MET-high and indicate actionable targets for OMM patients. To guide clinical decision-making, we developed immunohistochemical diagnostic tools that distinguish between transcriptomic subgroups. We found that OMM had conserved transcriptomic subtypes and biological similarity between human and canine OMM, with significant implications for patient classification, treatment, and clinical trial design. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kelly L Bowlt Blacklock
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Kevin Donnelly
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Yuting Lu
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jorge Del Pozo
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, Edinburgh, UK
| | - Laura Glendinning
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, Edinburgh, UK
| | - Gerry Polton
- North Downs Specialist Referrals, Bletchingley, UK
| | - Laura Selmic
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - Jean-Benoit Tanis
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Neston, UK
| | - David Killick
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological Science, University of Liverpool, Neston, UK
| | - Maciej Parys
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, Edinburgh, UK
| | | | | | - Stefano Zago
- The Ralph Veterinary Referral Centre, Marlow, UK
| | | | - Darren J Shaw
- Royal (Dick) School of Veterinary Studies and the Roslin Institute, Edinburgh, UK
| | - Michael S Tivers
- Paragon Veterinary Referrals, Paragon Point, Red Hall Crescent, Wakefield, UK
| | - Davide Malucelli
- Paragon Veterinary Referrals, Paragon Point, Red Hall Crescent, Wakefield, UK
| | | | - Katarzyna Purzycka
- Anderson Moores Veterinary Specialists, The Granary, Bunstead Barns, Hampshire, UK
| | - Matteo Cantatore
- Anderson Moores Veterinary Specialists, The Granary, Bunstead Barns, Hampshire, UK
| | - Marie E Mathers
- Department of Pathology, Western General Hospital, Edinburgh, UK
| | - Mark Stares
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Centre, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Alison Meynert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - E Elizabeth Patton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research, CRUK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Mapendano CK, Nøhr AK, Sønderkær M, Pagh A, Carus A, Lörincz T, Haslund CA, Poulsen LØ, Ernst A, Bødker JS, Dahl SC, Sunde L, Brügmann AH, Vesteghem C, Pedersen IS, Ladekarl M. Longer survival with precision medicine in late-stage cancer patients. ESMO Open 2025; 10:104089. [PMID: 39754975 DOI: 10.1016/j.esmoop.2024.104089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND In a per-protocol analysis of molecularly profiled patients with treatment-refractory, end-stage cancer discussed at the National Molecular Tumor Board (NMTB), we aimed to assess the overall survival (OS) outcome of targeted treatment compared with no targeted treatment. MATERIALS AND METHODS Patients were prospectively included at a single oncological center. Whole exome and RNA sequencing (tumor-normal) were carried out, and cases were presented at the NMTB for discussion of targeted treatment. Treatment was available through a basket trial, by compassionate use or in early clinical trials. RESULTS One hundred and ninety-six patients were included from 2020 to 2023. In all but three patients a driver variant was disclosed, while 42% had simultaneous affection of more than three oncogenic pathways. In 42% of patients a druggable target was identified but two-thirds did not receive the suggested treatment. The fraction of patients initiating treatment yearly rose from 8% to 22%. For patients treated (N = 30), the clinical benefit rate was 44% and median time on treatment was 3.5 months. Druggable targets were enriched in lung cancers, while patients receiving or not receiving targeted treatment had similar clinical characteristics. The median OS was longer for patients receiving targeted treatment (15 months), but similar for patients with no druggable target and suggested targeted treatment not initiated (5 and 6 months, respectively) (P = 0.004). In multivariate analysis, targeted treatment (hazard ratio 0.43, confidence interval 0.25-0.72), few metastatic sites, and adenocarcinoma histology were predictive of improved OS while alterations of the RTK/RAS pathway were prognostically unfavorable. CONCLUSIONS Tissue-agnostic targeted treatment based on molecular tumor profiling is possible in an increasing fraction of end-stage cancer patients. In those who receive targeted treatment, results strongly suggest a significant survival benefit.
Collapse
Affiliation(s)
- C K Mapendano
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A K Nøhr
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | - M Sønderkær
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - A Pagh
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A Carus
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - T Lörincz
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - C A Haslund
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - L Ø Poulsen
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - A Ernst
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - J S Bødker
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - S C Dahl
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | - L Sunde
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Clinical Genetics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A H Brügmann
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - C Vesteghem
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - I S Pedersen
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - M Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
5
|
Zhu W, Wu J, Lai W, Li F, Zeng H, Li X, Su H, Liu B, Zhao X, Zou C, Xiao H, Luo Y. Harnessing machine learning and multi-omics to explore tumor evolutionary characteristics and the role of AMOTL1 in prostate cancer. Int J Biol Macromol 2025; 286:138402. [PMID: 39643184 DOI: 10.1016/j.ijbiomac.2024.138402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Although recent advancements have shed light on the crucial role of coordinated evolution among cell subpopulations in influencing disease progression, the full potential of these insights has not yet been fully harnessed in the clinical application of personalized precision medicine for prostate cancer (PCa). In this study, we utilized single-cell sequencing to identify the evolutionary characteristics of tumoral cell states and employed comprehensive bulk RNA sequencing to evaluate their potential as prognostic indicators and therapeutic targets. Leveraging advancements in artificial intelligence, we integrated machine learning with multi-omics to develop and validate the tumor evolutionary characteristic predictive indicator (TECPI). TECPI not only demonstrated superior prognostic performance compared to traditional clinical predictors and 81 previously published models but also improved patient outcomes by accurately identifying individuals who would benefit from immunotherapy and targeted therapies. Furthermore, we experimentally validated the critical role of AMOTL1 in PCa pharmacodynamics through its interaction with AR, pivotal for modulating the sensitivity to AR antagonist. Additionally, we demonstrated the generalizability and applicability of TECPI across pan-cancers. In summary, this study emphasizes the importance of understanding cellular diversity and dynamics within the tumor microenvironment to predict PCa progression and to guide targeted therapy effectively.
Collapse
Affiliation(s)
- Weian Zhu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Jianjie Wu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Wenjie Lai
- Department of Urology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510317, Guangdong, China
| | - Fengao Li
- Department of Urology, Shaoxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Shaoxing 312000, Zhejiang, China
| | - Hengda Zeng
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Xiaoyang Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Huabin Su
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Bohao Liu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Xiao Zhao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Chen Zou
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, Guangdong, China.
| |
Collapse
|
6
|
Ahuja S, Zaheer S. Advancements in pathology: Digital transformation, precision medicine, and beyond. J Pathol Inform 2025; 16:100408. [DOI: 10.1016/j.jpi.2024.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
|
7
|
Jacobs JJL, Beekers I, Verkouter I, Richards LB, Vegelien A, Bloemsma LD, Bongaerts VAMC, Cloos J, Erkens F, Gradowska P, Hort S, Hudecek M, Juan M, Maitland-van der Zee AH, Navarro-Velázquez S, Ngai LL, Rafiq QA, Sanges C, Tettero J, van Os HJA, Vos RC, de Wit Y, van Dijk S. A data management system for precision medicine. PLOS DIGITAL HEALTH 2025; 4:e0000464. [PMID: 39787064 PMCID: PMC11717228 DOI: 10.1371/journal.pdig.0000464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 01/12/2025]
Abstract
Precision, or personalised medicine has advanced requirements for medical data management systems (MedDMSs). MedDMS for precision medicine should be able to process hundreds of parameters from multiple sites, be adaptable while remaining in sync at multiple locations, real-time syncing to analytics and be compliant with international privacy legislation. This paper describes the LogiqSuite software solution, aimed to support a precision medicine solution at the patient care (LogiqCare), research (LogiqScience) and data science (LogiqAnalytics) level. LogiqSuite is certified and compliant with international medical data and privacy legislations. This paper evaluates a MedDMS in five types of use cases for precision medicine, ranging from data collection to algorithm development and from implementation to integration with real-world data. The MedDMS is evaluated in seven precision medicine data science projects in prehospital triage, cardiovascular disease, pulmonology, and oncology. The P4O2 consortium uses the MedDMS as an electronic case report form (eCRF) that allows real-time data management and analytics in long covid and pulmonary diseases. In an acute myeloid leukaemia, study data from different sources were integrated to facilitate easy descriptive analytics for various research questions. In the AIDPATH project, LogiqCare is used to process patient data, while LogiqScience is used for pseudonymous CAR-T cell production for cancer treatment. In both these oncological projects the data in LogiqAnalytics is also used to facilitate machine learning to develop new prediction models for clinical-decision support (CDS). The MedDMS is also evaluated for real-time recording of CDS data from U-Prevent for cardiovascular risk management and from the Stroke Triage App for prehospital triage. The MedDMS is discussed in relation to other solutions for privacy-by-design, integrated data stewardship and real-time data analytics in precision medicine. LogiqSuite is used for multi-centre research study data registrations and monitoring, data analytics in interdisciplinary consortia, design of new machine learning / artificial intelligence (AI) algorithms, development of new or updated prediction models, integration of care with advanced therapy production, and real-world data monitoring in using CDS tools. The integrated MedDMS application supports data management for care and research in precision medicine.
Collapse
Affiliation(s)
| | - Inés Beekers
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Inge Verkouter
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Levi B. Richards
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| | - Alexandra Vegelien
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
- Faculty of Mathematics, VU, Amsterdam, The Netherlands
| | | | - Vera A. M. C. Bongaerts
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
| | | | - Frederik Erkens
- Department Production Metrology, Fraunhofer Institute for Production Technology IPT, Aachen, Germany
| | - Patrycja Gradowska
- HOVON Foundation, Rotterdam, The Netherlands; Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Simon Hort
- Adaptive Produktionssteuerung, Fraunhofer Institute for Production Technology IPT, Aachen, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, University Clinic Würzburg, Würzburg, Germany
| | - Manel Juan
- Fundació Clínic per a la Recerca Biomèdica—Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Immunology department, Hospital Clinic of Barcelona, Barcelona, Spain
- HSJD-Clinic Immunotherapy platform, Barcelona, Spain
| | | | - Sergio Navarro-Velázquez
- Fundació Clínic per a la Recerca Biomèdica—Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Immunology department, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Lok Lam Ngai
- Department of Haematology, Amsterdam UMC, The Netherlands
| | - Qasim A. Rafiq
- Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
| | - Carmen Sanges
- Medizinische Klinik und Poliklinik II, University Clinic Würzburg, Würzburg, Germany
| | - Jesse Tettero
- Department of Haematology, Amsterdam UMC, The Netherlands
| | - Hendrikus J. A. van Os
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
- National eHealth Living Lab, Leiden, The Netherlands
| | - Rimke C. Vos
- Public Health & Primary Care, and Health Campus The Hague, Leiden University Medical Center, The Hague, The Netherlands
| | - Yolanda de Wit
- Department of Pulmonary Medicine, Amsterdam UMC, The Netherlands
| | - Steven van Dijk
- Clinical Care & Research, ORTEC B.V., Zoetermeer, The Netherlands
| |
Collapse
|
8
|
Mavroeidis L, Napolitano A, Huang P, Jones RL. Novel Therapeutics in Soft Tissue Sarcoma. Cancers (Basel) 2024; 17:10. [PMID: 39796641 PMCID: PMC11718850 DOI: 10.3390/cancers17010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
There has been noteworthy progress in molecular characterisation and therapeutics in soft tissue sarcomas. Novel agents have gained regulatory approval by the FDA. Examples are the tyrosine kinase inhibitors avapritinib and ripretinib in gastrointestinal stromal tumours (GIST), the immune check point inhibitor atezolizumab in alveolar soft part tissue sarcoma, the γ-secretase inhibitor nirogacestat in desmoid tumours, the NTRK inhibitors larotrectinib and entrectinib in tumours with NTRK fusions, the mTOR inhibitor nab-sirolimus in PEComa, and the EZH-2 inhibitor tazemetostat in epithelioid sarcoma. The FDA has also recently granted accelerated approval for autologous T-cell therapy with afami-cel in patients with HLA-A*02 and MAGE-A4-expressing synovial sarcoma. There are other promising treatments that are still investigational, such as MDM2 and CDK4/6 inhibitors in well-/dedifferentiated liposarcoma, immune checkpoint inhibitors in the head and neck angiosarcoma and a subset of patients with undifferentiated pleomorphic sarcoma, and PARP inhibitors in leiomyosarcoma. The challenges in drug development in soft tissue sarcoma are due to the rarity and the molecular heterogeneity of the disease and the fact that many subtypes are associated with complex karyotypes or non-targetable molecular alterations. We believe that progress maybe possible with a better understanding of the complex biology, the development of novel compounds for difficult targets such as proteolysis targeting chimeras (Protacs), the utilisation of modern clinical trial designs, and enhanced collaboration of academia with industry to develop treatments with a strong biologic rationale.
Collapse
Affiliation(s)
- Leonidas Mavroeidis
- Sarcoma Unit, The Royal Marsden Hospital and Institute of Cancer Research, London SW3 6JZ, UK
| | | | | | | |
Collapse
|
9
|
Birkenbihl C, de Jong J, Yalchyk I, Fröhlich H. Deep learning-based patient stratification for prognostic enrichment of clinical dementia trials. Brain Commun 2024; 6:fcae445. [PMID: 39713242 PMCID: PMC11660909 DOI: 10.1093/braincomms/fcae445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/20/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Dementia probably due to Alzheimer's disease is a progressive condition that manifests in cognitive decline and impairs patients' daily life. Affected patients show great heterogeneity in their symptomatic progression, which hampers the identification of efficacious treatments in clinical trials. Using artificial intelligence approaches to enable clinical enrichment trials serves a promising avenue to identify treatments. In this work, we used a deep learning method to cluster the multivariate disease trajectories of 283 early dementia patients along cognitive and functional scores. Two distinct subgroups were identified that separated patients into 'slow' and 'fast' progressing individuals. These subgroups were externally validated and independently replicated in a dementia cohort comprising 2779 patients. We trained a machine learning model to predict the progression subgroup of a patient from cross-sectional data at their time of dementia diagnosis. The classifier achieved a prediction performance of 0.70 ± 0.01 area under the receiver operating characteristic curve in external validation. By emulating a hypothetical clinical trial conducting patient enrichment using the proposed classifier, we estimate its potential to decrease the required sample size. Furthermore, we balance the achieved enrichment of the trial cohort against the accompanied demand for increased patient screening. Our results show that enrichment trials targeting cognitive outcomes offer improved chances of trial success and are more than 13% cheaper compared with conventional clinical trials. The resources saved could be redirected to accelerate drug development and expand the search for remedies for cognitive impairment.
Collapse
Affiliation(s)
- Colin Birkenbihl
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin 53757, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| | - Johann de Jong
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim 55216, Germany
| | - Ilya Yalchyk
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin 53757, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin 53757, Germany
- Bonn-Aachen International Center for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn 53115, Germany
| |
Collapse
|
10
|
Small SL. Precision neurology. Ageing Res Rev 2024; 104:102632. [PMID: 39657848 DOI: 10.1016/j.arr.2024.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Over the past several decades, high-resolution brain imaging, blood and cerebrospinal fluid analyses, and other advanced technologies have changed diagnosis from an exercise depending primarily on the history and physical examination to a computer- and online resource-aided process that relies on larger and larger quantities of data. In addition, randomized controlled trials (RCT) at a population level have led to many new drugs and devices to treat neurological disease, including disease-modifying therapies. We are now at a crossroads. Combinatorially profound increases in data about individuals has led to an alternative to population-based RCTs. Genotyping and comprehensive "deep" phenotyping can sort individuals into smaller groups, enabling precise medical decisions at a personal level. In neurology, precision medicine that includes prediction, prevention and personalization requires that genomic and phenomic information further incorporate imaging and behavioral data. In this article, we review the genomic, phenomic, and computational aspects of precision medicine for neurology. After defining biological markers, we discuss some applications of these "-omic" and neuroimaging measures, and then outline the role of computation and ultimately brain simulation. We conclude the article with a discussion of the relation between precision medicine and value-based care.
Collapse
Affiliation(s)
- Steven L Small
- Department of Neuroscience, University of Texas at Dallas, Dallas, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurology, The University of Chicago, Chicago, IL, USA; Department of Neurology, University of California, Irvine, Orange, CA, USA.
| |
Collapse
|
11
|
Lewandrowski KU, Li ZZ, Liu X, Xifeng Z, Kwon B, Dowling Á, Knight M, Terxeira de Carvalho PS, Kim C, Basil GW, Alonso Cuéllar GO, Morgenstern C, Ongulade J, Jiang Y, Ito K, Bergamaschi JP, Kim JSL, Ramirez JF, Oertel J, Elfar JC, Alhammoud A, Bonazza NA, Burkhardt BW, Alvim Fiorelli RK, Schmidt SL, Lorio MP. Insights on High-Value Procedures From the ISASS 4-Part Webinar Series on Current and Emerging Techniques in Endoscopic Spine Surgery Based on Surgeon Experience. Int J Spine Surg 2024; 18:S66-S82. [PMID: 39547675 PMCID: PMC11616408 DOI: 10.14444/8676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND The authors conducted a comprehensive review and integration of insights from 4 webinars hosted by the International Society for the Advancement of Spine Surgery (ISASS) to arrive at recommendations for best clinical practices for guideline development for endoscopic spine surgery. This perspective article discusses the limitations of traditional surgical trials and amalgamates surgeons' experience and research on various cutting-edge techniques. METHODS Data were extracted from surveys conducted during each webinar session involving 3639 surgeons globally. The polytomous Rasch model was employed to analyze responses, ensuring a robust statistical assessment of surgeon endorsements and educational impacts and focusing on operative nuances and experience-based outcomes. Bias detection was performed using the differential item functioning test. RESULTS The ISASS webinars provided a dynamic platform for discussing advances in endoscopic spine surgery, identifying a range of high-value procedures from basic discectomies to complex lumbar interbody fusions. Each high-value endoscopic spine surgery was highlighted in separate peer-reviewed publications, which form the basis for this summary document that synthesizes key takeaways from these webinars. High-value clinical applications of endoscopic spine surgery, primarily defined as higher-intensity endorsement transformation from the pre- to postwebinar survey with a shift to higher mean logit locations of test items both with unbiased and orderly threshold progression, were: (a) Percutaneous interlaminar endoscopic decompression for lateral canal stenosis, (b) transforaminal debridement of low-grade degenerative spondylolisthesis, (c) transforaminal full-endoscopic interbody fusion for hard disc herniation, (d) endoscopic standalone lumbar interbody fusion, (e) endoscopic debridement of spondylolytic spondylolisthesis, and (f) posterior cervical foraminotomy for herniated disc and bony stenosis. CONCLUSIONS The ISASS webinar series has significantly impacted surgeons' education and contributed to the identification of high-value endoscopic spine surgery practices that may serve as a cornerstone for surgeon training standards, policy, and guidelines development. Ongoing research on technological advancements and expansions of clinical indications combined with systematic review is expected to refine the recommendations on high-value endoscopic spinal surgeries recommended for enhanced reimbursement. CLINICAL RELEVANCE Assessing surgeon confidence and acceptance of endoscopic spinal surgeries using polytomous Rasch analysis. LEVEL OF EVIDENCE Level 2 (inferential) and 3 (observational) evidence because Rasch analysis provides statistical validation of instruments rather than direct clinical outcomes.
Collapse
Affiliation(s)
- Kai-Uwe Lewandrowski
- Division Personalized Pain Research and Education, Center for Advanced Spine Care of Southern Arizona, Tucson, AZ, USA
- Department of Orthopaedic Surgery, University of Arizona, Banner Medical Center, Tucson, AZ, USA
- Dr. honoris causa Department of Orthopedics at Hospital Universitário Gaffree Guinle, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Orthopedics, Hospital Universitário Gaffree Guinle Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Zhen-Zhou Li
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Zhang Xifeng
- Department of Orthopedics, First Medical Center, PLA General Hospital, Beijing, China
| | - Brian Kwon
- Department of Orthopaedics, New England Baptist Hospital, Boston, MA, USA
| | - Álvaro Dowling
- Department of Orthopedic Surgery, USP, Ribeirão Preto, Brazil
- Spine Center, Endoscopic Spine Clinic, Santiago, Chile
| | - Martin Knight
- The Spinal Foundation, Manchester University, The Weymouth Hospital, London, UK
| | - Paulo Sergio Terxeira de Carvalho
- Department of Neurosurgery, Gaffrée e Guinle University Hospital, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Choll Kim
- Department of Orthopaedic Surgery, UCSD Medical Center East Campus, Excell Spine Center, San Diego, CA, USA
| | - Gregory W Basil
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Gabriel Oswaldo Alonso Cuéllar
- Department of Orthopaedics, Clínica Reina Sofía - Clínica Colsanitas, Bogotá, D.C., Colombia
- Latin American Endoscopic Spine Surgeons LESS Invasiva Academy, Bogotá, D.C., Colombia
| | | | - John Ongulade
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yi Jiang
- Department of Orthopaedics, Beijing Haidian Hospital, Beijing, China
| | - Kenyu Ito
- Department of Spinal Surgery, Aichi Spine Institute, Fuso-cho Niwa-gun Aichi, Japan
| | | | - Jin-Sung L Kim
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jorge F Ramirez
- Department of Orthopaedic, Minimally Invasive Spine Center, Bogotá, D.C., Colombia
- Department of Orthopaedics, Reina Sofía Clinic, Bogotá, D.C., Colombia
- Department of Orthopaedics, Fundación Universitaria Sanitas, Bogotá, D.C., Colombia
| | - Joachim Oertel
- Neurosschirurgie, Universitätsklinikum des Saarlandes Neurosschirurgie, Homburg, Saarland, Germany
| | - John C Elfar
- Department of Orthopedics and Sports Medicine, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Abduljabbar Alhammoud
- Department of Orthopedic Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Nicholas A Bonazza
- Department of Orthopedic Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Benedikt W Burkhardt
- Wirbelsäulenzentrum/Spine Center, WSC Hirslanden Klinik Zurich, Zurich, Switzerland
| | - Rossano Kepler Alvim Fiorelli
- Department of Thoracic Surgery, Gaffrée e Guinle University Hospital, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Sergio Luis Schmidt
- Department of Neurology, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Morgan P Lorio
- Advanced Orthopedics, Altamonte Springs, Orlando, FL, USA
- Orlando College of Osteopathic Medicine, Orlando, FL, USA
| |
Collapse
|
12
|
Bollhagen A, Bodenmiller B. Highly Multiplexed Tissue Imaging in Precision Oncology and Translational Cancer Research. Cancer Discov 2024; 14:2071-2088. [PMID: 39485249 PMCID: PMC11528208 DOI: 10.1158/2159-8290.cd-23-1165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 05/24/2024] [Accepted: 08/13/2024] [Indexed: 11/03/2024]
Abstract
Precision oncology tailors treatment strategies to a patient's molecular and health data. Despite the essential clinical value of current diagnostic methods, hematoxylin and eosin morphology, immunohistochemistry, and gene panel sequencing offer an incomplete characterization. In contrast, highly multiplexed tissue imaging allows spatial analysis of dozens of markers at single-cell resolution enabling analysis of complex tumor ecosystems; thereby it has the potential to advance our understanding of cancer biology and supports drug development, biomarker discovery, and patient stratification. We describe available highly multiplexed imaging modalities, discuss their advantages and disadvantages for clinical use, and potential paths to implement these into clinical practice. Significance: This review provides guidance on how high-resolution, multiplexed tissue imaging of patient samples can be integrated into clinical workflows. It systematically compares existing and emerging technologies and outlines potential applications in the field of precision oncology, thereby bridging the ever-evolving landscape of cancer research with practical implementation possibilities of highly multiplexed tissue imaging into routine clinical practice.
Collapse
Affiliation(s)
- Alina Bollhagen
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Liu XY, Yu TJ, Shao ZM. Precision medicine for breast cancer: advances and challenges. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:35. [PMID: 39534585 PMCID: PMC11557152 DOI: 10.21037/tbcr-24-35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Xin-Yi Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tian-Jian Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Breast Cancer, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
14
|
Klas K, Strzebonska K, Zaborowska L, Krawczyk T, Włodarczyk A, Bąk-Kuchejda U, Polak M, Van Wambeke S, Waligora M. Risk and Benefit for Basket Trials in Oncology: A Systematic Review and Meta-Analysis. Target Oncol 2024:10.1007/s11523-024-01107-3. [PMID: 39455508 DOI: 10.1007/s11523-024-01107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Oncology research is increasingly adopting new clinical trial models that implement the concept of precision medicine. One of these is the basket clinical trial design. Basket clinical trials allow new treatments to be evaluated across multiple tumor types. Patients recruited to basket clinical trials share certain molecular characteristics of their cancer that are predictive of clinical benefit from the experimental treatment. OBJECTIVE Our aim was to describe the risks and benefits of basket clinical trials in oncology. METHODS Our study was prospectively registered in PROSPERO (CRD42023406401). We systematically searched PubMed, Embase, and ClinicalTrials.gov for reports of basket clinical trials in oncology published between 1 January, 2001, and 14 June, 2023. We measured the risk by treatment-related adverse events (grades 3, 4, and 5), and the benefit by objective response rate. We also extracted and analyzed data on progression-free survival and overall survival. When possible, data were meta-analyzed. RESULTS We included 126 arms of 75 basket clinical trials accounting for 7659 patients. The pooled objective response rate was 18.0% (95% confidence interval [CI] 14.8-21.1). The rate of treatment-related death was 0.7% (95% CI 0.4-1.0), while 30.4% (95% CI 24.2-36.7) of patients experienced grade 3/4 drug-related toxicity. The median progression-free survival was 3.1 months (95% CI 2.6-3.9), and the median overall survival was 8.9 months (95% CI 6.7-10.2). CONCLUSIONS Our results provide an empirical basis for communicating about the risks and benefits of basket clinical trials and for refining new models of clinical trials applied in precision medicine.
Collapse
Affiliation(s)
- Katarzyna Klas
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Karolina Strzebonska
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
| | - Lucja Zaborowska
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Krawczyk
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Alicja Włodarczyk
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
| | - Urszula Bąk-Kuchejda
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | - Maciej Polak
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland
- Department of Epidemiology and Population Studies, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University Medical College, Kraków, Poland
| | | | - Marcin Waligora
- Research Ethics in Medicine Study Group (REMEDY), Department of Bioethics, Faculty of Health Sciences, Jagiellonian University Medical College, Michalowskiego 12, 31-126, Kraków, Poland.
| |
Collapse
|
15
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
16
|
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, Tan Y, Liu Z. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater 2024:e2401843. [PMID: 39397335 DOI: 10.1002/adhm.202401843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/25/2024] [Indexed: 10/15/2024]
Abstract
The development of personalized precision medicine has become a pivotal focus in modern healthcare. Organoids-on-a-Chip (OoCs), a groundbreaking fusion of organoid culture and microfluidic chip technology, has emerged as a promising approach to advancing patient-specific treatment strategies. In this review, the diverse applications of OoCs are explored, particularly their pivotal role in personalized precision medicine, and their potential as a cutting-edge technology is highlighted. By utilizing patient-derived organoids, OoCs offer a pathway to optimize treatments, create precise disease models, investigate disease mechanisms, conduct drug screenings, and individualize therapeutic strategies. The emphasis is on the significance of this technological fusion in revolutionizing healthcare and improving patient outcomes. Furthermore, the transformative potential of personalized precision medicine, future prospects, and ongoing advancements in the field, with a focus on genomic medicine, multi-omics integration, and ethical frameworks are discussed. The convergence of these innovations can empower patients, redefine treatment approaches, and shape the future of healthcare.
Collapse
Affiliation(s)
- Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
17
|
Doga H, Bose A, Sahin ME, Bettencourt-Silva J, Pham A, Kim E, Andress A, Saxena S, Parida L, Robertus JL, Kawaguchi H, Soliman R, Blankenberg D. How can quantum computing be applied in clinical trial design and optimization? Trends Pharmacol Sci 2024; 45:880-891. [PMID: 39317621 DOI: 10.1016/j.tips.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/26/2024]
Abstract
Clinical trials are necessary for assessing the safety and efficacy of treatments. However, trial timelines are severely delayed with minimal success due to a multitude of factors, including imperfect trial site selection, cohort recruitment challenges, lack of efficacy, absence of reliable biomarkers, etc. Each of these factors possesses a unique computational challenge, such as data management, trial simulations, statistical analyses, and trial optimization. Recent advancements in quantum computing offer a promising opportunity to overcome these hurdles. In this opinion we uniquely explore the application of quantum optimization and quantum machine learning (QML) to the design and execution of clinical trials. We examine the current capabilities and limitations of quantum computing and outline its potential to streamline clinical trials.
Collapse
Affiliation(s)
- Hakan Doga
- IBM Quantum, Almaden Research Center, San Jose, CA, USA.
| | | | - M Emre Sahin
- The Hartree Centre, STFC, Sci-Tech Daresbury, Warrington, UK
| | | | - Anh Pham
- Deloitte Consulting LLP, Atlanta, GA, USA
| | | | | | | | | | - Jan Lukas Robertus
- Imperial College London and Royal Brompton and Harefield Hospitals, London, UK
| | | | | | | |
Collapse
|
18
|
Niazmand VR, Raheb MA, Eqra N, Vatankhah R, Farrokhi A. Deep reinforcement learning control of combined chemotherapy and anti-angiogenic drug delivery for cancerous tumor treatment. Comput Biol Med 2024; 181:109041. [PMID: 39180855 DOI: 10.1016/j.compbiomed.2024.109041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
By virtue of the chronic and dangerous nature of cancer, researchers have explored various approaches to managing the abnormal cell growth associated with this disease using novel treatment methods. This study introduces a control system based on normalized advantage function reinforcement learning. It aims to boost the body's immune response against cancer cell proliferation. This control approach is applied to provide a combination of both chemotherapy and anti-angiogenic drugs for the first time without the need for complex, predefined mathematical models. It employs a model-free reinforcement learning technique that adaptively adjusts to individual patients to determine optimal drug administration with minimum injection rates. In this regard, a comprehensive and realistic simulation and training environment is employed, with the concentrations of normal cells, cancer cells, and endothelial cells, as well as the levels of chemotherapy and anti-angiogenic agents, as state variables. Furthermore, high levels of disturbances are considered in the simulation to investigate the robustness of the proposed method against probable uncertainties in the treatment process or patient parameters. A practical reward function has also been devised in alignment with medical objectives to ensure effective and safe treatment outcomes. The results demonstrate robustness and superior performance compared to the existing methods. Simulations show that the proposed approach is a dependable strategy for effectively reducing the concentration of cancer cells in the shortest duration using minimal doses of chemotherapy and anti-angiogenic drugs.
Collapse
Affiliation(s)
- Vahid Reza Niazmand
- Department of Computer Science, Memorial University of Newfoundland, St John's, Canada
| | - Mohammad Ali Raheb
- Department of Mechanical Engineering, Politecnico di Milano, Milan, Italy
| | - Navid Eqra
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Ramin Vatankhah
- School of Mechanical Engineering, Shiraz University, Shiraz, Iran
| | - Amirmohammad Farrokhi
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Van der Speeten K, Kusamura S, Villeneuve L, Piso P, Verwaal VJ, González-Moreno S, Glehen O. The 2022 PSOGI International Consensus on HIPEC Regimens for Peritoneal Malignancies: HIPEC Technologies. Ann Surg Oncol 2024; 31:7090-7110. [PMID: 39037523 DOI: 10.1245/s10434-024-15513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/09/2024] [Indexed: 07/23/2024]
Abstract
This manuscript reports the results of an international consensus on technologies of hyperthermic intraperitoneal perioperative chemotherapy (HIPEC) performed with the following goals: To provide recommendations for the technological parameters to perform HIPEC. To identify the role of heat and its application forms in treating peritoneal metastases. To provide recommendations regarding the correct dosimetry of intraperitoneal chemotherapy drugs and their carrier solutions. To identify for each intraperitoneal chemotherapy regimen the best dosimetry and fractionation. To identify areas of future research pertaining to HIPEC technology and regimens. This consensus was performed by the Delphi technique and comprised two rounds of voting. In total, 96 of 102 eligible panelists replied to both Delphi rounds (94.1%) with a consensus of 39/51 questions on HIPEC technical aspects. Among the recommendations that met with the strongest consensus were those concerning the dose of HIPEC drug established in mg/m2, a target temperature of at least 42°C, and the use of at least three temperature probes to pursue hyperthermia. Ninety minutes as the ideal HIPEC duration seemed to make consensus. These results should be considered when designing new clinical trials in patients with peritoneal surface malignancies.
Collapse
Affiliation(s)
- Kurt Van der Speeten
- Department of Surgical Oncology, Ziekenhuis Oost-Limburg, Genk, Belgium.
- Faculty of Life Sciences, BIOMED Research Institute, University Hasselt, Hasselt, Belgium.
| | - Shigeki Kusamura
- Department of Surgical Oncology, PSM unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laurent Villeneuve
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| | - Pompiliu Piso
- Department of General and Visceral Surgery, Hospital Barmherzige Brüder, Regensburg, Germany
| | - Vic J Verwaal
- Peritoneal Surface Malignancy and HIPEC Institute for Regional Sundhedforskning, Syddansk University, Odense, Sweden
| | | | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-sud, Lyon, France
| |
Collapse
|
20
|
Vitorino R. Transforming Clinical Research: The Power of High-Throughput Omics Integration. Proteomes 2024; 12:25. [PMID: 39311198 PMCID: PMC11417901 DOI: 10.3390/proteomes12030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
High-throughput omics technologies have dramatically changed biological research, providing unprecedented insights into the complexity of living systems. This review presents a comprehensive examination of the current landscape of high-throughput omics pipelines, covering key technologies, data integration techniques and their diverse applications. It looks at advances in next-generation sequencing, mass spectrometry and microarray platforms and highlights their contribution to data volume and precision. In addition, this review looks at the critical role of bioinformatics tools and statistical methods in managing the large datasets generated by these technologies. By integrating multi-omics data, researchers can gain a holistic understanding of biological systems, leading to the identification of new biomarkers and therapeutic targets, particularly in complex diseases such as cancer. The review also looks at the integration of omics data into electronic health records (EHRs) and the potential for cloud computing and big data analytics to improve data storage, analysis and sharing. Despite significant advances, there are still challenges such as data complexity, technical limitations and ethical issues. Future directions include the development of more sophisticated computational tools and the application of advanced machine learning techniques, which are critical for addressing the complexity and heterogeneity of omics datasets. This review aims to serve as a valuable resource for researchers and practitioners, highlighting the transformative potential of high-throughput omics technologies in advancing personalized medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
21
|
Zarbock A, Forni LG, Koyner JL, Bell S, Reis T, Meersch M, Bagshaw SM, Fuhmann DY, Liu KD, Pannu N, Arikan AA, Angus DC, Duquette D, Goldstein SL, Hoste E, Joannidis M, Jongs N, Legrand M, Mehta RL, Murray PT, Nadim MK, Ostermann M, Prowle J, See EJ, Selby NM, Shaw AD, Srisawat N, Ronco C, Kellum JA. Recommendations for clinical trial design in acute kidney injury from the 31st acute disease quality initiative consensus conference. A consensus statement. Intensive Care Med 2024; 50:1426-1437. [PMID: 39115567 PMCID: PMC11377501 DOI: 10.1007/s00134-024-07560-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Novel interventions for the prevention or treatment of acute kidney injury (AKI) are currently lacking. To facilitate the evaluation and adoption of new treatments, the use of the most appropriate design and endpoints for clinical trials in AKI is critical and yet there is little consensus regarding these issues. We aimed to develop recommendations on endpoints and trial design for studies of AKI prevention and treatment interventions based on existing data and expert consensus. METHODS At the 31st Acute Disease Quality Initiative (ADQI) meeting, international experts in critical care, nephrology, involving adults and pediatrics, biostatistics and people with lived experience (PWLE) were assembled. We focused on four main areas: (1) patient enrichment strategies, (2) prevention and attenuation studies, (3) treatment studies, and (4) innovative trial designs of studies other than traditional (parallel arm or cluster) randomized controlled trials. Using a modified Delphi process, recommendations and consensus statements were developed based on existing data, with > 90% agreement among panel members required for final adoption. RESULTS The panel developed 12 consensus statements for clinical trial endpoints, application of enrichment strategies where appropriate, and inclusion of PWLE to inform trial designs. Innovative trial designs were also considered. CONCLUSION The current lack of specific therapy for prevention or treatment of AKI demands refinement of future clinical trial design. Here we report the consensus findings of the 31st ADQI group meeting which has attempted to address these issues including the use of predictive and prognostic enrichment strategies to enable appropriate patient selection.
Collapse
Affiliation(s)
- Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Albert-Schweitzer Campus 1, Building A1, 48149, Münster, Germany.
- Outcomes Research Consortium, Cleveland, OH, USA.
| | - Lui G Forni
- Depatment of Critical Care, Royal Surrey Hospital Foundation Trust, Guildford, Surrey, UK
- School of Medicine, Kate Granger Building, University of Surrey, Guildford, Surrey, UK
| | - Jay L Koyner
- Section of Nephrology, University of Chicago, Chicago, IL, USA
| | - Samira Bell
- Division of Population Health and Genomics, University of Dundee, Dundee, UK
| | - Thiago Reis
- Hospital Sírio-Libanês, São Paulo, Brazil
- Fenix Nephrology, São Paulo, Brazil
- Laboratory of Molecular Pharmacology, University of Brasília, Brasília, Brazil
| | - Melanie Meersch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Albert-Schweitzer Campus 1, Building A1, 48149, Münster, Germany
| | - Sean M Bagshaw
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta and Alberta Health Services, Edmonton, AB, Canada
| | - Dana Y Fuhmann
- UPMC Children's Hospital of Pittsburgh, 4401 Penn Avenue, Suite 2000, Pittsburgh, PA, 15224, USA
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathleen D Liu
- Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, CA, USA
| | - Neesh Pannu
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ayse Akcan Arikan
- Division of Nephrology and Critical Care Medicine, Department of Pediatric, Baylor College of Medicine, Houston, TX, USA
| | - Derek C Angus
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - D'Arcy Duquette
- Critical Care Strategic Clinical Network, Alberta Health Services, Calgary, Canada
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Eric Hoste
- Intensive Care Unit, Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Niels Jongs
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Matthieu Legrand
- Department of Anesthesia and Perioperative Care, Division of Critical Care Medicine, UCSF, San Francisco, CA, USA
| | - Ravindra L Mehta
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | | | - Mitra K Nadim
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Marlies Ostermann
- Department of Intensive Care, King's College London, Thomas' Hospital, Guy's & St, London, UK
| | - John Prowle
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Emily J See
- Department of Critical Care, University of Melbourne, Parkville, Australia
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Nephrology, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Nicholas M Selby
- Centre for Kidney Research and Innovation, University of Nottingham, Nottingham, UK
| | - Andrew D Shaw
- Department of Intensive Care and Resuscitation, The Cleveland Clinic, Cleveland, OH, USA
| | - Nattachai Srisawat
- Division of Nephrology, Department of Medicine, Faculty of Medicine, and Center of Excellence in Critical Care Nephrology, Chulalongkorn University, Bangkok, Thailand
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padua, Italy
- International Renal Research Institute of Vicenza (IRRV), Vicenza, Italy
- Department of Nephrology, San Bortolo Hospital, Vicenza, Italy
| | - John A Kellum
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Chen H, Brannath W, Futschik A. Adaptive Multiple Comparisons With the Best. Biom J 2024; 66:e202300242. [PMID: 39126674 DOI: 10.1002/bimj.202300242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 08/12/2024]
Abstract
Subset selection methods aim to choose a nonempty subset of populations including a best population with some prespecified probability. An example application involves location parameters that quantify yields in agriculture to select the best wheat variety. This is quite different from variable selection problems, for instance, in regression. Unfortunately, subset selection methods can become very conservative when the parameter configuration is not least favorable. This will lead to a selection of many non-best populations, making the set of selected populations less informative. To solve this issue, we propose less conservative adaptive approaches based on estimating the number of best populations. We also discuss variants of our adaptive approaches that are applicable when the sample sizes and/or variances differ between populations. Using simulations, we show that our methods yield a desirable performance. As an illustration of potential gains, we apply them to two real datasets, one on the yield of wheat varieties and the other obtained via genome sequencing of repeated samples.
Collapse
Affiliation(s)
- Haoyu Chen
- Vetmeduni Vienna, Wien, Austria
- Vienna Graduate School of Population Genetics, Vienna, Austria
- Johannes Kepler University Linz, Linz, Austria
| | - Werner Brannath
- Kompetenzzentrum fur Klinische Studien, Universität Bremen, Bremen, Germany
| | | |
Collapse
|
23
|
Oualikene-Gonin W, Jaulent MC, Thierry JP, Oliveira-Martins S, Belgodère L, Maison P, Ankri J. Artificial intelligence integration in the drug lifecycle and in regulatory science: policy implications, challenges and opportunities. Front Pharmacol 2024; 15:1437167. [PMID: 39156111 PMCID: PMC11327028 DOI: 10.3389/fphar.2024.1437167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Artificial intelligence tools promise transformative impacts in drug development. Regulatory agencies face challenges in integrating AI while ensuring reliability and safety in clinical trial approvals, drug marketing authorizations, and post-market surveillance. Incorporating these technologies into the existing regulatory framework and agency practices poses notable challenges, particularly in evaluating the data and models employed for these purposes. Rapid adaptation of regulations and internal processes is essential for agencies to keep pace with innovation, though achieving this requires collective stakeholder collaboration. This article thus delves into the need for adaptations of regulations throughout the drug development lifecycle, as well as the utilization of AI within internal processes of medicine agencies.
Collapse
Affiliation(s)
- Wahiba Oualikene-Gonin
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
| | - Marie-Christine Jaulent
- INSERM, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances en e-Santé, LIMICS, Sorbonne Université, Paris, France
| | | | - Sofia Oliveira-Martins
- Faculty of Pharmacy of Lisbon University, Lisbon, Portugal
- CHRC – Comprehensive Health Research Center, Evora, Portugal
| | - Laetitia Belgodère
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
| | - Patrick Maison
- Agence Nationale de Sécurité des Médicaments et des Produits de Santé (ANSM) Saint-Denis, Saint-Denis, France
- EA 7379, Faculté de Santé, Université Paris-Est Créteil, Créteil, France
- CHI Créteil, Créteil, France
| | - Joël Ankri
- Université de Versailles St Quentin-Paris Saclay, Inserm U1018, Guyancourt, France
| | | |
Collapse
|
24
|
Angelucci F, Ai AR, Piendel L, Cerman J, Hort J. Integrating AI in fighting advancing Alzheimer: diagnosis, prevention, treatment, monitoring, mechanisms, and clinical trials. Curr Opin Struct Biol 2024; 87:102857. [PMID: 38838385 DOI: 10.1016/j.sbi.2024.102857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/15/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
The application of artificial intelligence (AI) in neurology is a growing field offering opportunities to improve accuracy of diagnosis and treatment of complicated neuronal disorders, plus fostering a deeper understanding of the aetiologies of these diseases through AI-based analyses of large omics data. The most common neurodegenerative disease, Alzheimer's disease (AD), is characterized by brain accumulation of specific pathological proteins, accompanied by cognitive impairment. In this review, we summarize the latest progress on the use of AI in different AD-related fields, such as analysis of neuroimaging data enabling early and accurate AD diagnosis; prediction of AD progression, identification of patients at higher risk and evaluation of new treatments; improvement of the evaluation of drug response using AI algorithms to analyze patient clinical and neuroimaging data; the development of personalized AD therapies; and the use of AI-based techniques to improve the quality of daily life of AD patients and their caregivers.
Collapse
Affiliation(s)
- Francesco Angelucci
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.
| | - Alice Ruixue Ai
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Lydia Piendel
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic; Augusta University/University of Georgia Medical Partnership, Medical College of Georgia, Athens, GA, USA
| | - Jiri Cerman
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic; INDRC, International Neurodegenerative Disorders Research Center, Prague, Czech Republic
| |
Collapse
|
25
|
Maggie Liu SY, Jin ZY, Deng JY, Zhong SM, Ahn MJ, Horinouchi H, Li Y, Wu YL. Drug development and evidence for lung cancer targeted therapy in Eastern Asia. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 49:101090. [PMID: 39381018 PMCID: PMC11459064 DOI: 10.1016/j.lanwpc.2024.101090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 10/10/2024]
Abstract
The development of targeted drugs in the Eastern Asia region is going through a flourishing stage. With the continuous advancement of technology and medical research, biotechnology companies and research institutions in the region have made significant progress in cancer field. The Eastern Asian region not only actively participates in clinical trials, but is also committed to developing personalized medical plans to meet the diverse genotypes and phenotypes of patients. The governments and enterprises are increasingly valuing innovation, strengthening international cooperation, and promoting drug development. This paper summarizes the development of genetic testing technology, targeted drugs approval, ongoing promising clinical trials in the field of lung cancer and the important progress made by governments in the Eastern Asian region, and proposed key factors that will contribute to the promising future prospects in the region. The targeted drug market in the Eastern Asian region is expected to drive the medical field forward.
Collapse
Affiliation(s)
- Si-Yang Maggie Liu
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Zhen-Yi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
| | - Jia-Yi Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Si-Min Zhong
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yangqiu Li
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Kurzrock R, Hong D. How to Build a Successful Phase I Clinical Trials Unit: Lessons Based on the MD Anderson Cancer Center Experience. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2024; 7:142-149. [PMID: 39220000 PMCID: PMC11361344 DOI: 10.36401/jipo-23-243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2024]
Affiliation(s)
- Razelle Kurzrock
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
- WIN Consortium, Paris, France
- University of Nebraska, Omaha, NB, USA
| | - David Hong
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
27
|
Arteaga-Henríquez G, Ramos-Sayalero C, Ibañez-Jimenez P, Karina Rosales-Ortiz S, Kilencz T, Schiweck C, Schnorr I, Siegl A, Arias-Vasquez A, Bitter I, Fadeuilhe C, Ferrer M, Lavebratt C, Matura S, Reif A, Réthelyi JM, Richarte V, Rommelse N, Antoni Ramos-Quiroga J. Efficacy of a synbiotic in the management of adults with Attention-Deficit and Hyperactivity Disorder and/or Borderline Personality Disorder and high levels of irritability: Results from a multicenter, randomized, placebo-controlled, "basket" trial. Brain Behav Immun 2024; 120:360-371. [PMID: 38885746 DOI: 10.1016/j.bbi.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Irritability worsens prognosis and increases mortality in individuals with Attention-Deficit and Hyperactivity Disorder (ADHD) and/or Borderline Personality Disorder (BPD). However, treatment options are still insufficient. The aim of this randomized, double blind, placebo-controlled study was to investigate the superiority of a synbiotic over placebo in the management of adults with ADHD and/or BPD and high levels of irritability. The study was conducted between February 2019 and October 2020 at three European clinical centers located in Hungary, Spain and Germany. Included were patients aged 18-65 years old diagnosed with ADHD and/or BPD and high levels of irritability (i.e., an Affectivity Reactivity Index (ARI-S) ≥ 5, plus a Clinical Global Impression-Severity Scale (CGI-S) score ≥ 4). Subjects were randomized 1(synbiotic):1(placebo); the agent was administered each day, for 10 consecutive weeks. The primary outcome measure was end-of-treatment response (i.e., a reduction ≥ 30 % in the ARI-S total score compared to baseline, plus a Clinical Global Impression-Improvement (CGI-I) total score of < 3 (very much, or much improved) at week 10). Between-treatment differences in secondary outcomes, as well as safety were also investigated. Of the 231 included participants, 180 (90:90) were randomized and included in the intention-to-treat-analyses. Of these, 117 (65 %) were females, the mean age was 38 years, ADHD was diagnosed in 113 (63 %), BPD in 44 (24 %), both in 23 (13 %). The synbiotic was well tolerated. At week 10, patients allocated to the synbiotic experienced a significantly higher response rate compared to those allocated to placebo (OR: 0.2, 95 % CI:0.1 to 0.7; P = 0.01). These findings suggest that that (add-on) treatment with a synbiotic may be associated with a clinically meaningful improvement in irritability in, at least, a subgroup of adults with ADHD and/or BPD. A superiority of the synbiotic over placebo in the management of emotional dysregulation (-3.6, 95 % CI:-6.8 to -0.3; P = 0.03), emotional symptoms (-0.6, 95 % CI:-1.2 to -0.05; P = 0.03), inattention (-1.8, 95 % CI: -3.2 to -0.4; P = 0.01), functioning (-2.7, 95 % CI: -5.2 to -0.2; P = 0.03) and perceived stress levels (-0.6, 95 % CI: -1.2 to -0.05; P = 0.03) was also suggested. Higher baseline RANK-L protein levels were associated with a significantly lower response rate, but only in the synbiotic group (OR: 0.1, 95 % CI: -4.3 to - 0.3, P = 0.02). In the placebo group, higher IL-17A levels at baseline were significantly associated with a higher improvement in in particular, emotional dysregulation (P = 0.04), opening a door for new (targeted) drug intervention. However, larger prospective studies are warranted to confirm the findings. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03495375.
Collapse
Affiliation(s)
- Gara Arteaga-Henríquez
- Department of Mental Health, Hospital Universitari Vall d́Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain; NCRR-The National Center for Register-Based Research, Aahrus University. Aahrus, Denmark.
| | - Carolina Ramos-Sayalero
- Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
| | - Pol Ibañez-Jimenez
- Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
| | - Silvia Karina Rosales-Ortiz
- Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain
| | - Tünde Kilencz
- Semmelweis University, Department of Psychiatry and Psychotherapy, Budapest, Hungary
| | - Carmen Schiweck
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Isabel Schnorr
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Anne Siegl
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Alejandro Arias-Vasquez
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands; Department of Psychiatry, Radboudd University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands
| | - István Bitter
- Semmelweis University, Department of Psychiatry and Psychotherapy, Budapest, Hungary
| | - Christian Fadeuilhe
- Department of Mental Health, Hospital Universitari Vall d́Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain
| | - Marc Ferrer
- Department of Mental Health, Hospital Universitari Vall d́Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autónoma de Barcelona, Barcelona, Catalonia, Spain
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Instituet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Silke Matura
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Andreas Reif
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - János M Réthelyi
- Semmelweis University, Department of Psychiatry and Psychotherapy, Budapest, Hungary
| | - Vanesa Richarte
- Department of Mental Health, Hospital Universitari Vall d́Hebron, Barcelona, Catalonia, Spain; Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autónoma de Barcelona, Barcelona, Catalonia, Spain
| | - Nanda Rommelse
- Karakter Child and Adolescent Psychiatry University Center, Stockholm, the Netherlands
| | - Josep Antoni Ramos-Quiroga
- Department of Mental Health, Hospital Universitari Vall d́Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health, and Addictions, Vall d́Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Biomedical Network Research Center on Mental Health (CIBERSAM), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autónoma de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
28
|
Zhou W, Xu Z, Liu S, Lou X, Liu P, Xie H, Zhang S, Liu X, Zhuo B, Huang H. Landscape of clinical drug development of ADCs used for the pharmacotherapy of cancers: an overview of clinical trial registry data from 2002 to 2022. BMC Cancer 2024; 24:898. [PMID: 39060958 PMCID: PMC11282866 DOI: 10.1186/s12885-024-12652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND To provide reference for clinical development of ADCs in the industry, we analyzed the landscape and characteristics of clinical trials about antibody-drug conjugates (ADCs). METHOD Clinical trials to study ADCs used for the pharmacotherapy of cancers initiated by the sponsor were searched in the Cite line Pharma Intelligence (Trialtrove database), and the landscape and characteristics of these clinical trials were analyzed from multiple perspectives, such as the number, phases, status, indications, and targets of the clinical trials. RESULT As of December 31, 2022, a total of 431 clinical trials have been initiated to study ADCs used for the pharmacotherapy of cancers, and the number of the last 10 years was 5.5 times as large as the first 11 years. These clinical trials involved 47 indications, including breast cancer, lymphoma (lymphoma, non-Hodgkin's and lymphoma, Hodgkin's), unspecified solid tumor, bladder cancer and lung cancer (lung, non-small cell cancer and lung, small cell cancer). As for each of these five indications, 50 + clinical trials have been carried out, accounting for as high as 48.50% (454/936). ADCs involve 38 targets, which are relatively concentrated. Among them, ERBB2 (HER2) and TNFRSF8 (CD30) involve in 100 + registered clinical trials, and TNFRSF17 (BCMA), NECTIN4 and CD19 in 10 + trials. The clinical trials for these five targets account for 79.02% (354/448) of the total number. Up to 93.97% (405/431) of these clinical trials explored the correlation between biomarkers and efficacy. Up to 45.91% (292/636) of Lots (lines of treatment) applied in the clinical trials were the second line. Until December 31, 2022, 54.52% (235/431) of the clinical trials have been completed or terminated. CONCLUSION ADCs are a hotspot of research and development in oncology clinical trials, but the indications, targets, phases, and Lot that have been registered are seemingly relatively concentrated at present. This study provides a comprehensive analysis which can assist researchers/developer quickly grasp relevant knowledge to assess a product and also providing new clues and ideas for future research.
Collapse
Affiliation(s)
- Wenjing Zhou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China.
| | - Zhiyuan Xu
- Department of Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shu Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiaohuan Lou
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Pengcheng Liu
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, 211198, P. R. China
| | - Huali Xie
- Department of Pharmacy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Shuiyan Zhang
- Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518000, P. R. China
| | - Xi Liu
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Baoshan Zhuo
- Department of Clinical Trials Center, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, 518000, P. R. China
| | - Hongbing Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
29
|
Qi W, Wu X, Feng Y, Cao Q. Letter to the editor about "Preoperative proteinuria correlates with renal function after partial nephrectomy for renal cell carcinoma". World J Urol 2024; 42:446. [PMID: 39060483 DOI: 10.1007/s00345-024-05149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Affiliation(s)
- Wei Qi
- School of Medicine, Macau University of Science and Technology, 999078, Macau, China
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, 650093, China
| | - Xinyan Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 610000, China
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, 650093, China
| | - Yangdongping Feng
- School of Medical Humanities, China Medical University, Shenyang, 110122, China
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, 650093, China
| | - Qiang Cao
- School of Medicine, Macau University of Science and Technology, 999078, Macau, China.
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, 650093, China.
| |
Collapse
|
30
|
Enoma D. Genomics in Clinical trials for Breast Cancer. Brief Funct Genomics 2024; 23:325-334. [PMID: 38146120 DOI: 10.1093/bfgp/elad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023] Open
Abstract
Breast cancer (B.C.) still has increasing incidences and mortality rates globally. It is known that B.C. and other cancers have a very high rate of genetic heterogeneity and genomic mutations. Traditional oncology approaches have not been able to provide a lasting solution. Targeted therapeutics have been instrumental in handling the complexity and resistance associated with B.C. However, the progress of genomic technology has transformed our understanding of the genetic landscape of breast cancer, opening new avenues for improved anti-cancer therapeutics. Genomics is critical in developing tailored therapeutics and identifying patients most benefit from these treatments. The next generation of breast cancer clinical trials has incorporated next-generation sequencing technologies into the process, and we have seen benefits. These innovations have led to the approval of better-targeted therapies for patients with breast cancer. Genomics has a role to play in clinical trials, including genomic tests that have been approved, patient selection and prediction of therapeutic response. Multiple clinical trials in breast cancer have been done and are still ongoing, which have applied genomics technology. Precision medicine can be achieved in breast cancer therapy with increased efforts and advanced genomic studies in this domain. Genomics studies assist with patient outcomes improvement and oncology advancement by providing a deeper understanding of the biology behind breast cancer. This article will examine the present state of genomics in breast cancer clinical trials.
Collapse
Affiliation(s)
- David Enoma
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 2500 University Dr NW, Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
31
|
de Pellegars-Malhortie A, Picque Lasorsa L, Mazard T, Granier F, Prévostel C. Why Is Wnt/β-Catenin Not Yet Targeted in Routine Cancer Care? Pharmaceuticals (Basel) 2024; 17:949. [PMID: 39065798 PMCID: PMC11279613 DOI: 10.3390/ph17070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
Collapse
Affiliation(s)
- Auriane de Pellegars-Malhortie
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Laurence Picque Lasorsa
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| | - Thibault Mazard
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
- Medical Oncology Department, ICM, University of Montpellier, CEDEX 5, 34298 Montpellier, France
| | | | - Corinne Prévostel
- IRCM (Montpellier Cancer Research Institute), University of Montpellier, Inserm, ICM (Montpellier Regional Cancer Institute), 34298 Montpellier, CEDEX 5, France; (A.d.P.-M.); (L.P.L.); (T.M.)
| |
Collapse
|
32
|
Lap CJ, Abrahim MS, Nassereddine S. Perspectives and challenges of small molecule inhibitor therapy for FLT3-mutated acute myeloid leukemia. Ann Hematol 2024; 103:2215-2229. [PMID: 37975931 DOI: 10.1007/s00277-023-05545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous clonal disease characterized overall by an aggressive clinical course. The underlying genetic abnormalities present in leukemic cells contribute significantly to the AML phenotype. Mutations in FMS-like tyrosine kinase 3 (FLT3) are one of the most common genetic abnormalities identified in AML, and the presence of these mutations strongly influences disease presentation and negatively impacts prognosis. Since mutations in FLT3 were identified in AML, they have been recognized as a valid therapeutic target resulting in decades of research to develop effective small molecule inhibitor treatment that could improve outcome for these patients. Despite the approval of several FLT3 inhibitors over the last couple of years, the treatment of patients with FLT3-mutated AML remains challenging and many questions still need to be addressed. This review will provide an up-to-date overview of our current understanding of FLT3-mutated AML and discuss what the current status is of the available FLT3 inhibitors for the day-to-day management of this aggressive disease.
Collapse
Affiliation(s)
- Coen J Lap
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Marwa Sh Abrahim
- The George Washington Cancer Center, George Washington University, Washington, DC, USA
| | - Samah Nassereddine
- The George Washington Cancer Center, George Washington University, Washington, DC, USA.
| |
Collapse
|
33
|
Farah E, Kenney M, Warkentin MT, Cheung WY, Brenner DR. Examining external control arms in oncology: A scoping review of applications to date. Cancer Med 2024; 13:e7447. [PMID: 38984669 PMCID: PMC11234289 DOI: 10.1002/cam4.7447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/11/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024] Open
Abstract
OBJECTIVES Randomized controlled trials (RCTs) are the gold standard for evaluating the comparative efficacy and safety of new cancer therapies. However, enrolling patients in control arms of clinical trials can be challenging for rare cancers, particularly in the context of precision oncology and targeted therapies. External Control Arms (ECAs) are a potential solution to address these challenges in clinical research design. We conducted a scoping review to explore the use of ECAs in oncology. METHODS We systematically searched four databases, namely MEDLINE, EMBASE, Web of Science, and Scopus. We screened titles, abstracts, and full texts for eligible articles focusing on patients undergoing therapy for cancer, employing ECAs, and reporting clinical outcomes. RESULTS Of the 629 articles screened, 23 were included in this review. The earliest included studies were published in 1996, while most studies were published in the past 5 years. 44% (10/23) of ECAs were employed in blood-related cancer studies. Geographically, 30% (7/23) of studies were conducted in the United States, 22% (5/23) in Japan, and 9% (2/23) in South Korea. The primary data sources used to construct the ECAs involved pooled data from previous trials (35%, 8/23), administrative health databases (17%, 4/23) and electronic medical records (17%, 4/23). While 52% (12/23) of the studies employed methods to align treatment and ECAs characteristics, 48% (11/23) lacked explicit strategies. CONCLUSION ECAs offer a valuable approach in oncology research, particularly when alternative designs are not feasible. However, careful methodological planning and detailed reporting are essential for meaningful and reliable results.
Collapse
Affiliation(s)
- Eliya Farah
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew Kenney
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Matthew T. Warkentin
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Winson Y. Cheung
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Darren R. Brenner
- Department of Oncology, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health Sciences, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
34
|
Jiang YC, Xu QT, Wang HB, Ren SY, Zhang Y. A novel prognostic signature related to programmed cell death in osteosarcoma. Front Immunol 2024; 15:1427661. [PMID: 39015570 PMCID: PMC11250594 DOI: 10.3389/fimmu.2024.1427661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Background Osteosarcoma primarily affects children and adolescents, with current clinical treatments often resulting in poor prognosis. There has been growing evidence linking programmed cell death (PCD) to the occurrence and progression of tumors. This study aims to enhance the accuracy of OS prognosis assessment by identifying PCD-related prognostic risk genes, constructing a PCD-based OS prognostic risk model, and characterizing the function of genes within this model. Method We retrieved osteosarcoma patient samples from TARGET and GEO databases, and manually curated literature to summarize 15 forms of programmed cell death. We collated 1621 PCD genes from literature sources as well as databases such as KEGG and GSEA. To construct our model, we integrated ten machine learning methods including Enet, Ridge, RSF, CoxBoost, plsRcox, survivalSVM, Lasso, SuperPC, StepCox, and GBM. The optimal model was chosen based on the average C-index, and named Osteosarcoma Programmed Cell Death Score (OS-PCDS). To validate the predictive performance of our model across different datasets, we employed three independent GEO validation sets. Moreover, we assessed mRNA and protein expression levels of the genes included in our model, and investigated their impact on proliferation, migration, and apoptosis of osteosarcoma cells by gene knockdown experiments. Result In our extensive analysis, we identified 30 prognostic risk genes associated with programmed cell death (PCD) in osteosarcoma (OS). To assess the predictive power of these genes, we computed the C-index for various combinations. The model that employed the random survival forest (RSF) algorithm demonstrated superior predictive performance, significantly outperforming traditional approaches. This optimal model included five key genes: MTM1, MLH1, CLTCL1, EDIL3, and SQLE. To validate the relevance of these genes, we analyzed their mRNA and protein expression levels, revealing significant disparities between osteosarcoma cells and normal tissue cells. Specifically, the expression levels of these genes were markedly altered in OS cells, suggesting their critical role in tumor progression. Further functional validation was performed through gene knockdown experiments in U2OS cells. Knockdown of three of these genes-CLTCL1, EDIL3, and SQLE-resulted in substantial changes in proliferation rate, migration capacity, and apoptosis rate of osteosarcoma cells. These findings underscore the pivotal roles of these genes in the pathophysiology of osteosarcoma and highlight their potential as therapeutic targets. Conclusion The five genes constituting the OS-PCDS model-CLTCL1, MTM1, MLH1, EDIL3, and SQLE-were found to significantly impact the proliferation, migration, and apoptosis of osteosarcoma cells, highlighting their potential as key prognostic markers and therapeutic targets. OS-PCDS enables accurate evaluation of the prognosis in patients with osteosarcoma.
Collapse
Affiliation(s)
- Yu-Chen Jiang
- Affiliated Zhongshan Hospital Of Dalian University, Dalian, China
| | - Qi-Tong Xu
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hong-Bin Wang
- Affiliated Zhongshan Hospital Of Dalian University, Dalian, China
| | - Si-Yuan Ren
- Affiliated Zhongshan Hospital Of Dalian University, Dalian, China
| | - Yao Zhang
- Affiliated Zhongshan Hospital Of Dalian University, Dalian, China
| |
Collapse
|
35
|
El-Sayed MM, Bianco JR, Li Y, Fabian Z. Tumor-Agnostic Therapy-The Final Step Forward in the Cure for Human Neoplasms? Cells 2024; 13:1071. [PMID: 38920700 PMCID: PMC11201516 DOI: 10.3390/cells13121071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.
Collapse
Affiliation(s)
| | | | | | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (M.M.E.-S.); (J.R.B.); (Y.L.)
| |
Collapse
|
36
|
Garg P, Pareek S, Kulkarni P, Salgia R, Singhal SS. Nanoengineering Solutions for Cancer Therapy: Bridging the Gap between Clinical Practice and Translational Research. J Clin Med 2024; 13:3466. [PMID: 38929995 PMCID: PMC11204592 DOI: 10.3390/jcm13123466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Nanoengineering has emerged as a progressive method in cancer treatment, offering precise and targeted delivery of therapeutic agents while concurrently reducing overall toxicity. This scholarly article delves into the innovative strategies and advancements in nanoengineering that bridge the gap between clinical practice and research in the field of cancer treatment. Various nanoengineered platforms such as nanoparticles, liposomes, and dendrimers are scrutinized for their capacity to encapsulate drugs, augment drug efficacy, and enhance pharmacokinetics. Moreover, the article investigates research breakthroughs that drive the progression and enhancement of nanoengineered remedies, encompassing the identification of biomarkers, establishment of preclinical models, and advancement of biomaterials, all of which are imperative for translating laboratory findings into practical medical interventions. Furthermore, the integration of nanotechnology with imaging modalities, which amplify cancer detection, treatment monitoring, and response assessment, is thoroughly examined. Finally, the obstacles and prospective directions in nanoengineering, including regulatory challenges and issues related to scalability, are examined. This underscores the significance of fostering collaboration among various entities in order to efficiently translate nanoengineered interventions into enhanced cancer therapies and patient management.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
37
|
Radu P, Kumar G, Cole A, Fameli A, Guthrie M, Annemans L, Geissler J, Italiano A, O’Rourke B, Xoxi E, Steuten L. Evolving assessment pathways for precision oncology medicines to improve patient access: a tumor-agnostic lens. Oncologist 2024; 29:465-472. [PMID: 38630538 PMCID: PMC11144967 DOI: 10.1093/oncolo/oyae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/07/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Genomic and molecular alterations are increasingly important in cancer diagnosis, and scientific advances are opening new treatment avenues. Precision oncology (PO) uses a patient's genomic profile to determine optimal treatment, promising fewer side effects and higher success rates. Within PO, tumor-agnostic (TA) therapies target genomic alterations irrespective of tumor location. However, traditional value frameworks and approval pathways pose challenges which may limit patient access to PO therapies. OBJECTIVES This study describes challenges in assessing PO and TA medicines, explores possible solutions, and provides actionable recommendations to facilitate an iterative life-cycle assessment of these medicines. METHODS After reviewing the published literature, we obtained insights from key stakeholders and European experts across a range of disciplines, through individual interviews and an industry workshop. The research was guided and refined by an international expert committee through 2 sounding board meetings. RESULTS The current challenges faced by PO and TA medicines are multiple and can be demonstrated through real-world examples of the current barriers and opportunities. A life-cycle approach to assessment should be taken, including key actions at the early stages of evidence generation, regulatory and reimbursement stage, as well as payment and adoption solutions that make use of the evolving evidence base. Working toward these solutions to maximize PO medicine value is a shared responsibility and stands to benefit all stakeholders. CONCLUSIONS Our call to action is to expand access to comprehensive genomic testing, foster a learning health care system, enable fast and equitable access to cost-effective treatments, and ultimately improve health outcomes.
Collapse
Affiliation(s)
| | | | - Amanda Cole
- Office of Health Economics, London, United Kingdom
| | | | - Mark Guthrie
- Global Access Strategy Oncology, Roche, San Francisco, CA, United States
| | - Lieven Annemans
- Department of Health Economics, Ghent University, Ghent, Belgium
| | | | - Antoine Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonié, Bordeaux, France
| | | | - Entela Xoxi
- Faculty of Economics, ALTEMS Università Cattolica del Sacro Cuore, Rome, Italy
| | | |
Collapse
|
38
|
Smokovski I, Steinle N, Behnke A, Bhaskar SMM, Grech G, Richter K, Niklewski G, Birkenbihl C, Parini P, Andrews RJ, Bauchner H, Golubnitschaja O. Digital biomarkers: 3PM approach revolutionizing chronic disease management - EPMA 2024 position. EPMA J 2024; 15:149-162. [PMID: 38841615 PMCID: PMC11147994 DOI: 10.1007/s13167-024-00364-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 06/07/2024]
Abstract
Non-communicable chronic diseases (NCDs) have become a major global health concern. They constitute the leading cause of disabilities, increased morbidity, mortality, and socio-economic disasters worldwide. Medical condition-specific digital biomarker (DB) panels have emerged as valuable tools to manage NCDs. DBs refer to the measurable and quantifiable physiological, behavioral, and environmental parameters collected for an individual through innovative digital health technologies, including wearables, smart devices, and medical sensors. By leveraging digital technologies, healthcare providers can gather real-time data and insights, enabling them to deliver more proactive and tailored interventions to individuals at risk and patients diagnosed with NCDs. Continuous monitoring of relevant health parameters through wearable devices or smartphone applications allows patients and clinicians to track the progression of NCDs in real time. With the introduction of digital biomarker monitoring (DBM), a new quality of primary and secondary healthcare is being offered with promising opportunities for health risk assessment and protection against health-to-disease transitions in vulnerable sub-populations. DBM enables healthcare providers to take the most cost-effective targeted preventive measures, to detect disease developments early, and to introduce personalized interventions. Consequently, they benefit the quality of life (QoL) of affected individuals, healthcare economy, and society at large. DBM is instrumental for the paradigm shift from reactive medical services to 3PM approach promoted by the European Association for Predictive, Preventive, and Personalized Medicine (EPMA) involving 3PM experts from 55 countries worldwide. This position manuscript consolidates multi-professional expertise in the area, demonstrating clinically relevant examples and providing the roadmap for implementing 3PM concepts facilitated through DBs.
Collapse
Affiliation(s)
- Ivica Smokovski
- University Clinic of Endocrinology, Diabetes and Metabolic Disorders, Skopje, North Macedonia
- Faculty of Medical Sciences, University Goce Delcev, Stip, North Macedonia
| | - Nanette Steinle
- Veteran Affairs Capitol Health Care Network, Linthicum, MD USA
- University of Maryland School of Medicine, Baltimore, MD USA
| | - Andrew Behnke
- Endocrinology Section, Carilion Clinic, Roanoke, VA USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA USA
| | - Sonu M. M. Bhaskar
- Department of Neurology, Division of Cerebrovascular Medicine and Neurology, National Cerebral and Cardiovascular Centre (NCVC), Suita, Osaka Japan
- Department of Neurology & Neurophysiology, Liverpool Hospital, Ingham Institute for Applied Medical Research and South Western Sydney Local Health District, Sydney, NSW Australia
- NSW Brain Clot Bank, Global Health Neurology Lab & NSW Health Pathology, Sydney, NSW Australia
| | - Godfrey Grech
- Department of Pathology, Faculty of Medicine & Surgery, University of Malta, Msida, Malta
| | - Kneginja Richter
- Faculty of Medical Sciences, University Goce Delcev, Stip, North Macedonia
- CuraMed Tagesklinik Nürnberg GmbH, Nuremberg, Germany
- Technische Hochschule Nürnberg GSO, Nuremberg, Germany
- University Clinic for Psychiatry and Psychotherapy, Paracelsus Medical University, Nuremberg, Germany
| | - Günter Niklewski
- University Clinic for Psychiatry and Psychotherapy, Paracelsus Medical University, Nuremberg, Germany
| | - Colin Birkenbihl
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Paolo Parini
- Cardio Metabolic Unit, Department of Medicine Huddinge, and Department of Laboratory Medicine, Karolinska Institute, and Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Russell J. Andrews
- Nanotechnology & Smart Systems Groups, NASA Ames Research Center, Aerospace Medical Association, Silicon Valley, CA USA
| | - Howard Bauchner
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalized (3P) Medicine, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
39
|
Khan MMM, Munir MM, Woldesenbet S, Khalil M, Endo Y, Katayama E, Altaf A, Dillhoff M, Obeng-Gyasi S, Pawlik TM. Disparities in clinical trial enrollment among patients with gastrointestinal cancer relative to minority-serving and safety-netting hospitals. J Gastrointest Surg 2024; 28:896-902. [PMID: 38555017 DOI: 10.1016/j.gassur.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/06/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND For results to be generalizable to all patients with cancer, clinical trials need to include a diverse patient demographic that is representative of the general population. We sought to characterize the effect of receiving care at a minority-serving hospital (MSH) and/or safety-net hospital on clinical trial enrollment among patients with gastrointestinal (GI) malignancies. METHODS Adult patients with GI cancer who underwent oncologic surgery and were enrolled in institutional-/National Cancer Institute-funded clinical trials between 2012 and 2019 were identified in the National Cancer Database. Multivariable regression was used to assess the relationship between MSH and safety-net status relative to clinical trial enrollment. RESULTS Among 1,112,594 patients, 994,598 (89.4%) were treated at a non-MSH, whereas 117,996 (10.6%) were treated at an MSH. Only 1857 patients (0.2%) were enrolled in a clinical trial; most patients received care at a non-MSH (1794 [96.6%]). On multivariable analysis, the odds of enrollment in a clinical trial were markedly lower among patients treated at an MSH vs non-MSH (odds ratio [OR], 0.32; 95% CI, 0.22-0.46). In addition, even after controlling for receipt of care at MSH, Black patients remained at lower odds of enrollment in a clinical trial than White patients (OR, 0.57; 95% CI, 0.45-0.73; both P < .05). CONCLUSION Overall, clinical trial participation among patients with GI cancer was extremely low. Patients treated at an MSH and high safety-net burden hospitals and Black individuals were much less likely to be enrolled in a clinical trial. Efforts should be made to improve trial enrollment and address disparities in trial representation.
Collapse
Affiliation(s)
- Muhammad Muntazir Mehdi Khan
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Muhammad Musaab Munir
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Selamawit Woldesenbet
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Mujtaba Khalil
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Yutaka Endo
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Erryk Katayama
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Abdullah Altaf
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Mary Dillhoff
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Samilia Obeng-Gyasi
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States
| | - Timothy M Pawlik
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, United States.
| |
Collapse
|
40
|
Nussinov R, Yavuz BR, Jang H. Anticancer drugs: How to select small molecule combinations? Trends Pharmacol Sci 2024; 45:503-519. [PMID: 38782689 PMCID: PMC11162304 DOI: 10.1016/j.tips.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Small molecules are at the forefront of anticancer therapies. Successive treatments with single molecules incur drug resistance, calling for combination. Here, we explore the tough choices oncologists face - not just which drugs to use but also the best treatment plans, based on factors such as target proteins, pathways, and gene expression. We consider the reality of cancer's disruption of normal cellular processes, highlighting why it's crucial to understand the ins and outs of current treatment methods. The discussion on using combination drug therapies to target multiple pathways sheds light on a promising approach while also acknowledging the hurdles that come with it, such as dealing with pathway crosstalk. We review options and provide examples and the mechanistic basis, altogether providing the first comprehensive guide to combinatorial therapy selection.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
41
|
Church AJ, Wakefield CE, Hetherington K, Shern JF. Promise and Perils of Precision Oncology for Patients With Pediatric and Young Adult Sarcomas. Am Soc Clin Oncol Educ Book 2024; 44:e432794. [PMID: 38924707 DOI: 10.1200/edbk_432794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
The completion of multiple national pediatric precision oncology platform trials and the incorporation of standardized molecular profiling into the diagnostic care of pediatric and young adult patients with sarcomas have proven the feasibility and potential of the approach. In this work, we explore the current state of the art of precision oncology for pediatric and young adults with sarcoma. We highlight important lessons learned and the challenges that should be addressed in the next generation of trials. The chapter outlines current efforts to improve standardization of molecular assays, harmonization of data collection, and novel molecular tools such as cell-free DNA analyses. Finally, we discuss the impacts and psychosocial outcomes experienced by patients and communication strategies for providers.
Collapse
Affiliation(s)
- Alanna J Church
- Department of Pathology, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Claire E Wakefield
- School of Clinical Medicine, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Kate Hetherington
- School of Clinical Medicine, Faculty of Medicine, UNSW Sydney, Randwick, NSW, Australia
- Behavioural Sciences Unit, Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Jack F Shern
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
42
|
Tufail M, Wan WD, Jiang C, Li N. Targeting PI3K/AKT/mTOR signaling to overcome drug resistance in cancer. Chem Biol Interact 2024; 396:111055. [PMID: 38763348 DOI: 10.1016/j.cbi.2024.111055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
This review comprehensively explores the challenge of drug resistance in cancer by focusing on the pivotal PI3K/AKT/mTOR pathway, elucidating its role in oncogenesis and resistance mechanisms across various cancer types. It meticulously examines the diverse mechanisms underlying resistance, including genetic mutations, feedback loops, and microenvironmental factors, while also discussing the associated resistance patterns. Evaluating current therapeutic strategies targeting this pathway, the article highlights the hurdles encountered in drug development and clinical trials. Innovative approaches to overcome resistance, such as combination therapies and precision medicine, are critically analyzed, alongside discussions on emerging therapies like immunotherapy and molecularly targeted agents. Overall, this comprehensive review not only sheds light on the complexities of resistance in cancer but also provides a roadmap for advancing cancer treatment.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China; Institute of Oral Precancerous Lesions, Central South University, Changsha, China; Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
43
|
Edsjö A, Russnes HG, Lehtiö J, Tamborero D, Hovig E, Stenzinger A, Rosenquist R. High-throughput molecular assays for inclusion in personalised oncology trials - State-of-the-art and beyond. J Intern Med 2024; 295:785-803. [PMID: 38698538 DOI: 10.1111/joim.13785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
In the last decades, the development of high-throughput molecular assays has revolutionised cancer diagnostics, paving the way for the concept of personalised cancer medicine. This progress has been driven by the introduction of such technologies through biomarker-driven oncology trials. In this review, strengths and limitations of various state-of-the-art sequencing technologies, including gene panel sequencing (DNA and RNA), whole-exome/whole-genome sequencing and whole-transcriptome sequencing, are explored, focusing on their ability to identify clinically relevant biomarkers with diagnostic, prognostic and/or predictive impact. This includes the need to assess complex biomarkers, for example microsatellite instability, tumour mutation burden and homologous recombination deficiency, to identify patients suitable for specific therapies, including immunotherapy. Furthermore, the crucial role of biomarker analysis and multidisciplinary molecular tumour boards in selecting patients for trial inclusion is discussed in relation to various trial concepts, including drug repurposing. Recognising that today's exploratory techniques will evolve into tomorrow's routine diagnostics and clinical study inclusion assays, the importance of emerging technologies for multimodal diagnostics, such as proteomics and in vivo drug sensitivity testing, is also discussed. In addition, key regulatory aspects and the importance of patient engagement in all phases of a clinical trial are described. Finally, we propose a set of recommendations for consideration when planning a new precision cancer medicine trial.
Collapse
Affiliation(s)
- Anders Edsjö
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Hege G Russnes
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Janne Lehtiö
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
- Cancer genomics and proteomics, Karolinska University Hospital, Solna, Sweden
| | - David Tamborero
- Department of Oncology and Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Eivind Hovig
- Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Albrecht Stenzinger
- Institute of Pathology, Division of Molecular Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics and Genomics, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
44
|
Phanthunane C, Pongcharoen S, Pannarunothai S, Roboon J, Phanthunane P, Nontarak J. Precision medicine in Asia enhanced by next-generation sequencing: Implications for Thailand through a scoping review and interview study. Clin Transl Sci 2024; 17:e13868. [PMID: 38924657 PMCID: PMC11197108 DOI: 10.1111/cts.13868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Next-generation sequencing (NGS) significantly enhances precision medicine (PM) by offering personalized approaches to diagnosis, treatment, and prevention of unmet medical needs. Little is known about the current situation of PM in Asia. Thus, we aimed to conduct an overview of the progress and gaps in PM in Asia and enrich it with in-depth insight into the possibilities of future PM in Thailand. This scoping review focused on Asian countries starting with non-cancer studies, including rare and undiagnosed diseases (RUDs), non-communicable diseases (NCDs), infectious diseases (IDs), and pharmacogenomics, with a focus on NGS. Subsequent in-depth interviews with experts in Thailand were performed, and a thematic analysis served as the main qualitative methodology. Out of 2898 searched articles, 387 studies were included after the review. Although most of the studies focused on cancer, 89 (23.0%) studies were related to RUDs (17.1%), NCDs (2.8%), IDs (1.8%), and pharmacogenomics (1.3%). Apart from medicine and related sciences, the studies were mostly composed of PM (61.8%), followed by genetics medicine and bioinformatics. Interestingly, 28% of articles were conducted exclusively within the fields of medicine and related sciences, emphasizing interdisciplinary integration. The experts emphasized the need for sustainability-driven political will, nurturing collaboration, reinforcing computational infrastructure, and expanding the bioinformatic workforce. In Asia, developments of NGS have made remarkable progress in PM. Thailand has extended PM beyond cancer and focused on clinical implementation. We summarized the PM challenges, including equity and efficiency targeting, guided research funding, sufficient sample size, integrated collaboration, computational infrastructure, and sufficient trained human resources.
Collapse
Affiliation(s)
- Chumut Phanthunane
- Division of Medical OncologyChulabhorn Hospital, Chulabhorn Royal AcademyBangkokThailand
| | - Sutatip Pongcharoen
- Department of Medicine, Faculty of MedicineNaresuan UniversityPhitsanulokThailand
| | | | - Jureepon Roboon
- Department of Anatomy, Faculty of Medical ScienceNaresuan UniversityPhitsanulokThailand
- Centre of Excellence in Medical BiotechnologyNaresuan UniversityPhitsanulokThailand
| | - Pudtan Phanthunane
- Department of Economics, Faculty of Business, Economics and CommunicationsNaresuan UniversityPhitsanulokThailand
| | - Jiraluck Nontarak
- Department of Epidemiology, Faculty of Public HealthMahidol UniversityBangkokThailand
| |
Collapse
|
45
|
Xu Y, Xin X, Tao T. Decoding the neurotoxic effects of propofol: insights into the RARα-Snhg1-Bdnf regulatory cascade. Am J Physiol Cell Physiol 2024; 326:C1735-C1752. [PMID: 38618701 PMCID: PMC11371332 DOI: 10.1152/ajpcell.00547.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/26/2023] [Accepted: 01/27/2024] [Indexed: 04/16/2024]
Abstract
The potential neurotoxic effects of propofol, an extensively utilized anesthetic, underline the urgency to comprehend its influence on neuronal health. Insights into the role of the retinoic acid receptor-α, small nucleolar RNA host gene 1, and brain-derived neurotrophic factor (RARα-Snhg1-Bdnf) network can offer significant advancements in minimizing these effects. The study targets the exploration of the RARα and Snhg1 regulatory network's influence on Bdnf expression in the realm of propofol-induced neurotoxicity. Harnessing the Gene Expression Omnibus (GEO) database and utilizing JASPAR and RNA-Protein Interaction Prediction (RPISeq) database for projections, the study embarks on an in-depth analysis employing both in vitro and in vivo models. The findings draw a clear link between propofol-induced neurotoxicity and the amplification of RAR signaling pathways, impacting hippocampal development and apoptosis and leading to increased RARα and Snhg1 and decreased Bdnf. Propofol is inferred to accentuate neurotoxicity by heightening RARα and Snhg1 interactions, culminating in Bdnf suppression.NEW & NOTEWORTHY This study aimed to decode propofol's neurotoxic effects on the regulatory cascade, provide insights into the RARα-Snhg1-Bdnf interaction, apply extensive validation techniques, provide a detailed analysis and exploration of propofol's neurotoxicity, and offer a comprehensive approach to understanding molecular interactions.
Collapse
Affiliation(s)
- Yuhai Xu
- Department of Anesthesiology, Air Force Medical Center, Beijing, People's Republic of China
| | - Xin Xin
- Department of Anesthesiology, Air Force Medical Center, Beijing, People's Republic of China
| | - Tianzhu Tao
- Department of Anesthesiology, Air Force Medical Center, Beijing, People's Republic of China
| |
Collapse
|
46
|
Zhou Z, Lin T, Chen S, Zhang G, Xu Y, Zou H, Zhou A, Zhang Y, Weng S, Han X, Liu Z. Omics-based molecular classifications empowering in precision oncology. Cell Oncol (Dordr) 2024; 47:759-777. [PMID: 38294647 DOI: 10.1007/s13402-023-00912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine. CONCLUSION This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ting Lin
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuang Chen
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Haijiao Zou
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Aoyang Zhou
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
47
|
Lee SY. Using Bayesian statistics in confirmatory clinical trials in the regulatory setting: a tutorial review. BMC Med Res Methodol 2024; 24:110. [PMID: 38714936 PMCID: PMC11077897 DOI: 10.1186/s12874-024-02235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Bayesian statistics plays a pivotal role in advancing medical science by enabling healthcare companies, regulators, and stakeholders to assess the safety and efficacy of new treatments, interventions, and medical procedures. The Bayesian framework offers a unique advantage over the classical framework, especially when incorporating prior information into a new trial with quality external data, such as historical data or another source of co-data. In recent years, there has been a significant increase in regulatory submissions using Bayesian statistics due to its flexibility and ability to provide valuable insights for decision-making, addressing the modern complexity of clinical trials where frequentist trials are inadequate. For regulatory submissions, companies often need to consider the frequentist operating characteristics of the Bayesian analysis strategy, regardless of the design complexity. In particular, the focus is on the frequentist type I error rate and power for all realistic alternatives. This tutorial review aims to provide a comprehensive overview of the use of Bayesian statistics in sample size determination, control of type I error rate, multiplicity adjustments, external data borrowing, etc., in the regulatory environment of clinical trials. Fundamental concepts of Bayesian sample size determination and illustrative examples are provided to serve as a valuable resource for researchers, clinicians, and statisticians seeking to develop more complex and innovative designs.
Collapse
Affiliation(s)
- Se Yoon Lee
- Department of Statistics, Texas A &M University, 3143 TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
48
|
Mack E, Horak P, Fröhling S, Neubauer A. [Precision oncology and molecular tumor boards]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:462-471. [PMID: 38652307 DOI: 10.1007/s00108-024-01689-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/25/2024]
Abstract
Precision oncology is a field of personalized medicine in which tumor biology forms the basis for tailored treatments. The preferred approach currently applied in clinical practice is based on the concept of malignant tumors as genetic diseases that are caused by mutations in oncogenes and tumor suppressors. On the one hand, these can be targeted by molecular drugs, while on the other hand, next-generation sequencing allows for comprehensive analysis of all relevant aberrations, thus enabling the matching of appropriate treatments across entities based on molecular information. Rational molecular therapies are developed and annotated with supporting evidence by molecular tumor boards, which have been established at various academic centers in recent years. Advancing precision oncology to a new standard of care requires improved applicability of personalized molecular therapies and thorough scientific evaluation of precision oncology programs.
Collapse
Affiliation(s)
- Elisabeth Mack
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg und Philipps-Universität Marburg, Marburg, Deutschland.
- Klinik für Hämatologie, Medizinische Onkologie und Palliativmedizin, St. Marienkrankenhaus Siegen, Siegen, Deutschland.
| | - Peter Horak
- Abteilung für Translationale Medizinische Onkologie, Heidelberg, Deutschland
| | - Stefan Fröhling
- Abteilung für Translationale Medizinische Onkologie, Heidelberg, Deutschland
| | - Andreas Neubauer
- Klinik für Hämatologie, Onkologie und Immunologie, Universitätsklinikum Gießen und Marburg, Standort Marburg und Philipps-Universität Marburg, Marburg, Deutschland
| |
Collapse
|
49
|
Hoo R, Chua KLM, Panda PK, Skanderup AJ, Tan DSW. Precision Endpoints for Contemporary Precision Oncology Trials. Cancer Discov 2024; 14:573-578. [PMID: 38571432 DOI: 10.1158/2159-8290.cd-24-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
SUMMARY Traditional endpoints such as progression-free survival and overall survival do not fully capture the pharmacologic and pharmacodynamic effects of a therapeutic intervention. Incorporating mechanism-driven biomarkers and validated surrogate proximal endpoints can provide orthogonal readouts of anti-tumor activity and delineate the relative contribution of treatment components on an individual level, highlighting the limitation of solely relying on aggregated readouts from clinical trials to facilitate go/no-go decisions for precision therapies.
Collapse
Affiliation(s)
- Regina Hoo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer and Therapeutics Research Laboratory, National Cancer Centre Singapore, Singapore
- Genome Institute of Singapore, Singapore
| | - Kevin L M Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Pankaj Kumar Panda
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| | | | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
- Cancer and Therapeutics Research Laboratory, National Cancer Centre Singapore, Singapore
- Genome Institute of Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore
| |
Collapse
|
50
|
Sathian B, van Teijlingen E, Simkhada P, Kabir R, Al Hamad H. Editorial: Evidence-based approaches in aging and public health. Front Public Health 2024; 12:1391432. [PMID: 38638469 PMCID: PMC11025452 DOI: 10.3389/fpubh.2024.1391432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Affiliation(s)
- Brijesh Sathian
- Geriatrics and Long Term Care Department, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
- Faculty of Health and Social Sciences, Bournemouth University, Bournemouth, United Kingdom
| | - Edwin van Teijlingen
- Faculty of Health and Social Sciences, Bournemouth University, Bournemouth, United Kingdom
| | - Padam Simkhada
- Faculty of Health and Social Sciences, Bournemouth University, Bournemouth, United Kingdom
- School of Human and Health Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Russell Kabir
- School of Allied Health, Anglia Ruskin University, Essex, United Kingdom
| | - Hanadi Al Hamad
- Geriatrics and Long Term Care Department, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|