1
|
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
|
2
|
Louie BH, Kato S, Lim JS, Kim KH, Lim HJ, Okamura R, Lee S, Kim L, Sicklick JK, Lippman SM, Kurzrock R. Molecular Tumor Board for Unicorns: Outcomes for rare and ultra-rare cancers using an N-of-One personalized treatment strategy. iScience 2024; 27:110465. [PMID: 39148716 PMCID: PMC11324991 DOI: 10.1016/j.isci.2024.110465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 05/06/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Treatment of rare/ultra-rare tumors is an unmet need due to a lack of standardized therapies and clinical trials. We developed the Molecular Tumor Board (MTB), a multidisciplinary team that integrates molecular profiling to generate personalized, N-of-One treatments for advanced cancers. This study evaluates 112 patients with rare/ultra-rare tumors who presented to the MTB and were evaluable for clinical therapeutic outcome. Overall, 46/112 patients (41%) received a treatment regimen with a high degree of matching between tumor molecular alterations and drugs given (reflected by a high Matching Score (≥50%)). Patients with a high versus low Matching Score experienced significantly longer progression-free survival (p = 0.005) and overall survival (p = 0.047), and higher rates of clinical benefit (stable disease ≥6 months, partial response, or complete response) (54% vs. 32% p = 0.027). The MTB facilitated personalized N-of-One matching of drugs to tumor molecular alterations, which was associated with improved clinical outcomes in patients with rare/ultra-rare cancers.
Collapse
Affiliation(s)
- Bryan H Louie
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jordan S Lim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Ki Hwan Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Hyo Jeong Lim
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Ryosuke Okamura
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Lisa Kim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, and Department of Pharmacology, UC San Diego Health Sciences, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, Paris, France
- Medical College of Wisconsin Cancer Center and Genomic Sciences and Precision Medicine Center, Milwaukee, WI, USA
- University of Nebraska, Omaha, NE, USA
| |
Collapse
|
3
|
D'Amico S, Dall’Olio L, Rollo C, Alonso P, Prada-Luengo I, Dall’Olio D, Sala C, Sauta E, Asti G, Lanino L, Maggioni G, Campagna A, Zazzetti E, Delleani M, Bicchieri ME, Morandini P, Savevski V, Arroyo B, Parras J, Zhao LP, Platzbecker U, Diez-Campelo M, Santini V, Fenaux P, Haferlach T, Krogh A, Zazo S, Fariselli P, Sanavia T, Della Porta MG, Castellani G. MOSAIC: An Artificial Intelligence-Based Framework for Multimodal Analysis, Classification, and Personalized Prognostic Assessment in Rare Cancers. JCO Clin Cancer Inform 2024; 8:e2400008. [PMID: 38875514 PMCID: PMC11371092 DOI: 10.1200/cci.24.00008] [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: 01/12/2024] [Revised: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 06/16/2024] Open
Abstract
PURPOSE Rare cancers constitute over 20% of human neoplasms, often affecting patients with unmet medical needs. The development of effective classification and prognostication systems is crucial to improve the decision-making process and drive innovative treatment strategies. We have created and implemented MOSAIC, an artificial intelligence (AI)-based framework designed for multimodal analysis, classification, and personalized prognostic assessment in rare cancers. Clinical validation was performed on myelodysplastic syndrome (MDS), a rare hematologic cancer with clinical and genomic heterogeneities. METHODS We analyzed 4,427 patients with MDS divided into training and validation cohorts. Deep learning methods were applied to integrate and impute clinical/genomic features. Clustering was performed by combining Uniform Manifold Approximation and Projection for Dimension Reduction + Hierarchical Density-Based Spatial Clustering of Applications with Noise (UMAP + HDBSCAN) methods, compared with the conventional Hierarchical Dirichlet Process (HDP). Linear and AI-based nonlinear approaches were compared for survival prediction. Explainable AI (Shapley Additive Explanations approach [SHAP]) and federated learning were used to improve the interpretation and the performance of the clinical models, integrating them into distributed infrastructure. RESULTS UMAP + HDBSCAN clustering obtained a more granular patient stratification, achieving a higher average silhouette coefficient (0.16) with respect to HDP (0.01) and higher balanced accuracy in cluster classification by Random Forest (92.7% ± 1.3% and 85.8% ± 0.8%). AI methods for survival prediction outperform conventional statistical techniques and the reference prognostic tool for MDS. Nonlinear Gradient Boosting Survival stands in the internal (Concordance-Index [C-Index], 0.77; SD, 0.01) and external validation (C-Index, 0.74; SD, 0.02). SHAP analysis revealed that similar features drove patients' subgroups and outcomes in both training and validation cohorts. Federated implementation improved the accuracy of developed models. CONCLUSION MOSAIC provides an explainable and robust framework to optimize classification and prognostic assessment of rare cancers. AI-based approaches demonstrated superior accuracy in capturing genomic similarities and providing individual prognostic information compared with conventional statistical methods. Its federated implementation ensures broad clinical application, guaranteeing high performance and data protection.
Collapse
Affiliation(s)
- Saverio D'Amico
- Humanitas Clinical and Research Center—IRCCS, Milan, Italy
- Train s.r.l., Milan, Italy
| | | | - Cesare Rollo
- Computational Biomedicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Patricia Alonso
- Department of Signals, Systems and Radiocommunications, Polytechnic University of Madrid, Madrid, Spain
| | | | | | - Claudia Sala
- Experimental, Diagnostic and Specialty Medicine—DIMES, Bologna, Italy
| | | | - Gianluca Asti
- Humanitas Clinical and Research Center—IRCCS, Milan, Italy
| | - Luca Lanino
- Humanitas Clinical and Research Center—IRCCS, Milan, Italy
| | | | | | - Elena Zazzetti
- Humanitas Clinical and Research Center—IRCCS, Milan, Italy
| | | | | | | | | | - Borja Arroyo
- Department of Signals, Systems and Radiocommunications, Polytechnic University of Madrid, Madrid, Spain
| | - Juan Parras
- Department of Signals, Systems and Radiocommunications, Polytechnic University of Madrid, Madrid, Spain
| | - Lin Pierre Zhao
- Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/University Paris 7, Paris, France
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Maria Diez-Campelo
- Hematology Department, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Valeria Santini
- Hematology, Azienda Ospedaliero-Universitaria Careggi & University of Florence, Florence, Italy
| | - Pierre Fenaux
- Hematology and Bone Marrow Transplantation, Hôpital Saint-Louis/University Paris 7, Paris, France
| | | | | | - Santiago Zazo
- Department of Signals, Systems and Radiocommunications, Polytechnic University of Madrid, Madrid, Spain
| | - Piero Fariselli
- Computational Biomedicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tiziana Sanavia
- Computational Biomedicine Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Matteo Giovanni Della Porta
- Humanitas Clinical and Research Center—IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy (DIFA), Bologna, Italy
- Experimental, Diagnostic and Specialty Medicine—DIMES, Bologna, Italy
| |
Collapse
|
4
|
Wani AK, Prakash A, Sena S, Akhtar N, Singh R, Chopra C, Ariyanti EE, Mudiana D, Yulia ND, Rahayu F. Unraveling molecular signatures in rare bone tumors and navigating the cancer pathway landscapes for targeted therapeutics. Crit Rev Oncol Hematol 2024; 196:104291. [PMID: 38346462 DOI: 10.1016/j.critrevonc.2024.104291] [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: 10/15/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Rare cancers (RCs), which account for over 20% of cancer cases, face significant research and treatment challenges due to their limited prevalence. This results in suboptimal outcomes compared to more common malignancies. Rare bone tumors (RBTs) constitute 5-10% of rare cancer cases and pose unique diagnostic complexities. The therapeutic potential of anti-cancer drugs for RBTs remains largely unexplored. Identifying molecular alterations in cancer-related genes and their associated pathways is essential for precision medicine in RBTs. Small molecule inhibitors and monoclonal antibodies targeting specific RBT-associated proteins show promise. Ongoing clinical trials aim to define RBT biomarkers, subtypes, and optimal treatment contexts, including combination therapies and immunotherapeutic agents. This review addresses the challenges in diagnosing, treating, and studying RBTs, shedding light on the current state of RBT biomarkers, potential therapeutic targets, and promising inhibitors. Rare cancers demand attention and innovative solutions to improve clinical outcomes.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India.
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Saikat Sena
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar 144411, India
| | - Esti Endah Ariyanti
- Research Center for Applied Botany, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Deden Mudiana
- Research Center for Ecology and Ethnobiology, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Nina Dwi Yulia
- Research Center for Applied Botany, National Research and Innovation Agency, Bogor 16911, Indonesia
| | - Farida Rahayu
- Research Center for Genetic Engineering, National Research and Innovation Agency, Bogor 16911, Indonesia
| |
Collapse
|
5
|
Xue J, Lyu Q. Challenges and opportunities in rare cancer research in China. SCIENCE CHINA. LIFE SCIENCES 2024; 67:274-285. [PMID: 38036799 DOI: 10.1007/s11427-023-2422-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/15/2023] [Indexed: 12/02/2023]
Abstract
Cancer is one of the major public health challenges in China. Rare cancers collectively account for a considerable proportion of all malignancies. The lack of awareness of rare cancers among healthcare professionals and the general public, the typically complex and delayed diagnosis, and limited access to clinical trials are key challenges. Recent years have witnessed an increase in funding for research related to rare cancers in China. In this review, we provide a comprehensive overview of rare cancers and summarize the status of research on rare cancers in China and overseas, including the trends of funding and publications. We also highlight the challenges and perspectives regarding rare cancers in China.
Collapse
Affiliation(s)
- Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Natural Science Foundation of China, Beijing, 100085, China
| | - Qunyan Lyu
- National Natural Science Foundation of China, Beijing, 100085, China.
| |
Collapse
|
6
|
Moreno L, DuBois SG, Glade Bender J, Mauguen A, Bird N, Buenger V, Casanova M, Doz F, Fox E, Gore L, Hawkins DS, Izraeli S, Jones DT, Kearns PR, Molenaar JJ, Nysom K, Pfister S, Reaman G, Smith M, Weigel B, Vassal G, Zwaan CM, Paoletti X, Iasonos A, Pearson AD. Combination Early-Phase Trials of Anticancer Agents in Children and Adolescents. J Clin Oncol 2023; 41:3408-3422. [PMID: 37015036 PMCID: PMC10414747 DOI: 10.1200/jco.22.02430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/07/2023] [Indexed: 04/06/2023] Open
Abstract
PURPOSE There is an increasing need to evaluate innovative drugs for childhood cancer using combination strategies. Strong biological rationale and clinical experience suggest that multiple agents will be more efficacious than monotherapy for most diseases and may overcome resistance mechanisms and increase synergy. The process to evaluate these combination trials needs to maximize efficiency and should be agreed by all stakeholders. METHODS After a review of existing combination trial methodologies, regulatory requirements, and current results, a consensus among stakeholders was achieved. RESULTS Combinations of anticancer therapies should be developed on the basis of mechanism of action and robust preclinical evaluation, and may include data from adult clinical trials. The general principle for combination early-phase studies is that, when possible, clinical trials should be dose- and schedule-confirmatory rather than dose-exploratory, and every effort should be made to optimize doses early. Efficient early-phase combination trials should be seamless, including dose confirmation and randomized expansion. Dose evaluation designs for combinations depend on the extent of previous knowledge. If not previously evaluated, limited evaluation of monotherapy should be included in the same clinical trial as the combination. Randomized evaluation of a new agent plus standard therapy versus standard therapy is the most effective approach to isolate the effect and toxicity of the novel agent. Platform trials may be valuable in the evaluation of combination studies. Patient advocates and regulators should be engaged with investigators early in a proposed clinical development pathway and trial design must consider regulatory requirements. CONCLUSION An optimized, agreed approach to the design and evaluation of early-phase pediatric combination trials will accelerate drug development and benefit all stakeholders, most importantly children and adolescents with cancer.
Collapse
Affiliation(s)
- Lucas Moreno
- Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Steven G. DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | | | | | - Nick Bird
- Solving Kids' Cancer UK, London, United Kingdom
| | - Vickie Buenger
- Coalition Against Childhood Cancer (CAC2), Philadelphia, PA
| | | | - François Doz
- Université Paris Cité, Paris, France
- SIREDO Centre (Care, Innovation Research in Pediatric, Adolescent and Young Adults Oncology), Institut Curie, Paris, France
| | | | - Lia Gore
- Children's Hospital Colorado, Aurora, CO
- University of Colorado, Aurora, CO
| | | | - Shai Izraeli
- Rina Zaizov Pediatric Hematology Oncology Division, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Hematological Malignancies Centre of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David T.W. Jones
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- NIHR Birmingham Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Pamela R. Kearns
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Pharmaceutical Sciences Utrecht University, Utrecht, the Netherlands
| | - Jan J. Molenaar
- Division of Pediatric Neurooncology, DKFZ, KiTZ
- Righospitalet, Copenhagen, Denmark
| | - Karsten Nysom
- Clinical Trial Unit and Childhood Brain Tumors, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Pfister
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | - Gilles Vassal
- Innovative Therapies for Children with Cancer, Paris, France
- ACCELERATE, Brussels, Belgium
- Gustave Roussy Cancer Centre, Paris, France
| | - Christian Michel Zwaan
- Righospitalet, Copenhagen, Denmark
- Department of Pediatric Oncology, Hematology, Erasmus MC, Sophia Children’s Hospital, the Netherlands
| | | | | | - Andrew D.J. Pearson
- Innovative Therapies for Children with Cancer, Paris, France
- ACCELERATE, Brussels, Belgium
| |
Collapse
|
7
|
Zheng Y, Peng H, Hu X, Ou Y, Wang D, Wang H, Ren S. Progress and prospects of targeted therapy and immunotherapy for urachal carcinoma. Front Pharmacol 2023; 14:1199395. [PMID: 37324454 PMCID: PMC10267743 DOI: 10.3389/fphar.2023.1199395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Urachal carcinoma (UrC) is a rare and aggressive disease. Systematic chemotherapy shows limited efficacy in patients with advanced disease, while targeted therapy and immunotherapy may provide a reasonable alternative for specific populations. The molecular pattern of colorectal cancer (CRC) have recently been identified; this understanding has significantly influenced the clinical management of CRC in terms of molecular-targeted therapy. Although some genetic alterations have been associated with UrC, there is still no systematic overview of the molecular profile of this rare malignancy. Methods: In this review, we comprehensively discuss the molecular profile of UrC and further identify potential targets for the personalized treatment of UrC as well as immune checkpoint inhibitors that represent underlying biomarkers. A systematic literature search was carried out by searching the PubMed, EMBASE, and Web of Science databases to identify all literature related to targeted therapy and immunotherapy in urachal carcinoma from inception to February 2023. Results: A total of 28 articles were eligible, and most studies included were case report sand retrospective case series. Furthermore, 420 cases of UrC were identified to analyze the association between mutations and UrC. The most commonly mutated gene in UrC was TP53 with the prevalence of 70%, followed by KRAS mutations in 28.3%, MYC mutations in 20.3%, SMAD4 mutations in 18.2% and GNAS mutations in 18%, amongst other genes. Discussion: The molecular patterns of UrC and CRC are similar yet distinct. Notably, targeted therapy, especially EGFR-targeting therapy, might provide curative efficacy for patients with UrC by applying specific molecular markers. Additional potential biomarkers for the immunotherapy of UrC are mismatch repair (MMR) status and PD-L1 expression profile. In addition, combined regimens featuring targeted agents and immune checkpoint blockers might increase antitumor activity and exert better efficacy in UrC patients with specific mutational burden.
Collapse
Affiliation(s)
- Yang Zheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Heling Peng
- Medical Administration Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Ou
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Dong Wang
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Han Wang
- Department of Gastroenterology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
| | - Shangqing Ren
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
8
|
Subbiah V, Kreitman RJ, Wainberg ZA, Gazzah A, Lassen U, Stein A, Wen PY, Dietrich S, de Jonge MJA, Blay JY, Italiano A, Yonemori K, Cho DC, de Vos FYFL, Moreau P, Fernandez EE, Schellens JHM, Zielinski CC, Redhu S, Boran A, Passos VQ, Ilankumaran P, Bang YJ. Dabrafenib plus trametinib in BRAFV600E-mutated rare cancers: the phase 2 ROAR trial. Nat Med 2023; 29:1103-1112. [PMID: 37059834 PMCID: PMC10202803 DOI: 10.1038/s41591-023-02321-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/27/2023] [Indexed: 04/16/2023]
Abstract
BRAFV600E alterations are prevalent across multiple tumors. Here we present final efficacy and safety results of a phase 2 basket trial of dabrafenib (BRAF kinase inhibitor) plus trametinib (MEK inhibitor) in eight cohorts of patients with BRAFV600E-mutated advanced rare cancers: anaplastic thyroid carcinoma (n = 36), biliary tract cancer (n = 43), gastrointestinal stromal tumor (n = 1), adenocarcinoma of the small intestine (n = 3), low-grade glioma (n = 13), high-grade glioma (n = 45), hairy cell leukemia (n = 55) and multiple myeloma (n = 19). The primary endpoint of investigator-assessed overall response rate in these cohorts was 56%, 53%, 0%, 67%, 54%, 33%, 89% and 50%, respectively. Secondary endpoints were median duration of response (DoR), progression-free survival (PFS), overall survival (OS) and safety. Median DoR was 14.4 months, 8.9 months, not reached, 7.7 months, not reached, 31.2 months, not reached and 11.1 months, respectively. Median PFS was 6.7 months, 9.0 months, not reached, not evaluable, 9.5 months, 5.5 months, not evaluable and 6.3 months, respectively. Median OS was 14.5 months, 13.5 months, not reached, 21.8 months, not evaluable, 17.6 months, not evaluable and 33.9 months, respectively. The most frequent (≥20% of patients) treatment-related adverse events were pyrexia (40.8%), fatigue (25.7%), chills (25.7%), nausea (23.8%) and rash (20.4%). The encouraging tumor-agnostic activity of dabrafenib plus trametinib suggests that this could be a promising treatment approach for some patients with BRAFV600E-mutated advanced rare cancers. ClinicalTrials.gov registration: NCT02034110 .
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Robert J Kreitman
- Laboratory of Molecular Biology, National Institutes of Health, Bethesda, MD, USA
| | - Zev A Wainberg
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy Cancer Institute, Villejuif, France
| | - Ulrik Lassen
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Stein
- Department of Internal Medicine II (Oncology Center), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jean-Yves Blay
- Center Leon Berard & University Claude Bernard Lyon I, Lyon, France
| | - Antoine Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | | | | | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | | | - Elena Elez Fernandez
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | | | | | - Suman Redhu
- Global Program Biostatistics, Novartis Oncology, Cambridge, MA, USA
| | - Aislyn Boran
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Vanessa Q Passos
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Palanichamy Ilankumaran
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Wedderburn LR, Ramanan AV, Croft AP, Hyrich KL, Dick AD. Towards molecular-pathology informed clinical trials in childhood arthritis to achieve precision medicine in juvenile idiopathic arthritis. Ann Rheum Dis 2023; 82:449-456. [PMID: 36600186 PMCID: PMC10086280 DOI: 10.1136/ard-2022-222553] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
In childhood arthritis, collectively known as Juvenile idiopathic arthritis (JIA), the rapid rise of available licensed biological and targeted small molecule treatments in recent years has led to improved outcomes. However, real-world data from multiple countries and registries show that despite a large number of available drugs, many children and young people continue to suffer flares and experience significant periods of time with active disease for many years. More than 50% of young people with JIA require ongoing immune suppression well into adult life, and they may have to try multiple different treatments in that time. There are currently no validated tools with which to select specific treatments, nor biomarkers of response to assist in such choices, therefore, current management uses essentially a trial-and-error approach. A further consequence of recent progress is a reducing pool of available children or young people who are eligible for new trials. In this review we consider how progress towards a molecular based approach to defining treatment targets and informing trial design in JIA, combined with novel approaches to clinical trials, could provide strategies to maximise discovery and progress, in order to move towards precision medicine for children with arthritis.
Collapse
Affiliation(s)
- Lucy R Wedderburn
- UCL GOS Institute of Child Health, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, UCL, London, UK
- National Institute of Health Research Biomedical Research Centre at GOSH London UK, Great Ormond Street Hospital, London, UK
| | - Athimalaipet V Ramanan
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Adam P Croft
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute of Health Research Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Kimme L Hyrich
- Centre for Epidemiology Versus Arthritis, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- National Institute of Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew D Dick
- Translational Health Sciences, University of Bristol, Bristol, UK
- UCL Institute of Ophthalmology, University College London, London, UK
- National Institute of Health Research Biomedical Research Centre, Moorfields and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
10
|
Tang M, Pearson SA, Simes RJ, Chua BH. Harnessing Real-World Evidence to Advance Cancer Research. Curr Oncol 2023; 30:1844-1859. [PMID: 36826104 PMCID: PMC9955401 DOI: 10.3390/curroncol30020143] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Randomized controlled trials (RCTs) form a cornerstone of oncology research by generating evidence about the efficacy of therapies in selected patient populations. However, their implementation is often resource- and cost-intensive, and their generalisability to patients treated in routine practice may be limited. Real-world evidence leverages data collected about patients receiving clinical care in routine practice outside of clinical trial settings and provides opportunities to identify and address gaps in clinical trial evidence. This review outlines the strengths and limitations of real-world and RCT evidence and proposes a framework for the complementary use of the two bodies of evidence to advance cancer research. There are challenges to the implementation of real-world research in oncology, including heterogeneity of data sources, timely access to high-quality data, and concerns about the quality of methods leveraging real-world data, particularly causal inference. Improved understanding of the strengths and limitations of real-world data and ongoing efforts to optimise the conduct of real-world evidence research will improve its reliability, understanding and acceptance, and enable the full potential of real-world evidence to be realised in oncology practice.
Collapse
Affiliation(s)
- Monica Tang
- Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick 2031, Australia
- Correspondence:
| | | | - Robert J. Simes
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown 2050, Australia
| | - Boon H. Chua
- Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Randwick 2031, Australia
- Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| |
Collapse
|
11
|
Sase K, Mukai M, Fujiwara Y. Clinical Practice Guidelines in Cardio-Oncology: A Sea of Opportunity. JACC CardioOncol 2023; 5:145-148. [PMID: 36875908 PMCID: PMC9982292 DOI: 10.1016/j.jaccao.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kazuhiro Sase
- Clinical Pharmacology and Regulatory Science, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Institute for Medical Regulatory Science, Organization for University Research Initiatives, Waseda University, Tokyo, Japan
| | - Mikio Mukai
- Department of Medical Check-up, Osaka International Cancer Institute, Osaka Prefectural Hospital Organization, Osaka, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.,Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| |
Collapse
|
12
|
Do Early Phase Oncology Trials Predict Clinical Efficacy in Subsequent Biomarker-Enriched Phase III Randomized Trials? Target Oncol 2022; 17:665-674. [PMID: 36197635 DOI: 10.1007/s11523-022-00920-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 10/10/2022]
Abstract
BACKGROUND Promising early phase trial results of biomarker-targeted therapies have occasionally led to regulatory approval. OBJECTIVE We examined if early phase trials were predictive of efficacy in randomized controlled trials (RCTs) with matching treatment settings. PATIENTS AND METHODS Cancer drug RCTs conducted between January 2006 and March 2021 were identified through Clinicaltrials.gov. Biomarker-enriched RCTs and associated matching early phase trials were included. Trial pairs were compared using objective response rate (ORR) and progression-free survival (PFS). We examined whether early phase trials results were associated with RCT results using logistic regression. RESULTS The search yielded 2157 unique RCTs and 27 RCTs pairing with early phase trials were included. Based on average difference of trial pairs, ORR was similar (1.6%; 95% confidence interval (CI) - 2.5 to 5.6, p = 0.50) and median PFS was higher in early phase trials (2.0 months; 95% CI 0.9-3.0, p < 0.05). On an individual pair basis, there was large variability in difference for ORR (range - 23.9 to 20.2%) and median PFS (range - 0.8 to 7.4 months). The probability of the RCT meeting its primary endpoint is 95% (95% prediction interval (PI) 72.8-99.3%) when the early phase trial ORR is 77.7%. CONCLUSIONS Overall, in early phase trials, ORR has minimal bias and median PFS appears to be slightly overestimated. Substantial variability between trials suggests early phase trial results may be inconsistent with subsequent RCT. Early phase trial results may be associated with RCTs meeting their primary endpoint when ORR is very high; however, caution must be exercised when using early phase trials as representative of RCTs.
Collapse
|
13
|
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
|
14
|
Dooms M, Saesen R, Steemans I, Lansens J, Huys I. Characteristics of Early Phase Clinical Trials for Rare Cancers: Insights From Interviews With Stakeholders. Front Pharmacol 2022; 13:775217. [PMID: 35586057 PMCID: PMC9108391 DOI: 10.3389/fphar.2022.775217] [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: 09/13/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Rare cancers occur with an incidence of no more than six cases per 100,000 people according to the definition used by the Surveillance of Rare Cancers in Europe project. For a variety of reasons (low prevalence, cytotoxicity), it is challenging to perform the necessary clinical studies to investigate the safety and efficacy of investigational medicines against such rare malignancies, reformulating even at the earliest stages of the drug development process. This article investigates the differences between phase I rare cancer trials performed in commercial (companies) and non-commercial settings (academic hospitals).Materials and Methods: The differences were explored through the conduct of semi-structured interviews with three different stakeholder groups: representatives from academia (n = 7), representatives from companies (n = 4) and representatives from patient organizations (n = 4). All the interviews were transcribed verbatim and analyzed in NVivo using the framework method.Results: According to the interviewees, the academic and commercial stakeholders collaborate in the majority of phase I rare cancer trials. In general, the commercial partner finances the trial, whereas academia is responsible for the execution of the study procedures. The average cost of undertaking these trials is difficult to estimate because it depends on what is specifically requested during the trial. The 3 + 3 study design remains the most widely used design and the use of expansion cohorts is controversial. With regard to the regulatory aspects of phase I rare cancer trials, it was expressed that a good regulatory framework facilitates the conduct of these studies, but that increased regulation and oversight also has drawbacks, e.g., differences in standards between different ethics committees, over interpretation of the rules, insufficient availability of qualified personnel and higher workloads. The patient organization representatives claimed that patients experience no differences in terms of accommodation, compensation and paperwork between the academic and commercial settings or the degree of follow-up. They also believed that the direct input of patients can bring added value to such studies not only with regard to the recruitment process and the feasibility of the study but also the legibility of the informed consent forms.Conclusion: The growing need for first-in-man trials in rare malignancies needs to be highlighted, as difficult as they are to undertake and to co-develop, not only because rare cancer patients deserve an appropriate treatment, but also because these medicines represent the future of cancer therapy in the precision medicine era. Cooperation of commercial and academic sites are needed. Patient organizations need to be educated to take part in this process.
Collapse
|
15
|
Koehler M, Hoppe S, Kropf S, Lux A, Bartsch R, Holzner B, Krauter J, Florschütz A, Jentsch-Ullrich K, Frommer J, Flechtner HH, Fischer T. Randomized Trial of a Supportive Psychotherapy for Parents of Adolescents and Young Adults With Hematologic Malignancies. J Natl Compr Canc Netw 2022; 20:jnccn20614. [PMID: 35405661 DOI: 10.6004/jnccn.2021.7075] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 06/17/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Cancer regularly disrupts health and developmental trajectories in adolescents and young adults (AYAs). Parents have been shown to have a substantial impact on the health and cancer survivorship activities of AYA patients in the form of symptom management. However, no randomized controlled trial has evaluated a coping support intervention (CSI) program for parents of AYAs with cancer aged 18 to 40 years. PATIENTS AND METHODS From November 30, 2012, to August 29, 2016, parents of AYAs with hematologic malignancies were randomized in a phase III controlled trial (1:1 ratio, stratified sampling) to either the research-based CSI AYA-Parents group (CSI group; n=82) or the standard care (SC) group (n=70). CSI consisted of 5 sessions to achieve the enhancement of parental adaptive coping as the primary outcome (per the adaptive coping scale of the 28-item Brief COPE, a validated multidimensional self-assessment-questionnaire recommended for clinical cancer research). Measures of adaptive coping, depression, and mental health were collected at pre-CSI (measurement date T1), at the end of the intervention sessions (measurement date T2), and at follow-up (3 months). We calculated mean change scores in outcomes and estimated intervention effect sizes (Cohen's d) for changes from T1 to T2/T3, with 0.2 indicating a small effect, 0.5 a medium effect, and 0.8 a large effect. All statistical tests were 2-sided. RESULTS In the intention-to-treat analysis, the CSI group significantly improved their adaptive coping compared with the SC group (95% CI, 0.30-2.54; P=.013; d=0.405), whereas adaptive coping in the SC group deteriorated. The CSI group also experienced a significant decrease in depressive symptoms and improved mental health with clinical significance (95% CI, -1.98 to -0.30; P=.008; d=0.433, and 95% CI, -0.19 to 3.97; P=.074; d=0.292, respectively). Sensitivity analyses confirmed the robustness of the main intention-to-treat analysis. CONCLUSIONS CSI improved effectively adaptive coping and depression in parents of AYAs with hematologic malignancies. It may represent a novel family-based approach in AYA oncology care.
Collapse
Affiliation(s)
- Michael Koehler
- 1Department of Hematology and Oncology, University Hospital Magdeburg.,2Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, and
| | - Susanne Hoppe
- 1Department of Hematology and Oncology, University Hospital Magdeburg.,2Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, and
| | - Siegfried Kropf
- 3Institute for Biometry and Medical Informatics, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anke Lux
- 3Institute for Biometry and Medical Informatics, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Rainer Bartsch
- 1Department of Hematology and Oncology, University Hospital Magdeburg.,2Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, and
| | - Bernhard Holzner
- 4Department of Psychiatry, Psychotherapy and Psychosomatic, Innsbruck Medical University, Innsbruck, Austria
| | - Juergen Krauter
- 5Department of Hematology and Oncology, Braunschweig Municipal Hospital, Braunschweig, Germany
| | - Axel Florschütz
- 6Department of Internal Medicine, Dessau Municipal Hospital, Dessau-Roßlau, Germany
| | | | - Joerg Frommer
- 8Department of Psychosomatic Medicine and Psychotherapy, University Hospital Magdeburg, and
| | - Hans-Henning Flechtner
- 9Department of Child and Adolescent Psychiatry, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas Fischer
- 1Department of Hematology and Oncology, University Hospital Magdeburg.,2Gesundheitscampus Immunologie, Infektiologie und Inflammation (GCI3), Medical Center, and
| |
Collapse
|
16
|
Casali P, Licitra L, Frezza A, Trama A. “Rare cancers”: not all together in clinical studies! Ann Oncol 2022; 33:463-465. [DOI: 10.1016/j.annonc.2022.01.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/23/2021] [Accepted: 01/30/2022] [Indexed: 11/01/2022] Open
|
17
|
Rantala ES, Hernberg MM, Piperno-Neumann S, Grossniklaus HE, Kivelä TT. Metastatic uveal melanoma: The final frontier. Prog Retin Eye Res 2022; 90:101041. [PMID: 34999237 DOI: 10.1016/j.preteyeres.2022.101041] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Treatment of primary intraocular uveal melanoma has developed considerably, its driver genes are largely unraveled, and the ways to assess its risk for metastases are very precise, being based on an international staging system and genetic data. Unfortunately, the risk of distant metastases, which emerge in approximately one half of all patients, is unaltered. Metastases are the leading single cause of death after uveal melanoma is diagnosed, yet no consensus exists regarding surveillance, staging, and treatment of disseminated disease, and survival has not improved until recently. The final frontier in conquering uveal melanoma lies in solving these issues to cure metastatic disease. Most studies on metastatic uveal melanoma are small, uncontrolled, retrospective, and do not report staging. Meta-analyses confirm a median overall survival of 10-13 months, and a cure rate that approaches nil, although survival exceeding 5 years is possible, estimated 2% either with first-line treatment or with best supportive care. Hepatic ultrasonography and magnetic resonance imaging as surveillance methods have a sensitivity of 95-100% and 83-100%, respectively, to detect metastases without radiation hazard according to prevailing evidence, but computed tomography is necessary for staging. No blood-based tests additional to liver function tests are generally accepted. Three validated staging systems predict, each in defined situations, overall survival after metastasis. Their essential components include measures of tumor burden, liver function, and performance status or metastasis free interval. Age and gender may additionally influence survival. Exceptional mutational events in metastases may make them susceptible to checkpoint inhibitors. In a large meta-analysis, surgical treatment was associated with 6 months longer median overall survival as compared to conventional chemotherapy and, recently, tebentafusp as first-line treatment at the first interim analysis of a randomized phase III trial likewise provided a 6 months longer median overall survival compared to investigator's choice, mostly pembrolizumab; these treatments currently apply to selected patients. Promoting dormancy of micrometastases, harmonizing surveillance protocols, promoting staging, identifying predictive factors, initiating controlled clinical trials, and standardizing reporting will be critical steppingstones in reaching the final frontier of curing metastatic uveal melanoma.
Collapse
Affiliation(s)
- Elina S Rantala
- Ocular Oncology Service, Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4 C, PL 220, FI-00029, HUS, Helsinki, Finland.
| | - Micaela M Hernberg
- Comprehensive Cancer Center, Department of Oncology, Helsinki University Hospital and University of Helsinki, Paciuksenkatu 3, PL 180, FI-00029, HUS, Helsinki, Finland.
| | | | - Hans E Grossniklaus
- Section of Ocular Oncology, Emory Eye Center, 1365 Clifton Road B, Atlanta, GA, 30322, USA.
| | - Tero T Kivelä
- Ocular Oncology Service, Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4 C, PL 220, FI-00029, HUS, Helsinki, Finland.
| |
Collapse
|
18
|
Jo H, Yagishita S, Hayashi Y, Ryu S, Suzuki M, Kohsaka S, Ueno T, Matsumoto Y, Horinouchi H, Ohe Y, Watanabe SI, Motoi N, Yatabe Y, Mano H, Takahashi K, Hamada A. Comparative study on the efficacy and exposure of molecular target agents in non-small cell lung cancer PDX models with driver genetic alterations. Mol Cancer Ther 2021; 21:359-370. [PMID: 34911818 DOI: 10.1158/1535-7163.mct-21-0371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/11/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
Abstract
Patient-derived xenografts (PDXs) can adequately reflect clinical drug efficacy. However, the methods for evaluating drug efficacy are not fully established. We selected five non-small cell lung cancer (NSCLC) PDXs with genetic alterations from established PDXs and the corresponding molecular targeted therapy was administered orally for 21 consecutive days. Genetic analysis, measurement of drug concentrations in blood and tumors using liquid chromatography and tandem mass spectrometry, and analysis of drug distribution in tumors using matrix-assisted laser desorption/ionization mass spectrometry were performed. Fifteen (20%) PDXs were established using samples collected from 76 NSCLC patients with genetic alterations. The genetic alterations observed in original patients were largely maintained in PDXs. We compared the drug efficacy in original patients and PDX models; the efficacies against certain PDXs correlated with the clinical effects, while those against the others did not. We determined blood and intratumor concentrations in the PDX model, but both concentrations were low, and no evident correlation with the drug efficacy could be observed. The intratumoral spatial distribution of the drugs was both homogeneous and heterogeneous for each drug, and the distribution was independent of the expression of the target protein. The evaluation of drug efficacy in PDXs enabled partial reproduction of the therapeutic effect in original patients. A more detailed analysis of systemic and intratumoral pharmacokinetics may help clarify the mode of action of drugs. Further development of evaluation methods and indices to improve the prediction accuracy of clinical efficacy is warranted.
Collapse
Affiliation(s)
- Hitomi Jo
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| | - Yoshiharu Hayashi
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| | - Shoraku Ryu
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| | - Mikiko Suzuki
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute
| | | | | | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital
| | | | - Noriko Motoi
- Department of Pathology, National Cancer Center Hospital
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital
| | | | | | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute
| |
Collapse
|
19
|
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
|
20
|
Hollis RL, Stillie LJ, Hopkins S, Bartos C, Churchman M, Rye T, Nussey F, Fegan S, Nirsimloo R, Inman GJ, Herrington CS, Gourley C. Clinicopathological Determinants of Recurrence Risk and Survival in Mucinous Ovarian Carcinoma. Cancers (Basel) 2021; 13:5839. [PMID: 34830992 PMCID: PMC8616033 DOI: 10.3390/cancers13225839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Mucinous ovarian carcinoma (MOC) is a unique form of ovarian cancer. MOC typically presents at early stage but demonstrates intrinsic chemoresistance; treatment of advanced-stage and relapsed disease is therefore challenging. We harness a large retrospective MOC cohort to identify factors associated with recurrence risk and survival. A total of 151 MOC patients were included. The 5 year disease-specific survival (DSS) was 84.5%. Risk of subsequent recurrence after a disease-free period of 2 and 5 years was low (8.3% and 5.6% over the next 10 years). The majority of cases were FIGO stage I (35.6% IA, 43.0% IC). Multivariable analysis identified stage and pathological grade as independently associated with DSS (p < 0.001 and p < 0.001). Grade 1 stage I patients represented the majority of cases (53.0%) and demonstrated exceptional survival (10 year DSS 95.3%); survival was comparable between grade I stage IA and stage IC patients, and between grade I stage IC patients who did and did not receive adjuvant chemotherapy. At 5 years following diagnosis, the proportion of grade 1, 2 and 3 patients remaining disease free was 89.5%, 74.9% and 41.7%; the corresponding proportions for FIGO stage I, II and III/IV patients were 91.1%, 76.7% and 19.8%. Median post-relapse survival was 5.0 months. Most MOC patients present with low-grade early-stage disease and are at low risk of recurrence. New treatment options are urgently needed to improve survival following relapse, which is associated with extremely poor prognosis.
Collapse
Affiliation(s)
- Robert L. Hollis
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| | - Lorna J. Stillie
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, Scotland, UK;
| | - Samantha Hopkins
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh EH4 2LF, Scotland, UK; (S.H.); (F.N.); (R.N.)
| | - Clare Bartos
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| | - Michael Churchman
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| | - Tzyvia Rye
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| | - Fiona Nussey
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh EH4 2LF, Scotland, UK; (S.H.); (F.N.); (R.N.)
| | - Scott Fegan
- The Simpson Centre for Reproductive Health, Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh EH16 4TJ, Scotland, UK;
| | - Rachel Nirsimloo
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh EH4 2LF, Scotland, UK; (S.H.); (F.N.); (R.N.)
| | - Gareth J. Inman
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, Scotland, UK;
| | - C. Simon Herrington
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| | - Charlie Gourley
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, Scotland, UK; (L.J.S.); (C.B.); (M.C.); (T.R.); (C.S.H.); (C.G.)
| |
Collapse
|
21
|
Clinical performance in ERN eUROGEN for penile, testicular, adrenal and soft tissue cancers. Eur J Surg Oncol 2021; 48:680-686. [PMID: 34893364 DOI: 10.1016/j.ejso.2021.11.014] [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: 10/06/2021] [Accepted: 11/14/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND European Reference Network (ERN) eUROGEN is a cross-border collaboration set up by the European Commission in 2017 aimed at tackling rare urogenital conditions, including cancers. OBJECTIVE This report aims to assess ERN eUROGEN's operational activity with a focus on rare urogenital cancers. DESIGN, SETTING AND PARTICIPANTS Data for descriptive analyses were collected retrospectively between 2013 and 2017, and prospectively between 2018 and 2020. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Operational indicators were set by the European Commission from 2018. Additionally, in 2019/20 centres self-assessed clinical service provision and provided clinical metrics for rare cancer specialist centres as established by experts. RESULTS AND LIMITATIONS Results revealed that the cumulative rare urogenital cancer population increased 519.8% from 1,631 in 2013 to 10,109 in 2020. This may provide opportunities for research and creation of a large cancer registry. In total, ten centres met the clinical requirements for rare cancer specialist centres providing evidence of high-volume. Differences in data collection methods between centres limit further analyses. Other rare cancer data identified 39 panel discussions, three webinars, and eight publications. CONCLUSIONS Whilst limitations to data analysis remain, ERN eUROGEN has demonstrated excellent operational performance with promising opportunities for rare cancer research.
Collapse
|
22
|
Mittra A, Takebe N, Florou V, Chen AP, Naqash AR. The emerging landscape of immune checkpoint inhibitor based clinical trials in adults with advanced rare tumors. Hum Vaccin Immunother 2021; 17:1935-1939. [PMID: 33325769 PMCID: PMC8189105 DOI: 10.1080/21645515.2020.1854604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
"Rare cancers" are a diverse collection of cancers that collectively account for approximately 20% of all adult cancers in the United States. Their rarity has caused an underrepresentation of these cancers in preclinical research and clinical trials, leading to fewer (and often no) treatment options for patients backed by robust clinical evidence. The recent advent of immune checkpoint inhibitors (ICIs) into the oncologist's armamentarium, while revolutionizing the treatment of many common cancers, has also started to make gradual inroads into the treatment of certain rare cancers. One reason is that the efficacy of ICIs depends more on factors intrinsic to the tumor cells and the tumor microenvironment and less on tumor histology. Recent years have seen ICI approvals in many rare cancers, and many trials are being designed using ICIs as single agents or in combination. In this commentary, we present an overview of the emerging role of ICIs in some rare cancers.
Collapse
Affiliation(s)
- Arjun Mittra
- Division of Medical Oncology, The Ohio State University James Cancer Hospital, Columbus, OH, USA
| | - Naoko Takebe
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis. National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Vaia Florou
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alice P Chen
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis. National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Abdul Rafeh Naqash
- Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis. National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| |
Collapse
|
23
|
Reinke DK. Meaningful engagement of the patient in rare cancer research: sarcoma as an exemplar. Curr Probl Cancer 2021; 45:100772. [PMID: 34289946 DOI: 10.1016/j.currproblcancer.2021.100772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 10/21/2022]
Abstract
Patient advocates who understand scientific methods and proper research processes can bring valuable perspectives to modern research. This is particularly important in rare cancers like sarcoma as each patient becomes a precious source of information to better diagnose, understand the biology and the effect of treatment. Reviewing approaches used by other cancer patient advocates can provide valuable insights to develop effective research advocates in rare cancers such as sarcoma.
Collapse
Affiliation(s)
- Denise K Reinke
- SARC (Sarcoma Alliance for Research through Collaboration), Ann Arbor, MI.
| |
Collapse
|
24
|
The design of a Bayesian platform trial to prevent and eradicate inhibitors in patients with hemophilia. Blood Adv 2021; 4:5433-5441. [PMID: 33156923 DOI: 10.1182/bloodadvances.2020002789] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [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
|
25
|
Edenfield WJ, Chung K, O'Rourke M, Cull E, Martin J, Bowers H, Smith W, Gluck WL. A Phase II Study of Durvalumab in Combination with Tremelimumab in Patients with Rare Cancers. Oncologist 2021; 26:e1499-e1507. [PMID: 33893692 DOI: 10.1002/onco.13798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/13/2021] [Indexed: 01/29/2023] Open
Abstract
LESSONS LEARNED Disease control with signals of response were demonstrated, which should lead to future validating clinical trials using checkpoint inhibitors in this underserved rare malignancy population. Although the study of single types of rare cancers is practically challenging, clinical trial designs that aggregate such patients into cohorts treated similarly are feasible, even in the community setting. BACKGROUND Patients with rare cancers are an underserved population with limited access to clinical trials aside from phase I trials in the refractory setting. Treatment of these patients is often based on collections of anecdotes and small denominator review articles. Despite broad evidence of efficacy of combined immune checkpoint blockade across multiple tumor types, patients with rare tumors have not been afforded the opportunity for these therapies. METHODS A phase II, investigator-initiated, single institution trial using durvalumab (1,500 mg every [Q]4 weeks × 13) and tremelimumab (75 mg Q4 weeks × 7, then Q12 weeks × 2) is reported. The population included 50 patients with advanced rare solid tumors (incidence <6/100,000 per year). The phase II dose and safety profile were defined in prior phase I trials. All patients had exhausted standard therapy options and all had received at least one prior line of systemic therapy (n = 49) unless a standard treatment option did not exist (n = 1). RESULTS A complete response was demonstrated in one patient with anal cancer. Striking partial responses were seen in four patients. Prolonged disease stability was noted in 18 patients. Thirteen patients experienced disease progression. Patients were considered unevaluable if unable to initiate therapy (n = 6) or unable to complete two cycles of therapy (n = 8). In all cases, patients were unevaluable because of clinical deterioration. The toxicity profile paralleled prior published studies. Toxicities were manageable and without new signals. There were two events of grade 4 immune-mediated hepatitis and one death from pneumonitis. CONCLUSION This single-cohort basket trial demonstrated clinical activity from combined checkpoint blockade in 23 of the 36 evaluable patients. Patients with rare cancers, not eligible for immunotherapy via conventional clinical trial mechanisms, should be considered for this therapy through compassionate use, further clinical trials, and national registry programs.
Collapse
Affiliation(s)
| | - Ki Chung
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | - Mark O'Rourke
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | - Elizabeth Cull
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | - Julie Martin
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | - Heather Bowers
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | - Wesley Smith
- Prisma Health Cancer Institute, Greenville, South Carolina, USA
| | | |
Collapse
|
26
|
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
|
27
|
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: 2.7] [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
|
28
|
Lansu J, Bovée JVMG, Braam P, van Boven H, Flucke U, Bonenkamp JJ, Miah AB, Zaidi SH, Thway K, Bruland ØS, Baldini EH, Jebsen NL, Scholten AN, van den Ende PLA, Krol ADG, Ubbels JF, van der Hage JA, van Werkhoven E, Klomp HM, van der Graaf WTA, van Coevorden F, Schrage Y, van Houdt WJ, Haas RL. Dose Reduction of Preoperative Radiotherapy in Myxoid Liposarcoma: A Nonrandomized Controlled Trial. JAMA Oncol 2021; 7:e205865. [PMID: 33180100 PMCID: PMC7662477 DOI: 10.1001/jamaoncol.2020.5865] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
IMPORTANCE Currently, preoperative radiotherapy for all soft-tissue sarcomas is identical at a 50-Gy dose level, which can be associated with morbidity, particularly wound complications. The observed clinical radiosensitivity of the myxoid liposarcoma subtype might offer the possibility to reduce morbidity. OBJECTIVE To assess whether a dose reduction of preoperative radiotherapy for myxoid liposarcoma would result in comparable oncological outcome with less morbidity. DESIGN, SETTING, AND PARTICIPANTS The Dose Reduction of Preoperative Radiotherapy in Myxoid Liposarcomas (DOREMY) trial is a prospective, single-group, phase 2 nonrandomized controlled trial being conducted in 9 tertiary sarcoma centers in Europe and the US. Participants include adults with nonmetastatic, biopsy-proven and translocation-confirmed myxoid liposarcoma of the extremity or trunk who were enrolled between November 24, 2010, and August 1, 2019. Data analyses, using both per-protocol and intention-to-treat approaches, were conducted from November 24, 2010, to January 31, 2020. INTERVENTIONS The experimental preoperative radiotherapy regimen consisted of 36 Gy in once-daily 2-Gy fractions, with subsequent definitive surgical resection after an interval of 4 or more weeks. MAIN OUTCOMES AND MEASURES As a short-term evaluable surrogate for local control, the primary end point was centrally reviewed pathologic treatment response. The experimental regimen was regarded as a success when 70% or more of the resection specimens showed extensive treatment response, defined as 50% or greater of the tumor volume containing treatment effects. Morbidity outcomes consisted of wound complications and late toxic effects. RESULTS Among the 79 eligible patients, 44 (56%) were men and the median (interquartile range) age was 45 (39-56) years. Two patients did not undergo surgical resection because of intercurrent metastatic disease. Extensive pathological treatment response was observed in 70 of 77 patients (91%; posterior mean, 90.4%; 95% highest probability density interval, 83.8%-96.4%). The local control rate was 100%. The rate of wound complication requiring intervention was 17%, and the rate of grade 2 or higher toxic effects was 14%. CONCLUSIONS AND RELEVANCE The findings of the DOREMY nonrandomized clinical trial suggest that deintensification of preoperative radiotherapy dose is effective and oncologically safe and is associated with less morbidity than historical controls, although differences in radiotherapy techniques and follow-up should be considered. A 36-Gy dose delivered in once-daily 2-Gy fractions is proposed as a dose-fractionation approach for myxoid liposarcoma, given that phase 3 trials are logistically impossible to execute in rare cancers. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02106312.
Collapse
Affiliation(s)
- Jules Lansu
- Sarcoma Unit, Department of Radiotherapy, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Pètra Braam
- Department of Radiotherapy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hester van Boven
- Sarcoma Unit, Department of Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Uta Flucke
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Aisha B. Miah
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden Hospital, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Shane H. Zaidi
- Sarcoma Unit, Department of Radiotherapy and Physics, The Royal Marsden Hospital, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Khin Thway
- The Institute of Cancer Research, London, United Kingdom
- Sarcoma Unit, Department of Pathology, The Royal Marsden Hospital, London, United Kingdom
| | - Øyvind S. Bruland
- Department of Medical Oncology and Radiotherapy, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elizabeth H. Baldini
- Department of Radiation Oncology, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
| | - Nina L. Jebsen
- Center for Bone and Soft Tissue Tumors, Department of Oncology, Department of Orthopedic Surgery, Haukeland University Hospital, Bergen, Norway
- Center for Cancer Biomarkers, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Astrid N. Scholten
- Sarcoma Unit, Department of Radiotherapy, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Piet L. A. van den Ende
- Department of Radiotherapy, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Augustinus D. G. Krol
- Department of Radiotherapy, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan F. Ubbels
- Department of Radiotherapy, Groningen University Medical Center, Groningen, the Netherlands
| | - Jos A. van der Hage
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Erik van Werkhoven
- Department of Biometrics, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Houke M. Klomp
- Sarcoma Unit, Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Frits van Coevorden
- Sarcoma Unit, Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Yvonne Schrage
- Sarcoma Unit, Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Winan J. van Houdt
- Sarcoma Unit, Department of Surgery, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rick L. Haas
- Sarcoma Unit, Department of Radiotherapy, the Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Radiotherapy, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
29
|
De Vita A, Bongiovanni A, Blay JY, Ibrahim T. Editorial: New Insights Into the Landscape of Rare Tumors: Translational and Clinical Research Perspective. Front Oncol 2020; 10:593785. [PMID: 33194758 PMCID: PMC7659953 DOI: 10.3389/fonc.2020.593785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/23/2020] [Indexed: 11/17/2022] Open
Affiliation(s)
- Alessandro De Vita
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Meldola, Italy
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.,Unicancer, Paris, France.,EURACAN (European network for Rare adult solid Cancer) EC 739521, Lyon, France
| | - Toni Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Meldola, Italy
| |
Collapse
|
30
|
Basket trials: From tumour gnostic to tumour agnostic drug development. Cancer Treat Rev 2020; 90:102082. [DOI: 10.1016/j.ctrv.2020.102082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
|
31
|
Mäkelä R, Arjonen A, Suryo Rahmanto A, Härmä V, Lehtiö J, Kuopio T, Helleday T, Sangfelt O, Kononen J, Rantala JK. Ex vivo assessment of targeted therapies in a rare metastatic epithelial-myoepithelial carcinoma. Neoplasia 2020; 22:390-398. [PMID: 32645560 PMCID: PMC7341452 DOI: 10.1016/j.neo.2020.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/30/2022]
Abstract
Epithelial-myoepithelial carcinoma (EMC) is a rare subtype of salivary gland neoplasms. Since the initial description of the cancer, just over 300 cases have been reported. EMCs occupy a biphasic cellular differentiation-state defined by the constitution of two cell types representing epithelial and myoepithelial lineages, yet the functional consequence of the differentiation-state heterogeneity with respect to therapy resistance of the tumors remains unclear. The reported local recurrence rate of the cases is approximately 30%, and while distant metastases are rare, a significant fraction of these cases are reported to receive no survival benefit from radio- or chemotherapy given in addition to surgery. Moreover, no targeted therapies have been reported for these neoplasms. We report here the first use and application of ex vivo drug screening together with next generation sequencing to assess targeted treatment strategies for a rare metastatic epithelial-myoepithelial carcinoma. Results of the ex vivo drug screen demonstrate significant differential therapeutic sensitivity between the epithelial and myoepithelial intra-tumor cell lineages suggesting that differentiation-state heterogeneity within epithelial-myoepithelial carcinomas may present an outlet to partial therapeutic responses to targeted therapies including MEK and mTOR inhibitors. These results suggest that the intra-tumor lineage composition of EMC could be an important factor to be assessed when novel treatments are being evaluated for management of metastatic EMC.
Collapse
Affiliation(s)
| | | | | | - Ville Härmä
- Misvik Biology Oy, Turku, Finland; University of Sheffield, Department of Oncology and Metabolism, South Yorkshire, Sheffield, UK.
| | - Janne Lehtiö
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden.
| | - Teijo Kuopio
- Central Finland Health Care District, Jyväskylä Medical Centre, Jyväskylä, Finland.
| | - Thomas Helleday
- University of Sheffield, Department of Oncology and Metabolism, South Yorkshire, Sheffield, UK
| | - Olle Sangfelt
- Karolinska Institutet, Department of Cell and Molecular Biology, Stockholm, Sweden.
| | - Juha Kononen
- Central Finland Health Care District, Jyväskylä Medical Centre, Jyväskylä, Finland; Docrates Hospital, Helsinki, Finland.
| | - Juha K Rantala
- Misvik Biology Oy, Turku, Finland; University of Sheffield, Department of Oncology and Metabolism, South Yorkshire, Sheffield, UK.
| |
Collapse
|
32
|
Clinical development of cell therapies for cancer: The regulators' perspective. Eur J Cancer 2020; 138:41-53. [PMID: 32836173 DOI: 10.1016/j.ejca.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/04/2020] [Indexed: 11/21/2022]
Abstract
Novel cell therapies for haematological malignancies and solid tumours address pressing clinical need while offering potentially paradigm shifts in efficacy. However, innovative development risks outflanking information on statutory frameworks, regulatory guidelines and their working application. Meeting this challenge, regulators offer wide-ranging expertise and experience in confidential scientific and regulatory advice. We advocate early incorporation of regulatory perspectives to support strategic development of clinical programmes. We examine critical issues and key advances in clinical oncology trials to highlight practical approaches to optimising the clinical development of cell therapies. We recommend early consideration of collaborative networks, early-access schemes, reducing bias in single-arm trials, adaptive trials, clinical end-points supporting risk/benefit and cost/benefit analyses, companion diagnostics, real-world data and common technical issues. This symbiotic approach between developers and regulators should reduce development risk, safely expedite marketing authorisation, and promote early, wider availability of potentially transformative cell therapies for cancer.
Collapse
|
33
|
Effect size estimates from umbrella designs: Handling patients with a positive test result for multiple biomarkers using random or pragmatic subtrial allocation. PLoS One 2020; 15:e0237441. [PMID: 32797088 PMCID: PMC7428134 DOI: 10.1371/journal.pone.0237441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Umbrella trials have been suggested to increase trial conduct efficiency when investigating different biomarker-driven experimental therapies. An overarching platform is used for patient screening and subsequent subtrial allocation according to patients’ biomarker status. Two subtrial allocation schemes for patients with a positive test result for multiple biomarkers are (i) the pragmatic allocation to the eligible subtrial with the currently fewest included patients and (ii) the random allocation to one of the eligible subtrials. Obviously, the subtrials compete for such patients which are consequently underrepresented in the subtrials. To address questions of the impact of an umbrella design in general as well as with respect to subtrial allocation and analysis method, we investigate an umbrella trial with two parallel group subtrials and discuss generalisations. First, we analytically quantify the impact of the umbrella design with random allocation on the number of patients needed to be screened, the biomarker status distribution and treatment effect estimates compared to the corresponding gold standard of an independent parallel group design. Using simulations and real data, we subsequently compare both allocation schemes and investigate weighted linear regression modelling as possible analysis method for the umbrella design. Our results show that umbrella designs are more efficient than the gold standard. However, depending on the biomarker status distribution in the disease population, an umbrella design can introduce differences in estimated treatment effects in the presence of an interaction between treatment and biomarker status. In principle, weighted linear regression together with the random allocation scheme can address this difference though it is difficult to assess if such an approach is applicable in practice. In any case, caution is required when using treatment effect estimates derived from umbrella designs for e.g. future trial planning or meta-analyses.
Collapse
|
34
|
Keech J, Dai WF, Trudeau M, Mercer RE, Naipaul R, Wright FC, Ferguson SE, Darling G, Gavura S, Eisen A, Kouroukis CT, Beca J, Chan KKW. Impact of rarity on Canadian oncology health technology assessment and funding. Int J Technol Assess Health Care 2020; 36:1-6. [PMID: 32779560 DOI: 10.1017/s0266462320000483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVES The pan-Canadian Oncology Drug Review (pCODR) evaluates new cancer drugs for public funding recommendations. While pCODR's deliberative framework evaluates overall clinical benefit and includes considerations for exceptional circumstances, rarity of indication is not explicitly addressed. Given the high unmet need that typically accompanies these indications, we explored the impact of rarity on oncology HTA recommendations and funding decisions. METHODS We examined pCODR submissions with final recommendations from 2012 to 2017. Incidence rates were calculated using pCODR recommendation reports and statistics from the Canadian Cancer Society. Indications were classified as rare if the incidence rate was lower than 1/100,000 diagnoses, a definition referenced by the Canadian Agency for Drugs and Technologies in Health. Each pCODR final report was examined for the funding recommendation/justification, level of supporting evidence (presence of a randomized control trial [RCT]), and time to funding (if applicable). RESULTS Of the ninety-six pCODR reviews examined, 16.6 percent were classified as rare indications per above criteria. While the frequency of positive funding recommendations were similar between rare and nonrare indication (78.6 vs. 75 percent), rare indications were less likely to be presented with evidence from RCT (50 vs. 90 percent). The average time to funding did not differ significantly across provinces. CONCLUSION Rare indications appear to be associated with weaker clinical evidence. There appears to be no association between rarity, positive funding recommendations, and time to funding. Further work will evaluate factors associated with positive recommendations and the real-world utilization of funded treatments for rare indications.
Collapse
Affiliation(s)
- James Keech
- Cancer Care Ontario & Canadian Centre for Applied Research in Cancer Control, Toronto, Ontario, Canada
| | - Wei Fang Dai
- Cancer Care Ontario & Canadian Centre for Applied Research in Cancer Control, Toronto, Ontario, Canada
| | - Maureen Trudeau
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Rebecca E Mercer
- Cancer Care Ontario & Canadian Centre for Applied Research in Cancer Control, Toronto, Ontario, Canada
| | | | - Frances C Wright
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | | - Gail Darling
- University Health Network, Toronto, Ontario, Canada
| | - Scott Gavura
- Cancer Care Ontario & Canadian Centre for Applied Research in Cancer Control, Toronto, Ontario, Canada
| | - Andrea Eisen
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - C Tom Kouroukis
- Juravinski Cancer Centre, Hamilton Health Sciences Centre, Hamilton, Ontario, Canada
| | - Jaclyn Beca
- Cancer Care Ontario & Canadian Centre for Applied Research in Cancer Control, Toronto, Ontario, Canada
| | - Kelvin K W Chan
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
35
|
The Evolution of Master Protocol Clinical Trial Designs: A Systematic Literature Review. Clin Ther 2020; 42:1330-1360. [DOI: 10.1016/j.clinthera.2020.05.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/10/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
|
36
|
Arjonen A, Mäkelä R, Härmä V, Rintanen N, Kuopio T, Kononen J, Rantala JK. Image-based ex vivo drug screen to assess targeted therapies in recurrent thymoma. Lung Cancer 2020; 145:27-32. [DOI: 10.1016/j.lungcan.2020.04.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/07/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022]
|
37
|
Nannini M, Valerio DS, Gruppioni E, Altimari A, Chiusole B, Saponara M, Pantaleo MA, Brunello A. Complete radiological response to first-line regorafenib in a patient with abdominal relapse of BRAF V600E mutated GIST. Therap Adv Gastroenterol 2020; 13:1756284820927305. [PMID: 32647535 PMCID: PMC7328213 DOI: 10.1177/1756284820927305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 04/21/2020] [Indexed: 02/04/2023] Open
Abstract
Up to 13% of KIT and PDGFRA wild-type (WT) gastrointestinal stromal tumours (GIST) harbour a BRAF mutation, mostly involving the exon 15 V600E hot spot. Even if BRAF mutation is recognized as druggable target in other solid tumours, currently advanced BRAF-mutant GIST share the same therapeutical algorithm of KIT/PDGFRA mutants. We report a complete radiological response in a 51-year-old woman with V600E BRAF-mutated metastatic GIST who was treated with regorafenib (REG) as first-line therapy. REG represents the standard third-line therapy for advanced GIST patients progressing on or failing to respond to imatinib and sunitinib. However, according to its wide spectrum of action, with MAPK signalling pathway blockade at different levels, metastatic KIT/PDGFRA WT, including BRAF mutants, may benefit from REG upfront in first line.
Collapse
Affiliation(s)
| | - Di Scioscio Valerio
- Department of Radiology, S.Orsola Malpighi Hospital,
University of Bologna, Italy
| | - Elisa Gruppioni
- Laboratory of Oncologic Molecular Pathology,
S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Annalisa Altimari
- Laboratory of Oncologic Molecular Pathology,
S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Benedetta Chiusole
- Medical Oncology 1 Unit, Department of Oncology, Istituto
Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Maristella Saponara
- Department of Specialized, Experimental and Diagnostic
Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Specialized, Experimental and Diagnostic
Medicine, S.Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy,“Giorgio Prodi” Cancer Research Center, University of
Bologna, Bologna, Italy
| | - Antonella Brunello
- Medical Oncology 1 Unit, Department of Oncology, Istituto
Oncologico Veneto IOV - IRCCS, Padova, Italy
| |
Collapse
|
38
|
Mäkelä R, Arjonen A, Härmä V, Rintanen N, Paasonen L, Paprotka T, Rönsch K, Kuopio T, Kononen J, Rantala JK. Ex vivo modelling of drug efficacy in a rare metastatic urachal carcinoma. BMC Cancer 2020; 20:590. [PMID: 32576176 PMCID: PMC7313172 DOI: 10.1186/s12885-020-07092-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/19/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Ex vivo drug screening refers to the out-of-body assessment of drug efficacy in patient derived vital tumor cells. The purpose of these methods is to enable functional testing of patient specific efficacy of anti-cancer therapeutics and personalized treatment strategies. Such approaches could prove powerful especially in context of rare cancers for which demonstration of novel therapies is difficult due to the low numbers of patients. Here, we report comparison of different ex vivo drug screening methods in a metastatic urachal adenocarcinoma, a rare and aggressive non-urothelial bladder malignancy that arises from the remnant embryologic urachus in adults. METHODS To compare the feasibility and results obtained with alternative ex vivo drug screening techniques, we used three different approaches; enzymatic cell viability assay of 2D cell cultures and image-based cytometry of 2D and 3D cell cultures in parallel. Vital tumor cells isolated from a biopsy obtained in context of a surgical debulking procedure were used for screening of 1160 drugs with the aim to evaluate patterns of efficacy in the urachal cancer cells. RESULTS Dose response data from the enzymatic cell viability assay and the image-based assay of 2D cell cultures showed the best consistency. With 3D cell culture conditions, the proliferation rate of the tumor cells was slower and potency of several drugs was reduced even following growth rate normalization of the responses. MEK, mTOR, and MET inhibitors were identified as the most cytotoxic targeted drugs. Secondary validation analyses confirmed the efficacy of these drugs also with the new human urachal adenocarcinoma cell line (MISB18) established from the patient's tumor. CONCLUSIONS All the tested ex vivo drug screening methods captured the patient's tumor cells' sensitivity to drugs that could be associated with the oncogenic KRASG12V mutation found in the patient's tumor cells. Specific drug classes however resulted in differential dose response profiles dependent on the used cell culture method indicating that the choice of assay could bias results from ex vivo drug screening assays for selected drug classes.
Collapse
Affiliation(s)
- Rami Mäkelä
- Misvik Biology Ltd, Karjakatu 35 B, FI-20520, Turku, Finland
| | - Antti Arjonen
- Misvik Biology Ltd, Karjakatu 35 B, FI-20520, Turku, Finland.,Brinter Ltd, Turku, Finland
| | - Ville Härmä
- Misvik Biology Ltd, Karjakatu 35 B, FI-20520, Turku, Finland.,University of Sheffield, Sheffield, UK
| | - Nina Rintanen
- Central Finland Health Care District, Jyväskylä, Finland
| | | | - Tobias Paprotka
- Eurofins Genomics Europe Sequencing GmbH, Constance, Germany
| | - Kerstin Rönsch
- Eurofins Genomics Europe Sequencing GmbH, Constance, Germany
| | - Teijo Kuopio
- Central Finland Health Care District, Jyväskylä, Finland
| | - Juha Kononen
- Central Finland Health Care District, Jyväskylä, Finland.,Docrates Hospital, Helsinki, Finland
| | - Juha K Rantala
- Misvik Biology Ltd, Karjakatu 35 B, FI-20520, Turku, Finland. .,University of Sheffield, Sheffield, UK.
| |
Collapse
|
39
|
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: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
|
40
|
Everett CC, Reynolds C, Fernandez C, Stocken DD, Sharples LD, Sathyapalan T, Heller S, Storey RF, Ajjan RA. Rationale and design of the LIBERATES trial: Protocol for a randomised controlled trial of flash glucose monitoring for optimisation of glycaemia in individuals with type 2 diabetes and recent myocardial infarction. Diab Vasc Dis Res 2020; 17:1479164120957934. [PMID: 33081502 PMCID: PMC7919208 DOI: 10.1177/1479164120957934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hyperglycaemia in individuals with type 2 diabetes (T2D) and myocardial infarction (MI) is associated with guarded clinical prognosis. Studies improving glucose levels in T2D following MI relied on HbA1c as the main glycaemic marker, failing to address potential adverse effects of hypoglycaemia and glucose variability. We describe the design of the LIBERATES trial that investigates the role of flash glucose monitoring in optimising glycaemic markers in high vascular risk individuals with T2D. This multicentre trial is designed to recruit up to 150 insulin and/or sulphonylurea-treated T2D patients, within 5 days of a proven MI. Individuals will be randomised 1:1 into intervention and control groups using flash glucose monitoring sensors and traditional self-monitoring of blood glucose, respectively. The control group will also wear a blinded continuous glucose monitoring sensor. The primary outcome is the difference in time spent in euglycaemia (defined as glucose levels between 3.9-10.0 mmol/l), comparing study groups 3 months following recruitment, assessed daily for 14 days and as an average. Secondary and exploratory end points include time spent in hypoglycaemia and hyperglycaemia, HbA1c, quality of life measures, major adverse cardiac events and cost-effectiveness of the intervention. This study will establish the role of flash glucose monitoring in glycaemic management of individuals with T2D sustaining a cardiac event.(Trial Registration: ISRCTN14974233, registered 12th June 2017).
Collapse
Affiliation(s)
- Colin C Everett
- Clinical Trials Research Unit, University of Leeds, Leeds, West Yorkshire, UK
| | - Catherine Reynolds
- Clinical Trials Research Unit, University of Leeds, Leeds, West Yorkshire, UK
| | - Catherine Fernandez
- Clinical Trials Research Unit, University of Leeds, Leeds, West Yorkshire, UK
| | - Deborah D Stocken
- Clinical Trials Research Unit, University of Leeds, Leeds, West Yorkshire, UK
| | - Linda D Sharples
- London School of Hygiene and Tropical Medicine, University of London, Bloomsbury, London, UK
| | | | - Simon Heller
- Department of Oncology and Metabolism, Sheffield Teaching Hospitals Trust, Sheffield, South Yorkshire, UK
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Ramzi A Ajjan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, West Yorkshire, UK
- Department of Diabetes and Endocirnology, Leeds Teaching Hospitals Trust, Leeds, West Yorkshire, UK
- Ramzi A Ajjan, Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, The LIGHT Laboratories, Clarendon Way, Leeds, West Yorkshire LS2 9JT, UK.
| |
Collapse
|
41
|
Precision medicine in rare tumors and the need for multicenter trials and international collaboratives: Sinonasal cancer as paradigm. Oral Oncol 2020; 109:104737. [PMID: 32340860 DOI: 10.1016/j.oraloncology.2020.104737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 01/11/2023]
|
42
|
|
43
|
Cullen M, Huddart R, Joffe J, Gardiner D, Maynard L, Hutton P, Mazhar D, Shamash J, Wheater M, White J, Goubar A, Porta N, Witts S, Lewis R, Hall E. The 111 Study: A Single-arm, Phase 3 Trial Evaluating One Cycle of Bleomycin, Etoposide, and Cisplatin as Adjuvant Chemotherapy in High-risk, Stage 1 Nonseminomatous or Combined Germ Cell Tumours of the Testis. Eur Urol 2020; 77:344-351. [PMID: 31901440 PMCID: PMC7026695 DOI: 10.1016/j.eururo.2019.11.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 11/26/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Standard management in the UK for high-risk stage 1 nonseminoma germ cell tumours of the testis (NSGCTT) is two cycles of adjuvant bleomycin, etoposide (360 mg/m2), and cisplatin (BE360P) chemotherapy, or surveillance. OBJECTIVE To test whether one cycle of BE500P achieves similar recurrence rates to two cycles of BE360P. DESIGN, SETTING, AND PARTICIPANTS A total of 246 patients with vascular invasion-positive stage 1 NSGCTT or combined seminoma + NSGCTT were centrally registered in a single-arm prospective study. INTERVENTION One cycle comprising bleomycin 30000 IU on days 1, 8, and 15, etoposide 165 mg/m2 on days 1-3, and cisplatin 50 mg/m2 on days 1-2, plus antibacterial and granulocyte colony stimulating factor prophylaxis. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was 2-yr malignant recurrence (MR); the aim was to exclude a rate of ≥5%. Participants had regular imaging and tumour marker (TM) assessment for 5 yr. RESULTS AND LIMITATIONS The median follow-up was 49 mo (interquartile range 37-60). Ten patients with rising TMs at baseline were excluded. Four patients had MR at 6, 7, 13, and 27 mo; all received second-line chemotherapy and surgery and three remained recurrence-free at 5 yr. The 2-yr MR rate was 1.3% (95% confidence interval 0.3-3.7%). Three patients developed nonmalignant recurrences with localised teratoma differentiated, rendered disease-free after surgery. Grade 3-4 febrile neutropenia occurred in 6.8% of participants. CONCLUSIONS BE500P is safe and the 2-yr MR rate is consistent with that seen following two BE360P cycles. The 111 study is the largest prospective trial investigating one cycle of adjuvant BE500P in high-risk stage 1 NSGCTT. Adoption of one cycle of BE500P as standard would reduce overall exposure to chemotherapy in this young population. PATIENT SUMMARY Removing the testicle fails to cure many patients with high-risk primary testicular cancer since undetectable cancers are often present elsewhere. A standard additional treatment in Europe is two cycles of chemotherapy to eradicate these. This trial shows one cycle has few adverse effects and comparable outcomes to those seen with two cycles.
Collapse
Affiliation(s)
- Michael Cullen
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Robert Huddart
- The Institute Of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK.
| | | | - Deborah Gardiner
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Lauren Maynard
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Paul Hutton
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Danish Mazhar
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Matthew Wheater
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jeff White
- Beatson West of Scotland Cancer Centre, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Aicha Goubar
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Stephanie Witts
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Rebecca Lewis
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| |
Collapse
|
44
|
Wallach JD, Wang K, Zhang AD, Cheng D, Grossetta Nardini HK, Lin H, Bracken MB, Desai M, Krumholz HM, Ross JS. Updating insights into rosiglitazone and cardiovascular risk through shared data: individual patient and summary level meta-analyses. BMJ 2020; 368:l7078. [PMID: 32024657 PMCID: PMC7190063 DOI: 10.1136/bmj.l7078] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To conduct a systematic review and meta-analysis of the effects of rosiglitazone treatment on cardiovascular risk and mortality using multiple data sources and varying analytical approaches with three aims in mind: to clarify uncertainties about the cardiovascular risk of rosiglitazone; to determine whether different analytical approaches are likely to alter the conclusions of adverse event meta-analyses; and to inform efforts to promote clinical trial transparency and data sharing. DESIGN Systematic review and meta-analysis of randomized controlled trials. DATA SOURCES GlaxoSmithKline's (GSK's) ClinicalStudyDataRequest.com for individual patient level data (IPD) and GSK's Study Register platforms, MEDLINE, PubMed, Embase, Web of Science, Cochrane Central Registry of Controlled Trials, Scopus, and ClinicalTrials.gov from inception to January 2019 for summary level data. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Randomized, controlled, phase II-IV clinical trials that compared rosiglitazone with any control for at least 24 weeks in adults. DATA EXTRACTION AND SYNTHESIS For analyses of trials for which IPD were available, a composite outcome of acute myocardial infarction, heart failure, cardiovascular related death, and non-cardiovascular related death was examined. These four events were examined independently as secondary analyses. For analyses including trials for which IPD were not available, myocardial infarction and cardiovascular related death were examined, which were determined from summary level data. Multiple meta-analyses were conducted that accounted for trials with zero events in one or both arms with two different continuity corrections (0.5 constant and treatment arm) to calculate odds ratios and risk ratios with 95% confidence intervals. RESULTS 33 eligible trials were identified from ClinicalStudyDataRequest.com for which IPD were available (21 156 patients). Additionally, 103 trials for which IPD were not available were included in the meta-analyses for myocardial infarction (23 683 patients), and 103 trials for which IPD were not available contributed to the meta-analyses for cardiovascular related death (22 772 patients). Among 29 trials for which IPD were available and that were included in previous meta-analyses using GSK's summary level data, more myocardial infarction events were identified by using IPD instead of summary level data for 26 trials, and fewer cardiovascular related deaths for five trials. When analyses were limited to trials for which IPD were available, and a constant continuity correction of 0.5 and a random effects model were used to account for trials with zero events in only one arm, patients treated with rosiglitazone had a 33% increased risk of a composite event compared with controls (odds ratio 1.33, 95% confidence interval 1.09 to 1.61; rosiglitazone population: 274 events among 11 837 patients; control population: 219 events among 9319 patients). The odds ratios for myocardial infarction, heart failure, cardiovascular related death, and non-cardiovascular related death were 1.17 (0.92 to 1.51), 1.54 (1.14 to 2.09), 1.15 (0.55 to 2.41), and 1.18 (0.60 to 2.30), respectively. For analyses including trials for which IPD were not available, odds ratios for myocardial infarction and cardiovascular related death were attenuated (1.09, 0.88 to 1.35, and 1.12, 0.72 to 1.74, respectively). Results were broadly consistent when analyses were repeated using trials with zero events across both arms and either of the two continuity corrections was used. CONCLUSIONS The results suggest that rosiglitazone is associated with an increased cardiovascular risk, especially for heart failure events. Although increased risk of myocardial infarction was observed across analyses, the strength of the evidence varied and effect estimates were attenuated when summary level data were used in addition to IPD. Because more myocardial infarctions and fewer cardiovascular related deaths were reported in the IPD than in the summary level data, sharing IPD might be necessary when performing meta-analyses focused on safety. SYSTEMATIC REVIEW REGISTRATION OSF Home https://osf.io/4yvp2/.
Collapse
Affiliation(s)
- Joshua D Wallach
- Department of Environmental Health Sciences, Yale School of Public Health, 60 College Street, New Haven, CT 06510, USA
- Collaboration for Research Integrity and Transparency, Yale School of Medicine, New Haven, CT, USA
| | - Kun Wang
- Center for Outcomes Research and Evaluation, Yale-New Haven Health System, New Haven, CT, USA
| | - Audrey D Zhang
- Center for Outcomes Research and Evaluation, Yale-New Haven Health System, New Haven, CT, USA
- New York University School of Medicine, New York, NY, USA
| | - Deanna Cheng
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | | | - Haiqun Lin
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Michael B Bracken
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Mayur Desai
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Harlan M Krumholz
- Center for Outcomes Research and Evaluation, Yale-New Haven Health System, New Haven, CT, USA
- Section of Cardiovascular Medicine and the National Clinician Scholars Program, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| | - Joseph S Ross
- Collaboration for Research Integrity and Transparency, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale-New Haven Health System, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
- Section of General Medicine and the National Clinician Scholars Program, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
45
|
Glaspole IN. Acute exacerbations in IPF: A clarion call for collaborative research. Respirology 2020; 25:572-573. [PMID: 31997468 DOI: 10.1111/resp.13770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/13/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Ian N Glaspole
- Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Respiratory Medicine, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Precision Medicine in Soft Tissue Sarcoma Treatment. Cancers (Basel) 2020; 12:cancers12010221. [PMID: 31963219 PMCID: PMC7017346 DOI: 10.3390/cancers12010221] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Soft tissue sarcoma (STS) is a rare component of malignant diseases. STS includes various histological subtypes, and there are some important differences among the different histological subtypes regarding the mutation profile and sensitivity to antitumor agents. Many clinical trials of STS incorporating many different histological subtypes in various populations have been conducted; it is difficult to compare the findings and make conclusions about clinical efficacy. Targeted therapies focusing on specific histological subtypes and precision therapy focusing on the specific genetic mutation(s) of each STS patient are being investigated. Since STS patients are a small population, new clinical trial designs are required to evaluate and establish new targeted therapies for each histological subtype that has a limited number of patients, and preclinical investigations are needed to detect targetable mutations. Now that cancer genome profiling is used in clinical practice, it is urgently necessary to connect the genome profiling data obtained in clinical settings to the optimal clinical treatment strategies. Herein we review the development and challenges of precision therapy in the management of STS patients.
Collapse
|
47
|
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: 0.4] [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
|
48
|
Precision Oncology-The Quest for Evidence. J Pers Med 2019; 9:jpm9030043. [PMID: 31492009 PMCID: PMC6789813 DOI: 10.3390/jpm9030043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/07/2019] [Accepted: 08/22/2019] [Indexed: 12/17/2022] Open
Abstract
The molecular characterization of patient tumors provides a rational and highly promising approach for guiding oncologists in treatment decision-making. Notwithstanding, genomic medicine still remains in its infancy, with innovators and early adopters continuing to carry a significant portion of the clinical and financial risk. Numerous innovative precision oncology trials have emerged globally to address the associated need for evidence of clinical utility. These studies seek to capitalize on the power of predictive biomarkers and/or treatment decision support analytics, to expeditiously and cost-effectively demonstrate the positive impact of these technologies on drug resistance/response, patient survival, and/or quality of life. Here, we discuss the molecular foundations of these approaches and highlight the diversity of innovative trial strategies that are capitalizing on this emergent knowledge. We conclude that, as increasing volumes of clinico-molecular outcomes data become available, in future, we will begin to transition away from expert systems for treatment decision support (TDS), towards the power of AI-assisted TDS-an evolution that may truly revolutionize the nature and success of cancer patient care.
Collapse
|
49
|
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: 0.4] [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
|
50
|
Opportunities and challenges of personalized therapy of patients with HR ALL. Hemasphere 2019; 3:HEMASPHERE-2019-0052. [PMID: 35309807 PMCID: PMC8925720 DOI: 10.1097/hs9.0000000000000229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/21/2019] [Indexed: 11/29/2022] Open
|