1
|
Sherry AD, Lim B, Ludmir EB, Msaouel P. Misunderstood trial design: randomisation compels comparison. Lancet Oncol 2025; 26:e297. [PMID: 40449502 DOI: 10.1016/s1470-2045(25)00166-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 06/03/2025]
Affiliation(s)
- Alexander D Sherry
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Wang S, Perucca E, Berkovic SF, Perucca P. Precision therapies for genetic epilepsies in 2025: Promises and pitfalls. Epilepsia Open 2025. [PMID: 40411479 DOI: 10.1002/epi4.70065] [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: 02/14/2025] [Revised: 05/04/2025] [Accepted: 05/12/2025] [Indexed: 05/26/2025] Open
Abstract
By targeting the underlying etiology, precision therapies offer an exciting paradigm shift to improve the stagnant outcomes of drug-resistant epilepsies, including developmental and epileptic encephalopathies. Unlike conventional antiseizure medications (ASMs) which only treat the symptoms (seizures) but have no effect on the underlying disease, precision therapies have the potential to suppress not only the seizures but also disabling comorbidities, including cognitive and behavioral abnormalities, which share the same causative mechanisms. Monogenic epilepsies are an attractive target for precision therapies because of their well-defined molecular mechanisms which can be tested in vitro and can be counteracted by specific drugs. Unfortunately, however, for the vast majority of proposed precision therapies, the evidence for their clinical efficacy is either non-existent or limited to uncontrolled observational accounts. Everolimus is the sole precision therapy with a seizure-related indication with class I evidence of efficacy, highlighting the practical and ethical challenges in obtaining high-level evidence. Here, we review the evidence landscape for candidate precision therapies, including repurposed and innovative treatments currently in development, discuss lessons learned from their use, and highlight strategies to improve their application and evaluation in the clinical setting. PLAIN LANGUAGE SUMMARY: Precision therapies offer a new approach to treat drug-resistant monogenic epilepsies, that is, epilepsies caused by a defect in a single gene. Unlike traditional antiseizure medications, precision therapies target the cause of the disease and have the potential to improve not only seizure control but also concomitant conditions such as cognitive and behavioral disorders. To date, the evidence derived from the clinical use of most proposed precision therapies is limited. This review explores current evidence and strategies to advance their development.
Collapse
Affiliation(s)
- Shuyu Wang
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Victoria, Australia
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Melbourne, Victoria, Australia
| | - Emilio Perucca
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Victoria, Australia
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Samuel F Berkovic
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Victoria, Australia
| | - Piero Perucca
- Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Victoria, Australia
- Bladin-Berkovic Comprehensive Epilepsy Program, Department of Neurology, Austin Health, Melbourne, Victoria, Australia
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Neurology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Jiménez-Labaig P, Lorini L, Gurizzan C, Kinloch E, Burton S, Forster MD, Metcalf R, Ferrarotto R, Bossi P, O Leary B, Hanna G, Felip E, Garcia IB, Harrington KJ. Clinical trials for patients with salivary gland cancers: A systematic review of worldwide registers and an evaluation of current challenges. Crit Rev Oncol Hematol 2025; 211:104747. [PMID: 40294876 DOI: 10.1016/j.critrevonc.2025.104747] [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: 12/02/2024] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Clinical trials (CT) are crucial for generating scientific evidence and improving clinical outcomes, but they can be challenging in the context of rare cancers. Salivary gland cancers (SGC) are rare and heterogeneous tumors, without standard-of-care approved systemic therapies. We analyzed completed and ongoing CTs to assess the current state of clinical research activity in the field. METHODS ClinicalTrials.gov, WHO-ICTRP, HealthCanadaCT were searched for antineoplastic pharmacological and interventional CT involving patients with SGC from the trials database creation until August 6th, 2024. CT characteristics and status were collected. RESULTS 134 clinical trials met inclusion criteria. Of these, 78 % were sponsored by non-industry entities. 49 % were conducted at only one site, and 61 % at up to five centers. Only 25 trials (19 %) were multinational, being 15 industry-sponsored, a significantly higher proportion compared to non-industry-sponsored trials(p < 0.01). 16 % CTs were umbrella or basket, and 6 % were randomized, again predominantly industry-sponsored(p < 0.01). Regarding SGC-specific trials, 32 % were open to all patients with SGC, regardless of specific histology. Patients with adenoid cystic, salivary duct, and mucoepidermoid carcinoma had access to 92 %, 66 % and 62 % of trials, respectively. 88 % CT targeted palliative setting, and 38 % incorporated predictive biomarkers. Tyrosine kinase inhibitors were the most studied therapy(26 %), followed by immunotherapy(15 %), chemotherapy and antibody-drug conjugate(12 % each) and androgen-blockade(8 %), among others. CONCLUSION Clinical research for patients with SGC relies mainly in non-industry organisations, most of them limited to run trials in one to five sites, in a single country. Further collaboration between investigators is needed, as well as reconsidering inclusion criteria and trial designs.
Collapse
Affiliation(s)
- Pablo Jiménez-Labaig
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom; Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Luigi Lorini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Cristina Gurizzan
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Emma Kinloch
- Salivary Gland Cancer UK, London, United Kingdom
| | - Sarah Burton
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The International Centre for Recurrent Head and Neck Cancer, United Kingdom
| | - Martin D Forster
- UCL Cancer Institute, London, United Kingdom; University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Robert Metcalf
- Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom; The Cancer Research UK Manchester Institute, United Kingdom
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, United States
| | - Paolo Bossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano, MI, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan 20072, Italy
| | - Ben O Leary
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| | - Glenn Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, United States
| | - Enriqueta Felip
- Lung and Head & Neck Tumors Unit. Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Irene Braña Garcia
- Lung and Head & Neck Tumors Unit. Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Kevin J Harrington
- Head and Neck Unit. The Royal Marsden NHS Foundation Trust, London, United Kingdom; The Institute of Cancer Research, Division of Radiotherapy and Imaging, London, United Kingdom
| |
Collapse
|
4
|
Cooper S, Nicholson S, Crook J, Watkin N, Pettaway C, Barber J, Mitra A, Woodley O, Millin A, Hall E, Pathmanathan A, Penegar S, Burnett S, Spiess P, Miles E, Hoffman K, Yang H, Tree AC. Standardization of radiation therapy to Inguinal and Pelvic Lymph Nodes in Locally Advanced Cancer of the Penis, as Defined by the International Penile Advanced Cancer Trial (InPACT). Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00255-X. [PMID: 40180061 DOI: 10.1016/j.ijrobp.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE InPACT addresses the optimal management of locally advanced penile cancer, aiming to prospectively evaluate the relative benefits and sequencing of surgery, chemotherapy, and chemoradiotherapy. At trial inception, radiation therapy protocols for this rare cancer lacked consistency and standardization, necessitating multicenter, international collaboration to develop comprehensive radiation therapy planning, delivery, and quality assurance guidelines. METHODS AND MATERIALS InPACT has 2 main aims; to establish the efficacy of neoadjuvant chemotherapy or chemoradiotherapy in patients with macroscopically-involved inguinal nodes. Second, to compare prophylactic pelvic lymph node dissection plus chemoradiation to the inguinal and pelvic fields versus chemoradiation alone in patients whose inguinal node histology predicts a high risk of occult pelvic node involvement. The primary outcome measure for the trial is survival time. An international group was convened to achieve consensus on radiation therapy contouring, planning, dose, fractionation, and delivery for this rare cancer. These guidelines have been used throughout the conduct of the trial to date and form part of the radiation therapy quality assurance for each participating center. RESULTS International consensus radiation therapy guidelines were established, encompassing risk status assessment and indications for each treatment region based on radiological and pathologic risk status of nodal basins. Guidance provides a nodal contouring atlas, addresses prepubic fat coverage, and specifies dose fractionation for both neoadjuvant and adjuvant settings, including recommendations for macroscopic disease. Trial recruitment is ongoing. Oncological and toxicity outcomes will be reported in due course. CONCLUSIONS The InPACT radiation therapy guidelines offer a step toward international consensus on contouring for inguino-pelvic radiation therapy in penile cancer.
Collapse
Affiliation(s)
- Sian Cooper
- The Royal Marsden Hospital, London, United Kingdom; The Institute of Cancer Research, London, United Kingdom.
| | - Steve Nicholson
- The Institute of Cancer Research, London, United Kingdom; Mid and South Essex NHS Foundation Trust, United Kingdom
| | - Juanita Crook
- University of British Columbia, BCCancer Center for the Southern Interior, Kelowna, British Columbia
| | - Nick Watkin
- St George's University Hospitals NHS Foundation Trust, United Kingdom
| | | | - Jim Barber
- Velindre University NHS Trust, Cardiff, United Kingdom
| | - Anita Mitra
- University College London Hospitals NHS FT, London, United Kingdom
| | - Owain Woodley
- Velindre University NHS Trust, Cardiff, United Kingdom; National Radiotherapy Trials QA (RTTQA) Group, Northwood, United Kingdom
| | | | - Emma Hall
- The Institute of Cancer Research, London, United Kingdom
| | - Angela Pathmanathan
- The Royal Marsden Hospital, London, United Kingdom; The Institute of Cancer Research, London, United Kingdom
| | - Steven Penegar
- The Institute of Cancer Research, London, United Kingdom
| | | | - Philippe Spiess
- Department of GU Oncology, Moffitt Cancer Center; Tampa, Florida
| | - Elizabeth Miles
- National Radiotherapy Trials QA (RTTQA) Group, Northwood, United Kingdom; East and North Hertfordshire NHS Trust, United Kingdom
| | - Karen Hoffman
- The University of Texas MD Anderson Cancer Center, Texas
| | - Huiqi Yang
- Department of Oncology, Addenbrooke's Hospital NHS Foundation Trust, Hills Rd, Cambridge, UK
| | - Alison C Tree
- The Royal Marsden Hospital, London, United Kingdom; The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
5
|
Lopez-Rey BG, Carot-Sans G, Ouchi D, Torres F, Pontes C. Use of Bayesian approaches in oncology clinical trials: A cross-sectional analysis. Front Pharmacol 2025; 16:1548997. [PMID: 40201693 PMCID: PMC11975924 DOI: 10.3389/fphar.2025.1548997] [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: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Purpose Bayesian approaches may improve the efficiency of trials and accelerate decision-making, but reluctance to depart from traditional frequentist statistics may limit their use. Because oncology trials generally involve severe conditions with no or limited therapeutic options, they are well-suited to applying Bayesian methodologies and are perceived as using these methods often in early phases. Objectives In this study, we aim to describe the use of Bayesian methods and designs in oncology clinical trials in the last 20 years. Method A cross-sectional observational study was conducted to identify oncology clinical trials using Bayesian approaches registered in clinicaltrials.gov between 2004 and 2024. Trials were searched in clinicaltrials.gov, PubMed, and through manual search of cross-references. Results Bayesian trials were retrieved, and their main characteristics were extracted using R and verified manually. Between 2004 and 2024, 384,298 trials were registered in clinicaltrials.gov; we identified 84,850 oncology clinical trials (22%), of which 640 (0.75%) used Bayesian approaches. The adoption of Bayesian trials increased significantly after 2011, but while half of all Bayesian studies started in the last 5 years, this paralleled the overall increase in oncology research rather than an increase in the proportion of Bayesian trials. The majority of Bayesian trials were phase 1 and phase 2 studies, and two-thirds of Bayesian trials with efficacy objectives had single-arm designs, often utilizing binary endpoints, such as overall response, as the primary measure. Conclusion The uptake of Bayesian methods in oncology clinical trials has flattened and is still scarce, and is mostly applied to the analysis of treatment efficacy in single-arm trials with binary endpoints. There is room for further uptake and use of their potential advantages in settings with small populations and severe conditions with unmet needs.
Collapse
Affiliation(s)
- Borja G. Lopez-Rey
- Spanish Agency of Medicines and Medical Devices (AEMPS), Madrid, Spain
- Biostatistics Unit, Medical School, Department of Paediatrics, Obstetrics and Gynaecology and Preventive Medicine and Public Health, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gerard Carot-Sans
- Catalan Health Service, Barcelona, Spain
- Digitalization for the Sustainability of the Healthcare System (DS3), Barcelona, Spain
| | - Dan Ouchi
- Biostatistics Unit, Medical School, Department of Paediatrics, Obstetrics and Gynaecology and Preventive Medicine and Public Health, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ferran Torres
- Biostatistics Unit, Medical School, Department of Paediatrics, Obstetrics and Gynaecology and Preventive Medicine and Public Health, Universitat Autònoma de Barcelona, Barcelona, Spain
- Digitalization for the Sustainability of the Healthcare System (DS3), Barcelona, Spain
| | - Caridad Pontes
- Digitalization for the Sustainability of the Healthcare System (DS3), Barcelona, Spain
- Departament de Farmacologia, de Toxicología i de Terapèutica, Universitat Autònoma de Barcelona, Barcelona, Spain
- Servei de Farmacologia Clínica, Hospital de la Santa Creu i de Sant Pau, Barcelona, Spain
| |
Collapse
|
6
|
Baudin E, Hadoux J, Berruti A, Lamartina L, Texier M, Fassnacht M. Sunitinib for the treatment of phaeochromocytomas and paragangliomas - Authors' reply. Lancet 2025; 405:381-382. [PMID: 39892908 DOI: 10.1016/s0140-6736(24)02783-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025]
Affiliation(s)
- Eric Baudin
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif F-94805, France.
| | - Julien Hadoux
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif F-94805, France
| | - Alfredo Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences, and Publi, Azienda Ospedaliera Spedali Civili di Brescia, Brescia, Italy
| | - Livia Lamartina
- Department of Imaging, Endocrine Oncology Unit, Gustave Roussy, University Paris Saclay, Villejuif F-94805, France
| | - Matthieu Texier
- Office of Biostatistics and Epidemiology, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Inserm, Université Paris-Saclay, CESP U1018, Oncostat, labeled Ligue Contre le Cancer, Villejuif, France
| | - Martin Fassnacht
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Würzburg, Würzburg, Germany; Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Faron M, Naman A, Delahousse J, Hescot S, Hadoux J, Castinetti F, Drui D, Renoult-Pierre P, Libe R, Lamartina L, Leboulleux S, Al-Ghuzlan A, Lombès M, Paci A, Baudin E. Prognostic value of total, free and lipoprotein fraction-bound plasma mitotane levels in advanced adrenocortical carcinoma: a prospective study of the ENDOCAN-COMETE-Cancer network. J Endocrinol Invest 2025; 48:357-367. [PMID: 39172357 DOI: 10.1007/s40618-024-02439-7] [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: 03/26/2024] [Accepted: 08/03/2024] [Indexed: 08/23/2024]
Abstract
PURPOSE Mitotane is the only approved treatment for metastatic adrenocortical carcinoma (ACC). Monitoring plasma levels is recommended, but its predictive value is insufficient. METHODS This prospective study of the French ENDOCAN-COMETE network aimed to investigate the prognostic role of plasma mitotane levels pharmacokinetics and free or bound to lipoprotein fraction measurements during six consecutive months. Lipoprotein fractions were isolated by ultracentrifugation, and mitotane level was determined by HPLC-UV. Total, free, and lipoprotein fraction bound plasma mitotane were monitored every two months for six months with morphological assessment. The primary endpoint was overall survival (OS). RESULTS 21 patients with metastatic ACC were included. Median overall survival was 23 months. The median free mitotane level per patient was 12% (± 7%), and the majority (88%) was bound to lipoprotein fractions. Several pharmacokinetics measures of total mitotane were related to OS: first level at one month (p = 0.026), mean level (p = 0.055), and area under the curve (AUC) (p = 0.048), with higher exposure associated to longer OS. Free mitotane (not bounded) and mitotane bounded to lipoprotein subfraction added no prognostic values. The relationship between the mitotane level and OS suggested a minimum "effective" threshold of 10-15 mg/L or an area under the curve above 100 mg/L/month with no individualized maximum value. CONCLUSION This prospective study did not identify any added prognostic value of free mitotane level over the total level. Early total mitotane level measurements (before 3-6 months) were related to OS with a higher and faster exposure related to more prolonged survival.
Collapse
Affiliation(s)
- M Faron
- Department of Surgical Oncology, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
- INSERM 1018 CESP ONCOSTAT Team, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France.
| | - A Naman
- Nuclear Medicine and Endocrine Unit, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - J Delahousse
- Pharmacology Department, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - S Hescot
- Nuclear Medicine Unit, Institut Curie, 35 Rue Dailly, 92210, Saint Cloud, France
| | - J Hadoux
- Nuclear Medicine and Endocrine Unit, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - F Castinetti
- Department of Endocrinology, Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de La Conception, 147 Boulevard Baille, 13005, Marseille, France
| | - D Drui
- Department of Endocrinology, L'institut du Thorax, CHU Nantes, Bd J Monod Saint Herblain, 44093, Nantes Cedex 1, France
| | - P Renoult-Pierre
- CHRU de Tours Hopital Bretonneau, 2 Boulevard Tonnellée, 37000, Tours, France
| | - R Libe
- Endocan-Comete Network Coordinator, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, 27 Rue du Faubourg Saint Jacques, 75014, Paris, France
| | - L Lamartina
- Nuclear Medicine and Endocrine Unit, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - S Leboulleux
- Nuclear Medicine and Endocrine Unit, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - A Al-Ghuzlan
- Department of Biology and Pathology, Institut Gustave Roussy, 94805, Villejuif, France
| | - M Lombès
- INSERM UMR_S U1185, Fac Med Paris Sud, Université Paris-Saclay, 63 Rue Gabriel Péri, 94276, Le Kremlin Bicêtre, France
| | - A Paci
- Pharmacology Department, Gustave Roussy, 114 Rue Edouard Vaillant, 94805, Villejuif, France
| | - E Baudin
- Nuclear Medicine and Endocrine Unit, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| |
Collapse
|
8
|
Weijers JAM, Verhaegh GW, Lassche G, van Engen-van Grunsven ACH, Driessen CML, van Erp NP, Jonker MA, Schalken JA, van Herpen CML. A randomized phase II trial on the addition of dutasteride to combined androgen blockade therapy versus combined androgen blockade therapy alone in patients with advanced or metastatic salivary duct carcinoma - the DUCT study protocol. BMC Cancer 2024; 24:1174. [PMID: 39304797 PMCID: PMC11415984 DOI: 10.1186/s12885-024-12889-0] [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: 07/25/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Salivary duct carcinoma (SDC) is a rare and aggressive subtype of salivary gland cancer, frequently associated with incurable recurrences and distant metastases (R/M). Proliferation of SDC relies on androgen receptor (AR) signalling, prompting the use of combined androgen blockade (CAB, i.e., luteinizing hormone-releasing hormone agonist and/or AR antagonists) to R/M SDC patients. However, only a subset of patients benefits from such treatments. We have shown that response to CAB is associated with steroid 5α-reductase 1 (SRD5A1) mRNA expression. SRD5A1 catalyses the intracellular conversion of testosterone into the more potent AR-agonist dihydrotestosterone. This conversion can be inhibited by dutasteride, a potent SRD5A1-inhibitor, which is currently prescribed for benign prostatic hyperplasia. We hypothesize that repurposing dutasteride to target AR signalling in SDC could enhance therapeutic response and clinical outcome in SDC patients. METHODS This prospective, open-label, randomized controlled phase II clinical trial, is designed to investigate whether dutasteride as an adjunct drug to CAB improves response rate and clinical outcome in patients with AR-positive R/M SDC. Patients are divided in two cohorts based on their prior systemic treatments. In cohort A, CAB-naïve patients (n = 74) will be randomly assigned to either a control arm (Arm 1) receiving CAB (goserelin 10.8 mg/3m and bicalutamide 50 mg/OD) or an experimental arm (Arm 2) where dutasteride (0.5 mg/OD) is added to the CAB regimen. In cohort B, patients with disease progression after adjuvant or first-line palliative CAB therapy (max. n = 24) will receive goserelin, bicalutamide, and dutasteride to assess whether the addition of dutasteride can overcome therapy resistance. The primary endpoints are the objective response rate and duration of response. Secondary endpoints are progression-free survival, overall survival, clinical benefit rate, quality of life, and safety. Translational research will be performed to explore molecular target expression differences and their correlation with clinical outcome. DISCUSSION The DUCT study addresses an unmet medical need by investigating the repurposing of dutasteride to enhance treatment response and improve clinical outcome for patients with R/M SDC, especially those with limited alternative treatment options, such as HER2-negative cases. By repurposing a registered low-cost drug, this trial's findings could be readily applied into clinical practice. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT05513365. Date of registration: August 24, 2022. PROTOCOL VERSION Current protocol version 4.0, February 21, 2024.
Collapse
Affiliation(s)
- Jetty A M Weijers
- Department of Medical Oncology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, 6525 GA, The Netherlands
| | - Gerald W Verhaegh
- Department of Urology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen, The Netherlands
| | - G Lassche
- Department of Medical Oncology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, 6525 GA, The Netherlands
| | - Adriana C H van Engen-van Grunsven
- Department of Pathology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen, The Netherlands
| | - Chantal M L Driessen
- Department of Medical Oncology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, 6525 GA, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen, The Netherlands
| | - Marianne A Jonker
- Science Department IQ Health, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen, The Netherlands
| | - Jack A Schalken
- Department of Urology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA, Nijmegen, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud Institute for Medical Innovation, Radboud University Medical Center, Geert Grooteplein Zuid 8, Nijmegen, 6525 GA, The Netherlands.
| |
Collapse
|
9
|
Arima M, Inoue H, Misumi A, Tsukamoto S, Matsushita I, Araki S, Ohta M, Takahashi K, Imazato M, Goto T, Aoki Y, Tagawa K, Hirose M, Fujita Y, Yoshida N, Nakao S, Kondo H, Kusuhara K, Kimura K, Hasegawa S, Ikeda Y, Kodama Y, Moritake H, Ochiai M, Ohga S, Kishimoto J, Todaka K, Ieiri I, Sonoda KH. Safety and efficacy of ripasudil eye drops in preterm infants with retinopathy of prematurity: phase 1/2, open label, single-arm trial. Jpn J Ophthalmol 2024; 68:490-499. [PMID: 39060675 DOI: 10.1007/s10384-024-01100-3] [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: 10/27/2023] [Accepted: 05/29/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE To assess the safety and efficacy of ripasudil for retinopathy of prematurity (ROP). STUDY DESIGN Phase 1/2, multicenter, open-label, single-arm, 12-week clinical trial. METHODS Infants born with gestational age (GA) of ≤ 32 weeks or weight of ≤ 1500 g with zone I or II, ≥ stage 1, ROP in both eyes were enrolled. Ripasudil eye drops were administered to patients in both eyes. Phase 1 was a dose-escalation study (once daily for 1 week, then twice daily for 2 weeks); an additional dosing up to 9 weeks was allowed if no safety issues occurred. In phase 2, ripasudil was administered twice daily for up to 12 weeks. Adverse events were assessed. The proportion of patients with type 1 ROP progression, number of days for type 1 ROP progression, and progression to the most advanced ROP stage were estimated. RESULTS Twenty-four infants were enrolled (phase 1, n = 3; phase 2, n = 21). Nineteen and four patients experienced systemic and ocular adverse events, respectively. Efficacy endpoints were not different between the ripasudil and historical control groups. However, in the GA ≤ 27 weeks subgroup, fewer patients progressed to type 1 ROP in the ripasudil than in the historical control group (P = 0.09). In the GA ≤ 27 weeks subgroups, the 25th percentile for the number of days for type 1 ROP progression was 22 days in the historical control group and 44 days in the ripasudil group. CONCLUSION Ripasudil was safe and inhibited/delayed type 1 ROP progression, especially in infants with short GA.
Collapse
Affiliation(s)
- Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan.
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan.
| | - Hirosuke Inoue
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Misumi
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Shoko Tsukamoto
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| | - Itsuka Matsushita
- Department of Ophthalmology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shunsuke Araki
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Manami Ohta
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Kazumasa Takahashi
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Miyuki Imazato
- Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tomoko Goto
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoshinori Aoki
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Koshiro Tagawa
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Masayuki Hirose
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Yuito Fujita
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Noriko Yoshida
- Clinical Research Center, Saga University Hospital, Saga, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, National Kyushu Medical Center, Fukuoka, Japan
| | - Hiroyuki Kondo
- Department of Ophthalmology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yasuhiro Ikeda
- Department of Ophthalmology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yuki Kodama
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Hiroshi Moritake
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Masayuki Ochiai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Junji Kishimoto
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Koji Todaka
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka City, Fukuoka, 812-8582, Japan
| |
Collapse
|
10
|
Creutzberg CL, Kim JW, Eminowicz G, Allanson E, Eberst L, Kim SI, Nout RA, Park JY, Lorusso D, Mileshkin L, Ottevanger PB, Brand A, Mezzanzanica D, Oza A, Gebski V, Pothuri B, Batley T, Gordon C, Mitra T, White H, Howitt B, Matias-Guiu X, Ray-Coquard I, Gaffney D, Small W, Miller A, Concin N, Powell MA, Stuart G, Bookman MA. Clinical research in endometrial cancer: consensus recommendations from the Gynecologic Cancer InterGroup. Lancet Oncol 2024; 25:e420-e431. [PMID: 39214113 DOI: 10.1016/s1470-2045(24)00192-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: 02/23/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 09/04/2024]
Abstract
The Gynecologic Cancer InterGroup (GCIG) Endometrial Cancer Consensus Conference on Clinical Research (ECCC) was held in Incheon, South Korea, Nov 2-3, 2023. The aims were to develop consensus statements for future trials in endometrial cancer to achieve harmonisation on design elements, select important questions, and identify unmet needs. All 33 GCIG member groups participated in the development, refinement, and finalisation of 18 statements within four topic groups, addressing adjuvant treatment in high-risk disease; treatment for metastatic and recurrent disease; trial designs for rare endometrial cancer subgroups and special circumstances; and specific methodology and adaptation for trials in low-resource settings. In addition, eight areas of unmet need were identified. This was the first GCIG Consensus Conference to include patient advocates and an expert on inclusion, diversity, equity, and access to take part in all aspects of the process and output. Four early-career investigators were also selected for participation, ensuring that they represented different GCIG member groups and regions. Unanimous consensus was obtained for 16 of the 18 statements, with 97% concordance for the remaining two. Using the described methodology from previous Ovarian Cancer Consensus Conferences, this conference did not require even one minority statement. The high acceptance rate following active involvement in the preparation, discussion, and refinement of the statements by all representatives confirmed the consensus progress within a global academic setting, and the expectation that the ECCC will lead to greater harmonisation, actualisation, inclusion, and resolution of unmet needs in clinical research for individuals living with and beyond endometrial cancer worldwide.
Collapse
Affiliation(s)
- Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, Netherlands.
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Gemma Eminowicz
- Department of Clinical Oncology, University College London Hospital, London, UK
| | - Emma Allanson
- Division of Obstetrics and Gynaecology, Medical School, University of Western Australia, Perth, WA, Australia
| | - Lauriane Eberst
- Department of Medical Oncology, Institut de Cancérologie de Strasbourg, Strasbourg, France
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Remi A Nout
- Department of Radiation Oncology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Jeong-Yeol Park
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Humanitas San Pio X, Milan, Italy; Humanitas University Rozzano, Milan, Italy
| | - Linda Mileshkin
- Department of Medical Oncology, Peter MacCallum Cancer Centre and University of Melbourne, VIC, Australia
| | | | - Alison Brand
- Department of Gynaecological Oncology, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Delia Mezzanzanica
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Amit Oza
- Division of Medical Oncology and Hematology, UHN - Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Val Gebski
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - Bhavana Pothuri
- Perlmutter Cancer Center, NYU Langone Health, New York, NJ, USA; Diversity and Health Equity for Clinical Trials, GOG-Foundation, Philadelphia, PA USA
| | - Tania Batley
- Ko Ngai Tūhoe te iwi, Kaitauwhiro Mātātahi Mokopuna Ora, Te Pūtahitanga o Te Waipounamu, Christchurch, New Zealand
| | - Carol Gordon
- Canadian Cancer Trials Group, London, ON, Canada
| | - Tina Mitra
- Kolkata Gynecological Oncology Trials and Translational Research Group (KolGOTrg), New Town, Kolkata, India
| | - Helen White
- Peaches Womb Cancer Trust, Manchester, UK; Cancer Research Advocates Forum, London, UK
| | - Brooke Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xavier Matias-Guiu
- Department of Pathology, Hospital U de Bellvitge, University of Barcelona, Barcelona, Spain; Department of Pathology, Hospital U Arnau de Vilanova, University of Lleida, Lleida, Spain
| | | | - David Gaffney
- Huntsman Cancer Institute, Department of Radiation Oncology, University of Utah, Salt Lake City, UT, USA
| | - William Small
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernadin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Austin Miller
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nicole Concin
- Department of Gynaecology and Gynaecological Oncology, Medical University of Vienna, Vienna, Austria; Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthew A Powell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, MO, USA
| | - Gavin Stuart
- Department of Obstetrics and Gynecology, University of British Colombia, Vancouver, BC, Canada
| | | |
Collapse
|
11
|
Stacchiotti S, Bouche G, Herold R, Pantziarka P, Schuster K, Wilson R, Pignatti F, Kasper B. How to develop new systemic treatments in ultra-rare cancers with high unmet needs? The case of alveolar soft-part sarcoma. Eur J Cancer 2024; 202:114003. [PMID: 38479120 DOI: 10.1016/j.ejca.2024.114003] [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: 12/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/21/2024]
Abstract
Developing new drugs or generating evidence for existing drugs in new indications for ultra-rare cancers is complex and carries a high-risk of failure. This gets even harder in ultra-rare tumours, which have an annual incidence of 1 per 1,000,000 population or less. Here, we illustrate the problem of adequate evidence generation in ultra-rare tumours, using Alveolar Soft-Part Sarcomas (ASPS) - an ultra-rare sarcoma newly diagnosed in approximately 60 persons a year in the European Union - as an exemplar case showing challenges in development despite being potentially relevant for classes of agents. We discuss some possible approaches for addressing such challenges, especially focussing on constructive collaboration between academic groups, patients and advocates, drug manufacturers, and regulators to optimise drug development in ultra-rare cancers. This article, written by various European stakeholders, proposes a way forward to ultimately get better options for patients with ultra-rare cancers.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | | | | | | | | | - Roger Wilson
- Sarcoma Patient Advocacy Global Network, Wölfersheim, Germany
| | | | - Bernd Kasper
- Sarcoma Unit, Mannheim Cancer Center (MCC), Mannheim University Medical Center, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
12
|
Yoon KW, Jo J, Lee D. Small intestinal adenocarcinoma accompanied by lynch syndrome: A case report. Medicine (Baltimore) 2023; 102:e35323. [PMID: 37773826 PMCID: PMC10545008 DOI: 10.1097/md.0000000000035323] [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: 06/20/2023] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
RATIONALE Lynch syndrome is caused by germline mutations of DNA mismatch repair genes. A significant risk increase for several types of cancer is one of the characteristics of lynch syndrome. PATIENT CONCERNS A 45-year-old female presented to the emergency department with abdominal pain that had persisted for a month. DIAGNOSES The abdominal and pelvic computed tomography scan showed edematous and thickening of the proximal small bowel wall, as well as dilatation of the proximal bowel and stomach. INTERVENTIONS Tumor resection of the small bowel was performed, and adenocarcinoma was confirmed pathologically. Microsatellite instability was also confirmed. OUTCOMES Postoperative imaging revealed soft tissue lesions with potential for tumor seeding. Two months after the first surgery, a secondary surgery was performed as a result of cancer recurrence. The patient received chemotherapy with capecitabine. The latest computed tomography scan, performed 19 months after the cessation of chemotherapy, did not show any recurrence. LESSONS In the rare incidence of small bowel cancer genetic mutation testing and detailed family history should be actively considered.
Collapse
Affiliation(s)
- Kyoung Won Yoon
- Division of Critical Care, Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Jaemin Jo
- Division of Hemato-Oncology, Department of Internal Medicine, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Republic of Korea
| | - Donghyoun Lee
- Department of Surgery, Jeju National University Hospital, Jeju National University College of Medicine, Republic of Korea
| |
Collapse
|
13
|
Rosenberg N, Stolwijk NN, van den Berg S, Heus JJ, van der Wel V, van Gelder T, Bosch AM, de Visser SJ, Hollak CEM. Development of medicines for rare diseases and inborn errors of metabolism: Toward novel public-private partnerships. J Inherit Metab Dis 2023; 46:806-816. [PMID: 36938792 DOI: 10.1002/jimd.12605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
Medicine development for rare diseases, including inborn errors of metabolism (IEMs) is challenging. Many academic innovations fail to reach the patient, either by stranding in the translational stage or due to suboptimal patient access related to pricing or uncertain effectiveness. Expanding and solidifying the role of the academic in public-private partnerships (PPPs) may present an innovative solution to help overcome these complexities. This narrative review explores the literature on traditional and novel collaborative approaches to medicine development for rare diseases and analyzes examples of PPPs, with a specific focus on IEMs. Several academic institutions have introduced guidelines for socially responsible licensing of innovations for private development. The PPP model offers a more integrative approach toward academic involvement of medicine development. By sharing risks and rewards, failures in the translational stage can be mutually absorbed. If socially responsible terms are not included, however, high pricing can impede patient access. Therefore, we propose a framework for socially responsible PPPs aimed at medicine development for metabolic disorders. This socially responsible PPP framework could stimulate successful and accessible medicine development for IEMs as well as other rare diseases if the establishment of such collaborations includes terms securing joint data ownership and evidence generation, fast access, and socially responsible pricing.
Collapse
Affiliation(s)
- Noa Rosenberg
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Nina N Stolwijk
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Sibren van den Berg
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J Heus
- Innovation eXchange Amsterdam (IXA) Office Amsterdam UMC, Amsterdam, The Netherlands
| | - Vincent van der Wel
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Orfenix B.V., Leiden, The Netherlands
| | - Teun van Gelder
- Department of Clinical Pharmacy & Toxicology, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Annet M Bosch
- Department of Pediatrics, Division of Metabolic Diseases, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Saco J de Visser
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Centre for Future Affordable & Sustainable Therapy development (FAST), The Hague, The Netherlands
| | - Carla E M Hollak
- Medicines for Society (Medicijn voor de Maatschappij), Platform at Amsterdam UMC - University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for Inborn Errors of Metabolism, MetabERN, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Kaiser MF, Hall A, Walker K, Sherborne A, De Tute RM, Newnham N, Roberts S, Ingleson E, Bowles K, Garg M, Lokare A, Messiou C, Houlston RS, Jackson G, Cook G, Pratt G, Owen RG, Drayson MT, Brown SR, Jenner MW. Daratumumab, Cyclophosphamide, Bortezomib, Lenalidomide, and Dexamethasone as Induction and Extended Consolidation Improves Outcome in Ultra-High-Risk Multiple Myeloma. J Clin Oncol 2023; 41:3945-3955. [PMID: 37315268 DOI: 10.1200/jco.22.02567] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 04/05/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
PURPOSE The multicenter OPTIMUM (MUKnine) phase II trial investigated daratumumab, low-dose cyclophosphamide, lenalidomide, bortezomib, and dexamethasone (Dara-CVRd) before and after autologous stem-cell transplant (ASCT) in newly diagnosed patients with molecularly defined ultra-high-risk (UHiR) multiple myeloma (NDMM) or plasma cell leukemia (PCL). To provide clinical context, progression-free survival (PFS) and overall survival (OS) were referenced to contemporaneous outcomes seen in patients with UHiR NDMM treated in the recent Myeloma XI (MyeXI) trial. METHODS Transplant-eligible all-comers NDMM patients were profiled for UHiR disease, defined by presence of ≥2 genetic risk markers t(4;14)/t(14;16)/t(14;20), del(1p), gain(1q), and del(17p), and/or SKY92 gene expression risk signature. Patients with UHiR MM/PCL were offered treatment with Dara-CVRd induction, V-augmented ASCT, extended Dara-VR(d) consolidation, and Dara-R maintenance. UHiR patients treated in MyeXI with carfilzomib, lenalidomide, dexamethasone, and cyclophosphamide, or lenalidomide, dexamethasone, and cyclophosphamide, ASCT, and R maintenance or observation were identified by mirrored molecular screening. OPTIMUM PFS at 18 months (PFS18m) was compared against MyeXI using a Bayesian framework, and patients were followed up to the end of consolidation for PFS and OS. RESULTS Of 412 screened NDMM OPTIMUM patients, 103 were identified as UHiR or PCL and subsequently treated on trial with Dara-CVRd; 117 MyeXI patients identified as UHiR formed the external comparator arm, with comparable clinical and molecular characteristics to OPTIMUM. Comparison of PFS18m per Bayesian framework resulted in a 99.5% chance of OPTIMUM being superior to MyeXI. At 30 months' follow-up, PFS was 77% for OPTIMUM versus 39.8% for MyeXI, and OS 83.5% versus 73.5%, respectively. Extended post-ASCT Dara-VRd consolidation therapy was highly deliverable, with limited toxicity. CONCLUSION Our results suggest that Dara-CVRd induction and extended post-ASCT Dara-VRd consolidation markedly improve PFS for UHiR NDMM patients over conventional management, supporting further evaluation of this strategy.
Collapse
Affiliation(s)
- Martin F Kaiser
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Department of Haematology, The Royal Marsden Hospital, London, United Kingdom
| | - Andrew Hall
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Katrina Walker
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Amy Sherborne
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Ruth M De Tute
- Haematological Malignancy Diagnostic Service, Leeds Cancer Centre, Leeds Teaching Hospitals Trust, Leeds, United Kingdom
| | - Nicola Newnham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sadie Roberts
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Emma Ingleson
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Kristian Bowles
- Department of Haematology, Norfolk and Norwich University Hospitals NHS Trust, Norwich, United Kingdom
| | - Mamta Garg
- Department of Haematology, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Anand Lokare
- Department of Haematology, Birmingham Heartlands, Birmingham, United Kingdom
| | - Christina Messiou
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Department of Haematology, The Royal Marsden Hospital, London, United Kingdom
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Graham Jackson
- Department of Haematology, Newcastle University, Newcastle, United Kingdom
| | - Gordon Cook
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
- Leeds Cancer Centre, Leeds Teaching Hospitals Trust, Leeds, United Kingdom
| | - Guy Pratt
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Roger G Owen
- Haematological Malignancy Diagnostic Service, Leeds Cancer Centre, Leeds Teaching Hospitals Trust, Leeds, United Kingdom
| | - Mark T Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sarah R Brown
- Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Matthew W Jenner
- Department of Haematology, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
15
|
Wex J, Szkultecka-Debek M, Drozd M, King S, Zibelnik N. Exploring the feasibility of using the ICER Evidence Rating Matrix for Comparative Clinical Effectiveness in assessing treatment benefit and certainty in the clinical evidence on orphan therapies for paediatric indications. Orphanet J Rare Dis 2023; 18:193. [PMID: 37474954 PMCID: PMC10360248 DOI: 10.1186/s13023-023-02701-w] [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: 01/04/2023] [Accepted: 04/06/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND The evaluation of clinical evidence takes account of health benefit (efficacy and safety) and the degree of certainty in the estimate of benefit. In orphan indications practical and ethical challenges in conducting clinical trials, particularly in paediatric patients, often limit the available evidence, rendering structured evaluation challenging. While acknowledging the paucity of evidence, regulators and reimbursement authorities compare the efficacy and safety of alternative treatments for a given indication, often in the context of the benefits of other treatments for similar or different conditions. This study explores the feasibility of using the Institute for Clinical and Economic Review (ICER) Evidence Rating Matrix for Comparative Clinical Effectiveness in structured assessment of both the magnitude of clinical benefit (net health benefit, NHB) and the certainty of the effect estimate in a sample of orphan therapies for paediatric indications. RESULTS Eleven systemic therapies with European Medicines Agency (EMA) orphan medicinal product designation, licensed for 16 paediatric indications between January 2017 and March 2020 were identified using OrphaNet and EMA databases and were selected for evaluation with the ICER Evidence Rating Matrix: burosumab; cannabidiol; cerliponase alfa; chenodeoxycholic acid (CDCA); dinutuximab beta; glibenclamide; metreleptin; nusinersen; tisagenlecleucel; velmanase alfa; and vestronidase alfa. EMA European Public Assessment Reports, PubMed, EMBASE, the Cochrane Library, Clinical Key, and conference presentations from January 2016 to April 2021 were searched for evidence on efficacy and safety. Two of the identified therapies were graded as "substantial" NHB: dinutuximab beta (neuroblastoma maintenance) and nusinersen (Type I SMA), and one as "comparable" NHB (CDCA). The NHB grade of the remaining therapies fell between "comparable" and "substantial". No therapies were graded as having negative NHB. The certainty of the estimate ranged from "high" (dinutuximab beta in neuroblastoma maintenance) to "low" (CDCA, metreleptin and vestronidase alfa). The certainty of the other therapies was graded between "low" and "high". The ICER Evidence Rating Matrix overall rating "A" (the highest) was given to two therapies, "B+" to 6 therapies, "C+" to five therapies, and "I" (the lowest) to three therapies. The scores varied between rating authors with mean agreement over all indications of 71.9% for NHB, 56.3% for certainty and 68.8% for the overall rating. CONCLUSIONS Using the ICER Matrix to grade orphan therapies according to their treatment benefit and certainty is feasible. However, the assessment involves subjective judgements based on heterogenous evidence. Tools such as the ICER Matrix might aid decision makers to evaluate treatment benefit and its certainty when comparing therapies across indications.
Collapse
Affiliation(s)
- Jaro Wex
- Global Market Access & HEOR, EUSA Pharma Ltd, Third Floor, Breakspear Park, Breakspear Way, Hemel Hempstead, HP2 4TZ, UK.
| | | | | | | | | |
Collapse
|
16
|
Powell G, Gagnon M, Komarova S, Rauch F, Veilleux LN. Delivering a Home-Based Exercise Program to Youth With Osteogenesis Imperfecta: Protocol for a Comparative-Approach Study. JMIR Res Protoc 2023; 12:e40262. [PMID: 37399052 PMCID: PMC10365614 DOI: 10.2196/40262] [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: 06/13/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is a rare bone fragility disorder associated with muscle weakness. Individuals with OI may therefore benefit from exercise interventions aiming to improve muscle and bone strength. Given the rarity of OI, many patients do not have access to exercise specialists who are familiar with the disorder. As such, telemedicine, the provision of health care through technology to provide care at a distance, may be well suited for this population. OBJECTIVE The main objectives are (1) to investigate the feasibility and cost-effectiveness of 2 telemedicine approaches for the delivery of an exercise intervention for youth with OI and (2) to assess the impact of the exercise intervention on muscle function and cardiopulmonary fitness in youth with OI. METHODS Patients with OI type I (the mildest form of OI; n=12, aged 12-16 years) from a pediatric orthopedic tertiary hospital will be randomized to receive a 12-week remote exercise intervention in either (1) a supervised group (n=6), monitored every session, or (2) a follow-up group (n=6), receiving monthly progress update appointments. Participants will undergo the following pre- and postintervention evaluations: sit-to-stand test, push-up test, sit-up test, single-legged balance test, and a heel-rise test. Both groups will be given the same 12-week exercise regimen, which includes cardiovascular, resistance, and flexibility training. For each exercise training session involving the supervised group, a kinesiologist will provide instructions to participants through live video sessions using a teleconferencing application. On the other hand, the follow-up group will discuss their progress with the kinesiologist every 4 weeks over a teleconferencing video call. Feasibility will be assessed by recruitment, adherence, and completion rates. A cost-effectiveness analysis of both approaches will be computed. Changes in muscle function and cardiopulmonary fitness will be examined between the 2 groups, pre- and postintervention. RESULTS It is anticipated that the supervised group will have higher adherence and completion rates compared to the follow-up group, which may be associated with greater physiological benefits; however, it may not be as cost-effective compared to the follow-up approach. CONCLUSIONS By determining the most feasible telemedicine approach, this study may serve as a basis for providing increased access to specialized adjunct therapies for individuals with rare disorders. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/40262.
Collapse
Affiliation(s)
- Georgia Powell
- Department of Surgery, Division Surgical and Interventional Sciences, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| | - Marianne Gagnon
- Department of Surgery, Division Surgical and Interventional Sciences, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| | - Svetlana Komarova
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Frank Rauch
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
- Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - Louis-Nicolas Veilleux
- Department of Surgery, Division Surgical and Interventional Sciences, McGill University, Montreal, QC, Canada
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| |
Collapse
|
17
|
Hamid M, Joyce CM, Carroll HK, Kenneally C, Mulcahy S, O'Neill MK, Coulter J, O'Reilly S. Challenging gestational trophoblastic disease cases and mimics: An exemplar for the management of rare tumours. Eur J Obstet Gynecol Reprod Biol 2023; 286:76-84. [PMID: 37224702 DOI: 10.1016/j.ejogrb.2023.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVE Rare tumour management is challenging for clinicians as evidence bases are limited and clinical trials are difficult to conduct. It is even more difficult for patients where self-reliance alone is insufficient to overcome the challenges of navigating care which is often poorly evidence based. In Ireland, a national Gestational Trophoblastic Disease (GTD) service was established as one of 3 initiatives for rare tumours by the National Cancer Control Programme. The service has a national clinical lead, a dedicated supportive nursing service and a clinical biochemistry liaison team. This study sought to assess the impact of a GTD centre using national clinical guidelines and integrating and networking with European and International GTD groups on the clinical management of challenging GTD cases and to consider the application of this model of care to other rare tumour management. STUDY DESIGN In this article, we analyse the impact of a national GTD service on five challenging cases, and review how the service affects patient management in this rare tumour type. These cases were selected from a cohort of patients who were voluntarily registered in the service based on the diagnostic management dilemma they posed. RESULTS Case management was impacted by the identification of GTD mimics, the provision of lifesaving treatment of metastatic choriocarcinoma with brain metastasis, networking with international colleagues, the identification of early relapse, the use of genetics to differentiate treatment pathways and prognosis, and supportive supervision of treatment courses of up to 2 years of therapy in a cohort of patients starting or completing families. CONCLUSION The National GTD service could be an exemplar for the management of rare tumours (such as cholangiocarcinoma) in our jurisdiction which could benefit from a similar constellation of supports. Our study demonstrates the importance of a nominated national clinical lead, dedicated nurse navigator support, registration of cases and networking. The impact of our service would be greater if registration was mandatory rather than voluntary. Such a measure would also ensure equity of access for patients to the service, assist in quantifying the need for resourcing and facilitate research to improve outcomes.
Collapse
Affiliation(s)
- M Hamid
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - C M Joyce
- Pregnancy Loss Research Group, Department of Obstetrics & Gynaecology, University College Cork, Cork, Ireland; Department of Biochemistry and Cell Biology, University College Cork, Cork, Ireland; Department of Clinical Biochemistry, Cork University Hospital, Cork, Ireland.
| | - H K Carroll
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland
| | - C Kenneally
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - S Mulcahy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Mary-Kate O'Neill
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - J Coulter
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - S O'Reilly
- Department of Medical Oncology, Cork University Hospital, Cork, Ireland; Cancer Research @UCC, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Beishuizen A, Mellgren K, Andrés M, Auperin A, Bacon CM, Bomken S, Burke GAA, Burkhardt B, Brugieres L, Chiang AKS, Damm-Welk C, d'Amore E, Horibe K, Kabickova E, Khanam T, Kontny U, Klapper W, Lamant L, Le Deley MC, Loeffen J, Macintyre E, Mann G, Meyer-Wentrup F, Michgehl U, Minard-Colin V, Mussolin L, Oschlies I, Patte C, Pillon M, Reiter A, Rigaud C, Roncery L, Salaverria I, Simonitsch-Klupp I, Uyttebroeck A, Verdu-Amoros J, Williams D, Woessmann W, Wotherspoon A, Wrobel G, Zimmermann M, Attarbaschi A, Turner SD. Improving outcomes of childhood and young adult non-Hodgkin lymphoma: 25 years of research and collaboration within the framework of the European Intergroup for Childhood Non-Hodgkin Lymphoma. Lancet Haematol 2023; 10:e213-e224. [PMID: 36858678 DOI: 10.1016/s2352-3026(22)00374-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/01/2022] [Accepted: 11/18/2022] [Indexed: 03/03/2023]
Abstract
The European Intergroup for Childhood Non-Hodgkin Lymphoma (EICNHL) was established 25 years ago with the goal to facilitate clinical trials and research collaborations in the field both within Europe and worldwide. Since its inception, much progress has been made whereby major improvements in outcomes have been achieved. In this Review, we describe the different diagnostic entities of non-Hodgkin lymphoma in children and young adults describing key features of each entity and outlining clinical achievements made in the context of the EICNHL framework. Furthermore, we provide an overview of advances in biopathology with an emphasis on the role of biological studies and how they have shaped available treatments. Finally, for each entity, we describe future goals, upcoming clinical trials, and highlight areas of research that require our focus going forward.
Collapse
Affiliation(s)
- Auke Beishuizen
- Division of Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands; The Netherlands and Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Karin Mellgren
- Department of Paediatric Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mara Andrés
- Department of Pediatric Oncology, University Hospital Le Fe, Valencia, Spain
| | - Anne Auperin
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Chris M Bacon
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Wolfson Childhood Cancer Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Bomken
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Wolfson Childhood Cancer Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - G A Amos Burke
- Department of Paediatric Haematology, Oncology and Palliative Care, Cambridge University Hospitals NHS Foundation Trust, Cambridge
| | - Birgit Burkhardt
- Department of Pediatric Hematology, Oncology, and BMT, University Hospital Muenster, Münster, Germany
| | - Laurence Brugieres
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alan K S Chiang
- Department of Pediatrics & AdolescentMedicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Christine Damm-Welk
- Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Emanuele d'Amore
- Department of Pathological Anatomy, San Bortolo Hospital, Vicenza, Italy
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Edita Kabickova
- Department of Pediatric Hematology and Oncology, Charles University & University Hospital Motol, Prague, Czech Republic
| | - Tasneem Khanam
- Department of Paediatric Haematology, Oncology and Palliative Care, Cambridge University Hospitals NHS Foundation Trust, Cambridge
| | - Udo Kontny
- Section of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatric and Adolescent Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section, University of Schleswig-Holstein, Kiel, Germany
| | - Laurence Lamant
- Université Toulouse III-Paul Sabatier, Laboratoire d'Excellence Toulouse Cancer-TOUCAN, Équipe Labellisée La Ligue Contre Le Cancer, Inserm, Toulouse, France
| | | | - Jan Loeffen
- Division of Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elizabeth Macintyre
- Onco-hematology, Université Paris Cité and Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Georg Mann
- Pediatric Hematology and Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Friederike Meyer-Wentrup
- Division of Hemato-Oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Ulf Michgehl
- Department of Paediatric Haematology, Oncology and Palliative Care, Cambridge University Hospitals NHS Foundation Trust, Cambridge
| | - Veronique Minard-Colin
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lara Mussolin
- Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy; Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, Padova University Hospital, Padova, Italy
| | - Ilske Oschlies
- Institute of Pathology, Hematopathology Section, University of Schleswig-Holstein, Kiel, Germany
| | - Catherine Patte
- Department of Pediatric and Adolescent Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Marta Pillon
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Maternal and Child Health Department, Padova University Hospital, Padova, Italy
| | - Alfred Reiter
- Department of Pediatric Hematology and Oncology, Justus Liebig-University Giessen, Giessen, Germany
| | - Charlotte Rigaud
- Department of Pediatric Hematology, Oncology, and BMT, University Hospital Muenster, Münster, Germany
| | - Leila Roncery
- St Anna Children's Hospital, Department of Paediatric Haematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Itziar Salaverria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Anne Uyttebroeck
- Department of Pediatric Hematology and Oncology, University Hospital Leuven,KU Leuven, Leuven, Belgium
| | - Jaime Verdu-Amoros
- Department of Pediatric Hematology and Oncology, University Hospital Valencia, Valencia, Spain
| | - Denise Williams
- Wolfson Childhood Cancer Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Wilhelm Woessmann
- Pediatric Hematology and Oncology, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Grazyna Wrobel
- Bone Marrow Transplantation and Pediatric Hematology and Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Martin Zimmermann
- Hannover Medical School, Department of Pediatric Hematology and Oncology, Hannover, Germany
| | - Andishe Attarbaschi
- St Anna Children's Hospital, Department of Paediatric Haematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK; Central European Institute for Technology, Masaryk University, Brno, Czech Republic.
| | | |
Collapse
|
19
|
Accrual-Monitoring Practices for Various Disease Trials among AACI Member Cancer Centers. Clin Pract 2022; 12:692-700. [PMID: 36136866 PMCID: PMC9498538 DOI: 10.3390/clinpract12050072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Progress in the management of rare diseases, including rare cancers, is dependent upon clinical trials; however, as many as 32% of rare-disease trials go uncompleted or unpublished due to insufficient accrual. Monitoring practices may differ between institutions. We sought to survey the regulatory standards for various trial types among major U.S. cancer centers. A 10-question survey was designed using Qualtrics assessment software. The survey was sent via email to an internal server of member institutions of the Association of American Cancer Institutes (AACI). Of 103 AACI centers, 31% completed the survey (n = 32). Respondents differed in their definitions of a rare disease, minimum expectations for rare tumor studies, and frequency of accrual monitoring by their institutional Protocol Review and Monitoring Committee. Seventy-three percent of respondents did not close trials based on low accrual. Strategies to optimize accrual included investigator incentives for high accrual and penalties for low accrual in 37% and 13% of respondents, respectively.
Collapse
|
20
|
Azoulay L. Rationale, Strengths, and Limitations of Real-World Evidence in Oncology: A Canadian Review and Perspective. Oncologist 2022; 27:e731-e738. [PMID: 35762676 PMCID: PMC9438907 DOI: 10.1093/oncolo/oyac114] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/26/2022] [Indexed: 11/14/2022] Open
Abstract
Randomized controlled trials (RCTs) continue to be the basis for essential evidence regarding the efficacy of interventions such as cancer therapies. Limitations associated with RCT designs, including selective study populations, strict treatment regimens, and being time-limited, mean they do not provide complete information about an intervention’s safety or the applicability of the trial’s results to a wider range of patients seen in real-world clinical practice. For example, recent data from Alberta showed that almost 40% of patients in the province’s cancer registry would be trial-ineligible per common exclusion criteria. Real-world evidence (RWE) offers an opportunity to complement the RCT evidence base with this kind of information about safety and about use in wider patient populations. It is also increasingly recognized for being able to provide information about an intervention’s effectiveness and is considered by regulators as an important component of the evidence base in drug approvals. Here, we examine the limitations of RCTs in oncology research, review the different types of RWE available in this area, and discuss the strengths and limitations of RWE for complementing RCT oncology data.
Collapse
Affiliation(s)
- Laurent Azoulay
- Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montréal, Québec, Canada.,Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montréal, Québec, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montréal, Québec, Canada
| |
Collapse
|
21
|
Crook J, Black PC, Spiess PE, Pettaway C. The first International Penile Advanced Cancer Trial (InPACT) opens in Canada! Why additional centers are needed. Can Urol Assoc J 2022; 16:70-71. [PMID: 35133267 PMCID: PMC8932418 DOI: 10.5489/cuaj.7704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Penile Cancer is rare in Canada and the Western world. For this reason, penile cancer lags well behind most other malignancies in the development of evidence-based guidelines for management. This dilemma was the basis for the international cooperation to develop InPACT: the International Penile Advanced Cancer Trial (NCT02305654), sponsored by both the National Cancer Institute (lead by ECOG-ACRIN Cooperative Group) and Cancer Research of the United Kingdom (CRUK). The aim of InPACT is to determine prospectively the potential benefits of chemotherapy or chemoradiation in addition to lymph node dissection in patients with clinical evidence of inguinal lymph node metastases. InPACT is now open in Canada. We will review the eligibility and randomizations.
Collapse
Affiliation(s)
- Juanita Crook
- BCCancer, University of British Columbia, Kelowna, BC, Canada
| | - Peter C. Black
- Vancouver Prostate Center, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
22
|
Wang J, Zhang S, Ahn C. Sample size estimation for comparing rates of change in K-group repeated binary measurements studies. COMMUN STAT-THEOR M 2021. [DOI: 10.1080/03610926.2020.1736302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Jijia Wang
- Department of Applied Clinical Research, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Song Zhang
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Chul Ahn
- Department of Population and Data Sciences, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
23
|
Locati LD, Cavalieri S, Bergamini C, Resteghini C, Colombo E, Calareso G, Mariani L, Quattrone P, Alfieri S, Bossi P, Platini F, Capone I, Licitra L. Abiraterone Acetate in Patients With Castration-Resistant, Androgen Receptor-Expressing Salivary Gland Cancer: A Phase II Trial. J Clin Oncol 2021; 39:4061-4068. [PMID: 34597119 PMCID: PMC8677956 DOI: 10.1200/jco.21.00468] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The activity of androgen-deprivation therapy (ADT) in androgen receptor-positive (AR+) salivary gland carcinomas (SGCs) has been established in the past few years. Second-line treatment in castration-resistant patients is still unknown. We investigated the activity of abiraterone acetate as second-line treatment in ADT-resistant, AR+ patients with SGC. METHODS This was a single-institution phase II trial. A two-stage Simon's design was applied. The primary end point was confirmed objective response rate. Secondary end points were disease control rate, safety, progression-free survival, and overall survival. Patients were eligible when the following criteria were met: histologic diagnosis of AR-overexpressing SGC, measurable disease according to RECIST 1.1, clinical and/or radiologic progression on ADT, suppressed serum testosterone, and no limits for the number of previous chemotherapy lines. All patients received abiraterone 1 g daily plus prednisone 10 mg and luteinizing hormone-releasing hormone agonist until progression or unacceptable toxicities. RESULTS From 2015 to 2019, 24 AR+ patients with SGC (23 men; median age 65.8 years) were treated within the study. The overall response rate was 21% (5 partial responses), with a disease control rate of 62.5%. The median duration of response was 5.82 months. Median progression-free survival was 3.65 months (95% CI, 1.94 to 5.89), and median overall survival was 22.47 months (95% CI, 6.74 to not reached). Objective response to previous ADT did not correlate with the activity of abiraterone. Adverse events (AEs) were recorded in 22 cases (92%) with grade 3 AEs in six patients (25%): fatigue (two), flushing (one), supraventricular tachycardia (one), and two non-drug-related AEs. No drug-related grade 4 or 5 AEs were recorded. CONCLUSION Abiraterone plus luteinizing hormone-releasing hormone agonist is active and safe as a second-line option in AR-expressing, castration-resistant SGC.
Collapse
Affiliation(s)
- Laura D Locati
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cristiana Bergamini
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Resteghini
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Colombo
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppina Calareso
- Radiology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luigi Mariani
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Pasquale Quattrone
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Bossi
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Platini
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Iolanda Capone
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Guren MG, Sebag-Montefiore D, Franco P, Johnsson A, Segelov E, Deutsch E, Rao S, Spindler KLG, Arnold D. Treatment of Squamous Cell Carcinoma of the Anus, Unresolved Areas and Future Perspectives for Research: Perspectives of Research Needs in Anal Cancer. Clin Colorectal Cancer 2021; 20:279-287. [PMID: 34645589 DOI: 10.1016/j.clcc.2021.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023]
Abstract
Anal cancer is a relatively rare, mostly HPV-related cancer. The curative treatment consists of concurrent chemoradiation delivered with modern radiotherapy techniques. The prognosis for most patients with early localized disease is very favourable; however patients with locally advanced disease and/or HPV negative tumours are at higher risk of locoregional and distant treatment failure. Tailored approaches are presently being investigated to determine the most suitable regimen in terms of radiotherapy dose prescription, target volume selection, normal tissue avoidance, and combination therapy. Metastatic anal cancer is treated with chemotherapy aiming at prolonged survival. The role of immune therapy in the clinical setting is being investigated. There is little knowledge on the biology of anal cancer, and an urgent need for more clinical and translational research dedicated to this disease. In this article, the evidence-base for the current treatment is briefly reviewed, and perspectives on future research needs are high-lighted.
Collapse
Affiliation(s)
| | | | - Pierfrancesco Franco
- Department of Translational Medicine, University of Eastern Piedmont and Department of Radiation Oncology, AOU ''Maggiore della Carità,'' Novara, Italy
| | - Anders Johnsson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Eva Segelov
- School of Clinical Sciences, Faculty of Medicine, Monash University, Clayton, Australia and Department of Oncology, Monash Health Clayton, Australia
| | | | - Sheela Rao
- GI Unit, Royal Marsden Hospital, London, UK
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, AK Altona, Hamburg, Germany
| |
Collapse
|
25
|
Lainé A, Hanvic B, Ray-Coquard I. Importance of guidelines and networking for the management of rare gynecological cancers. Curr Opin Oncol 2021; 33:442-446. [PMID: 34172594 DOI: 10.1097/cco.0000000000000760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Guidelines are essential to support appropriate medical management. The objective of our paper is to highlight the need for such recommendations, to reinforce strategies in place and to promote the creation of multidisciplinary networks to provide the most appropriate care to patients and to improve it. RECENT FINDINGS Gynecological rare cancers are not that rare since they represent around 50% of all gynecological cancers. Surgery remains the cornerstone of management for all subtypes. Apart from malignant ovarian germ-cell tumors for which conventional chemotherapy was proven to be efficient, response to standard systemic treatment for other gynecological cancers are disappointing. Better understanding of these pathologies is needed and requires more adapted management. SUMMARY Rare cancers are substantially heterogeneous but raise the need of integrating new cases in dedicated networks to enhance and homogenize medical practices. Centralized diagnosis, improved medical practice based on regularly updated international guidelines, and inclusion in innovative clinical trials linked to preclinical studies are essential to contribute to the promotion of improvement in patient care.
Collapse
Affiliation(s)
| | - Brunhilde Hanvic
- Centre Léon Bérard, University Claude Bernard Lyon I, Lyon
- Institut de cancérologie de Lorraine, Nancy, France
| | | |
Collapse
|
26
|
Gagnon M, Marino Merlo G, Yap R, Collins J, Elfassy C, Sawatzky B, Marsh J, Hamdy R, Veilleux LN, Dahan-Oliel N. Using Telerehabilitation to Deliver a Home Exercise Program to Youth With Arthrogryposis: Single Cohort Pilot Study. J Med Internet Res 2021; 23:e27064. [PMID: 34255680 PMCID: PMC8292936 DOI: 10.2196/27064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/13/2021] [Accepted: 05/06/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Arthrogryposis multiplex congenita (AMC) is characterized by joint contractures and muscle weakness, which limit daily activities. Youths with AMC require frequent physical therapeutic follow-ups to limit the recurrence of contractures and maintain range of motion (ROM) and muscle strength; however, access to specialized care may be limited because of geographical distance. Telerehabilitation can offer a potential solution for delivering frequent follow-ups for youth with AMC, but research on the use of telerehabilitation in children with musculoskeletal disorders is scarce. OBJECTIVE The study aims to evaluate the feasibility of delivering a home exercise program (HEP) by using telerehabilitation for youth with AMC. We also aim to explore the effectiveness of the HEP as a secondary aim. METHODS Youths aged between 8 and 21 years with AMC were recruited at the Shriners Hospitals for Children-Canada. The participants completed baseline and post-HEP questionnaires (the Physical Activity Questionnaire for Adolescents, Pediatrics Outcomes Data Collection Instrument, and Adolescent and Pediatric Pain Tool), and clinicians assessed their active ROM using a virtual goniometer. Clinicians used the Goal Attainment Scale with the participants to identify individualized goals to develop a 12-week HEP and assess the achievement of these goals. Follow-ups were conducted every 3 weeks to adjust the HEP. Data on withdrawal rates and compliance to the HEP and follow-ups were collected to assess the feasibility of this approach. The interrater reliability of using a virtual goniometer was assessed using the intraclass correlation coefficient and associated 95% CI. Nonparametric tests were used to evaluate feasibility and explore the effectiveness of the HEP. RESULTS Of the 11 youths who were recruited, 7 (median age: 16.9 years) completed the HEP. Of the 47 appointments scheduled, 5 had to be rescheduled in ≤24 hours. The participants performed their HEP 2.04 times per week (95% CI 1.25-4.08) and reported good satisfaction with the approach. A general intraclass correlation coefficient of 0.985 (95% CI 0.980-0.989) was found for the web-based ROM measurement. Individualized goals were related to pain management; endurance in writing, standing, or walking; sports; and daily activities. In total, 12 of the 15 goals set with the participants were achieved. Statistically significant improvements were observed in the pain and comfort domain of the Pediatrics Outcomes Data Collection Instrument (preintervention: median 71; 95% CI 34-100; postintervention: median 85; 95% CI 49-100; P=.08) and Physical Activity Questionnaire for Adolescents (preintervention: median 1.62; 95% CI 1.00-2.82; postintervention: median 2.32; 95% CI 1.00-3.45; P=.046). CONCLUSIONS The remote delivery of an HEP for youth with AMC is feasible. Promising results were found for the effectiveness of the HEP in helping youths with AMC to achieve their goals. The next step will be to assess the effectiveness of this exercise intervention in a randomized controlled trial. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR2-10.2196/18688.
Collapse
Affiliation(s)
- Marianne Gagnon
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,Department of Surgery, McGill University, Montreal, QC, Canada
| | | | - Rita Yap
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| | - Jessica Collins
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| | - Caroline Elfassy
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,School of Physical and Occupational Therapy, McGill University, Montreal, QC, Canada
| | - Bonita Sawatzky
- Department of Orthopedics, University of British Columbia, Vancouver, BC, Canada
| | - Jacquelyn Marsh
- School of Physical Therapy, Western University, London, ON, Canada
| | - Reggie Hamdy
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,Division of Paediatric Orthopaedics, Department of Paediatric Surgery, Montreal Children Hospital, Montreal, QC, Canada
| | - Louis-Nicolas Veilleux
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,Department of Surgery, McGill University, Montreal, QC, Canada
| | - Noémi Dahan-Oliel
- Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,School of Physical and Occupational Therapy, McGill University, Montreal, QC, Canada
| |
Collapse
|
27
|
Spreafico A, Hansen AR, Abdul Razak AR, Bedard PL, Siu LL. The Future of Clinical Trial Design in Oncology. Cancer Discov 2021; 11:822-837. [PMID: 33811119 PMCID: PMC8099154 DOI: 10.1158/2159-8290.cd-20-1301] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
Clinical trials represent a fulcrum for oncology drug discovery and development to bring safe and effective medicines to patients in a timely manner. Clinical trials have shifted from traditional studies evaluating cytotoxic chemotherapy in largely histology-based populations to become adaptively designed and biomarker-driven evaluations of molecularly targeted agents and immune therapies in selected patient subsets. This review will discuss the scientific, methodological, practical, and patient-focused considerations to transform clinical trials. A call to action is proposed to establish the framework for next-generation clinical trials that strikes an optimal balance of operational efficiency, scientific impact, and value to patients. SIGNIFICANCE: The future of cancer clinical trials requires a framework that can efficiently transform scientific discoveries to clinical utility through applications of innovative technologies and dynamic design methodologies. Next-generation clinical trials will offer individualized strategies which ultimately contribute to globalized knowledge and collective learning, through the joint efforts of all key stakeholders including investigators and patients.
Collapse
Affiliation(s)
- Anna Spreafico
- Division of Medical Oncology and Hematology, Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Aaron R Hansen
- Division of Medical Oncology and Hematology, Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Albiruni R Abdul Razak
- Division of Medical Oncology and Hematology, Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
28
|
Colen RR, Rolfo C, Ak M, Ayoub M, Ahmed S, Elshafeey N, Mamindla P, Zinn PO, Ng C, Vikram R, Bakas S, Peterson CB, Rodon Ahnert J, Subbiah V, Karp DD, Stephen B, Hajjar J, Naing A. Radiomics analysis for predicting pembrolizumab response in patients with advanced rare cancers. J Immunother Cancer 2021; 9:e001752. [PMID: 33849924 PMCID: PMC8051405 DOI: 10.1136/jitc-2020-001752] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We present a radiomics-based model for predicting response to pembrolizumab in patients with advanced rare cancers. METHODS The study included 57 patients with advanced rare cancers who were enrolled in our phase II clinical trial of pembrolizumab. Tumor response was evaluated using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 and immune-related RECIST (irRECIST). Patients were categorized as 20 "controlled disease" (stable disease, partial response, or complete response) or 37 progressive disease). We used 3D-slicer to segment target lesions on standard-of-care, pretreatment contrast enhanced CT scans. We extracted 610 features (10 histogram-based features and 600 second-order texture features) from each volume of interest. Least absolute shrinkage and selection operator logistic regression was used to detect the most discriminatory features. Selected features were used to create a classification model, using XGBoost, for the prediction of tumor response to pembrolizumab. Leave-one-out cross-validation was performed to assess model performance. FINDINGS The 10 most relevant radiomics features were selected; XGBoost-based classification successfully differentiated between controlled disease (complete response, partial response, stable disease) and progressive disease with high accuracy, sensitivity, and specificity in patients assessed by RECIST (94.7%, 97.3%, and 90%, respectively; p<0.001) and in patients assessed by irRECIST (94.7%, 93.9%, and 95.8%, respectively; p<0.001). Additionally, the common features of the RECIST and irRECIST groups also highly predicted pembrolizumab response with accuracy, sensitivity, specificity, and p value of 94.7%, 97%, 90%, p<0.001% and 96%, 96%, 95%, p<0.001, respectively. CONCLUSION Our radiomics-based signature identified imaging differences that predicted pembrolizumab response in patients with advanced rare cancer. INTERPRETATION Our radiomics-based signature identified imaging differences that predicted pembrolizumab response in patients with advanced rare cancer.
Collapse
Affiliation(s)
- Rivka R Colen
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Christian Rolfo
- Department of Thoracic Medical Oncology, University of Maryland, Baltimore, Maryland, USA
| | - Murat Ak
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mira Ayoub
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sara Ahmed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nabil Elshafeey
- Department of Breast Imaging, Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Priyadarshini Mamindla
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Pascal O Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Chaan Ng
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Raghu Vikram
- Abdominal Imaging Department, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Spyridon Bakas
- Radiology, Pathology, and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christine B Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joud Hajjar
- Section of Immunology, Allergy and Retrovirology, Baylor College of Medicine, Houston, Texas, USA
- William T Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
29
|
Krendyukov A, Singhvi S, Zabransky M. Value of Adaptive Trials and Surrogate Endpoints for Clinical Decision-Making in Rare Cancers. Front Oncol 2021; 11:636561. [PMID: 33763370 PMCID: PMC7982798 DOI: 10.3389/fonc.2021.636561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Despite high-level endorsement, the number of adaptive Phase II/III trials in rare cancers needs to be improved, with better understanding of their value for clinical decisions in daily practice. This paper describes approaches to trial design in rare cancers, which has been supplemented by a search of ClinicalTrials.gov for adaptive trial designs in rare cancer. In addition, an online survey of 3,200 oncologists was conducted. Practicing physicians were questioned on the importance of different evidence levels, types of adaptive trial design, and categories of surrogate endpoints for clinical decision making. The results of the online survey revealed that evidence from Phase II/III trials with an adaptive design and relatively small sample size was considered high value in rare cancer by 97% of responders, similar to the randomized controlled trial rating (82%). Surrogate clinical endpoints were considered valuable alternatives to overall survival by 80% of oncologists. Preferred adaptive designs were futility analysis, interim analysis, adaptive sample size, and adaptive randomization. In conclusion, rare cancer oncologists rate evidence from adaptive clinical trials with as high a value and importance for clinical decision making processes as conventional randomized controlled trials. All stakeholders have a vested interest in advances in clinical trial designs to ensure efficient and timely development of innovative medicinal products to allow more patients faster access to the pivotal treatment.
Collapse
|
30
|
Hart RI, Boyle D, Cameron DA, Cowie FJ, Hayward L, Heaney NB, Jesudason AB, Lawton J. Strategies for improving access to clinical trials by teenagers and young adults with cancer: A qualitative study of health professionals' views. Eur J Cancer Care (Engl) 2021; 30:e13408. [PMID: 33474755 DOI: 10.1111/ecc.13408] [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: 07/13/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Few teenagers and young adults (TYA) with cancer participate in clinical trials. Lack of opportunity has been identified as a major barrier. We canvassed health professionals' views on how TYA's access to trials might be improved. METHODS We interviewed 35 professionals with responsibility for delivering or facilitating cancer care and/or clinical trials. We analysed data using a qualitative descriptive approach. RESULTS Interviewees viewed improving TYA's access to trials as challenging, but possible. They reframed the problem as one of rare disease and surmised that modifying the organisation, administration and resourcing of research (and care) might expand opportunities for both TYA and other patients with low volume conditions. Proposals coalesced around four themes: consolidating the pool of patients; streamlining bureaucratic requirements; investing in the research workforce; and promoting pragmatism in trial design. CONCLUSION Accounts suggest there is scope to improve access to trials by TYA with cancer and other patients with rare diseases. Though re-configuring care, research and resource frameworks would present substantial challenges, doing nothing would also have costs. Change will require the support of a range of stakeholders, and agreement as to the best way forward. Further work, such as priority setting exercises, may be necessary to reach a consensus.
Collapse
Affiliation(s)
- Ruth I Hart
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Dorothy Boyle
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | - David A Cameron
- NHS Research Scotland Cancer Lead and Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | | | - Larry Hayward
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | | | - Angela B Jesudason
- Department of Paediatric Haematology and Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | - Julia Lawton
- Usher Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
31
|
Tingley K, Coyle D, Graham ID, Chakraborty P, Wilson K, Potter BK. Stakeholder perspectives on clinical research related to therapies for rare diseases: therapeutic misconception and the value of research. Orphanet J Rare Dis 2021; 16:26. [PMID: 33436030 PMCID: PMC7805116 DOI: 10.1186/s13023-020-01624-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/20/2020] [Indexed: 01/22/2023] Open
Abstract
Background For many rare diseases, few treatments are supported by strong evidence. Patients, family members, health care providers, and policy-makers thus have to consider whether to accept, recommend, or fund treatments with uncertain clinical effectiveness. They must also consider whether and how to contribute to clinical research that may involve receiving or providing the therapy being evaluated. Objective To understand why and how patients and families with rare metabolic diseases, specialist metabolic physicians, and health policy advisors choose whether to participate in studies and how they use and value research. Methods We conducted separate focus group interviews with each stakeholder group (three groups in total); two groups were conducted by telephone and the third was held in-person. Participants were recruited using purposive sampling. We analyzed each interview transcript sequentially using a qualitative description approach to inductively identify key themes. Several strategies to ensure credibility and trustworthiness were used including debriefing sessions after each focus group and having multiple team members review transcripts. Results Four patients/caregivers, six physicians, and three policy advisors participated. Our findings did not support conventional perspectives that therapeutic misconception (gaining access to treatment) is the main motivating factor for patients/caregivers to participate in clinical research. Rather, patients’/caregivers’ expressed reasons for participating in research included advancing science for the next generation and having an opportunity to share their experiences. Patients/caregivers and physicians described the difficulties in weighing risks versus benefits of accepting treatments not well-supported by evidence. Physicians also reported feeling conflicted in their dual role as patient advisor/advocate and evaluator of the evidence. Policy advisors were primarily focused on critically appraising the evidence to make recommendations for the health system. Conclusions Stakeholders differ in their perspectives on rare disease research but share concerns about the risks versus benefits of therapies when making individual- and population-level decisions.
Collapse
Affiliation(s)
- Kylie Tingley
- School of Epidemiology and Public Health, University of Ottawa, 600 Peter Morand Crescent, Ottawa, ON, K1G 5Z3, Canada.
| | - Doug Coyle
- School of Epidemiology and Public Health, University of Ottawa, 600 Peter Morand Crescent, Ottawa, ON, K1G 5Z3, Canada
| | - Ian D Graham
- School of Epidemiology and Public Health, University of Ottawa, 600 Peter Morand Crescent, Ottawa, ON, K1G 5Z3, Canada.,Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Pranesh Chakraborty
- Metabolics and Newborn Screening, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada.,Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada.,Newborn Screening Ontario, Ottawa, ON, Canada
| | - Kumanan Wilson
- School of Epidemiology and Public Health, University of Ottawa, 600 Peter Morand Crescent, Ottawa, ON, K1G 5Z3, Canada.,Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Bruyère Research Institute, Ottawa, ON, Canada.,Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Beth K Potter
- School of Epidemiology and Public Health, University of Ottawa, 600 Peter Morand Crescent, Ottawa, ON, K1G 5Z3, Canada
| | | |
Collapse
|
32
|
Pollack M, Keating K, Wissinger E, Jackson L, Sarnes E, Cuffel B. Transforming approaches to treating TRK fusion cancer: historical comparison of larotrectinib and histology-specific therapies. Curr Med Res Opin 2021; 37:59-70. [PMID: 33148054 DOI: 10.1080/03007995.2020.1847057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The results from basket trials utilized to gain regulatory approval of tumor-agnostic therapies can be difficult to interpret without the context of a comparator arm. We describe the role and efficacy of histology-based treatments to provide a historical comparison with larotrectinib. METHODS A systematic literature review (SLR) was conducted on the clinical outcomes of current histology-based standard of care treatments used in non-small cell lung cancer, colorectal cancer, thyroid cancer, gliomas, soft tissue sarcoma, salivary gland cancer, and infantile fibrosarcoma (7 of the 21 tumor histologies in the larotrectinib trials). The review focused on advanced stage/metastatic disease to make a historical comparison with larotrectinib. RESULTS Larotrectinib provides positive outcomes in both adult and pediatric patients with advanced or metastatic solid tumors known to harbor NTRK gene fusions across a wide range of tumor types. Although the numbers of patients per tumor type are limited, the results of this historical comparison demonstrated that larotrectinib is an efficacious treatment option when naïvely indirectly compared with historical treatments across all 7 reviewed tumor types, especially in comparison to later lines of therapy. CONCLUSIONS Utilizing larotrectinib as a case example across these types of historical comparisons shows that larotrectinib provides positive efficacy outcomes in TRK fusion cancer across tumor histologies known to harbor NTRK gene fusions that may be preferable to historical treatments.
Collapse
Affiliation(s)
| | - Karen Keating
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Louis Jackson
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| | | | - Brian Cuffel
- Bayer Healthcare Pharmaceuticals Inc, Whippany, NJ, USA
| |
Collapse
|
33
|
Rao S. Reply to S. Kim et al. J Clin Oncol 2020; 38:3974-3975. [DOI: 10.1200/jco.20.02643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Sheela Rao
- Sheela Rao, MBBS, Royal Marsden Hospital, Sutton, United Kingdom
| |
Collapse
|
34
|
Bertolet M, Brooks MM, Ragni MV. The design of a Bayesian platform trial to prevent and eradicate inhibitors in patients with hemophilia. Blood Adv 2020; 4:5433-5441. [PMID: 33156923 PMCID: PMC7656929 DOI: 10.1182/bloodadvances.2020002789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/22/2020] [Indexed: 02/02/2023] Open
Abstract
Among individuals with the rare congenital bleeding disorder hemophilia A, the major challenge is inhibitor formation, which is associated with significant morbidity and cost. Yet, as the optimal approach to prevent and eradicate inhibitors is not known, we are at equipoise. Because classic trial design is not practical in a rare disease setting, we designed 2 48-week randomized trials comparing ELOCTATE and emicizumab to prevent and eradicate inhibitors. To achieve statistical efficiency, we incorporated historic data (Bayesian priors) on inhibitor formation to allow preferential randomization to emicizumab, piecewise exponential survival models to determine mean and 95% confidence interval for inhibitor formation in each arm, and simulations to determine the best model design to optimize power. To achieve administrative efficiency, the trials will be performed with the same sites, staff, visit frequency, blood sampling, laboratories, and laboratory assays, with streamlined enrollment so patients developing inhibitors in the first trial may be enrolled on the second trial. The primary end point is the probability of inhibitor formation or inhibitor eradication, respectively. The design indicates early stopping rules for overwhelming evidence of superiority of the emicizumab arms. Simulations indicate that, with 66 subjects, the Prevention Trial will have 84% power to detect noninferiority of emicizumab to ELOCTATE with a margin of 10% if emicizumab is truly 10% superior to ELOCTATE; with 90 subjects, the Eradication Trial will have 80% power to detect 15% superiority of ELOCTATE immune tolerance induction with vs without emicizumab. Thus, a platform design provides statistical and administrative efficiency to conduct INHIBIT trials.
Collapse
Affiliation(s)
| | - Maria M Brooks
- Department of Epidemiology
- Department of Biostatistics, and
| | - Margaret V Ragni
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA; and
- Hemophilia Center of Western PA, Pittsburgh, PA
| |
Collapse
|
35
|
Krishna D, Rittié L, Tran H, Zheng X, Chen-Rogers CE, McGillivray A, Clay T, Ketkar A, Tarnowski J. Short Time to Market and Forward Planning Will Enable Cell Therapies to Deliver R&D Pipeline Value. Hum Gene Ther 2020; 32:433-445. [PMID: 33023309 DOI: 10.1089/hum.2020.212] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
There is considerable industry excitement about the curative potential of cell and gene therapies, but significant challenges remain in designing cost-effective treatments that are accessible globally. We have taken a modeling-based approach to define the cost and value drivers for cell therapy assets during pharmaceutical drug development. We have created a model development program for a lentiviral modified ex vivo autologous T cell therapy for Oncology indications. Using internal and external benchmarks, we have estimated the total out-of-pocket cost of development for an Oncology cell therapy asset from target identification to filing of marketing application to be $500-600 million. Our model indicates that both clinical and Chemistry Manufacturing and Controls (CMC) cost of development for cell therapies are higher due to unique considerations of ex vivo autologous cell therapies. We have computed a threshold revenue-generating patient number for our model asset that enables selection of assets that can address high unmet medical need and generate pipeline value. Using statistical approaches, we identified that short time to market (<5 years) and reduced commercial cost of goods (<$65,000 per dose) will be essential in developing competitive assets and we propose solutions to reduce both. We emphasize that teams must proactively plan alternate development scenarios with clear articulation of path to value generation and greater patient access. We recommend using a modeling-based approach to enable data driven go/no-go decisions during multigenerational cell therapy development.
Collapse
Affiliation(s)
- Delfi Krishna
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Laure Rittié
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Hoang Tran
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Xuan Zheng
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Chia-En Chen-Rogers
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Amanda McGillivray
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Timothy Clay
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Amol Ketkar
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| | - Joseph Tarnowski
- GlaxoSmithKline Pharmaceutical Research and Development, Collegeville, Pennsylvania, USA
| |
Collapse
|
36
|
Mollica V, Rizzo A, Massari F. Re: Toni K. Choueiri, Daniel Y.C. Heng, Jae Lyun Lee, et al. Efficacy of Savolitinib vs Sunitinib in Patients With MET-Driven Papillary Renal Cell Carcinoma: The SAVOIR Phase 3 Randomized Clinical Trial. JAMA Oncol. In press. https://doi.org/10.1001/jamaoncol.2020.2218: SAVOIR: From Own Goal to Winning Goal? Eur Urol Oncol 2020; 3:561-562. [PMID: 32653414 DOI: 10.1016/j.euo.2020.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 01/31/2023]
Affiliation(s)
- Veronica Mollica
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Alessandro Rizzo
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | | |
Collapse
|
37
|
Current status and perspectives of patient-derived rare cancer models. Hum Cell 2020; 33:919-929. [DOI: 10.1007/s13577-020-00391-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
|
38
|
Park JH, Lee JS, Koo H, Kim JE, Ahn JH, Ryu MH, Park SR, Yoon SK, Lee JC, Hong YS, Kim SY, Kim KP, Yoo CH, Hong JY, Lee JL, Jung KH, Rhyoo BY, Kim TW. How Cancer Patients Perceive Clinical Trials (CTs) in the Era of CTs: Current Perception and Its Differences Between Common and Rare Cancers. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2020; 35:545-556. [PMID: 30820925 DOI: 10.1007/s13187-019-01494-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Perception has recently been highlighted as a critical determinant for participation in clinical trials (CTs) among cancer patients. We evaluated cancer patients' current perceptions of CTs using the PARTAKE questionnaires, focusing on differences between patients with common and rare cancers. From November 2015 to May 2017, we prospectively surveyed patients who had received anti-cancer treatment at Asan Medical Center. Among 333 respondents, 70.9% had common and 29.1% had rare cancers. In the cohort, 87.7% of patients with common cancers and 75.3% of patients with rare cancers answered that they heard of and knew about CTs. However, willingness to participate in CTs was expressed only in approximately 56% of patients, although it was significantly associated with awareness and perception. Surprisingly, patients with rare cancers when compared with patients with common cancers showed significantly lower levels of awareness and perception (64.2% vs 79.9%, p = 0.003 and 77.3% vs 91.9%, p < 0.001), and consequently less willingness to participate in CTs (47.4% vs 58.9%, p = 0.06). In addition, cancer patients still harbored fear and concerns about safety and reward of CTs, and demonstrated substantial lack of knowledge about the voluntary nature of CTs, which was more obvious in patients with rare cancers. We identified relatively modest willingness of cancer patients to participate in CTs regardless of generally favorable perception. These findings are highlighted by the more negative perception of CTs among patients with rare cancers relative to those with common cancers. Further education and encouragement by research and public entities seem essential to improve motivation of CTs in cancer patients beyond good perception, especially for patients with rare cancers.
Collapse
Affiliation(s)
- Ji Hyun Park
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
- Department of Hemato-oncology, Konkuk Medical Center, University of Konkuk College of Medicine, Seoul, South Korea
| | - Ji Sung Lee
- Clinical Research Center, Asan Institute of Life Sciences, Asan Medical Center, Seoul, South Korea
| | - HaYeong Koo
- Clinical Research Center, Asan Institute of Life Sciences, Asan Medical Center, Seoul, South Korea
- Korea Clinical Trials Global Initiative, Seoul, South Korea
| | - Jeong Eun Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jin-Hee Ahn
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Min-Hee Ryu
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Sook-Ryun Park
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Shin-Kyo Yoon
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jae Cheol Lee
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Yong-Sang Hong
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Sun Young Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Kyo-Pyo Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Chang-Hoon Yoo
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jung Yong Hong
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jae Lyun Lee
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Kyung Hae Jung
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Baek-Yeol Rhyoo
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Tae Won Kim
- Departments of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
39
|
Nishikawa Y, Hoshino N, Horimatsu T, Funakoshi T, Hida K, Sakai Y, Muto M, Nakayama T. Chemotherapy for patients with unresectable or metastatic small bowel adenocarcinoma: a systematic review. Int J Clin Oncol 2020; 25:1441-1449. [PMID: 32448950 DOI: 10.1007/s10147-020-01703-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/27/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND There is no standard chemotherapy available for unresectable or metastatic small bowel adenocarcinoma (SBA) because of its rarity. This systematic review aims to assess the efficacy and safety of chemotherapy for patients with unresectable or metastatic SBA. METHODS In accordance with the PRISMA statements, literature search was conducted using PubMed, Scopus, and the Cochrane Central Register of Controlled Trials. The included studies were prospective randomized, nonrandomized, or observational studies. Risk of bias was assessed the ROBINS-I tool. RESULTS Seven prospective single-arm Phase II studies were included in this review. Six of them were assessed as having a moderate risk of bias and one as having a serious risk of bias. A meta-analysis was not performed, because the studies were single-arm. Systemic chemotherapy based on fluoropyrimidine regimens achieved favorable outcomes with acceptable adverse effects as a first therapy; however, the regimens differed in each study. The object response rate was 18-50%, and the disease control rate was 29-87%. With 5-fluorouracil, adriamycin, and mitomycin-C regimen, one treatment-related death occurred. A second line of therapy including chemotherapy with nab-paclitaxel also showed favorable efficacy. The object response rate was 20%, and the disease control rate was 50%. CONCLUSIONS Systemic chemotherapy based on fluoropyrimidine regimens was mainly used for unresectable or metastatic SBA. While it may achieve favorable outcomes with acceptable adverse effects, further evidence is needed.
Collapse
Affiliation(s)
- Yoshitaka Nishikawa
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
- Department of Health Informatics, Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Nobuaki Hoshino
- Department of Health Informatics, Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Department of Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takahiro Horimatsu
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Funakoshi
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koya Hida
- Department of Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
40
|
Beaulieu‐Jones BK, Finlayson SG, Yuan W, Altman RB, Kohane IS, Prasad V, Yu K. Examining the Use of Real-World Evidence in the Regulatory Process. Clin Pharmacol Ther 2020; 107:843-852. [PMID: 31562770 PMCID: PMC7093234 DOI: 10.1002/cpt.1658] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
The 21st Century Cures Act passed by the United States Congress mandates the US Food and Drug Administration to develop guidance to evaluate the use of real-world evidence (RWE) to support the regulatory process. RWE has generated important medical discoveries, especially in areas where traditional clinical trials would be unethical or infeasible. However, RWE suffers from several issues that hinder its ability to provide proof of treatment efficacy at a level comparable to randomized controlled trials. In this review article, we summarized the advantages and limitations of RWE, identified the key opportunities for RWE, and pointed the way forward to maximize the potential of RWE for regulatory purposes.
Collapse
Affiliation(s)
| | - Samuel G. Finlayson
- Department of Biomedical InformaticsHarvard Medical SchoolBostonMassachusettsUSA
| | - William Yuan
- Department of Biomedical InformaticsHarvard Medical SchoolBostonMassachusettsUSA
| | - Russ B. Altman
- Departments of Bioengineering, Genetics, Medicine, and Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - Isaac S. Kohane
- Department of Biomedical InformaticsHarvard Medical SchoolBostonMassachusettsUSA
| | - Vinay Prasad
- Division of Hematology OncologyDepartment of Public Health and Preventive MedicineCenter for Health Care EthicsKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Kun‐Hsing Yu
- Department of Biomedical InformaticsHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
41
|
|
42
|
Dehbi HM, Hackshaw A. Sample size calculation in randomised phase II selection trials using a margin of practical equivalence. Trials 2020; 21:301. [PMID: 32228674 PMCID: PMC7106856 DOI: 10.1186/s13063-020-04248-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 03/12/2020] [Indexed: 11/23/2022] Open
Abstract
Background In rare cancers or subtypes of common cancers, a comparison of multiple promising treatments may be required. The selected treatment can then be assessed against the standard of care (if it exists) or used as a backbone for combinations with new, possibly targeted, agents. There could be different experimental therapies or different doses of the same therapy, and either can be done in combination with standard treatments. A ’pick-the-winner’ design is often used, which focuses on efficacy to select the most promising treatment. However, a treatment with a slightly lower efficacy compared to another treatment may actually be preferred if it has a better toxicity or quality of life profile, is easier to administer, or cheaper. Methods By pre-defining a margin of practical equivalence in order to calculate the sample size, a more flexible assessment can be made of whether the treatments have very different effects or are sufficiently close so that other factors can be used to choose between them. Using exact binomial probabilities, we calculated the sample size for two- and three-arm randomised selection trials including a margin of practical equivalence with a variety of input parameters. Results We explain conceptually the margin of practical equivalence in this paper, and provide a free user-friendly web application to calculate the required sample size for a variety of input parameters. Conclusion The web application should help promote the randomised selection design with a margin of practical equivalence, which provides greater flexibility than the ’pick-the-winner’ approach in assessing the results of selection trials.
Collapse
Affiliation(s)
- Hakim-Moulay Dehbi
- Comprehensive Clinical Trials Unit at University College London (UCL), 90 High Holborn, London, WC1V 6LJ, UK.
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, 90 Tottenham Court Road, London, W1T 4TJ, UK
| |
Collapse
|
43
|
Koonrungsesomboon N, Ngamphaiboon N, Townamchai N, Teeyakasem P, Charoentum C, Charoenkwan P, Natesirinilkul R, Sathitsamitphong L, Ativitavas T, Chaiyawat P, Klangjorhor J, Hongeng S, Pruksakorn D. Phase II, multi-center, open-label, single-arm clinical trial evaluating the efficacy and safety of Mycophenolate Mofetil in patients with high-grade locally advanced or metastatic osteosarcoma (ESMMO): rationale and design of the ESMMO trial. BMC Cancer 2020; 20:268. [PMID: 32228535 PMCID: PMC7106788 DOI: 10.1186/s12885-020-06751-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/12/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Clinical outcomes of patients with osteosarcoma remain unsatisfactory, with little improvement in a 5-year overall survival over the past three decades. There is a substantial need for further research and development to identify and develop more efficacious agents/regimens in order to improve clinical outcomes of patients for whom the prognosis is unfavorable. Recently, mycophenolate mofetil, a prodrug of mycophenolic acid, has been found to have anticancer activity against osteosarcoma in both in vitro and animal experiments, so that further investigation in humans is warranted. METHODS A total of 27 patients with high-grade locally advanced or metastatic osteosarcoma will be enrolled into this phase II, multi-center, open-label, single-arm, two-stage clinical trial. The main objectives of this study are to determine the efficacy and safety of mycophenolate mofetil in the patients. The primary endpoint is progression-free survival at 16 weeks; the secondary endpoints include progression-free survival, overall survival, overall response rate, safety parameters, pharmacokinetic parameters, biomarkers, pain score, and quality of life. Mycophenolate mofetil at the initial dose of 5 g/day or lower will be administered for 4 cycles (28 days/cycle) or until disease progression or unacceptable toxicity. The dose of mycophenolate mofetil may be reduced by 1-2 g/day or withheld for some Grade 3 or Grade 4 toxicities whenever clinically needed. The duration of study participation is approximately 4-5 months, with a minimum of 12 study visits. If mycophenolate mofetil proves beneficial to some patients, as evidenced by stable disease or partial response at 16 weeks, administration of mycophenolate mofetil will continue in the extension period. DISCUSSION This trial is the first step in the translation of therapeutic potential of mycophenolate mofetil emerging from in vitro and animal studies into the clinical domain. It is designed to assess the efficacy and safety of mycophenolate mofetil in patients with high-grade locally advanced or metastatic osteosarcoma. The results will provide important information about whether or not mycophenolate mofetil is worth further development. TRIAL REGISTRATION This trial was prospectively registered on Thai Clinical Trials Registry (registration number: TCTR20190701001). The posted information will be updated as needed to reflect protocol amendments and study progress.
Collapse
Affiliation(s)
- Nut Koonrungsesomboon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Muscoloskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, Thailand
| | - Nuttapong Ngamphaiboon
- Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Nakhon Pathom, Thailand
| | - Natavudh Townamchai
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pimpisa Teeyakasem
- Muscoloskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiyut Charoentum
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Departmnet of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Touch Ativitavas
- Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Nakhon Pathom, Thailand
| | - Parunya Chaiyawat
- Muscoloskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, Thailand
| | - Jeerawan Klangjorhor
- Muscoloskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Nakhon Pathom, Thailand
| | - Dumnoensun Pruksakorn
- Muscoloskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, Thailand.
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros, Sriphoom, Muang, Chiang Mai, 50200, Thailand.
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
44
|
Cafferty FH, White JD, Shamash J, Hennig I, Stenning SP, Huddart RA. Long-term outcomes with intensive induction chemotherapy (carboplatin, bleomycin, vincristine and cisplatin/bleomycin, etoposide and cisplatin) and standard bleomycin, etoposide and cisplatin in poor prognosis germ cell tumours: A randomised phase II trial (ISRCTN53643604). Eur J Cancer 2020; 127:139-149. [PMID: 32007714 PMCID: PMC7045084 DOI: 10.1016/j.ejca.2019.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 11/30/2022]
Abstract
Background Up to 50% of men with poor prognosis, non-seminoma germ cell tumours (GCTs) die with standard BEP (bleomycin, etoposide and cisplatin) chemotherapy. An intensive regimen, CBOP/BEP (carboplatin, bleomycin, vincristine and cisplatin/BEP), met response targets in a randomised, phase II trial (74% complete response or partial response marker negative, 90% confidence interval (CI) 61%–85%). Aim To assess long-term outcomes and late toxicity associated with CBOP/BEP. Methods Patients with poor prognosis extracranial GCT were randomised to 4xBEP or CBOP/BEP (2xCBOP, 2xBO, 3xBEP with 15,000iu of bleomycin). Low-dose, stabilising chemotherapy before entry was permitted. Response rates (primary outcome) were reported previously. Here, we report secondary outcomes: progression-free survival (PFS), overall survival (OS) and late toxicity. Prognostic factors and the impact of marker decline are assessed in exploratory analysis. Results Eighty-nine patients (43 CBOP/BEP) were randomised. After median 63 months follow-up, 3-year PFS is 55.7% (95% CI: 39.7%, 69.0%) for CBOP/BEP and 38.7% (95% CI: 24.7%, 52.4%) for BEP (hazard ratio [HR]: 0.59 (0.33, 1.06), p = 0.079). Three-year OS is 65.0% (48.8%, 77.2%) and 58.5% (43.0%, 71.2%), respectively (HR: 0.79 (0.41, 1.52), p = 0.49). Twelve-month toxicity was affected by subsequent treatments, with no clear differences between arms. Stabilising chemotherapy was associated with poorer PFS (HR: 2.09 (1.14, 3.81), p = 0.017), whereas unfavourable marker decline, in 60 (70%) patients, was not. Conclusion Although not powered for PFS, results for CBOP/BEP are promising. Impact on OS was less clear (and will be affected by subsequent therapy). Further study in an international phase III trial is warranted. Trial registration ISRCTN 53643604. Intensive induction chemotherapy met response targets in poor prognosis germ cell tumours. Three-year progression-free survival was 56% for CBOP/BEP and 39% for BEP. Impact on survival was less clear and will be affected by subsequent therapy. Further study in an international phase III trial is warranted.
Collapse
Affiliation(s)
- Fay H Cafferty
- MRC Clinical Trials Unit at UCL, 90 High Holborn, London, WC1V 6LJ, UK.
| | - Jeff D White
- Beatson West of Scotland Cancer Centre, 1053 Great Western Road, Glasgow, G12 0YN, UK
| | - Jonathan Shamash
- St Bartholomew's Hospital, West Smithfield, London, EC1A 7BE, UK
| | - Ivo Hennig
- Nottingham University Hospitals NHS Trust, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Sally P Stenning
- MRC Clinical Trials Unit at UCL, 90 High Holborn, London, WC1V 6LJ, UK
| | - Robert A Huddart
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT, UK
| | | |
Collapse
|
45
|
Rastogi S, Rasheed A, Varshney A. Sarcoma and gastrointestinal stromal tumor update: Lessons from the American Society of Clinical Oncology 2020 and implications for practice. CANCER RESEARCH, STATISTICS, AND TREATMENT 2020. [DOI: 10.4103/crst.crst_242_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
46
|
Renfro LA, Ji L, Piao J, Onar-Thomas A, Kairalla JA, Alonzo TA. Trial Design Challenges and Approaches for Precision Oncology in Rare Tumors: Experiences of the Children's Oncology Group. JCO Precis Oncol 2019; 3:PO.19.00060. [PMID: 32923863 PMCID: PMC7446492 DOI: 10.1200/po.19.00060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
In the United States, cancer remains the leading cause of disease-related death in children. Although survival from any pediatric cancer has improved dramatically during past decades, a number of cancers continue to yield dismal prognoses, which has motivated the continued study of novel therapeutic strategies. Furthermore, even patients cured of pediatric cancer often experience severe adverse effects of treatment and other long-term health implications, such as cardiotoxicity or loss of fertility. For these patients, improved risk stratification to identify those who could safely receive alternate or less-intensive therapy without affecting prognosis is a key objective. Fortunately, pediatric cancers are rare overall, but even among patients with the same narrow cancer type, there is often broad heterogeneity in terms of prognosis, molecular features or pathology, current treatment strategies, and scientific objectives. As a result, the design of clinical trials in the pediatric cancer setting is challenged by a number of practical issues that must be addressed to ensure trial feasibility for this vulnerable group of patients. In this review, we discuss some of the unique trial design considerations often encountered in any rare tumor setting through the lens of our experiences as faculty statisticians for the Children's Oncology Group, the largest organization in the world dedicated exclusively to pediatric cancer research and clinical trials. These topics include risk stratification within individual trials, relaxation of trial operating characteristics and parameters, use of historical controls, and address of noninferiority-type objectives in small cohorts. We review each in terms of practical motivation, present challenges, and potential solutions described in the literature and implemented in selected example trials from the Children's Oncology Group.
Collapse
Affiliation(s)
| | - Lingyun Ji
- University of Southern California, Los Angeles, CA
| | - Jin Piao
- University of Southern California, Los Angeles, CA
| | | | | | | |
Collapse
|
47
|
Canter DJ, Nicholson S, Watkin N, Hall E, Pettaway C. The International Penile Advanced Cancer Trial (InPACT): Rationale and Current Status. Eur Urol Focus 2019; 5:706-709. [PMID: 31160252 DOI: 10.1016/j.euf.2019.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/17/2019] [Accepted: 05/14/2019] [Indexed: 01/18/2023]
Abstract
The historic international penile cancer trial InPACT is now open and accruing patients in the UK and USA. The trial is geared to answer important questions for patients with evidence of inguinal lymph node disease at presentation. This clinical trial update provides an overview of the study and progress to date.
Collapse
Affiliation(s)
| | - Steve Nicholson
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Nick Watkin
- Department of Urology, St. George's Teaching Hospital, London, UK
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Curtis Pettaway
- Department of Urology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA.
| |
Collapse
|
48
|
Bayar MA, Le Teuff G, Koenig F, Le Deley MC, Michiels S. Group sequential adaptive designs in series of time-to-event randomised trials in rare diseases: A simulation study. Stat Methods Med Res 2019; 29:1483-1498. [PMID: 31354106 DOI: 10.1177/0962280219862313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In rare diseases, fully powered large trials may not be doable in a reasonable time frame even with international collaborations. In a previous work, we proposed an approach based on a series of smaller parallel group two-arm randomised controlled trials (RCT) performed over a long research horizon. Within the series of trials, the treatment selected after each trial becomes the control treatment of the next one. We concluded that running more trials with smaller sample sizes and relaxed α-levels leads in the long term and under reasonable assumptions to larger survival benefits with a moderate increase of risk as compared to traditional designs based on larger but fewer trials designed to meet stringent evidence criteria. We now extend this quantitative framework with more 'flexible' designs including interim analyses for futility and/or efficacy, and three-arm adaptive designs with treatment selection at interim. In the simulation study, we considered different disease severities, accrual rates, and hypotheses of how treatments improve over time. For each design, we estimated the long-term survival benefit as the relative difference in hazard rates between the end and the start of the research horizon, and the risk defined as the probability of selecting at the end of the research horizon a treatment inferior to the initial control. We assessed the impact of the α-level and the choice of the stopping rule on the operating characteristics. We also compared the performance of series based on two- vs. three-arm trials. We show that relaxing α-levels within the limit of 0.1 is associated with larger survival gains and moderate increase of risk which remains within acceptable ranges. Including an interim analysis with a futility rule is associated with an additional survival gain and a better risk control as compared to series with no interim analysis, when the α-level is below or equal to 0.1, whereas the benefit of including an interim analysis is rather small for higher α-levels. Including an interim analysis for efficacy yields almost no additional gain. Series based on three-arm trials are associated with a systematic improvement in terms of survival gain and risk control as compared to series of two-arm trials.
Collapse
Affiliation(s)
- Mohamed Amine Bayar
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Villejuif, France.,CESP, Faculté de médecine - Université Paris-Sud, Faculté de médecine - INSERM, Université Paris-Saclay, Villejuif, France
| | - Gwénaël Le Teuff
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Villejuif, France.,CESP, Faculté de médecine - Université Paris-Sud, Faculté de médecine - INSERM, Université Paris-Saclay, Villejuif, France
| | - Franz Koenig
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Marie-Cécile Le Deley
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Villejuif, France.,CESP, Faculté de médecine - Université Paris-Sud, Faculté de médecine - INSERM, Université Paris-Saclay, Villejuif, France.,Unité de Méthodologie et Biostatistique, Centre Oscar Lambret, Lille, France
| | - Stefan Michiels
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Villejuif, France.,CESP, Faculté de médecine - Université Paris-Sud, Faculté de médecine - INSERM, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
49
|
Ahmed MA, Okour M, Brundage R, Kartha RV. Orphan drug development: the increasing role of clinical pharmacology. J Pharmacokinet Pharmacodyn 2019; 46:395-409. [PMID: 31338634 DOI: 10.1007/s10928-019-09646-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Over the last few decades there has been a paradigm shift in orphan drug research and development. The development of the regulatory framework, establishment of rare disease global networks that support drug developments, and advances in technology, has resulted in tremendous growth in orphan drug development. Nevertheless, several challenges during orphan drug development such as economic constraints; insufficient clinical information; fewer patients and thus inadequate power; etc. still exist. While the standard regulatory requirements for drug approval stays the same, applications of scientific judgment and regulatory flexibility is significantly important to help meeting some of the immense unmet medical need in rare diseases. Clinical pharmacology presents a vital role in accelerating orphan drug development and overcoming some of these challenges. This review highlights the critical contributions of clinical pharmacology in orphan drug development; for example, dose finding, optimizing clinical trial design, indication expansion, and population extrapolation. Examples of such applications are reviewed in this article.
Collapse
Affiliation(s)
- Mariam A Ahmed
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Twin Cities, MN, USA.
- , 10903 New Hampshire Ave, Silver Spring, MD, 20993, USA.
| | - Malek Okour
- Clinical Pharmacology Modeling and Simulation (CPMS), GlaxoSmithKline, Upper Providence, PA, USA
| | - Richard Brundage
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Twin Cities, MN, USA
- Center for Orphan Drug Research, University of Minnesota, Twin Cities, MN, USA
| | - Reena V Kartha
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Twin Cities, MN, USA
- Center for Orphan Drug Research, University of Minnesota, Twin Cities, MN, USA
| |
Collapse
|
50
|
Fortpied C, Vinches M. The Statistical Evaluation of Treatment and Outcomes in Head and Neck Squamous Cell Carcinoma Clinical Trials. Front Oncol 2019; 9:634. [PMID: 31355146 PMCID: PMC6640189 DOI: 10.3389/fonc.2019.00634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/26/2019] [Indexed: 11/13/2022] Open
Abstract
The purpose of this article is two-fold: to help statisticians confronted with the design, implementation and analysis of clinical trials and new to the field of head and neck cancer; but also to sensitize research physicians with the role, the tasks and the challenges faced by the medical statisticians. These two purposes altogether will hopefully encourage and enable fluid communication between the research physician and the medical statistician and the understanding of each other's field and concerns. In particular, the methodological challenges resulting from the heterogeneity of the head and neck cancer, the complexity of the treatments and the associated comorbidities are presented with examples borrowed from medical literature and from the practical experience of the authors in this field.
Collapse
|