1
|
Zhang H, Jiang X. Importance of clinical trials and contributions to contemporary medicine: commentary. Ann Med 2025; 57:2451190. [PMID: 39781895 PMCID: PMC11721763 DOI: 10.1080/07853890.2025.2451190] [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: 10/06/2023] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Clinical trials are crucial to contemporary medicine, offering valuable insights into the safety, efficacy, and appropriate use of new treatments. They contribute to public health improvements, reduce healthcare costs, uncover new understandings of disease biology, and facilitate the development of therapies for rare conditions. Investing in clinical trials is therefore essential for advancing medical science and enhancing patient outcomes globally.
Collapse
Affiliation(s)
- Haofuzi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Institute of Neurosurgery of People’s Liberation Army of China (PLA), PLA’s Key Laboratory of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Institute of Neurosurgery of People’s Liberation Army of China (PLA), PLA’s Key Laboratory of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
2
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
3
|
Campbell J, Cambrosio A, Basik M. Histology agnosticism: Infra-molecularizing disease? STUDIES IN HISTORY AND PHILOSOPHY OF SCIENCE 2024; 104:14-22. [PMID: 38377771 DOI: 10.1016/j.shpsa.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 11/22/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
The term "molecularization" has been used by historians and sociologists of science to describe the transition from an anatomic view of the body to a submicroscopic one, where health and illness, indeed life itself, are increasingly defined in terms of an individual's "genetic landscape." Here we introduce the notion of the infra-molecular as a way of extending and nuancing the molecularization trope as it applies to the domain of (post)genomic oncology. In particular we look at how infra-molecularity is enacted in practice as part of the so-called "histology-agnostic" turn in clinical cancer research and care. Drawing on fieldwork in North American oncology settings, we analyze how histology agnosticism partially reconfigures knowledge and practice across the linked domains of drug development and clinical trials, therapeutic decision making, and regulation, and the implications of this for an ongoing revision of how we understand the biopathology and temporality of cancer. We show how, in practice, the inframolecular gaze entails a "return" of histology as a modulator of histology-agnostic drugs and background for interpretation of mutational complexity.
Collapse
Affiliation(s)
- Jonah Campbell
- Department of Social Studies of Medicine, McGill University, Montreal, QC, Canada.
| | - Alberto Cambrosio
- Department of Social Studies of Medicine, McGill University, Montreal, QC, Canada.
| | - Mark Basik
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
| |
Collapse
|
4
|
Lee TW, Hunter FW, Tsai P, Print CG, Wilson WR, Jamieson SMF. Clonal dynamics limits detection of selection in tumour xenograft CRISPR/Cas9 screens. Cancer Gene Ther 2023; 30:1610-1623. [PMID: 37684549 PMCID: PMC10721547 DOI: 10.1038/s41417-023-00664-5] [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: 05/31/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023]
Abstract
Transplantable in vivo CRISPR/Cas9 knockout screens, in which cells are edited in vitro and inoculated into mice to form tumours, allow evaluation of gene function in a cancer model that incorporates the multicellular interactions of the tumour microenvironment. To improve our understanding of the key parameters for success with this method, we investigated the choice of cell line, mouse host, tumour harvesting timepoint and guide RNA (gRNA) library size. We found that high gRNA (80-95%) representation was maintained in a HCT116 subline transduced with the GeCKOv2 whole-genome gRNA library and transplanted into NSG mice when tumours were harvested at early (14 d) but not late time points (38-43 d). The decreased representation in older tumours was accompanied by large increases in variance in gRNA read counts, with notable expansion of a small number of random clones in each sample. The variable clonal dynamics resulted in a high level of 'noise' that limited the detection of gRNA-based selection. Using simulated datasets derived from our experimental data, we show that considerable reductions in count variance would be achieved with smaller library sizes. Based on our findings, we suggest a pathway to rationally design adequately powered in vivo CRISPR screens for successful evaluation of gene function.
Collapse
Affiliation(s)
- Tet Woo Lee
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Oncology Therapeutic Area, Janssen Research and Development, Spring House, PA, USA
| | - Peter Tsai
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Cristin G Print
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Raghani NR, Shah DD, Shah TS, Chorawala MR, Patel RB. Combating relapsed and refractory Mantle cell lymphoma with novel therapeutic armamentarium: Recent advances and clinical prospects. Crit Rev Oncol Hematol 2023; 190:104085. [PMID: 37536448 DOI: 10.1016/j.critrevonc.2023.104085] [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: 02/14/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023] Open
Abstract
Mantle cell lymphoma (MCL) is a rare, aggressive subtype of non-Hodgkin's lymphoma (NHL), accounting for 5% of all cases. Due to its virulence factor, it is an incurable disease and keeps relapsing despite an intensive treatment regimen. Advancements in research and drug discovery have shifted the treatment strategy from conventional chemotherapy to targeted agents and immunotherapies. The establishment of the role of Bruton tyrosine kinase led to the development of ibrutinib, a first-generation BTK inhibitor, and its successors. A conditioning regimen based immunotherapeutic agent like ibritumumob, has also demonstrated a viable response with a favorable toxicity profile. Brexucabtagene Autoleucel, the only approved CAR T-cell therapy, has proven advantageous for relapsed/refractory MCL in both children and adults. This article reviews certain therapies that could help update the current approach and summarizes a few miscellaneous agents, which, seldom studied in trials, could alleviate the regression observed in traditional therapies. DATA AVAILABILITY: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Neha R Raghani
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Disha D Shah
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Tithi S Shah
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Rakesh B Patel
- Department of Internal Medicine, Division of Hematology and Oncology, UI Carver College of Medicine: The University of Iowa Roy J and Lucille A Carver College of Medicine, 375 Newton Rd, Iowa City, IA 52242, USA.
| |
Collapse
|
6
|
Gristina V, Pisapia P, Barraco N, Pepe F, Iacono F, La Mantia M, Peri M, Galvano A, Incorvaia L, Badalamenti G, Bazan V, Troncone G, Russo A, Malapelle U. The significance of tissue-agnostic biomarkers in solid tumors: the more the merrier? Expert Rev Mol Diagn 2023; 23:851-861. [PMID: 37552548 DOI: 10.1080/14737159.2023.2245752] [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: 04/22/2023] [Accepted: 08/04/2023] [Indexed: 08/10/2023]
Abstract
INTRODUCTION To date, several emerging biomarkers have gained considerable interest in the field of predictive molecular oncology. The advent of precision medicine has led to the development of innovative drugs targeting rare molecular pathways independently from histology, defined as tissue-agnostic drugs. AREAS COVERED Although there is a lot of promise for this new tissue-agnostic model in the oncological scenario, crucial issues from both the diagnostic and therapeutic standpoint are emerging. This review aims to critically examine the role of tissue-agnostic biomarkers in different solid tumors, focusing on the prevalence and methods of detection of agnostic biomarkers together with drug approvals to guide clinicians in this evolving landscape. EXPERT OPINION To strengthen the framework for tissue-agnostic approvals, the dialogue between regulatory, industrial, and academic parties should be intensified. Critical questions include the development of an efficient network system that can overcome the heterogeneity of patients' inclusion criteria along with the increasingly difficult interpretation of next-generation sequencing (NGS) profiling technologies. Cost-effectiveness and risk-benefit studies are needed in the national context considering the modalities of access to diagnostic tests and reimbursement of treatments.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Nadia Barraco
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Federica Iacono
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Maria La Mantia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Marta Peri
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| |
Collapse
|
7
|
Delgado A, Enkemann S. Three Layers of Personalized Medicine in the Use of Sirolimus and Its Derivatives for the Treatment of Cancer. J Pers Med 2023; 13:jpm13050745. [PMID: 37240915 DOI: 10.3390/jpm13050745] [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: 03/08/2023] [Revised: 04/05/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Rapamycin and its derivatives are mTOR inhibitors which are FDA-approved for use as immunosuppressants and chemotherapeutic agents. These agents are currently approved to treat renal cell carcinomas, soft tissue sarcomas, and other rare tumors. As tumor treatment paradigms are moving away from organ-based drug selection and moving towards tumor characteristics for individualized treatment it is important to identify as many properties as possible that impact the efficacy of the rapalogues. A review of the current literature was conducted to identify enzymes involved in the metabolism of Sirolimus, Everolimus, Ridaforolimus, and Temsirolimus along with characteristics of tumors that predict the efficacy of these agents. This review also sought to establish whether the genetic characteristics of the patient might influence the activity of the rapalogues or lead to side effects from these agents. Current evidence suggests that tumors with mutations in the mTOR signal transduction pathway are sensitive to rapalogue treatment; the rapalogues are metabolized by cytochromes such as CYP3A4, CYP3A5, and CYP2C8 and transported by ABC transporters that are known to vary in activity in individuals; and that tumors can express these transporters and detoxifying enzymes. This results in three levels of genetic analysis that could impact the effectiveness of the mTOR inhibitors.
Collapse
Affiliation(s)
- Andres Delgado
- Aultman Hospital/NEOMED Program 1, Canton, OH 44710, USA
| | - Steven Enkemann
- Edward Via College of Osteopathic Medicine, 350 Howard St., Spartanburg, SC 29303, USA
| |
Collapse
|
8
|
Mansinho A, Fernandes RM, Carneiro AV. Histology-Agnostic Drugs: A Paradigm Shift-A Narrative Review. Adv Ther 2023; 40:1379-1392. [PMID: 36418841 PMCID: PMC10070286 DOI: 10.1007/s12325-022-02362-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022]
Abstract
Cancer diagnosis and therapeutics have been traditionally based on pathologic classification at the organ of origin. The availability of an unprecedented amount of clinical and biologic data provides a unique window of opportunity for the development of new drugs. What was once treated as a homogeneous disease with a one-size-fits-all approach was shown to be a rather heterogeneous condition, with multiple targetable mutations that can vary during the course of the disease. Clinical trial designs have had to adapt to the exponential growth of targetable mechanisms and new agents, with ensuing challenges that are closer to those experienced with rare diseases and orphan medicines. To face these problems, precision/enrichment and other novel trial designs have been developed, and the concept of histology-agnostic targeted therapeutic agents has emerged. Patients are selected for a specific agent based on specific genomic or molecular alterations, with the same compound used to potentially treat a multiplicity of cancers, granted that the actionable driver alteration is present. There are currently approved drugs for such indications, but this approach has raised issues on multiple levels. This review aims to address the challenges of this new concept and provide insights into possible solutions and frameworks on how to tackle them.
Collapse
Affiliation(s)
- André Mansinho
- Serviço de Oncologia Médica, Centro Hospitalar Universitário Lisboa Norte, Hospital de Santa Maria, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ricardo Miguel Fernandes
- Laboratório de Farmacologia Clínica e Terapêutica, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - António Vaz Carneiro
- Instituto de Saúde Baseada na Evidência, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisbon, Portugal.
- BC/CDI, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
9
|
Efficacy Endpoints in Phase II Clinical Trials for Meningioma: An Analysis of Recent Clinical Trials. Ther Innov Regul Sci 2023; 57:603-610. [PMID: 36602756 DOI: 10.1007/s43441-022-00494-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Response Evaluation Criteria in Solid Tumors (RECIST)-based response rates are commonly used as efficacy endpoints in phase II clinical trials for solid tumors. However, no consensus has been reached concerning adequate efficacy endpoints for phase II clinical trials targeting meningioma. Irregularity of lesions after resection, and varying degrees of dysplasia and histologic subtypes make establishing an appropriate efficacy evaluation difficult. METHODS We analyzed primary efficacy endpoints (PEEs) and background factors from 48 trials retrieved from ClinicalTrials.gov ( https://clinicaltrials.gov/ ) using the search criteria "meningioma," "interventional," "phase II," and "study start 4/1/2001 to 3/31/2021." Primary purpose of the study was efficacy endpoint setting in overall population and three subgroups. RESULTS Among 45 PEEs set in the 39 trials included; 33 trials with single PEE, and six trials with double PEEs, 17/45 (38%) trials adopted progression-free survival (PFS) rate, 15/45 (33%) trials response rate (seven Macdonald criteria or modified, three RECIST, three volumetric estimation, one RANO criteria, one unknown), 10/45 (22%) PFS, 1/45 (2%) OS, and 2/45 (4%) other endpoints. Although 26 PEEs were time-to-event endpoints, 19 of the 26 PEEs were single-arm studies. CONCLUSIONS Time-to-event efficacy endpoints were often compared to historical data, and two-dimensional evaluation is more suitable than one-dimensional one. Accumulation of prognostic data is essential to standardize time-to-event efficacy endpoints. Considering the difficulty of setting design for phase II clinical studies targeting meningioma, evaluation might be done with multiple efficacy endpoints.
Collapse
|
10
|
Horgan D, Spanic T, Apostolidis K, Curigliano G, Chorostowska-Wynimko J, Dauben HP, Lal JA, Dziadziuszko R, Mayer-Nicolai C, Kozaric M, Jönsson B, Gutierrez-Ibarluzea I, Fandel MH, Lopert R. Towards Better Pharmaceutical Provision in Europe-Who Decides the Future? Healthcare (Basel) 2022; 10:1594. [PMID: 36011250 PMCID: PMC9408332 DOI: 10.3390/healthcare10081594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/25/2022] Open
Abstract
Significant progress has been achieved in human health in the European Union in recent years. New medicines, vaccines, and treatments have been developed to tackle some of the leading causes of disease and life-threatening illnesses. It is clear that investment in research and development (R&D) for innovative medicines and treatments is essential for making progress in preventing and treating diseases. Ahead of the legislative process, which should begin by the end of 2022, discussions focus on how Europe can best promote the huge potential benefits of new science and technology within the regulatory framework. The challenges in European healthcare were spelled out by the panellists at the roundtable organised by European Alliance for Personalised Medicine (EAPM). Outcomes from panellists' discussions have been summarized and re-arranged in this paper under five headings: innovation, unmet medical need, access, security of supply, adapting to progress, and efficiency. Some of the conclusions that emerged from the panel are a call for a better overall holistic vision of the future of pharmaceuticals and health in Europe and a collaborative effort among all stakeholders, seeing the delivery of medicines as part of a broader picture of healthcare.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
| | - Tanja Spanic
- Europa Donna, The European Breast Cancer Coalition, 20149 Milan, Italy
| | | | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - Joanna Chorostowska-Wynimko
- Department of Genetics and Clinical Immunology, National Institute of Tuberculosis and Lung Diseases, 26 Plocka Str., 01-138 Warsaw, Poland
| | | | - Jonathan A. Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, GROW School of Oncology and Developmental Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, P.O. Box 616, 6211 LK Maastricht, The Netherlands
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy of the Medical University of Gdansk, 80-214 Gdansk, Poland
| | | | - Marta Kozaric
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
| | - Bengt Jönsson
- Department of Economics, Stockholm School of Economics (SSE), P.O. Box 6501, 113 83 Stockholm, Sweden
| | - Iñaki Gutierrez-Ibarluzea
- Department of Knowledge Management and Evaluation of the Basque Foundation for Health Innovation and Research (BIOEF), 48902 Barakaldo, Spain
| | | | - Ruth Lopert
- Organisation for Economic Co-Operation and Development, OECD, 75775 Paris, France
| |
Collapse
|
11
|
Cho D, Cheyne S, Lord SJ, Simes J, Lee CK. Extrapolating evidence for molecularly targeted therapies from common to rare cancers: a scoping review of methodological guidance. BMJ Open 2022; 12:e058350. [PMID: 35820758 PMCID: PMC9274540 DOI: 10.1136/bmjopen-2021-058350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/23/2022] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Cancer is increasingly classified according to biomarkers that drive tumour growth and therapies developed to target them. In rare biomarker-defined cancers, randomised controlled trials to adequately assess targeted therapies may be infeasible. Extrapolating existing evidence of targeted therapy from common cancers to rare cancers sharing the same biomarker may reduce evidence requirements for regulatory approval in rare cancers. It is unclear whether guidelines exist for extrapolation. We sought to identify methodological guidance for extrapolating evidence from targeted therapies used for common cancers to rare biomarker-defined cancers. DESIGN Scoping review. DATA SOURCES Websites of health technology assessment agencies, regulatory bodies, research groups, scientific societies and industry. EBM Reviews-Cochrane Methodology Register and Health Technology Assessment, Embase and MEDLINE databases (1946 to 11 May 2022). ELIGIBILITY CRITERIA Papers proposing a framework or recommendations for extrapolating evidence for rare cancers, small populations and biomarker-defined cancers. DATA EXTRACTION AND SYNTHESIS We extracted framework details where available and guidance for components of extrapolation. We used these components to structure and summarise recommendations. RESULTS We identified 23 papers. One paper provided an extrapolation framework but was not cancer specific. Extrapolation recommendations addressed six distinct components: strategies for grouping cancers as the same biomarker-defined disease; analytical validation requirements of a biomarker test to use across cancer types; strategies to generate control data when a randomised concurrent control arm is infeasible; sources to inform biomarker clinical utility assessment in the absence of prospective clinical evidence; requirements for surrogate endpoints chosen for the rare cancer; and assessing and augmenting safety data in the rare cancer. CONCLUSIONS In the absence of an established framework, our recommendations for components of extrapolation can be used to guide discussions about interpreting evidence to support extrapolation. The review can inform the development of an extrapolation framework for biomarker-targeted therapies in rare cancers.
Collapse
Affiliation(s)
- Doah Cho
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Saskia Cheyne
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Sarah J Lord
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- School of Medicine, The University of Notre Dame Australia, Sydney Campus, Darlinghurst, New South Wales, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, Camperdown, New South Wales, Australia
- Cancer Care Centre, St George Hospital, Kogarah, New South Wales, Australia
| |
Collapse
|
12
|
Personalised selection of experimental treatment in patients with advanced solid cancer is feasible using whole-genome sequencing. Br J Cancer 2022; 127:776-783. [PMID: 35606463 PMCID: PMC9381598 DOI: 10.1038/s41416-022-01841-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Biomarker-guided therapy in an experimental setting has been suggested to improve patient outcomes. However, trial-specific pre-screening tests are time and tissue consuming and complicate the personalised treatment of patients eligible for early-phase clinical trials. In this study the feasibility of whole-genome sequencing (WGS) as a one-test-for-all for guided inclusion in early-phase trials was investigated. METHODS Phase I Molecular Tumor Board (MTB) at the Erasmus MC Cancer Institute reviewed patients with advanced cancer without standard-of-care treatment (SOC) options for a 'fresh-frozen' (FF) tumour biopsy for WGS based on clinical-pathological features. Clinical grade WGS was performed by Hartwig Medical Foundation. MTB matched the patient with a trial, if available. RESULTS From September 2019-March 2021, 31 patients with highly diverse tumour types underwent a tumour biopsy for WGS. The median turnaround time (TAT) was 15 days [10-42 days]. At least one actionable event was found in 84% of the patients (26/31). One-third of the patients (11/31) received matched experimental treatment. CONCLUSIONS WGS on fresh FF biopsies is a feasible tool for the selection of personalised experimental therapy in patients with advanced cancer without SOC options. WGS is now possible in an acceptable TAT and thus could fulfil the role of a universal genomic pre-screening test.
Collapse
|
13
|
Russo A, Incorvaia L, Malapelle U, Del Re M, Capoluongo E, Vincenzi B, Chiari R, Cortesi L, Danesi R, Florena AM, Fontanini G, Gori S, Marchetti A, Normanno N, Pinto C, Sangiolo D, Silvestris N, Tagliaferri P, Tallini G, Cinieri S, Beretta GD. The Tumor-Agnostic Treatment For Patients with Solid Tumors: a Position Paper on behalf of the AIOM- SIAPEC/IAP-SIBIOC-SIF Italian Scientific Societies. Crit Rev Oncol Hematol 2021; 165:103436. [PMID: 34371157 DOI: 10.1016/j.critrevonc.2021.103436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/04/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The personalized medicine is in a rapidly evolving scenario. The identification of actionable mutations is revolutionizing the therapeutic landscape of tumors. The morphological and histological tumor features are enriched by the extensive genomic profiling, and the first tumor-agnostic drugs have been approved regardless of tumor histology, guided by predictive and druggable genetic alterations. This new paradigm of "mutational oncology", presents a great potential to change the oncologic therapeutic scenario, but also some critical aspects need to be underlined. A process governance is mandatory to ensure the genomic testing accuracy and homogeneity, the economic sustainability, and the regulatory issues, ultimately granting the possibility of translating this model in the "real world". In this position paper, based on experts' opinion, the AIOM-SIAPEC-IAP-SIBIOC-SIF Italian Scientific Societies revised the new agnostic biomarkers, the diagnostic technologies available, the current availability of agnostic drugs and their present indication.
Collapse
Affiliation(s)
- Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy.
| | - Lorena Incorvaia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80138, Naples, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ettore Capoluongo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy; CEINGE, Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80131, Naples, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University, 00128, Rome, Italy
| | - Rita Chiari
- Medical Oncology, AULSS 6 Euganea, South Padova Hospital, Monselice, PD, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ada Maria Florena
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Stefania Gori
- Department of Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, University-Foundation, CeSI Biotech Chieti, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Centre, IRCCS-AUSL di Reggio Emilia, Reggio Emilia, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology, Department of Internal Medicine and Oncology (DIMO), University of Bari, Bari, Italy
| | - Pierosandro Tagliaferri
- Medical and Translational Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | | |
Collapse
|