1
|
Rahadi A, Mahardya RTK, Listiani P, Herlinawaty E, Nugraha RR, Budiman DR, Suharlim C. Calibration of transition probabilities to model survival of adjuvant trastuzumab for early breast cancer in Indonesia. Int J Technol Assess Health Care 2025; 41:e18. [PMID: 40135279 PMCID: PMC11955306 DOI: 10.1017/s0266462325000157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVES Cost-effectiveness models fully informed by real-world epidemiological parameters yield the best results, but they are costly to obtain. Model calibration using real-world data/evidence (RWD/E) on routine health indicators can provide an alternative to improve the validity and acceptability of the results. We calibrated the transition probabilities of the reference chemotherapy treatment using RWE on patient overall survival (OS) to model the survival benefit of adjuvant trastuzumab in Indonesia. METHODS A Markov model comprising four health states was initially parameterized using the reference-treatment transition probabilities, obtained from published international evidence. We then calibrated these probabilities, targeting a 2-year OS of 86.11 percent from the RWE sourced from hospital registries. We compared projected OS duration and life-years gained (LYG) before and after calibration for the Nelder-Mead, Bound Optimization BY Quadratic Approximation, and generalized reduced gradient (GRG) nonlinear optimization methods. RESULTS The pre-calibrated transition probabilities overestimated the 2-year OS (92.25 percent). GRG nonlinear performed best and had the smallest difference with the RWD/E OS. After calibration, the projected OS duration was significantly lower than their pre-calibrated estimates across all optimization methods for both standard chemotherapy (~7.50 vs. 11.00 years) and adjuvant trastuzumab (~9.50 vs. 12.94 years). LYG measures were, however, similar (~2 years) for the pre-calibrated and calibrated models. CONCLUSIONS RWD/E calibration resulted in realistically lower survival estimates. Despite the little difference in LYG, calibration is useful to adapt external evidence commonly used to derive transition probabilities to the policy context, thereby enhancing the validity and acceptability of the modeling results.
Collapse
Affiliation(s)
- Arie Rahadi
- Management Sciences for Health, Arlington, VA, USA
| | | | - Putri Listiani
- Center for Health Financing Policy and Insurance Management, Gadjah Mada University, Sleman, Yogyakarta, Indonesia
| | - Eva Herlinawaty
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | | - Dani Ramdhani Budiman
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | |
Collapse
|
2
|
Rippin G, Sanz H, Hoogendoorn WE, Ballarini NM, Largent JA, Demas E, Postmus D, Framke T, Dávila LMA, Quinten C, Pignatti F. Examining the Effect of Missing Data and Unmeasured Confounding on External Comparator Studies: Case Studies and Simulations. Drug Saf 2024; 47:1245-1263. [PMID: 39102176 PMCID: PMC11554740 DOI: 10.1007/s40264-024-01467-9] [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] [Accepted: 07/04/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND AND OBJECTIVE Missing data and unmeasured confounding are key challenges for external comparator studies. This work evaluates bias and other performance characteristics depending on missingness and unmeasured confounding by means of two case studies and simulations. METHODS Two case studies were constructed by taking the treatment arms from two randomised controlled trials and an external real-world data source that exhibited substantial missingness. The indications of the randomised controlled trials were multiple myeloma and metastatic hormone-sensitive prostate cancer. Overall survival was taken as the main endpoint. The effects of missing data and unmeasured confounding were assessed for the case studies by reporting estimated external comparator versus randomised controlled trial treatment effects. Based on the two case studies, simulations were performed broadening the settings by varying the underlying hazard ratio, the sample size, the sample size ratio between the experimental arm and the external comparator, the number of missing covariates and the percentage of missingness. Thereby, bias and other performance metrics could be quantified dependent on these factors. RESULTS For the multiple myeloma external comparator study, results were in line with the randomised controlled trial, despite missingness and potential unmeasured confounding, while for the metastatic hormone-sensitive prostate cancer case study missing data led to a low sample size, leading overall to inconclusive results. Furthermore, for the metastatic hormone-sensitive prostate cancer study, missing data in important eligibility criteria led to further limitations. Simulations were successfully applied to gain a quantitative understanding of the effects of missing data and unmeasured confounding. CONCLUSIONS This exploratory study confirmed external comparator strengths and limitations by quantifying the impact of missing data and unmeasured confounding using case studies and simulations. In particular, missing data in key eligibility criteria were seen to limit the ability to derive the external comparator target analysis population accurately, while simulations demonstrated the magnitude of bias to expect for various settings.
Collapse
Affiliation(s)
- Gerd Rippin
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany.
| | - Héctor Sanz
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | | | | | - Joan A Largent
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | - Eleni Demas
- IQVIA, Unterschweinstiege 2-14, 60549, Frankfurt, Germany
| | - Douwe Postmus
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| | - Theodor Framke
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
- Institute for Biostatistics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Chantal Quinten
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| | - Francesco Pignatti
- European Medicines Agency, Domenico Scarlattilaan 6, Amsterdam, 1083 HS, The Netherlands
| |
Collapse
|
3
|
Chatain A, Fenioux C, Lamé G, Bouras A, Babai S, Ahmed ES, Monard A, Manuceau G, Tournigand C, Albin N, Kempf E. Clinical relevance of reports on early access programs for checkpoint inhibitors in cancer patients: a French retrospective nationwide cohort study. ESMO Open 2024; 9:103711. [PMID: 39395267 PMCID: PMC11693425 DOI: 10.1016/j.esmoop.2024.103711] [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: 06/07/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND To accelerate access to new drugs, France operated an early access program known as Temporary Authorizations for Use (ATUs) until 2021. We analyzed clinical reports submitted under ATUs for immune checkpoint inhibitors (ICIs) and assessed their clinical relevance regarding the approval of ICIs in oncology. METHODS We included all ICIs granted an ATU by the French drug safety agency, Agence nationale de sécurité du médicament et des produits de santé (ANSM; French National Agency for the Safety of Medicines and Health Products), for patients with cancer between 1 January 2010 and 31 December 2020. We collected patients' clinical and pharmacovigilance data from ATU reports submitted by pharmaceutical companies and compared these data with those from corresponding pivotal clinical trials (CTs). RESULTS The ATUs provided early access to 5807 patients with seven ICIs across 11 cancer indications, 1 of which had no corresponding ATU report. Of the 10 available ATU reports, only 1 included all required data. Clinical follow-up forms were available for 40.5% of patients. Differences in data reporting prevented us from comparing serious adverse events between the CTs and ATU reports. Clinicians and pharmaceutical companies often disagreed on whether ICIs caused 163 permanent treatment discontinuations, with Cohen's bias- and prevalence-adjusted κ = 0.52, 95% CI 0.33-0.68. Although agreement was almost perfect for 93 nonprogressive tumor deaths (κ = 0.88, 95% CI 0.66-0.97), 29% of ATU patient deaths remained unexplained and were reported as unrelated to treatment by the pharmaceutical companies. CONCLUSION French ATUs facilitated early access to new ICIs for many patients with cancer. However, data attrition hindered effective real-world monitoring.
Collapse
Affiliation(s)
- A Chatain
- Department of Medical Oncology, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France; Sorbonne University, Paris, France
| | - C Fenioux
- Department of Medical Oncology, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France
| | - G Lamé
- Paris Saclay University, Centrale Supélec, Industrial Engineering Laboratory, Gif-sur-Yvette, France
| | - A Bouras
- Department of Medical Oncology, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France
| | - S Babai
- Department of Pharmacovigilance, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France
| | - E S Ahmed
- Department of Medical Oncology, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France
| | - A Monard
- Department of Oncology, GHM Grenoble Non-Profit Hospital, Institut Daniel Hollard, Grenoble, France
| | - G Manuceau
- Department of Medical Oncology, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Créteil, France
| | - C Tournigand
- Department of Medical Oncology, Paris-East Créteil University, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Creteil, France
| | - N Albin
- Department of Oncology, GHM Grenoble Non-Profit Hospital, Institut Daniel Hollard, Grenoble, France
| | - E Kempf
- Department of Medical Oncology, Paris-East Créteil University, Henri Mondor and Albert Chenevier Teaching Hospital, AP-HP, Creteil, France.
| |
Collapse
|
4
|
Verkerk K, Voest EE. Generating and using real-world data: A worthwhile uphill battle. Cell 2024; 187:1636-1650. [PMID: 38552611 DOI: 10.1016/j.cell.2024.02.012] [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: 11/03/2023] [Revised: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 04/02/2024]
Abstract
The precision oncology paradigm challenges the feasibility and data generalizability of traditional clinical trials. Consequently, an unmet need exists for practical approaches to test many subgroups, evaluate real-world drug value, and gather comprehensive, accessible datasets to validate novel biomarkers. Real-world data (RWD) are increasingly recognized to have the potential to fill this gap in research methodology. Established applications of RWD include informing disease epidemiology, pharmacovigilance, and healthcare quality assessment. Currently, concerns regarding RWD quality and comprehensiveness, privacy, and biases hamper their broader application. Nonetheless, RWD may play a pivotal role in supplementing clinical trials, enabling conditional reimbursement and accelerated drug access, and innovating trial conduct. Moreover, purpose-built RWD repositories may support the extension or refinement of drug indications and facilitate the discovery and validation of new biomarkers. This perspective explores the potential of leveraging RWD to advance oncology, highlights its benefits and challenges, and suggests a path forward in this evolving field.
Collapse
Affiliation(s)
- K Verkerk
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - E E Voest
- Department of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, the Netherlands.
| |
Collapse
|
5
|
Luu VP, Fiorini M, Combes S, Quemeneur E, Bonneville M, Bousquet PJ. Challenges of artificial intelligence in precision oncology: public-private partnerships including national health agencies as an asset to make it happen. Ann Oncol 2024; 35:154-158. [PMID: 37769849 DOI: 10.1016/j.annonc.2023.09.3106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/13/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023] Open
Affiliation(s)
- V P Luu
- Epidemiology and innovation Unit, Artificial Intelligence and Cancers Association, Paris, France.
| | - M Fiorini
- Artificial Intelligence and Cancers Association, Paris, France
| | | | - E Quemeneur
- France Biotech, Paris, France; Transgene S.A., Illkirch-Graffenstaden, France
| | - M Bonneville
- Alliance pour la Recherche et l'Innovation des Industries de Santé, Paris, France; Institut Mérieux, Lyon, France
| | - P J Bousquet
- Health Survey, Data-Science, Assessment Division, Institut National du Cancer, Boulogne Billancourt, France; Aix Marseille University, INSERM, IRD, Economics and Social Sciences Applied to Health & Analysis of Medical Information (SESSTIM), Marseille, France
| |
Collapse
|
6
|
Macabeo B, Rotrou T, Millier A, François C, Laramée P. The Acceptance of Indirect Treatment Comparison Methods in Oncology by Health Technology Assessment Agencies in England, France, Germany, Italy, and Spain. PHARMACOECONOMICS - OPEN 2024; 8:5-18. [PMID: 38097828 PMCID: PMC10781913 DOI: 10.1007/s41669-023-00455-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/26/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Randomized controlled trials (RCTs) are the gold standard when comparing treatment effectiveness, and Health Technology Assessment (HTA) agencies state a clear preference for such direct comparisons. When these are not available, an indirect treatment comparison (ITC) is an alternative option. The objective of this study was to assess the acceptance of ITC methods by HTA agencies across England, France, Germany, Italy, and Spain, using oncology cases for a homogeneous sample of HTA evaluations. METHODS The study was conducted on the PrismAccess database in May 2021 to retrieve HTA evaluation reports for oncology treatments for solid tumors, in which an ITC was presented. The analysis was restricted to HTA evaluation reports published between April 2018 and April 2021 in England, France, Germany, Italy, and Spain. Identified HTA evaluation reports were screened and reviewed by two independent reviewers. For each ITC presented, the methodology and its acceptance by the HTA agency were analyzed. RESULTS Five hundred and forty-three HTA evaluation reports were identified, of which 120 (22%) presented an ITC. This proportion was the highest in England (51%) and lowest in France (6%). The overall acceptance rate of ITC methods was 30%, with the highest in England (47%) and lowest in France (0%). Network meta-analysis (NMA; 23%) was the most commonly used ITC technique, with a 39% acceptance rate overall, followed by Bucher ITC (19%; 43% acceptance rate) and matching-adjusted indirect comparison (13%; 33% acceptance rate). The most common criticisms of the ITC methods from HTA agencies related to data limitations (heterogeneity and lack of data; 48% and 43%, respectively) and the statistical methods used (41%). CONCLUSIONS The generally low acceptance rate of ITC methods by HTA agencies in oncology suggests that, whilst in the absence of a direct comparison ITCs may provide relevant evidence, this evidence is not widely considered sufficient for the purpose of HTA evaluations. The perception of ITC methods for the purpose of HTA evaluations varies substantially between countries. There is a need for further clarity on the properties of ITC techniques and the assessment of their results as ITC methods continue to evolve quickly and further techniques may become available in the future.
Collapse
Affiliation(s)
- Bérengère Macabeo
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France.
- Pierre Fabre Laboratories, Paris, France.
| | | | | | - Clément François
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
| | - Philippe Laramée
- Faculté des sciences Médicales et Paramédicales, Aix-Marseille University, 27 Boulevard Jean Moulin, 13005, Marseille, France
- Pierre Fabre Laboratories, Paris, France
| |
Collapse
|
7
|
Goldschmidt JH, Chou L, Chan PK, Chen L, Robert N, Kinsey J, Pitts K, Nestor M, Rock EP, Lazarus HM. Real-world outcomes of 18,186 metastatic solid tumor outpatients: Baseline blood cell counts correlate with survival after immune checkpoint inhibitor therapy. Cancer Med 2023; 12:20783-20797. [PMID: 37962239 PMCID: PMC10709745 DOI: 10.1002/cam4.6645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Patient survival in advanced/metastatic melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma (RCC) has improved with immune checkpoint inhibitors (ICI). Biomarkers' role in prognosis and treatment has been limited by conflicting trial results. METHODS This retrospective, observational study analyzed baseline demographic, clinical, laboratory, and treatment data versus outcomes of The US Oncology Network adult outpatients. Patients with advanced/metastatic melanoma, NSCLC, or RCC treated between January 1, 2015 and November 30, 2020 were given ICI monotherapy or combination therapy with ipilimumab, pembrolizumab, nivolumab, or atezolizumab. Treatment outcomes (overall survival [OS], time to treatment discontinuation, time to next treatment) were followed longitudinally until May 31, 2021, last patient record, or date of death. Baseline blood cell counts, including absolute monocyte count (AMC), absolute lymphocyte count (ALC), monocyte-to-lymphocyte ratio (MLR), absolute neutrophil count (ANC), and eosinophil count, were subdivided into quintiles for univariate and multivariable Cox regression analyses. RESULTS Data from 18,186 patients with advanced/metastatic melanoma (n = 3314), NSCLC (n = 12,416), and RCC (n = 2456) were analyzed. Better OS correlated with increased baseline serum albumin concentration, increased eosinophil and lymphocyte counts, and Western United States physician practice location. Decreased OS correlated with increased AMC, MLR, ANC, age, and worse Eastern Cooperative Oncology Group performance status. CONCLUSIONS To our knowledge, this study is the largest to date to associate baseline survival indicators and outcomes in outpatients with advanced/metastatic melanoma, NSCLC, or RCC and receiving ICIs. Results may inform disease-specific prognostic models and help providers identify patients most likely to benefit from ICI therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Joyce Kinsey
- Partner Therapeutics, IncLexingtonMassachusettsUSA
| | | | - Matt Nestor
- Partner Therapeutics, IncLexingtonMassachusettsUSA
| | | | - Hillard M. Lazarus
- Department of Medicine, Division of Hematology and OncologyCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
8
|
George G, Russell B, Rigg A, Coolen ACC, Van Hemelrijck M. Real World Data Studies of Antineoplastic Drugs: How Can They Be Improved to Steer Everyday Use in the Clinic? Pragmat Obs Res 2023; 14:95-100. [PMID: 37701044 PMCID: PMC10493103 DOI: 10.2147/por.s395959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
There is a growing interest in real world evidence when developing antineoplastic drugs owing to the shorter length of time and low costs compared to randomised controlled trials. External validity of studies in the regulatory phase can be enhanced by complementing randomised controlled trials with real world evidence. Furthermore, the use of real world evidence ensures the inclusion of patients often excluded from randomised controlled trials such as the elderly, certain ethnicities or those from certain geographical areas. This review explores approaches in which real world data may be integrated with randomised controlled trials. One approach is by using big data, especially when investigating drugs in the antineoplastic setting. This can even inform artificial intelligence thus ensuring faster and more precise diagnosis and treatment decisions. Pragmatic trials also offer an approach to examine the effectiveness of novel antineoplastic drugs without evading the benefits of randomised controlled trials. A well-designed pragmatic trial would yield results with high external validity by employing a simple study design with a large sample size and diverse settings. Although randomised controlled trials can determine efficacy of antineoplastic drugs, effectiveness in the real world may differ. The need for pragmatic trials to help guide healthcare decision-making led to the development of trials within cohorts (TWICs). TWICs make use of cohorts to conduct multiple randomised controlled trials while maintaining characteristics of real world data in routine clinical practice. Although real world data is often affected by incomplete data and biases such as selection and unmeasured biases, the use of big data and pragmatic approaches can improve the use of real world data in the development of antineoplastic drugs that can in turn steer decision-making in clinical practice.
Collapse
Affiliation(s)
- Gincy George
- Translational Oncology and Urology Research, King’s College London, London, UK
| | - Beth Russell
- Translational Oncology and Urology Research, King’s College London, London, UK
| | - Anne Rigg
- Guy’s Cancer Centre, Guy’s and St Thomas NHS Foundation Trust, London, UK
| | | | | |
Collapse
|
9
|
Nelli F, Virtuoso A, Giannarelli D, Fabbri A, Giron Berrios JR, Marrucci E, Fiore C, Ruggeri EM. Effects of Acetaminophen Exposure on Outcomes of Patients Receiving Immune Checkpoint Inhibitors for Advanced Non-Small-Cell Lung Cancer: A Propensity Score-Matched Analysis. Curr Oncol 2023; 30:8117-8133. [PMID: 37754504 PMCID: PMC10527930 DOI: 10.3390/curroncol30090589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
(1) Background: Several studies have investigated potential interactions between immune checkpoint inhibitors (ICIs) and commonly prescribed medications. Although acetaminophen (APAP) has not been considered susceptible to interaction with ICIs, recent research has shown that detectable plasma levels of this drug can hinder the efficacy of PD-1/PD-L1 blockade therapies. A reliable assessment of the potential interaction between APAP and ICIs in advanced non-small cell lung cancer (NSCLC) patients would be worthwhile since it is often prescribed in this condition. We sought to evaluate the impact of the concomitant use of APAP in patients with advanced NSCLC on PD-1/PD-L1 blockade using real-world evidence. (2) Methods: This study included consecutive patients with histologically proven stage IV NSCLC who underwent first-line therapy with pembrolizumab as a single agent or in combination with platinum-based chemotherapy, or second-line therapy with pembrolizumab, nivolumab, or atezolizumab. The intensity of APAP exposure was classified as low (therapeutic intake lasting less than 24 h or a cumulative intake lower than 60 doses of 1000 mg) or high (therapeutic intake lasting more than 24 h or a total intake exceeding 60 doses of 1000 mg). The favorable outcome of anti-PD-1/PD-L1 therapies was defined by durable clinical benefit (DCB). Progression-free survival (PFS) and overall survival (OS) were relevant to our efficacy analysis. Propensity score matching (PSM) methods were applied to adjust for differences between the APAP exposure subgroups. (3) Results: Over the course of April 2018 to October 2022, 80 patients were treated with first-line pembrolizumab either as single-agent therapy or in combination with platinum-based chemotherapy. During the period from June 2015 to November 2022, 145 patients were given anti-PD-1/PD-L1 blockade therapy as second-line treatment. Subsequent efficacy analyses relied on adjusted PSM populations in both treatment settings. Multivariate testing revealed that only the level of APAP and corticosteroid intake had an independent effect on DCB in both treatment lines. Multivariate Cox regression analysis confirmed high exposure to APAP and immunosuppressive corticosteroid therapy as independent predictors of shorter PFS and OS in both treatment settings. (4) Conclusions: Our findings would strengthen the available evidence that concomitant intake of APAP blunts the efficacy of ICIs in patients with advanced NSCLC. The detrimental effects appear to depend on the cumulative dose and duration of exposure to APAP. The inherent shortcomings of the current research warrant confirmation in larger independent series.
Collapse
Affiliation(s)
- Fabrizio Nelli
- Thoracic Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Antonella Virtuoso
- Thoracic Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Diana Giannarelli
- Biostatistics Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Agnese Fabbri
- Medical Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Julio Rodrigo Giron Berrios
- Medical Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Eleonora Marrucci
- Medical Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Cristina Fiore
- Medical Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| | - Enzo Maria Ruggeri
- Medical Oncology Unit, Department of Oncology and Hematology, Central Hospital of Belcolle, 01100 Viterbo, Italy
| |
Collapse
|
10
|
Fabbri A, Nelli F, Botticelli A, Giannarelli D, Marrucci E, Fiore C, Virtuoso A, Scagnoli S, Pisegna S, Alesini D, Sini V, Orlandi A, Fabi A, Piacentini F, Moscetti L, D’Auria G, Gamucci T, Mazzotta M, Pizzuti L, Vici P, Cretella E, Scavina P, La Cesa A, Persano M, Atzori F, Ruggeri EM. Pathologic response and survival after neoadjuvant chemotherapy with or without pertuzumab in patients with HER2-positive breast cancer: the Neopearl nationwide collaborative study. Front Oncol 2023; 13:1177681. [PMID: 37441419 PMCID: PMC10335743 DOI: 10.3389/fonc.2023.1177681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
Purpose Clinical trials have shown a significant increase in pathologic complete response (pCR) with the addition of pertuzumab to neoadjuvant chemotherapy for patients with early-stage HER-2 positive breast cancer. To date, limited studies have examined comparative outcomes of neoadjuvant pertuzumab in real-world setting. The Neopearl study aimed to assess comparative real-life efficacy and safety of neoadjuvant pertuzumab for these patients. Methods We conducted a nationwide retrospective analysis involving 17 oncology facilities with a certified multidisciplinary breast cancer treatment committee. We identified patients with HER-2 positive stage II-III breast cancer treated with neoadjuvant chemotherapy based on trastuzumab and taxanes with or without pertuzumab. All patients underwent breast surgery and received a comprehensive cardiologic evaluation at baseline and after neoadjuvant treatment. Patients who received the combination of pertuzumab, trastuzumab, and chemotherapy constituted case cohort (PTCT), whereas those treated with trastuzumab and chemotherapy accounted for control cohort (TCT). The pCR rate and 5-year event free survival (EFS) were the primary outcomes. Secondary end-points were rates of conversion from planned modified radical mastectomy (MRM) to breast conservation surgery (BCS) and cardiotoxicities. Results From March 2014 to April 2021, we included 271 patients, 134 (49%) and 137 (51%) in TCT and PTCT cohort, respectively. Positive axillary lymph nodes and stage III were more frequent in PTCT cohort. The pCR rate was significantly increased in patients who received pertuzumab (49% vs 62%; OR 1.74, 95%CI 1.04-2.89) and with HER-2 enriched subtypes (16% vs 85%; OR 2.94, 95%CI 1.60-5.41). After a median follow-up of 5 years, the 5-year EFS was significantly prolonged only in patients treated with pertuzumab (81% vs 93%; HR 2.22, 95%CI 1.03-4.79). The same analysis performed on propensity score matched population showed concordant results. On univariate analysis, only patients with positive lymph nodes were found to benefit from pertuzumab for both pCR and 5-year EFS. The rates of conversion from MRM to BCS and cardiologic toxicities did not differ between the cohorts. Conclusion Our findings support previous data on improved outcomes with the addition of pertuzumab to trastuzumab-based neoadjuvant chemotherapy. This benefit seems to be more significant in patients with clinically positive lymph nodes.
Collapse
Affiliation(s)
- Agnese Fabbri
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| | - Fabrizio Nelli
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| | - Andrea Botticelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Diana Giannarelli
- Biostatistics Unit, Scientific Directorate, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Eleonora Marrucci
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| | - Cristina Fiore
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| | - Antonella Virtuoso
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| | - Simone Scagnoli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Simona Pisegna
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Daniele Alesini
- UOSD Centro Oncologico S. Spirito e Nuovo Regina Margherita (SS-NRM), Ospedale Santo Spirito in Sassia, Rome, Italy
| | - Valentina Sini
- UOSD Centro Oncologico S. Spirito e Nuovo Regina Margherita (SS-NRM), Ospedale Santo Spirito in Sassia, Rome, Italy
| | - Armando Orlandi
- Department of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandra Fabi
- Precision Medicine Breast Unit, Scientific Directorate, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federico Piacentini
- Department of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Luca Moscetti
- Department of Medical Oncology, University Hospital of Modena, Modena, Italy
| | - Giuliana D’Auria
- Department of Medical Oncology, Medical Oncology Unit, Sandro Pertini Hospital, Rome, Italy
| | - Teresa Gamucci
- Department of Medical Oncology, Medical Oncology Unit, Sandro Pertini Hospital, Rome, Italy
| | - Marco Mazzotta
- Department of Medical Oncology, Medical Oncology Unit, Sandro Pertini Hospital, Rome, Italy
| | - Laura Pizzuti
- UOSD Sperimentazioni di fase IV, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizia Vici
- UOSD Sperimentazioni di fase IV, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Elisabetta Cretella
- Department of Medical Oncology, Medical Oncology Unit, Ospedale Di Bolzano, Azienda Sanitaria Alto Adige, Bolzano, Italy
| | - Paola Scavina
- Department of Medical Oncology, Medical Oncology Unit, San Giovanni Addolorata Hospital, Rome, Italy
| | - Annalisa La Cesa
- Department of Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Mara Persano
- Department of Medical Oncology, University Hospital and University of Cagliari, Cagliari, Italy
| | - Francesco Atzori
- Department of Medical Oncology, University Hospital and University of Cagliari, Cagliari, Italy
| | - Enzo Maria Ruggeri
- Department of Oncology and Hematology, Medical Oncology and Breast Unit, Central Hospital of Belcolle, Viterbo, Italy
| |
Collapse
|
11
|
Saesen R, Depreytere K, Krupianskaya K, Langeweg J, Verheecke J, Lacombe D, Huys I. Analysis of the characteristics and the degree of pragmatism exhibited by pragmatic-labelled trials of antineoplastic treatments. BMC Med Res Methodol 2023; 23:148. [PMID: 37355603 PMCID: PMC10290324 DOI: 10.1186/s12874-023-01975-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/10/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Pragmatic clinical trials (PCTs) are designed to reflect how an investigational treatment would be applied in clinical practice. As such, unlike their explanatory counterparts, they measure therapeutic effectiveness and are capable of generating high-quality real-world evidence. However, the conduct of PCTs remains extremely rare. The scarcity of such studies has contributed to the emergence of the efficacy-effectiveness gap and has led to calls for launching more of them, including in the field of oncology. This analysis aimed to identify self-labelled pragmatic trials of antineoplastic interventions and to evaluate whether their use of this label was justified. METHODS We searched PubMed® and Embase® for publications corresponding with studies that investigated antitumor therapies and that were tagged as pragmatic in their titles, abstracts and/or index terms. Subsequently, we consulted all available source documents for the included trials and extracted relevant information from them. The data collected were then used to appraise the degree of pragmatism displayed by the PCTs with the help of the validated PRECIS-2 tool. RESULTS The literature search returned 803 unique records, of which 46 were retained upon conclusion of the screening process. This ultimately resulted in the identification of 42 distinct trials that carried the 'pragmatic' label. These studies examined eight different categories of neoplasms and were mostly randomized, open-label, multicentric, single-country trials sponsored by non-commercial parties. On a scale of one (very explanatory) to five (very pragmatic), the median PCT had a PRECIS-2 score per domain of 3.13 (interquartile range: 2.57-3.53). The most and least pragmatic studies in the sample had a score of 4.44 and 1.57, respectively. Only a minority of trials were described in sufficient detail to allow them to be graded across all domains of the PRECIS-2 instrument. Many of the studies examined also had features that arguably precluded them from being pragmatic altogether, such as being monocentric or placebo-controlled in nature. CONCLUSION PCTs of antineoplastic treatments are generally no more pragmatic than they are explanatory.
Collapse
Affiliation(s)
- Robbe Saesen
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
- European Organisation for Research and Treatment of Cancer (EORTC), Avenue E. Mounier 83, 1200, Brussels, Belgium.
| | - Kevin Depreytere
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Karyna Krupianskaya
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Joël Langeweg
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Julie Verheecke
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Avenue E. Mounier 83, 1200, Brussels, Belgium
| | - Isabelle Huys
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Keloth VK, Banda JM, Gurley M, Heider PM, Kennedy G, Liu H, Liu F, Miller T, Natarajan K, V Patterson O, Peng Y, Raja K, Reeves RM, Rouhizadeh M, Shi J, Wang X, Wang Y, Wei WQ, Williams AE, Zhang R, Belenkaya R, Reich C, Blacketer C, Ryan P, Hripcsak G, Elhadad N, Xu H. Representing and utilizing clinical textual data for real world studies: An OHDSI approach. J Biomed Inform 2023; 142:104343. [PMID: 36935011 PMCID: PMC10428170 DOI: 10.1016/j.jbi.2023.104343] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 01/21/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Clinical documentation in electronic health records contains crucial narratives and details about patients and their care. Natural language processing (NLP) can unlock the information conveyed in clinical notes and reports, and thus plays a critical role in real-world studies. The NLP Working Group at the Observational Health Data Sciences and Informatics (OHDSI) consortium was established to develop methods and tools to promote the use of textual data and NLP in real-world observational studies. In this paper, we describe a framework for representing and utilizing textual data in real-world evidence generation, including representations of information from clinical text in the Observational Medical Outcomes Partnership (OMOP) Common Data Model (CDM), the workflow and tools that were developed to extract, transform and load (ETL) data from clinical notes into tables in OMOP CDM, as well as current applications and specific use cases of the proposed OHDSI NLP solution at large consortia and individual institutions with English textual data. Challenges faced and lessons learned during the process are also discussed to provide valuable insights for researchers who are planning to implement NLP solutions in real-world studies.
Collapse
Affiliation(s)
- Vipina K Keloth
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Juan M Banda
- Department of Computer Science, Georgia State University, Atlanta, GA, USA
| | - Michael Gurley
- Lurie Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Paul M Heider
- Biomedical Informatics Center, Medical University of South Carolina, Charleston, SC, USA
| | - Georgina Kennedy
- Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Feifan Liu
- Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Timothy Miller
- Computational Health Informatics Program, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Karthik Natarajan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Olga V Patterson
- VA Informatics and Computing Infrastructure, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA; Division of Epidemiology, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA; Verily Life Sciences, Mountain View, CA, USA
| | - Yifan Peng
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Kalpana Raja
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Ruth M Reeves
- TN Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Masoud Rouhizadeh
- Department of Pharmaceutical Outcomes & Policy, University of Florida, Gainesville, FL, USA; Biomedical Informatics and Data Science, Johns Hopkins University, Baltimore, MD, USA
| | - Jianlin Shi
- VA Informatics and Computing Infrastructure, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA; Division of Epidemiology, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA; Department of Biomedical Informatics, University of Utah, Salt Lake City, USA
| | - Xiaoyan Wang
- Sema4 Mount Sinai Genomics Incorporation, Stamford, CT, USA
| | - Yanshan Wang
- Department of Health Information Management, Department of Biomedical Informatics, and Intelligent Systems Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Rui Zhang
- Institute for Health Informatics, and Department of Pharmaceutical Care & Health Systems, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Clair Blacketer
- Janssen Pharmaceutical Research and Development LLC, Titusville, NJ, USA; Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Patrick Ryan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA; Janssen Pharmaceutical Research and Development LLC, Titusville, NJ, USA
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Noémie Elhadad
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA.
| | - Hua Xu
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
13
|
Saesen R, Van Hemelrijck M, Bogaerts J, Booth CM, Cornelissen JJ, Dekker A, Eisenhauer EA, Freitas A, Gronchi A, Hernán MA, Hulstaert F, Ost P, Szturz P, Verkooijen HM, Weller M, Wilson R, Lacombe D, van der Graaf WT. Defining the role of real-world data in cancer clinical research: The position of the European Organisation for Research and Treatment of Cancer. Eur J Cancer 2023; 186:52-61. [PMID: 37030077 DOI: 10.1016/j.ejca.2023.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
The emergence of the precision medicine paradigm in oncology has led to increasing interest in the integration of real-world data (RWD) into cancer clinical research. As sources of real-world evidence (RWE), such data could potentially help address the uncertainties that surround the adoption of novel anticancer therapies into the clinic following their investigation in clinical trials. At present, RWE-generating studies which investigate antitumour interventions seem to primarily focus on collecting and analysing observational RWD, typically forgoing the use of randomisation despite its methodological benefits. This is appropriate in situations where randomised controlled trials (RCTs) are not feasible and non-randomised RWD analyses can offer valuable insights. Nevertheless, depending on how they are designed, RCTs have the potential to produce strong and actionable RWE themselves. The choice of which methodology to employ for RWD studies should be guided by the nature of the research question they are intended to answer. Here, we attempt to define some of the questions that do not necessarily require the conduct of RCTs. Moreover, we outline the strategy of the European Organisation for Research and Treatment of Cancer (EORTC) to contribute to the generation of robust and high-quality RWE by prioritising the execution of pragmatic trials and studies set up according to the trials-within-cohorts approach. If treatment allocation cannot be left up to random chance due to practical or ethical concerns, the EORTC will consider undertaking observational RWD research based on the target trial principle. New EORTC-sponsored RCTs may also feature concurrent prospective cohorts composed of off-trial patients.
Collapse
|
14
|
Eskola SM, Leufkens HGM, Bate A, Bruin MLD, Gardarsdottir H. The Role of Real-World Data and Evidence in Oncology Medicines Approved in EU in 2018-2019. J Cancer Policy 2023; 36:100424. [PMID: 37116794 DOI: 10.1016/j.jcpo.2023.100424] [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: 02/10/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Use of Real-World data (RWD) has gained the interest of different stakeholders in cancer care. The aim of this study was to identify and describe the use of RWD/RWE during the pre-authorisation phase of products authorised by the EMA in 2018 and 2019 (n=111), with the focus on oncology medicines (n=24). Information was extracted from the European Public Assessment Report (EPAR) summaries and recorded for 5 stages (11 categories) of the drug development lifecycle (discovery, early development, clinical development, registration/market launch, lifecycle management). Specific chapters of full EPAR were reviewed to substantiate the findings on RWD/RWE use in clinical trial design, efficacy, safety, and effectiveness evaluation. RWD/RWE is present in all stages of the oncology drug development; 100.0% in discovery, 37.5% early development, 58.3% in clinical development, 62.5% in registration decision and 100.0% in post-authorization lifecycle management. Examples showed that trial design supported by RWD/RWE included use of open label/single arm studies; efficacy was about using either comparison of results to historical controls, supplying survey data obtained outside the clinical trial or utilizing expert panel advice; safety about including literature findings in evidence; and effectiveness on comparison of trial results of the given product to historical data or existing standard of care. The findings of this study provide specific insights into how RWD/RWE is used in development of cancer therapeutics, how it contributes to regulatory decision making and can guide further policy developments in this field.
Collapse
Affiliation(s)
- Sini M Eskola
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands; European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | - Hubertus G M Leufkens
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Andrew Bate
- Global Safety, GSK, Brentford, Middlesex, UK; Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Marie Louise De Bruin
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Helga Gardarsdottir
- Utrecht Centre for Pharmaceutical Policy and Regulation, Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Reykjavík, Iceland.
| |
Collapse
|
15
|
Sweeney SM, Hamadeh HK, Abrams N, Adam SJ, Brenner S, Connors DE, Davis GJ, Fiore L, Gawel SH, Grossman RL, Hanlon SE, Hsu K, Kelloff GJ, Kirsch IR, Louv B, McGraw D, Meng F, Milgram D, Miller RS, Morgan E, Mukundan L, O'Brien T, Robbins P, Rubin EH, Rubinstein WS, Salmi L, Schaller T, Shi G, Sigman CC, Srivastava S. Challenges to Using Big Data in Cancer. Cancer Res 2023; 83:1175-1182. [PMID: 36625843 PMCID: PMC10102837 DOI: 10.1158/0008-5472.can-22-1274] [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: 04/15/2022] [Revised: 07/29/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Big data in healthcare can enable unprecedented understanding of diseases and their treatment, particularly in oncology. These data may include electronic health records, medical imaging, genomic sequencing, payor records, and data from pharmaceutical research, wearables, and medical devices. The ability to combine datasets and use data across many analyses is critical to the successful use of big data and is a concern for those who generate and use the data. Interoperability and data quality continue to be major challenges when working with different healthcare datasets. Mapping terminology across datasets, missing and incorrect data, and varying data structures make combining data an onerous and largely manual undertaking. Data privacy is another concern addressed by the Health Insurance Portability and Accountability Act, the Common Rule, and the General Data Protection Regulation. The use of big data is now included in the planning and activities of the FDA and the European Medicines Agency. The willingness of organizations to share data in a precompetitive fashion, agreements on data quality standards, and institution of universal and practical tenets on data privacy will be crucial to fully realizing the potential for big data in medicine.
Collapse
Affiliation(s)
- Shawn M. Sweeney
- American Association for Cancer Research, Philadelphia, Pennsylvania
| | | | - Natalie Abrams
- Division of Cancer Prevention, Early Detection Research Network, National Cancer Institute, Rockville, Maryland
| | - Stacey J. Adam
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Sara Brenner
- Office of In Vitro Diagnostics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Dana E. Connors
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Gerard J. Davis
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Louis Fiore
- Boston University School of Medicine, Boston and New England Department of Veterans Affairs, Bedford, Massachusetts
| | - Susan H. Gawel
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Robert L. Grossman
- Center for Translational Data Science, The University of Chicago, Chicago, Illinois
| | - Sean E. Hanlon
- Center for Strategic Scientific Initiatives, National Cancer Institute, Bethesda, Maryland
| | | | - Gary J. Kelloff
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Bill Louv
- Project Data Sphere, Morrisville, North Carolina
| | - Deven McGraw
- Ciitizen Platform at Invitae, San Francisco, California
| | - Frank Meng
- Boston University and Veterans Administration Boston Healthcare System, Boston, Massachusetts
| | | | - Robert S. Miller
- CancerLinQ, American Society of Clinical Oncology, Alexandria, Virginia
| | - Emily Morgan
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | - Wendy S. Rubinstein
- Office of In Vitro Diagnostics, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, Maryland
| | - Liz Salmi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | - George Shi
- Abbott Diagnostics Division, Abbott Laboratories, Lake Forest, Illinois
| | - Caroline C. Sigman
- Boston University and Veterans Administration Boston Healthcare System, Boston, Massachusetts
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
16
|
Wang X, Dormont F, Lorenzato C, Latouche A, Hernandez R, Rouzier R. Current perspectives for external control arms in oncology clinical trials: Analysis of EMA approvals 2016-2021. J Cancer Policy 2023; 35:100403. [PMID: 36646208 DOI: 10.1016/j.jcpo.2023.100403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/17/2022] [Accepted: 01/08/2023] [Indexed: 01/15/2023]
Abstract
Leveraging external control data, especially real-world data (RWD), has drawn particular attention in recent years for facilitating oncology clinical development and regulatory decision-making. Medical regulators have published guidance on accelerating the use of RWD and external controls. However, few systematic discussions have been conducted on external controls in cancer drug submissions and regulatory feedback. This study aimed to identify European oncology drug approvals using external control data to demonstrate clinical efficacy. We included 18 eligible submissions employing 24 external controls and then discussed the use of external control, data sources, analysis methods, and regulators' feedback. The external controls have been actively submitted to the European Medical Agency (EMA) recently. We found that 17 % of the EMA-approved cancer drugs in 2016-2021 used external controls, among which 37 % of the cases leveraged RWD. However, nearly one-third of the external controls were not considered supportive evidence by EMA due to limitations regarding heterogeneous patient populations, missing outcome assessment in RWD, and inappropriate statistical analysis. This study highlighted that proper use of external controls requires a careful assessment of clinical settings, data availability, and statistical methodology. For better use of external controls in oncology clinical trials, we recommend: prospective study designs to avoid selection bias, sufficient baseline data to ensure the comparability of study populations, consistent endpoint measurements to enable outcome comparison, robust statistical methodology for comparative analysis, and collaborative efforts of sponsors and regulators to establish regulatory frameworks.
Collapse
Affiliation(s)
- Xiaomeng Wang
- INSERM, U900, Institut Curie, PSL Research University, Saint-Cloud, France; Department of Research and Development, Sanofi, Chilly-Mazarin, France.
| | - Flavio Dormont
- Department of Research and Development, Sanofi, Chilly-Mazarin, France
| | | | - Aurélien Latouche
- INSERM, U900, Institut Curie, PSL Research University, Saint-Cloud, France; Conservatoire National des Arts et Métiers, Paris, France
| | - Ramon Hernandez
- Department of Research and Development, Sanofi, Chilly-Mazarin, France
| | - Roman Rouzier
- Department of Surgical Oncology, Centre François Baclesse, Caen, France
| |
Collapse
|
17
|
Christopoulos P, Schlenk R, Kazdal D, Blasi M, Lennerz J, Shah R, Budczies J, Malek N, Fröhling S, Rosenquist R, Schirmacher P, Bozorgmehr F, Kuon J, Reck M, Thomas M, Stenzinger A. Real-world data for precision cancer medicine-A European perspective. Genes Chromosomes Cancer 2023. [PMID: 36852573 DOI: 10.1002/gcc.23135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/22/2023] [Indexed: 03/01/2023] Open
Abstract
Leveraging real-world data (RWD) for drug access is necessary to overcome a key challenge of modern precision oncology: tackling numerous low-prevalence oncogenic mutations across cancers. Withholding a potentially active medication in patients with rare mutations for the sake of control chemotherapy or "best" supportive care is neither practicable nor ethically justifiable anymore, particularly as RWD could meanwhile be used instead, according to scientific principles outlined by the US Food and Drug Administration, European Medicines Agency and other stakeholders. However, practical implementation varies, with occasionally opposite recommendations based on the same evidence in different countries. In the face of growing need for precision drugs, more transparency of evaluation, a priori availability of guidance for the academia and industry, as well as a harmonized framework for health technology assessment across the European Union (EU) are imperative. These could in turn trigger infrastructural changes in national and pan-European registries, cancer management guidelines (e.g., frequency of routine radiologic restaging, inclusion of patient-reported outcomes), and the health data space, to ensure conformity with declared standards and facilitate extraction of RWD sets (including patient-level data) suitable for approval and pricing with minimal effort. For an EU-wide unification of precision cancer medicine, collective negotiation of drug supply contracts and funding solidarity would additionally be required to handle the financial burden. According to experience from pivotal European programs, off-label use could potentially also be harmonized across EU-states to accelerate availability of novel drugs, streamline collection of valuable RWD, and mitigate related costs through wider partnerships with pharmaceutical companies.
Collapse
Affiliation(s)
- Petros Christopoulos
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany
| | - Richard Schlenk
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- German Center for Lung Research (DZL), Gießen, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Miriam Blasi
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Jochen Lennerz
- Machachussets General Hospital, Harvard University, Boston, USA
| | - Rajiv Shah
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jan Budczies
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nisar Malek
- Centers for Personalized Medicine (ZPM), Germany.,Department of Gastroenterology, Tübingen University Hospital, Tübingen, Germany
| | - Stefan Fröhling
- Centers for Personalized Medicine (ZPM), Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Peter Schirmacher
- Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| | - Farastuk Bozorgmehr
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Jonas Kuon
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany.,Department of Oncology, Lungenklinik Löwenstein, Löwenstein, Germany
| | - Martin Reck
- German Center for Lung Research (DZL), Gießen, Germany.,Department of Thoracic Oncology, Lungenclinic Großhansdorf, Großhansdorf, Germany
| | - Michael Thomas
- Department of Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Center for Lung Research (DZL), Gießen, Germany
| | - Albrecht Stenzinger
- German Center for Lung Research (DZL), Gießen, Germany.,Centers for Personalized Medicine (ZPM), Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Germany
| |
Collapse
|
18
|
Saesen R, Lacombe D, Huys I. Real-world data in oncology: a questionnaire-based analysis of the academic research landscape examining the policies and experiences of the cancer cooperative groups. ESMO Open 2023; 8:100878. [PMID: 36822113 PMCID: PMC10163156 DOI: 10.1016/j.esmoop.2023.100878] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/07/2023] [Accepted: 01/13/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Real-world data (RWD) have quickly emerged as an important source of information to address uncertainties about new treatments, including novel anticancer therapies. Many stakeholders are using such data and the evidence derived therefrom to answer the questions that remain about the safety and effectiveness of antitumor medicines after their approval by regulators. Our objective was to investigate the academic RWD study landscape and explore to what extent RWD are being integrated into investigator-initiated clinical research. MATERIALS AND METHODS We designed an online survey that was distributed between May and August 2022 to representatives of cancer cooperative groups active in Europe, North America, South America, Asia, and/or Oceania. RESULTS In total, 125 cooperative groups operating in 58 different countries and conducting research across 13 distinct cancer domains participated in the survey. While most of the responders (67.2%) did not have a formal policy in place to gather and utilize RWD, a majority (68.0%) had carried out studies involving the analysis of such data before, both for exploratory and confirmatory purposes. The groups that were experienced in capturing and interpreting RWD had mainly worked with observational RWD that were not predominantly prospective or retrospective in nature and which originated from disease registries, electronic health records, and patient questionnaires. They perceived the low costs and the large scale of RWD research to be its most significant benefits, and viewed the accompanying methodological and operational challenges as its biggest constraints. However, they did not have a common understanding of what RWD were. Despite their experience with analyzing RWD, their research portfolio still primarily comprised traditional clinical trials; 62.5% of the groups that had never undertaken any RWD studies were nonetheless planning to initiate them in the future. CONCLUSIONS Cancer cooperative groups are already incorporating RWD studies into their research agendas, but still lack knowledge and expertise in this regard, and do not agree on what RWD are. The conduct of conventional clinical trials continues to be their priority.
Collapse
Affiliation(s)
- R Saesen
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium; Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| | - D Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - I Huys
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Promise of Real-World Evidence for Patient Centricity in Gulf Cooperation Council Countries: Call to Action. Drugs Real World Outcomes 2022; 10:1-9. [PMID: 36394823 PMCID: PMC9944129 DOI: 10.1007/s40801-022-00336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/18/2022] Open
Abstract
Presently, Gulf Cooperation Council countries are lagging in the generation of real-world data and use of real-world evidence for patient-centered care compared with the global average. In a collaborative effort, experts from multiple domains of the healthcare environment from the Gulf Cooperation Council countries came together to present their views and recommended key action points for the generation of robust real-world data and leveraging real-world evidence in the countries. The opinions of the experts are presented, along with existing barriers to the effective generation of real-world evidence in the countries. The Gulf Cooperation Council countries are undergoing transformative changes paving the way for improved healthcare measures; however, the challenges in generating reliable, robust, accessible, and secure real-world evidence are persistent. Hence, ongoing public-private engagements, as well as collaborations between regulators, policymakers, healthcare professionals, insurance and pharmaceutical companies, and patients, are warranted. A few notable examples of real-world evidence studies highlighting the benefits of real-world evidence for gaining valuable insights into patient-centric decision making are also discussed. The actionable steps identified for successful real-world evidence generation would provide long-term, real-world evidence-based patient-centric benefits for the countries.
Collapse
|
20
|
Liu SV, Hu X, Li Y, Zhao B, Burke T, Velcheti V. Pembrolizumab-combination therapy for previously untreated metastatic nonsquamous NSCLC: Real-world outcomes at US oncology practices. Front Oncol 2022; 12:999343. [PMID: 36324586 PMCID: PMC9618586 DOI: 10.3389/fonc.2022.999343] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Objectives The availability of immunotherapies has expanded the options for treating metastatic NSCLC, but information is needed regarding outcomes of immunotherapy for patients treated outside of clinical trials. The aim of this retrospective study was to evaluate the outcomes of therapy with first-line pembrolizumab plus pemetrexed and carboplatin (pembrolizumab-combination) for patients with metastatic nonsquamous NSCLC in the real-world setting of oncology clinics in the United States (US). Methods Using deidentified, longitudinal patient records from a nationwide, electronic health record-derived US database, we identified patients with metastatic nonsquamous NSCLC, without EGFR/ALK/ROS1 genomic alterations, who had received no previous systemic anticancer therapy. Eligible patients had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and initiated first-line pembrolizumab-combination therapy from 11-May-2017 to 31-January-2019; data cutoff was 31-August-2020. Patients treated in a clinical trial were excluded. Manual chart review supplemented technology-enabled abstraction to identify disease progression and tumor response. Time-to-event endpoints from initiation of pembrolizumab-combination therapy were determined using Kaplan-Meier. Results Of 377 patients with metastatic nonsquamous NSCLC, 105 (28%), 104 (28%), and 103 (27%) had programmed death-ligand 1 (PD-L1) expression ≥50%, 1–49%, and <1%, respectively; PD-L1 expression was not documented for 65 patients (17%). Median age was 66 years, and 227 patients (60%) were men. Median follow-up time from first-line therapy initiation to data cutoff was 31.2 months (range, 19.0-39.6 months). Median pembrolizumab real-world time on treatment (rwToT) was 5.8 months (95% CI, 5.0-6.7); 12- and 24-month on-treatment rates for pembrolizumab were 28.0% and 14.9%, respectively. Median overall survival (OS) was 17.2 months (95% CI, 13.6-19.9). For patients in PD-L1 expression ≥50%, 1-49%, <1%, and unknown cohorts, the 12-month survival rates were 66.0%, 58.5%, 54.5%, and 58.3%, respectively, and 24-month survival rates were 43.1%, 37.2%, 35.6%, and 42.0%, respectively. Median real-world progression-free survival was 6.2 months (95% CI, 5.5-7.1); and the real-world response rate was 39.3%, with median duration of response of 13.1 months (95% CI, 10.5-16.8). Conclusions These findings demonstrate the benefits of first-line pembrolizumab-combination therapy for patients with EGFR/ALK-wild-type, metastatic nonsquamous NSCLC and good performance status who are treated at US community oncology clinics.
Collapse
Affiliation(s)
- Stephen V. Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
- *Correspondence: Stephen V. Liu,
| | - Xiaohan Hu
- Center for Observational and Real-world Evidence, Merck & Co., Inc., Rahway, NJ, United States
| | - Yeran Li
- Center for Observational and Real-world Evidence, Merck & Co., Inc., Rahway, NJ, United States
| | - Bin Zhao
- Clinical Research, Merck & Co., Inc., Rahway, NJ, United States
| | - Thomas Burke
- Center for Observational and Real-world Evidence, Merck & Co., Inc., Rahway, NJ, United States
| | - Vamsidhar Velcheti
- Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| |
Collapse
|
21
|
Schafer ES, Chao K, Stevens AM, Jo E, Hilsenbeck SG, Gossai NP, Doan A, Colace SI, Guinipero T, Otterson D, Kaplan JA, Hinson A, Pommert L, Wayne AS, Bhojwani D, Burke MJ. Real-world experience in treating pediatric relapsed/refractory or therapy-related myeloid malignancies with decitabine, vorinostat, and FLAG therapy based on a phase 1 study run by the TACL consortium. Pediatr Blood Cancer 2022; 69:e29812. [PMID: 35726868 DOI: 10.1002/pbc.29812] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 01/27/2023]
Abstract
Current therapies for relapsed/refractory (R/R) pediatric myeloid neoplasms are inadequately effective. Real-world data (RWD) can improve care by augmenting traditional studies and include individuals not eligible for clinical trials. The Therapeutic Advances in Childhood Leukemia and Lymphoma (TACL) consortium recently completed T2016-003, a phase 1 study of decitabine, vorinostat, fludarabine, cytarabine, and granulocyte colony-stimulating factor (G-CSF) in R/R acute myeloid leukemia (AML), which added epigenetic drugs to a cytotoxic backbone. We report results of RWD from six centers that treated 28 pediatric patients (26 with AML, two with other myeloid neoplasms) identically to the TACL study but who were not enrolled. This allowed unique analyses and the ability to compare data with the 35 TACL study patients. The overall response rate (ORR) (complete response [CR] plus CR with incomplete count recovery) among 26 RWD evaluable patients was 65%. The ORR of 13 patients with relapsed AML with epigenetic alterations was 69% (T2016-003 + RWD: 68%, n = 25), of eight patients with refractory AML was 38% (T2016-003 + RWD: 41%, n = 17) and of five patients with therapy-related AML (t-AML) was 80% (T2016-003 + RWD: 75%, n = 8). The mean number of Grade 3/4 toxicities experienced by the T2016-003-eligible RWD population (n = 22) (one per patient-cycle) was not meaningfully different than those (n = 6) who would have been TACL study-ineligible secondary to comorbidities (two per patient-cycle). Overall, this therapy was well tolerated and effective in pediatric patients with R/R myeloid neoplasms, particularly those with epigenetic alterations, t-AML, and refractory disease.
Collapse
Affiliation(s)
- Eric S Schafer
- Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Houston, Texas, USA
| | - Karen Chao
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Alexandra M Stevens
- Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Houston, Texas, USA
| | - Eunji Jo
- Baylor College of Medicine, Houston, Texas, USA
| | | | - Nathan P Gossai
- Center for Cancer and Blood Diseases, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Andrew Doan
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | - Joel A Kaplan
- Levine Children's Hospital/Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Ashley Hinson
- Levine Children's Hospital/Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Lauren Pommert
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Alan S Wayne
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Deepa Bhojwani
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
22
|
Saesen R, Kantidakis G, Marinus A, Lacombe D, Huys I. How do cancer clinicians perceive real-world data and the evidence derived therefrom? Findings from an international survey of the European Organisation for Research and Treatment of Cancer. Front Pharmacol 2022; 13:969778. [PMID: 36091761 PMCID: PMC9449152 DOI: 10.3389/fphar.2022.969778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The role of real-world evidence (RWE) in the development of anticancer therapies has been gradually growing over time. Regulators, payers and health technology assessment agencies, spurred by the rise of the precision medicine model, are increasingly incorporating RWE into their decision-making regarding the authorization and reimbursement of novel antineoplastic treatments. However, it remains unclear how this trend is viewed by clinicians in the field. This study aimed to investigate the opinions of these stakeholders with respect to RWE and its suitability for informing regulatory, reimbursement-related and clinical decisions in oncology.Methods: An online survey was disseminated to clinicians belonging to the network of the European Organisation for Research and Treatment of Cancer between May and July 2021.Results: In total, 557 clinicians across 30 different countries participated in the survey, representing 13 distinct cancer domains. Despite seeing the methodological challenges associated with its interpretation as difficult to overcome, the respondents mostly (75.0%) perceived RWE positively, and believed such evidence could be relatively strong, depending on the designs and data sources of the studies from which it is produced. Few (4.6%) saw a future expansion of its influence on decision-makers as a negative evolution. Furthermore, nearly all (94.0%) participants were open to the idea of sharing anonymized or pseudonymized electronic health data of their patients with external parties for research purposes. Nevertheless, most clinicians (77.0%) still considered randomized controlled trials (RCTs) to be the gold standard for generating clinical evidence in oncology, and a plurality (49.2%) thought that RWE cannot fully address the knowledge gaps that remain after a new antitumor intervention has entered the market. Moreover, a majority of respondents (50.7%) expressed that they relied more heavily on RCT-derived evidence than on RWE for their own decision-making.Conclusion: While cancer clinicians have positive opinions about RWE and want to contribute to its generation, they also continue to hold RCTs in high regard as sources of actionable evidence.
Collapse
Affiliation(s)
- Robbe Saesen
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
- *Correspondence: Robbe Saesen,
| | - Georgios Kantidakis
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Ann Marinus
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Denis Lacombe
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - Isabelle Huys
- Clinical Pharmacology and Pharmacotherapy Research Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Abstract
Randomized controlled trials (RCTs) are the gold standard design to establish the efficacy of new drugs and to support regulatory decision making. However, a marked increase in the submission of single-arm trials (SATs) has been observed in recent years, especially in the field of oncology due to the trend towards precision medicine contributing to the rise of new therapeutic interventions for rare diseases. SATs lack results for control patients, and information from external sources can be compiled to provide context for better interpretability of study results. External comparator arm (ECA) studies are defined as a clinical trial (most commonly a SAT) and an ECA of a comparable cohort of patients-commonly derived from real-world settings including registries, natural history studies, or medical records of routine care. This publication aims to provide a methodological overview, to sketch emergent best practice recommendations and to identify future methodological research topics. Specifically, existing scientific and regulatory guidance for ECA studies is reviewed and appropriate causal inference methods are discussed. Further topics include sample size considerations, use of estimands, handling of different data sources regarding differential baseline covariate definitions, differential endpoint measurements and timings. In addition, unique features of ECA studies are highlighted, specifically the opportunity to address bias caused by unmeasured ECA covariates, which are available in the SAT.
Collapse
|
24
|
Real-World Treatment with Nivolumab or Cabozantinib for Metastatic Renal Cell Carcinoma (mRCC) in the Veneto Region of Italy: Results of AMOUR Study. Target Oncol 2022; 17:467-474. [PMID: 35751733 PMCID: PMC9345814 DOI: 10.1007/s11523-022-00892-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
Background Second- or third-line treatment options for metastatic renal cell carcinoma (mRCC) have dramatically changed in the last few years. There are no criteria for the choice between nivolumab and cabozantinib, which both demonstrated overall survival (OS) gain in pivotal trials. Objective We conducted an analysis of oncological outcomes in patients treated in the Veneto Region (Italy), studying different sequences of TKI-nivolumab-cabozantinib or TKI-cabozantinib-nivolumab in a publicly funded healthcare system. Patients and Methods We conducted a retrospective, real-world analysis of all consecutive patients with mRCC treated with nivolumab or cabozantinib in 2017–2018 at 19 Oncology Units in the Veneto Region. Results We identified 170 patients, 73 % males, median age 68.4 years. All patients started second-line treatment, 59 % received a third-line therapy. Patients with NLR > 3 had a shorter OS (p < 0.0001). In the second-line treatment, nivolumab was administered to 108 patients (63 %), cabozantinib to 29 (17 %); in the third-line treatment nivolumab was administered to 42 patients (25 %), cabozantinib to 49 (29 %). Median OS and PFS in second line treatment were 28.4 and 6.6 months for nivolumab, 16.8 and 6.6 months for cabozantinib. Median OS and PFS in third-line treatment were 27 and 5.2 months for nivolumab, 16.6 and 7.5 months for cabozantinib. Median OS for nivolumab>cabozantinib sequence versus cabozantinib > nivolumab was 28.8 versus 19.9 months (p = 0.2); median PFS for both the sequences were similar at 5.7 months. A cost effectiveness per month of survival of the two sequences analysis was performed: the cost per month for the nivolumab > cabozantinib sequence was 1738.60whereas the cost for the other one was €1624.80. Conclusions In our real-world cohort, most patients received nivolumab as second-line treatment. Outcomes of single drugs are superimposable with those in the published literature. Both the sequences of nivolumab and cabozantinib appear to be viable, effective strategies from an OS and cost-effective perspective.
Collapse
|
25
|
Gately L, Drummond K, Rosenthal M, Harrup R, Dowling A, Gogos A, Lwin Z, Collins I, Campbell D, Ahern E, Phillips C, Gan HK, Bennett I, Sieber OM, Gibbs P. Beyond standard data collection – the promise and potential of BRAIN (Brain tumour Registry Australia INnovation and translation registry). BMC Cancer 2022; 22:604. [PMID: 35655179 PMCID: PMC9161524 DOI: 10.1186/s12885-022-09700-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Real-world data (RWD) is increasingly being embraced as an invaluable source of information to address clinical and policy-relevant questions that are unlikely to ever be answered by clinical trials. However, the largely unrealised potential of RWD is the value to be gained by supporting prospective studies and translational research. Here we describe the design and implementation of an Australian brain cancer registry, BRAIN, which is pursuing these opportunities.
Methods
BRAIN was designed by a panel of clinicians in conjunction with BIOGRID to capture comprehensive clinical data on patients diagnosed with brain tumours from diagnosis through treatment to recurrence or death. Extensive internal and external testing was undertaken, followed by implementation at multiple sites across Victoria and Tasmania.
Results
Between February 2021 and December 2021, a total of 350 new patients from 10 sites, including one private and two regional, were entered into BRAIN. Additionally, BRAIN supports the world’s first registry trial in neuro-oncology, EX-TEM, addressing the optimal duration of post-radiation temozolomide; and BioBRAIN, a dedicated brain tumour translational program providing a pipeline for biospecimen collection matched with linked clinical data.
Conclusions
Here we report on the first data collection effort in brain tumours for Australia, which we believe to be unique worldwide given the number of sites and patients involved and the extent to which the registry resource is being leveraged to support clinical and translational research. Further directions such as passive data flow and data linkages, use of artificial intelligence and inclusion of patient-entered data are being explored.
Collapse
|
26
|
van Laar SA, Gombert-Handoko KB, Groenwold RHH, van der Hulle T, Visser LE, Houtsma D, Guchelaar HJ, Zwaveling J. Real-World Metastatic Renal Cell Carcinoma Treatment Patterns and Clinical Outcomes in The Netherlands. Front Pharmacol 2022; 13:803935. [PMID: 35401238 PMCID: PMC8983834 DOI: 10.3389/fphar.2022.803935] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/18/2022] [Indexed: 12/28/2022] Open
Abstract
The number of treatment options for patients with metastatic renal cell carcinoma (mRCC) has significantly grown in the last 15 years. Although randomized controlled trials are fundamental in investigating mRCC treatment efficacy, their external validity can be limited. Therefore, the efficacy of the different treatment options should also be evaluated in clinical practice. We performed a chart review of electronic health records using text mining software to study the current treatment patterns and outcomes. mRCC patients from two large hospitals in the Netherlands, starting treatment between January 2015 and May 2020, were included. Data were collected from electronic health records using a validated text mining tool. Primary endpoints were progression-free survival (PFS) and overall survival (OS). Statistical analyses were performed using the Kaplan-Meier method. Most frequent first-line treatments were pazopanib (n = 70), sunitinib (n = 34), and nivolumab with ipilimumab (n = 28). The overall median PFS values for first-line treatment were 15.7 months (95% confidence interval [95%CI], 8.8-20.7), 16.3 months (95%CI, 9.3-not estimable [NE]) for pazopanib, and 6.9 months (95% CI, 4.4-NE) for sunitinib. The overall median OS values were 33.4 months (95%CI, 28.1-50.9 months), 39.3 months (95%CI, 29.5-NE) for pazopanib, and 28.1 months (95%CI, 7.0-NE) for sunitinib. For nivolumab with ipilimumab, median PFS and median OS were not reached. Of the patients who finished first- and second-line treatments, 64 and 62% received follow-up treatments, respectively. With most patients starting on pazopanib and sunitinib, these real-world treatment outcomes were most likely better than in pivotal trials, which may be due to extensive follow-up treatments.
Collapse
Affiliation(s)
- S A van Laar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - K B Gombert-Handoko
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - R H H Groenwold
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - T van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - L E Visser
- Department of Hospital Pharmacy, Haga Teaching Hospital, The Hague, Netherlands.,Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands.,Department of Hospital Pharmacy, Erasmus MC, Rotterdam, Netherlands
| | - D Houtsma
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, Netherlands
| | - H J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - J Zwaveling
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
27
|
Lorenzi M, Ferro A, Cecere F, Scattolin D, Del Conte A, Follador A, Pilotto S, Polo V, Santarpia M, Chiari R, Pavan A, Dal Maso A, Da Ros V, Targato G, Vari S, Indraccolo S, Calabrese F, Frega S, Bonanno L, Conte PF, Guarneri V, Pasello G. First-Line Osimertinib in Patients with EGFR-Mutant Advanced Non-Small Cell Lung Cancer: Outcome and Safety in the Real World: FLOWER Study. Oncologist 2022; 27:87-e115. [PMID: 35641222 PMCID: PMC9714585 DOI: 10.1002/onco.13951] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Osimertinib became the standard treatment for patients with untreated EGFR-mutant advanced non-small cell lung cancer (aNSCLC) following results reported in the phase III randomized FLAURA trial. Because of strict exclusion criteria, patient populations included in pivotal trials are only partially representative of real-world patients. METHODS We designed an observational, prospective, multicenter study enrolling patients with EGFR-mutant aNSCLC receiving first-line osimertinib to evaluate effectiveness, safety, and progression patterns in the real-world. RESULTS At data cutoff, 126 White patients from nine oncology centers were included. At diagnosis, 16 patients (12.7%) had a performance status (PS) ≥2 and 38 (30.2%) had brain metastases. Overall response rate (ORR) was 73%, disease control rate (DCR) 96.0%. After a median follow-up of 12.3 months, median time to treatment discontinuation (mTTD) was 25.3 months, median progression-free-survival (mPFS) was 18.9 months and median overall survival (mOS) was not reached (NR). One hundred and ten patients (87%) experienced adverse events (AEs), 42 (33%) of grade 3-4, with venous thromboembolism (VTE) as the most common (n = 10, 7.9%). No difference in rates of VTE was reported according to age, PS, comorbidity, and tumor load. We observed longer mTTD in patients without symptoms (NR vs. 18.8 months) and with fewer than three metastatic sites at diagnosis (NR vs. 21.4 months). Patients without brain metastases experienced longer mPFS (NR vs. 13.3 months). No difference in survival outcome was observed according to age, comorbidity, and type of EGFR mutation. Isolated progression and progression in fewer than three sites were associated with longer time to treatment discontinuation (TTD). CONCLUSION Osimertinib confirmed effectiveness and safety in the real world, although thromboembolism was more frequent than previously reported.
Collapse
Affiliation(s)
- Martina Lorenzi
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Alessandra Ferro
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Fabiana Cecere
- Oncology 1, Regina Elena National Cancer Institute – IRCCS,
Padova, Italy
| | - Daniela Scattolin
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
| | - Alessandro Del Conte
- Medical Oncology and Immunorelated Tumors, National Cancer Institute Centro di
Riferimento Oncologico (CRO) – IRCCS, Aviano (PN),
Italy
| | - Alessandro Follador
- Department of Medical Oncology, Azienda Sanitaria Universitaria Integrata of
Udine, Santa Maria della Misericordia Hospital,
Udine, Italy
| | - Sara Pilotto
- Oncology Department, Azienda Ospedaliera Universitaria Integrata di
Verona, Verona, Italy
| | - Valentina Polo
- Oncology Unit, Azienda Unità Locale Socio Sanitaria (AULSS 2) Marca Trevigiana,
Ca’ Foncello Hospital, Treviso, Italy
| | - Mariacarmela Santarpia
- Medical Oncology, Azienda Ospedaliera Policlinico Universitario “G.
Martino,”Messina, Italy
| | - Rita Chiari
- Medical Oncology, AULSS 6 Euganea, South Padua Hospital,
Monselice (PD), Italy
| | - Alberto Pavan
- Medical Oncology, AULSS 6 Euganea, South Padua Hospital,
Monselice (PD), Italy
| | - Alessandro Dal Maso
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Valentina Da Ros
- Medical Oncology and Immunorelated Tumors, National Cancer Institute Centro di
Riferimento Oncologico (CRO) – IRCCS, Aviano (PN),
Italy
| | - Giada Targato
- Department of Medical Oncology, Azienda Sanitaria Universitaria Integrata of
Udine, Santa Maria della Misericordia Hospital,
Udine, Italy
| | - Sabrina Vari
- Oncology 1, Regina Elena National Cancer Institute - IRCCS,
Rome, Italy
| | - Stefano Indraccolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV –
IRCCS, Padua, Italy
| | - Fiorella Calabrese
- Cardiovascular Pathology Unit, Department of Cardio-Thoracic and Vascular
Sciences, University of Padova, Padova, Italy
| | - Stefano Frega
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Laura Bonanno
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Pier Franco Conte
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| | - Giulia Pasello
- Department of Surgery, Oncology, and Gastroenterology, University of
Padova, Padova, Italy
- Division of Medical Oncology 2, Veneto Institute of Oncology -
IRCCS, Padova, Italy
| |
Collapse
|
28
|
Götte H, Kirchner M, Krisam J, Allignol A, Lamy F, Schüler A, Kieser M. An adaptive design for early clinical development including interim decision for single‐arm trial with external controls or randomized trial. Pharm Stat 2022; 21:625-640. [DOI: 10.1002/pst.2190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Heiko Götte
- Global Biostatistics, Epidemiology & Medical Writing Merck Healthcare KGaA Darmstadt Germany
| | - Marietta Kirchner
- Institute of Medical Biometry University of Heidelberg Heidelberg Germany
| | - Johannes Krisam
- Institute of Medical Biometry University of Heidelberg Heidelberg Germany
| | - Arthur Allignol
- Global Biostatistics, Epidemiology & Medical Writing Merck Healthcare KGaA Darmstadt Germany
| | - Francois‐Xavier Lamy
- Global Biostatistics, Epidemiology & Medical Writing Merck Healthcare KGaA Darmstadt Germany
| | - Armin Schüler
- Global Biostatistics, Epidemiology & Medical Writing Merck Healthcare KGaA Darmstadt Germany
| | - Meinhard Kieser
- Institute of Medical Biometry University of Heidelberg Heidelberg Germany
| |
Collapse
|
29
|
Yap TA, Jacobs I, Baumfeld Andre E, Lee LJ, Beaupre D, Azoulay L. Application of Real-World Data to External Control Groups in Oncology Clinical Trial Drug Development. Front Oncol 2022; 11:695936. [PMID: 35070951 PMCID: PMC8771908 DOI: 10.3389/fonc.2021.695936] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
Randomized controlled trials (RCTs) that assess overall survival are considered the "gold standard" when evaluating the efficacy and safety of a new oncology intervention. However, single-arm trials that use surrogate endpoints (e.g., objective response rate or duration of response) to evaluate clinical benefit have become the basis for accelerated or breakthrough regulatory approval of precision oncology drugs for cases where the target and research populations are relatively small. Interpretation of efficacy in single-arm trials can be challenging because such studies lack a standard-of-care comparator arm. Although an external control group can be based on data from other clinical trials, using an external control group based on data collected outside of a trial may not only offer an alternative to both RCTs and uncontrolled single-arm trials, but it may also help improve decision-making by study sponsors or regulatory authorities. Hence, leveraging real-world data (RWD) to construct external control arms in clinical trials that investigate the efficacy and safety of drug interventions in oncology has become a topic of interest. Herein, we review the benefits and challenges associated with the use of RWD to construct external control groups, and the relevance of RWD to early oncology drug development.
Collapse
Affiliation(s)
- Timothy A. Yap
- Department of Investigational Cancer Therapeutics (Phase I Program), Division of Cancer Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ira Jacobs
- Pfizer Inc., New York, NY, United States
| | | | | | | | - Laurent Azoulay
- Centre for Clinical Epidemiology Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Department of Epidemiology, Biostatistics, and Occupational Health and Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Pasello G, Lorenzi M, Calvetti L, Oliani C, Pavan A, Favaretto A, Palazzolo G, Giovanis P, Zustovich F, Bonetti A, Bernardi D, Mandarà M, Aprile G, Crivellaro G, Sinigaglia G, Tognazzo S, Morandi P, Bortolami A, Marino V, Bonanno L, Guarneri V, Conte P. OUP accepted manuscript. Oncologist 2022; 27:e484-e493. [PMID: 35429394 PMCID: PMC9177098 DOI: 10.1093/oncolo/oyac051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 01/28/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- Giulia Pasello
- Corresponding author: Giulia Pasello, University of Padova DiSCOG and Istituto Oncologico Veneto IRCCS, Via Gattamelata 64, 35128 Padova, Italy. Tel: +390498215608; Fax: +390498215932;
| | - Martina Lorenzi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Lorenzo Calvetti
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | | | - Alberto Pavan
- Medical Oncology Department, ULSS 3 Serenissima, Sant’Angelo General Hospital, Mestre and SS Giovanni e Paolo General Hospital, Venezia, Italy
| | - Adolfo Favaretto
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’Foncello Hospital, Treviso, Italy
| | - Giovanni Palazzolo
- Medical Oncology, AULSS 6 Euganea, Cittadella – Camposampiero Hospital, Camposampiero, Italy
| | - Petros Giovanis
- Department of Oncology, Unit of Oncology, Santa Maria del Prato Hospital, Azienda ULSS 1 Dolomiti, Feltre, Italy
| | - Fable Zustovich
- Clinical Oncology Department, AULSS 1 Dolomiti, San Martino Hospital, Belluno, Italy
| | - Andrea Bonetti
- Department of Oncology, AULSS 9 of the Veneto Region, Mater Salutis Hospital, Legnago, Italy
| | - Daniele Bernardi
- Medical Oncology, ULSS 4 “Veneto Orientale”, San Donà di Piave (VE), Italy
| | - Marta Mandarà
- Department of Medical Oncology, AULSS 9 Scaligera, Verona, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Giovanna Crivellaro
- Rete Oncologica Veneta (ROV), Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | | | - Sandro Tognazzo
- Rete Oncologica Veneta (ROV), Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Paolo Morandi
- Medical Oncology Department, ULSS 3 Serenissima, Sant’Angelo General Hospital, Mestre and SS Giovanni e Paolo General Hospital, Venezia, Italy
| | - Alberto Bortolami
- Rete Oncologica Veneta (ROV), Istituto Oncologico Veneto, IRCCS, Padova, Italy
| | - Valentina Marino
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’Foncello Hospital, Treviso, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | | | | |
Collapse
|
31
|
Flynn R, Plueschke K, Quinten C, Strassmann V, Duijnhoven RG, Gordillo-Marañon M, Rueckbeil M, Cohet C, Kurz X. Marketing Authorization Applications Made to the European Medicines Agency in 2018-2019: What was the Contribution of Real-World Evidence? Clin Pharmacol Ther 2021; 111:90-97. [PMID: 34689339 PMCID: PMC9299056 DOI: 10.1002/cpt.2461] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 10/13/2021] [Indexed: 11/21/2022]
Abstract
Information derived from routinely collected real‐world data has for a long time been used to support regulatory decision making on the safety of drugs and has more recently been used to support marketing authorization submissions to regulators. There is a lack of detailed information on the use and types of this real‐world evidence (RWE) as submitted to regulators. We used resources held by the European Medicines Agency (EMA) to describe the characteristics of RWE included in new marketing authorization applications (MAAs) and extensions of indication (EOIs) for already authorized products submitted to the EMA in 2018 and 2019. For MAAs, 63 of 158 products (39.9%) contained RWE with a total of 117 studies. For 31.7% of these products, the RWE submitted was derived from data collected before the planned authorization. The most common data sources were registries (60.3%) followed by hospital data (31.7%). RWE was mainly included to support safety (87.3%) and efficacy (49.2%) with cohort studies being the most frequently used study design (88.9%). For EOIs, 28 of 153 products (18.3%) contained RWE with a total of 36 studies. For 57.1% of these products, studies were conducted prior to the EOIs. RWE sources were mainly registries (35.6%) and hospital data (27.0%). RWE was typically used to support safety (82.1%) and efficacy (53.6%). Cohort studies were the most commonly used study design (87.6%). We conclude that there is widespread use of RWE to support evaluation of MAAs and EOIs submitted to the EMA and identify areas where further research is required.
Collapse
Affiliation(s)
- Robert Flynn
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands.,Medicines Monitoring Unit, University of Dundee, Dundee, UK
| | - Kelly Plueschke
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Chantal Quinten
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Valerie Strassmann
- Pharmacovigilance Office, Quality and Safety of Medicines, Human Medicines Division European Medicines Agency, Amsterdam, The Netherlands
| | - Ruben G Duijnhoven
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands.,Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Gordillo-Marañon
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands.,Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
| | - Marcia Rueckbeil
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands.,Department of Medical Statistics, University Hospital Aachen, Aachen, Germany
| | - Catherine Cohet
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| | - Xavier Kurz
- Data Analytics and Methods Task Force, European Medicines Agency, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Brown JP, Douglas IJ, Hanif S, Thwaites RMA, Bate A. Measuring the Effectiveness of Real-World Evidence to Ensure Appropriate Impact. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2021; 24:1241-1244. [PMID: 34452702 DOI: 10.1016/j.jval.2021.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/22/2021] [Accepted: 03/25/2021] [Indexed: 06/13/2023]
Abstract
The value of real-world evidence (RWE) in medicines regulation and health technology assessment has been increasingly emphasized. Nevertheless, although RWE is increasingly used, there has been limited systematic evidence of its value. A recent study that examined the role and impact of RWE in regulatory assessments conducted through the European Medicines Agency provided such evidence. Results of the study demonstrated RWE was important to decision making, particularly for certain questions such as the quantification of adverse events, the evaluation of risk minimization measures, and the assessment of product usage. The study suggested, however, that in many of the assessments further RWE would have been valuable and concluded that RWE has, as yet, played a limited role in hypothesis generation and in the assessment of medication effectiveness. This study had been possible only because of the transparency of the European Medicines Agency decision making. Ensuring transparency of RWE evidence collection, study design and conduct, and of decision making based on this evidence will facilitate further development of the uses and value of RWE. Keywords: benefit-risk assessment; medicines regulation; real-world evidence; regulatory decision making.
Collapse
Affiliation(s)
- Jeremy P Brown
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, England, UK.
| | - Ian J Douglas
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, England, UK
| | | | | | - Andrew Bate
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, England, UK; Global Safety, GSK, Brentford, Middlesex, England, UK
| |
Collapse
|
33
|
Ferrari SM, Elia G, Ragusa F, Paparo SR, Mazzi V, Miccoli M, Galdiero MR, Varricchi G, Foddis R, Guglielmi G, Spinelli C, La Motta C, Benvenga S, Antonelli A, Fallahi P. Lenvatinib: an investigational agent for the treatment of differentiated thyroid cancer. Expert Opin Investig Drugs 2021; 30:913-921. [PMID: 34428101 DOI: 10.1080/13543784.2021.1972971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Differentiated thyroid cancer (DTC; >90% of all TCs) derives from follicular cells. Surgery is the main therapeutic strategy, and radioiodine (RAI) is administered after thyroidectomy. When DTC progresses, it does not respond to RAI and thyroid-stimulating hormone (TSH)-suppressive thyroid hormone treatment, and other therapies (i.e. surgery, external beam radiation therapy and chemotherapy) do not lead to a better survival. Thanks to the understanding of the molecular pathways involved in TC progression, important advances have been done. Lenvatinib is a multitargeted tyrosine kinase inhibitor of VEGFR1-3, FGFR1-4, PDGFRα, RET, and KIT signaling networks implicated in tumor angiogenesis, approved in locally recurrent or metastatic, progressive, RAI-refractory DTC. Unmet needs regarding the patient clinical therapy responsiveness in aggressive RAI-refractory DTC still remain. AREAS COVERED We provide an overview from the literature of in vitro, in vivo and real-life studies regarding lenvatinib as an investigational agent for the treatment of aggressive TC. EXPERT OPINION According to the SELECT trial, the treatment should be initiated with a dosage of 24 mg/day, subsequently decreasing it in relation to the side effects. The decision making process in patients with aggressive RAI-refractory DTC should be personalized and the potential toxicity should be properly managed.
Collapse
Affiliation(s)
| | - Giusy Elia
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Valeria Mazzi
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Mario Miccoli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; WAO Center of Excellence, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR),Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; WAO Center of Excellence, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR),Naples, Italy
| | - Rudy Foddis
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Guglielmi
- U.O. Medicina Preventiva Del Lavoro, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Claudio Spinelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | | | - Salvatore Benvenga
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Master Program on Childhood, Adolescent and Women's Endocrine Health, University of Messina, Messina, Italy.,Interdepartmental Program of Molecular and Clinical Endocrinology and Women's Endocrine Health, Azienda Ospedaliera Universitaria Policlinico 'G. Martino', Messina, Italy
| | - Alessandro Antonelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Poupak Fallahi
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
34
|
Boyle JM, Hegarty G, Frampton C, Harvey-Jones E, Dodkins J, Beyer K, George G, Sullivan R, Booth C, Aggarwal A. Real-world outcomes associated with new cancer medicines approved by the Food and Drug Administration and European Medicines Agency: A retrospective cohort study. Eur J Cancer 2021; 155:136-144. [PMID: 34371443 PMCID: PMC8442759 DOI: 10.1016/j.ejca.2021.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Purpose Real-World Data (RWD) studies are increasingly used to support regulatory approvals, reimbursement decisions, and changes in clinical practice for novel cancer drugs. However, few studies have systematically appraised their quality or compared outcomes to pivotal trials. Methods All RWD studies (2010–2019) for drugs approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) from 2010 to 2015 for solid organ tumours in the non-curative setting were identified. Quality assessment was undertaken using the Newcastle Ottawa Scale. Survival differences between each RWD study and the pivotal trial were determined using a related sample Wilcoxon signed-rank test. Results 293 RWD studies for 45 of the 57 drug indications approved by the FDA/EMA were identified. The most common tumour types were prostate cancer (29%, n = 86) and melanoma (15%, n = 43). A quarter of the studies had industry funding. No high-quality studies were identified, and 78% were low quality. Comparative survival analysis between RWD and pivotal trials was possible for 224 studies (37 drug indications). Differences in median survival between the RWD studies and their corresponding trial ranged from −32 months to 21 months (IQR –4·2 months to 1·6 months). Low-quality studies were more likely to report superior survival outcomes (23%) compared to higher quality studies (8%) (p = 0.02). Conclusion RWD study quality for novel cancer drugs is low and of insufficient rigour to inform reimbursement decisions and clinical practice. RWD studies seeking publication should provide a completed quality assessment tool on submission. Greater investment in properly designed RWD studies is required. Study provides a systematic appraisal of FDA/EMA approved drugs in real-world practice. Most novel FDA/EMA cancer drugs have real-world data (RWD) studies, but the quality is low. Variability in survival outcomes exists, and findings should be applied cautiously. Most RWD studies reported inferior survival outcomes compared to the pivotal trial. Pre-publication critical appraisal checklists should be used for RWD studies.
Collapse
Affiliation(s)
- Jemma M Boyle
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | - Elizabeth Harvey-Jones
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Joanna Dodkins
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Katharina Beyer
- Translational and Oncology Research (TOUR), King's College London, United Kingdom
| | - Gincy George
- Translational and Oncology Research (TOUR), King's College London, United Kingdom
| | - Richard Sullivan
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; Institute of Cancer Policy, King's College London, United Kingdom
| | | | - Ajay Aggarwal
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom; Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom; Institute of Cancer Policy, King's College London, United Kingdom.
| |
Collapse
|
35
|
Multikinase inhibitors for the treatment of radioiodine refractory thyroid cancer: what have we learned from the 'real-world' experience? Curr Opin Oncol 2021; 33:3-8. [PMID: 33060402 DOI: 10.1097/cco.0000000000000693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE OF REVIEW Several molecularly targeted drugs for treating radioiodine resistant differentiated thyroid carcinomas (RAIR-DTC) have been identified. Among these, sorafenib and lenvatinib have been approved for clinical use in many countries. The present review will analyze efficacy and safety 'real-world' data (RWD) emerging after their commercialization. RECENT FINDINGS RWDs confirmed sorafenib and lenvatinib efficacy in terms of progression-free survival and, perhaps, overall survival improvement in patients with RAIR-DTC. Lenvatinib performance in RWDs appeared somehow lower than in randomized clinical trials (RCT), probably because the decision to start treatment in 'real life' was made when patients were in worse clinical conditions than in RCTs. Concerning safety, RWD studies corroborated RCT evidence of elevated overall and serious adverse event incidence. Notably, adverse events were manageable in most cases with appropriate treatment or dose reduction/interruption, so that the need for definitive withdrawal was limited. The suitability of multikinase inhibitors (MKI) as salvage therapy in RAIR-DTCs was also confirmed by RWD experience, at least for lenvatinib in the second-line setting. SUMMARY RWD analysis has corroborated RCT results in terms of MKI efficacy for both first-line and salvage treatment in patients with RAIR-DTC. The safety profiles emerging from RWDs seem to justify the caution recommended by most scientific guidelines.
Collapse
|
36
|
Fernandes LE, Epstein CG, Bobe AM, Bell JSK, Stumpe MC, Salazar ME, Salahudeen AA, Pe Benito RA, McCarter C, Leibowitz BD, Kase M, Igartua C, Huether R, Hafez A, Beaubier N, Axelson MD, Pegram MD, Sammons SL, O'Shaughnessy JA, Palmer GA. Real-world Evidence of Diagnostic Testing and Treatment Patterns in US Patients With Breast Cancer With Implications for Treatment Biomarkers From RNA Sequencing Data. Clin Breast Cancer 2021; 21:e340-e361. [PMID: 33446413 DOI: 10.1016/j.clbc.2020.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/27/2020] [Accepted: 11/13/2020] [Indexed: 01/21/2023]
Abstract
OBJECTIVE/BACKGROUND We performed a retrospective analysis of longitudinal real-world data (RWD) from patients with breast cancer to replicate results from clinical studies and demonstrate the feasibility of generating real-world evidence. We also assessed the value of transcriptome profiling as a complementary tool for determining molecular subtypes. METHODS De-identified, longitudinal data were analyzed after abstraction from records of patients with breast cancer in the United States (US) structured and stored in the Tempus database. Demographics, clinical characteristics, molecular subtype, treatment history, and survival outcomes were assessed according to strict qualitative criteria. RNA sequencing and clinical data were used to predict molecular subtypes and signaling pathway enrichment. RESULTS The clinical abstraction cohort (n = 4000) mirrored the demographics and clinical characteristics of patients with breast cancer in the US, indicating feasibility for RWE generation. Among patients who were human epidermal growth factor receptor 2-positive (HER2+), 74.2% received anti-HER2 therapy, with ∼70% starting within 3 months of a positive test result. Most non-treated patients were early stage. In this RWD set, 31.7% of patients with HER2+ immunohistochemistry (IHC) had discordant fluorescence in situ hybridization results recorded. Among patients with multiple HER2 IHC results at diagnosis, 18.6% exhibited intra-test discordance. Through development of a whole-transcriptome model to predict IHC receptor status in the molecular sequenced cohort (n = 400), molecular subtypes were resolved for all patients (n = 36) with equivocal HER2 statuses from abstracted test results. Receptor-related signaling pathways were differentially enriched between clinical molecular subtypes. CONCLUSIONS RWD in the Tempus database mirrors the overall population of patients with breast cancer in the US. These results suggest that real-time, RWD analyses are feasible in a large, highly heterogeneous database. Furthermore, molecular data may aid deficiencies and discrepancies observed from breast cancer RWD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mark D Pegram
- Stanford Comprehensive Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Sarah L Sammons
- Department of Medicine, Duke University Medical Center, Duke University, Durham, NC
| | | | | |
Collapse
|
37
|
De Leo S, Di Stefano M, Persani L, Fugazzola L, Colombo C. Lenvatinib as first-line treatment for advanced thyroid cancer: long progression-free survival. Endocrine 2021; 72:462-469. [PMID: 32885329 DOI: 10.1007/s12020-020-02477-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/23/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Lenvatinib (LEN) has been approved for the treatment of patients with progressive radioiodine-refractory differentiated thyroid cancer (RAI-R DTC). Real-life studies reported a lower progression-free survival (PFS) than the registration study, likely due to the more advanced stage of tumors, the more frequent pretreatment with other TKIs, the limited follow-up, and the worse clinical condition of the patients included. METHODS We evaluated the clinical data of our cohort of 13 consecutive patients, all receiving LEN as a first-line TKI treatment, and followed-up in a single tertiary Center. RESULTS All patients had an ECOG of 0-1 and regional or distant metastases were documented in 61.5% and 77% of patients, respectively. Median PFS was 22 months (95% CI 14-35) with partial response in 69% and stable disease in 31% of patients. All patients experienced at least one adverse event (AE), the most frequent being fatigue, anorexia, diarrhea, and hypertension. The daily dose was reduced in 70% of patients and only one patient (7.7%) discontinued the drug for AEs. CONCLUSION In this series of RAI-R DTC patients, with the unique features to have an ECOG 0 or 1 and to be naive for TKI treatments, PFS was the longest among all real-life published so far, with the highest rate of patients with partial response and one of the lowest drug discontinuation rate for AEs. The correct timing of treatment start, the tailoring of the dose, and a proper management of the AEs may have a significant impact on the treatment response to LEN.
Collapse
Affiliation(s)
- Simone De Leo
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Marta Di Stefano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Luca Persani
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Laura Fugazzola
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Carla Colombo
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
38
|
Yu SY, Mckavanagh D, McPherson I, Walpole E, Atkinson V, Hollingworth S. Survival of advanced melanoma patients treated with immunotherapy and targeted therapy: A real-world study. Pharmacoepidemiol Drug Saf 2021; 30:1371-1379. [PMID: 33840147 DOI: 10.1002/pds.5248] [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: 10/28/2020] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION We aimed to examine the survival outcomes plus patient and treatment characteristics of advanced melanoma patients treated with first-line immunotherapy (IT), targeted therapy (TT), and chemotherapy (CTH) and compare findings with information from pivotal trials for each therapy. MATERIALS AND METHODS We retrospectively reviewed the use of systematic IT, TT and CTH therapies in melanoma patients in four Queensland public hospitals. We estimated median duration of overall survival (OS) and survival rates (6 months, 1, and 2 years) using Kaplan-Meier methods. We compared our findings to those of clinical trials. RESULTS Five hundred three patients who met the inclusion criteria were divided into three groups based on the first-line treatment: IT 232; TT 157; and CTH 114. OS was 18 months with IT (95% CI 13, 22); 12 months with TT (95% CI 8, 15); and 5 months with CTH (95% CI 5, 6). The demographic characteristics, treatment protocols, and durations for IT and TT were generally consistent with trials but fewer patients in our study had subsequent therapy than in the trials. The OS in our study was slightly lower than the OS reported in trials. CONCLUSION The OS of novel cancer therapy in the real world was lower than seen in trials but is expected given these are patients who have a poorer prognosis. A future study could investigate the impact of prognostic factors on survival in the longer term. This study provides evidence that we can use routinely collected real-world data to evaluate the effectiveness of checkpoint and kinase inhibitors in patients with advanced melanoma.
Collapse
Affiliation(s)
- Su-Yeon Yu
- National Evidence-based Healthcare Collaborating Agency, Seoul, South Korea
| | - Dan Mckavanagh
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ian McPherson
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Euan Walpole
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | | | | |
Collapse
|
39
|
Dodwell D, Shakir R. Assessing New Drugs in Advanced Cancer: Beyond Randomised Evidence. Clin Oncol (R Coll Radiol) 2021; 33:e201-e202. [PMID: 33223383 DOI: 10.1016/j.clon.2020.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/09/2020] [Accepted: 11/06/2020] [Indexed: 11/19/2022]
Affiliation(s)
- D Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - R Shakir
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Vasiliadou I, Breik O, Baker H, Leslie I, Sim VR, Hegarty G, Michaelidou A, Nathan K, Hartley A, Good J, Sanghera P, Fong C, Urbano TG, Lei M, Petkar I, Ferreira MR, Nutting C, Wong KH, Newbold K, Harrington K, Bhide S, Kong A. Safety and Treatment Outcomes of Nivolumab for the Treatment of Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Retrospective Multicenter Cohort Study. Cancers (Basel) 2021; 13:cancers13061413. [PMID: 33808781 PMCID: PMC8003537 DOI: 10.3390/cancers13061413] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 01/19/2023] Open
Abstract
Nivolumab is an anti-PD-1 monoclonal antibody currently used as immunotherapy for patients with recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) with evidence of disease progression after platinum-based chemotherapy. This study evaluates real-world safety and treatment outcomes of non-trial nivolumab use. A retrospective multicenter cohort study of patients with recurrent/metastatic HNSCC treated with nivolumab between January 2017 and March 2020 was performed. Overall, 123 patients were included. The median age was 64 years, the majority of patients were male (80.5%) and had a smoking history (69.9%). Primary outcomes included overall response rate (ORR) of 19.3%, median progression-free survival (PFS) of 3.9 months, 1-year PFS rate of 16.8%, a median overall survival (OS) of 6.5 months and 1-year OS rate of 28.6%. These results are comparable to the CHECKMATE-141 study. Of 27 patients who had PD-L1 status tested, positive PD-L1 status did not significantly affect PFS (p = 0.86) or OS (p = 0.84). Nivolumab was well tolerated with only 15.1% experiencing immune-related toxicities (IRT) and only 6.7% of patients stopping due to toxicity. The occurrence of IRT appeared to significantly affect PFS (p = 0.01) but not OS (p = 0.07). Nivolumab in recurrent/metastatic HNSCC is well tolerated and may be more efficacious in patients who develop IRT.
Collapse
Affiliation(s)
- Ifigenia Vasiliadou
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Omar Breik
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
- Department of Oral and Maxillofacial Surgery, Royal Brisbane and Women’s Hospital, QLD 4029 Brisbane, Australia
| | - Holly Baker
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Isla Leslie
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
| | - Van Ren Sim
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Gemma Hegarty
- Kent Oncology Centre, Maidstone and Tunbridge Wells NHS Trust, Kent ME16 9QQ, UK; (G.H.); (A.M.); (K.N.)
| | - Andriana Michaelidou
- Kent Oncology Centre, Maidstone and Tunbridge Wells NHS Trust, Kent ME16 9QQ, UK; (G.H.); (A.M.); (K.N.)
| | - Kannon Nathan
- Kent Oncology Centre, Maidstone and Tunbridge Wells NHS Trust, Kent ME16 9QQ, UK; (G.H.); (A.M.); (K.N.)
| | - Andrew Hartley
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
| | - James Good
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
| | - Paul Sanghera
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
| | - Charles Fong
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
| | - Teresa Guerrero Urbano
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Mary Lei
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Imran Petkar
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Miguel Reis Ferreira
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
| | - Chris Nutting
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
| | - Kee Howe Wong
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
| | - Kate Newbold
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
| | - Kevin Harrington
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
| | - Shree Bhide
- Head and Neck Unit, Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (I.L.); (C.N.); (K.H.W.); (K.N.); (K.H.); (S.B.)
| | - Anthony Kong
- Guys Cancer Centre, Guy’s and St. Thomas NHS Foundation Trust, London SE1 9RT, UK; (I.V.); (V.R.S.); (T.G.U.); (M.L.); (I.P.); (M.R.F.)
- Department of Oncology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2WB, UK; (O.B.); (H.B.); (A.H.); (J.G.); (P.S.); (C.F.)
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Comprehensive Cancer Centre, King’s College London, Guy’s Campus, London SE1 1UL, UK
- Correspondence:
| |
Collapse
|
41
|
Jemielita T, Li XN, Piperdi B, Zhou W, Burke T, Chen C. Overall Survival With Second-Line Pembrolizumab in Patients With Non-Small-Cell Lung Cancer: Randomized Phase III Clinical Trial Versus Propensity-Adjusted Real-World Data. JCO Clin Cancer Inform 2021; 5:56-65. [PMID: 33439727 DOI: 10.1200/cci.20.00099] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To compare and characterize overall survival (OS) differences between clinical trial data from the KEYNOTE-010 trial and real-world data (RWD) from the Flatiron Health database in patients with programmed death ligand 1 (PD-L1)-expressing advanced non-small-cell lung cancer (NSCLC) who received second-line pembrolizumab monotherapy. METHODS Clinical trial data were from the randomized phase II/III KEYNOTE-010 trial that enrolled patients from August 28, 2013, to February 27, 2015. At data cutoff for KEYNOTE-010, the median survival follow-up time for pembrolizumab patients was 11.2 months. RWD were from Flatiron Health advanced NSCLC database and included patients who initiated second-line pembrolizumab from January 26, 2015, to February 28, 2019. At data cutoff for Flatiron, the median survival follow-up time for pembrolizumab-treated patients was 6.1 months. Clinical trial data from KEYNOTE-010 and RWD from Flatiron were analyzed without adjustment, with propensity adjustment, and filtered per the main KEYNOTE-010 eligibility criteria (EC) of histologically/cytologically confirmed PD-L1-positive NSCLC, Eastern Cooperative Oncology Group performance status of 0/1, no prior therapy with docetaxel for NSCLC, and laboratory values indicative of adequate organ function in addition to prior line of therapy requirements. RESULTS Among 243 patients from KEYNOTE-010 and 782 from Flatiron, median age was 63 v 68 years, and 64% v 54% were male, respectively. OS data from KEYNOTE-010 and Flatiron were similar without any adjustment (n = 782; hazard ratio [HR], 0.96; 95% CI, 0.80 to 1.15) and after both filtering and propensity adjustment (n = 221; HR, 0.99; 95% CI, 0.73 to 1.34). CONCLUSION Without any adjustment, as well as after applying similar EC and appropriate statistical methods, RWD demonstrated similar effectiveness for pembrolizumab in second-line NSCLC as observed in randomized clinical trials.
Collapse
Affiliation(s)
- Thomas Jemielita
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co, Inc, Kenilworth, NJ
| | - Xiaoyun Nicole Li
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co, Inc, Kenilworth, NJ
| | - Bilal Piperdi
- Oncology Clinical Development, Merck & Co, Inc, Kenilworth, NJ
| | - Wei Zhou
- Center for Observational and Real World Evidence (CORE), Merck & Co, Inc, Kenilworth, NJ
| | - Thomas Burke
- Center for Observational and Real World Evidence (CORE), Merck & Co, Inc, Kenilworth, NJ
| | - Cong Chen
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co, Inc, Kenilworth, NJ
| |
Collapse
|
42
|
Varnai P, Davé A, Farla K, Nooijen A, Petrosova L. The Evidence REVEAL Study: Exploring the Use of Real-World Evidence and Complex Clinical Trial Design by the European Pharmaceutical Industry. Clin Pharmacol Ther 2020; 110:1180-1189. [PMID: 33216976 PMCID: PMC8596609 DOI: 10.1002/cpt.2103] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/15/2020] [Indexed: 11/22/2022]
Abstract
The rapid evolution of science and technology allows innovative approaches to generate new types of evidence about the effectiveness of medical product development so as to speed up patients’ access to better diagnostics and treatment. Our study explored how two emerging approaches, the use of real‐world evidence (RWE) and complex clinical trial (CCT) design, are currently being used by the pharmaceutical industry to support premarketing authorization of medical product development and reviewed the international landscape for regulatory acceptance of such novel approaches. Combining evidence from a literature review, company survey, and interviews with international regulators and experts, we found that 80% of Europe‐based pharmaceutical companies have used RWE and 50% have used CCTs, in some capacity. Further, we present case examples of how companies are using these approaches and how international regulators are preparing for such developments. To conclude, we provide a set of recommendations for European industry and regulators to consider so that these novel approaches achieve their full potential within the EU regulatory system.
Collapse
|
43
|
Gini R, Sturkenboom MCJ, Sultana J, Cave A, Landi A, Pacurariu A, Roberto G, Schink T, Candore G, Slattery J, Trifirò G. Different Strategies to Execute Multi-Database Studies for Medicines Surveillance in Real-World Setting: A Reflection on the European Model. Clin Pharmacol Ther 2020; 108:228-235. [PMID: 32243569 PMCID: PMC7484985 DOI: 10.1002/cpt.1833] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Although postmarketing studies conducted in population-based databases often contain information on patients in the order of millions, they can still be underpowered if outcomes or exposure of interest is rare, or the interest is in subgroup effects. Combining several databases might provide the statistical power needed. A multi-database study (MDS) uses at least two healthcare databases, which are not linked with each other at an individual person level, with analyses carried out in parallel across each database applying a common study protocol. Although many MDSs have been performed in Europe in the past 10 years, there is a lack of clarity on the peculiarities and implications of the existing strategies to conduct them. In this review, we identify four strategies to execute MDSs, classified according to specific choices in the execution: (A) local analyses, where data are extracted and analyzed locally, with programs developed by each site; (B) sharing of raw data, where raw data are locally extracted and transferred without analysis to a central partner, where all the data are pooled and analyzed; (C) use of a common data model with study-specific data, where study-specific data are locally extracted, loaded into a common data model, and processed locally with centrally developed programs; and (D) use of general common data model, where all local data are extracted and loaded into a common data model, prior to and independent of any study protocol, and protocols are incorporated in centrally developed programs that run locally. We illustrate differences between strategies and analyze potential implications.
Collapse
Affiliation(s)
- Rona Gini
- Agenzia regionale di sanità della ToscanaFlorenceItaly
| | | | | | - Alison Cave
- European Medicines AgencyAmsterdamThe Netherlands
| | - Annalisa Landi
- Fondazione per la Ricerca Farmacologica Gianni Benzi OnlusValenzanoItaly
- Teddy European Network of Excellence for Paediatric Clinical ResearchPaviaItaly
| | | | | | - Tania Schink
- Leibniz Institute for Prevention Research and EpidemiologyBremenGermany
| | | | - Jim Slattery
- European Medicines AgencyAmsterdamThe Netherlands
| | - Gianluca Trifirò
- Department of Biomedical and Dental Sciences and Morphofunctional ImagingUniversità di MessinaMessinaItaly
| | | |
Collapse
|
44
|
van Laar SA, Gombert-Handoko KB, Guchelaar HJ, Zwaveling J. An Electronic Health Record Text Mining Tool to Collect Real-World Drug Treatment Outcomes: A Validation Study in Patients With Metastatic Renal Cell Carcinoma. Clin Pharmacol Ther 2020; 108:644-652. [PMID: 32575147 PMCID: PMC7484987 DOI: 10.1002/cpt.1966] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/15/2020] [Indexed: 12/28/2022]
Abstract
Real‐world evidence can close the inferential gap between marketing authorization studies and clinical practice. However, the current standard for real‐world data extraction from electronic health records (EHRs) for treatment evaluation is manual review (MR), which is time‐consuming and laborious. Clinical Data Collector (CDC) is a novel natural language processing and text mining software tool for both structured and unstructured EHR data and only shows relevant EHR sections improving efficiency. We investigated CDC as a real‐world data (RWD) collection method, through application of CDC queries for patient inclusion and information extraction on a cohort of patients with metastatic renal cell carcinoma (RCC) receiving systemic drug treatment. Baseline patient characteristics, disease characteristics, and treatment outcomes were extracted and these were compared with MR for validation. One hundred patients receiving 175 treatments were included using CDC, which corresponded to 99% with MR. Calculated median overall survival was 21.7 months (95% confidence interval (CI) 18.7–24.8) vs. 21.7 months (95% CI 18.6–24.8) and progression‐free survival 8.9 months (95% CI 5.4–12.4) vs. 7.6 months (95% CI 5.7–9.4) for CDC vs. MR, respectively. Highest F1‐score was found for cancer‐related variables (88.1–100), followed by comorbidities (71.5–90.4) and adverse drug events (53.3–74.5), with most diverse scores on international metastatic RCC database criteria (51.4–100). Mean data collection time was 12 minutes (CDC) vs. 86 minutes (MR). In conclusion, CDC is a promising tool for retrieving RWD from EHRs because the correct patient population can be identified as well as relevant outcome data, such as overall survival and progression‐free survival.
Collapse
Affiliation(s)
- Sylvia A van Laar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim B Gombert-Handoko
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Juliëtte Zwaveling
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
45
|
Real world data in the era of Immune Checkpoint Inhibitors (ICIs): Increasing evidence and future applications in lung cancer. Cancer Treat Rev 2020; 87:102031. [PMID: 32446182 DOI: 10.1016/j.ctrv.2020.102031] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022]
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed death 1 (PD-1) and PD-ligand 1 (PD-L1) quickly subverted the standard of treatment in Non-Small Cell Lung Cancer (NSCLC), where they were first introduced in all comers previously treated advanced/metastatic NSCLC patients and subsequently in the first line of PD-L1 selected cases of metastatic and locally advanced disease. Treatment algorithm is an evolving landscape, where the introduction of front-line ICIs, with or without chemotherapy, unavoidably influences the following treatment lines. In this context, medical oncologists are currently facing many unclear issues, which have been not clarified so far by available data. Effectiveness and safety in special populations underrepresented in clinical trials - such as elderly, poor PS, hepatitis or human immunodeficiency virus-affected patients - are only a part of the unexplored side of ICIs in the real world. Indeed, pivotal randomized clinical trials (RCTs) often lack of external validity because eligibility criteria exclude some patient subgroups commonly treated in real-world clinical practice. Similarly, cost-effectiveness and sustainability of these innovative agents are important issues to be considered in the real-world. Though affected by several limitations, real-world evidence (RWE) studies allow to collect data regarding overall treated patients in clinical practice according to local authority regulations, overcoming the intrinsic limits of RCTs. The present review focuses on RWE about ICIs in lung cancer treatment, with particular reference to special patient populations, and discusses potential application of real-world data in a potential innovative drug development model.
Collapse
|
46
|
Deluche E, Antoine A, Bachelot T, Lardy-Cleaud A, Dieras V, Brain E, Debled M, Jacot W, Mouret-Reynier MA, Goncalves A, Dalenc F, Patsouris A, Ferrero JM, Levy C, Lorgis V, Vanlemmens L, Lefeuvre-Plesse C, Mathoulin-Pelissier S, Petit T, Uwer L, Jouannaud C, Leheurteur M, Lacroix-Triki M, Courtinard C, Perol D, Robain M, Delaloge S. Contemporary outcomes of metastatic breast cancer among 22,000 women from the multicentre ESME cohort 2008-2016. Eur J Cancer 2020; 129:60-70. [PMID: 32135312 DOI: 10.1016/j.ejca.2020.01.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/26/2022]
Abstract
AIM Real-world data inform the outcome comparisons and help the development of new therapeutic strategies. To this end, we aimed to describe the full characteristics and outcomes in the Epidemiological Strategy and Medical Economics (ESME) cohort, a large national contemporary observational database of patients with metastatic breast cancer (MBC). METHODS Women aged ≥18 years with newly diagnosed MBC and who initiated MBC treatment between January 2008 and December 2016 in one of the 18 French Comprehensive Cancer Centers (N = 22,109) were included. We assessed the full patients' characteristics, first-line treatments, overall survival (OS) and first-line progression-free survival, as well as updated prognostic factors in the whole cohort and among the 3 major subtypes: hormone receptor positive and HER2-negative (HR+/HER2-, n = 13,656), HER2-positive (HER2+, n = 4017) and triple-negative (n = 2963) tumours. RESULTS The median OS of the whole cohort was 39.5 months (95% confidence interval [CI], 38.7-40.3). Five-year OS was 33.8%. OS differed significantly between the 3 subtypes (p < 0.0001) with a median OS of 43.3 (95% CI, 42.5-44.5) in HR+/HER2-; 50.1 (95% CI, 47.6-53.1) in HER2+; and 14.8 months (95% CI, 14.1-15.5) in triple-negative subgroups, respectively. Beyond performance status, the following variables had a constant significant negative prognostic impact on OS in the whole cohort and among subtypes: older age at diagnosis of metastases (except for the triple-negative subtype), metastasis-free interval between 6 and 24 months, presence of visceral metastases and number of metastatic sites ≥ 3. CONCLUSIONS The ESME program represents a unique large-scale real-life cohort on MBC. This study highlights important situations of high medical need within MBC patients. DATABASE REGISTRATION: clinicaltrials.gov Identifier NCT032753.
Collapse
Affiliation(s)
- Elise Deluche
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif, 94800, France; Department of Medical Oncology, CHU de Limoges, France
| | - Alison Antoine
- Department of Biostatistics, Centre Léon Bérard, 28 Promenade Léa et Napoléon Bullukian, Lyon, 69008, France
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, 28 Prom. Léa et Napoléon Bullukian, Lyon, 69008, France
| | - Audrey Lardy-Cleaud
- Department of Biostatistics, Centre Léon Bérard, 28 Promenade Léa et Napoléon Bullukian, Lyon, 69008, France
| | - Veronique Dieras
- Medical Oncology Department, Centre Eugéne Marquis, Avenue de La Bataille Flandres-Dunkerque, Rennes, 35000, France
| | - Etienne Brain
- Department of Medical Oncology, Institut Curie, 26 Rue D'Ulm, Paris & Saint-Cloud, 75005, France
| | - Marc Debled
- Department of Medical Oncology, Institut Bergonié, 229 Cours de L'Argonne, Bordeaux, 33000, France
| | - William Jacot
- Department of Medical Oncology, Institut Du Cancer de Montpellier, 208 Rue des Apothicaires, Montpellier, 34298, France
| | - Marie Ange Mouret-Reynier
- Department of Medical Oncology, Centre Jean Perrin, 58 Rue Montalembert, Clermont Ferrand, 63011, France
| | - Anthony Goncalves
- Department of Medical Oncology, Institut Paoli-Calmettes, 232 Boulevard de Sainte-Marguerite, Marseille, 13009, France
| | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius Regaud - IUCT Oncopole, 1 Avenue Irène-Joliot-Curie, Toulouse, 31059, France
| | - Anne Patsouris
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest Nantes & Angers, 15 Rue André Boquel, Angers, 49055, France
| | - Jean Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, 33 Avenue de Valambrose, Nice, 06189, France
| | - Christelle Levy
- Department of Medical Oncology, Centre François Baclesse, 3 Avenue Du Général Harris, Caen, 14000, France
| | - Veronique Lorgis
- Department of Medical Oncology, Institut de Cancérologie de Bourgogne, Dijon, 21079, France
| | - Laurence Vanlemmens
- Medical Oncology Department, Centre Oscar Lambret, 3 Rue Frédéric Combemale, Lille, 59000, France
| | - Claudia Lefeuvre-Plesse
- Medical Oncology Department, Centre Eugéne Marquis, Avenue de La Bataille Flandres-Dunkerque, Rennes, 35000, France
| | | | - Thierry Petit
- Department of Medical Oncology, Centre Paul Strauss, 3 Rue de La Porte de L'Hôpital, Strasbourg, 67000, France
| | - Lionel Uwer
- Medical Oncology Department, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy, 6 Avenue de Bourgogne, Vandœuvre-lès-Nancy, 54519, France
| | - Christelle Jouannaud
- Department of Medical Oncology, Institut de Cancérologie Jean-Godinot, 1 Rue Du Général Koenig, Reims, 51100, France
| | - Marianne Leheurteur
- Department of Medical Oncology, Centre Henri Becquerel, Rue D'Amiens, Rouen, 76000, France
| | - Magali Lacroix-Triki
- Department of BioPathology, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif, 94800, France
| | - Coralie Courtinard
- Department of Research and Development, R&D Unicancer, 101 Rue de Tolbiac, Paris, 75654, France
| | - David Perol
- Department of Biostatistics, Centre Léon Bérard, 28 Promenade Léa et Napoléon Bullukian, Lyon, 69008, France
| | - Mathieu Robain
- Department of Research and Development, R&D Unicancer, 101 Rue de Tolbiac, Paris, 75654, France
| | - Suzette Delaloge
- Department of Cancer Medicine, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif, 94800, France.
| |
Collapse
|
47
|
Moran M, Nickens D, Adcock K, Bennetts M, Charnley N, Fife K. Augmenting the randomized controlled trial with real-world data to aid clinical decision making in metastatic renal cell carcinoma: a systematic review and meta-analysis. Future Oncol 2019; 15:3987-4001. [DOI: 10.2217/fon-2019-0421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Aim: To evaluate how efficacy outcomes from real-world data (RWD) can support those from randomized controlled trials (RCTs), in the context of first-line tyrosine kinase inhibitor treatment of metastatic renal cell carcinoma. Patients & methods: PubMed, Ovid, MEDLINE and EMBASE were searched for RCTs and RWD studies with ≥50 adult patients per arm published in 2000–2017. Outcome measures were median progression-free survival, median overall survival and objective response rate. Results: A total of 13 RCTs and 22 RWD studies met eligibility criteria; 31, 28 and 25 studies, respectively, reported median progression-free survival, median overall survival and objective response rate. Summary outcome measures were similar in RWD and RCTs. Conclusion: RWD validates efficacy-based outcomes from RCTs and may provide supportive evidence to inform clinical decisions.
Collapse
Affiliation(s)
| | | | | | | | | | - Kate Fife
- Cambridge University Hospital, Cancer Services, Cambridge, UK
| |
Collapse
|
48
|
Kyr M, Polaskova K, Kuttnerova Z, Merta T, Neradil J, Berkovcova J, Horky O, Jezova M, Veselska R, Klement GL, Valik D, Sterba J. Individualization of Treatment Improves the Survival of Children With High-Risk Solid Tumors: Comparative Patient Series Analysis in a Real-Life Scenario. Front Oncol 2019; 9:644. [PMID: 31380281 PMCID: PMC6650566 DOI: 10.3389/fonc.2019.00644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/01/2019] [Indexed: 11/13/2022] Open
Abstract
Introduction: The individualization of treatment is attractive, especially in children with high-risk cancer. In such a rare and very heterogeneous group of diseases, large population-based clinical randomized trials are not feasible without international collaboration. We therefore propose comparative patient series analysis in a real-life scenario. Methods: Open cohort observational study, comparative analysis. Seventy patients with high-risk solid tumors diagnosed between 2003 and 2015 and in whom the treatment was individualized either empirically or based on biomarkers were analyzed. The heterogeneity of the cohort and repeated measurements were advantageously utilized to increase effective sample size using appropriate statistical tools. Results: We demonstrated a beneficial effect of empirically given low-dose metronomic chemotherapy (HR 0.46 for relapses, p = 0.017) as well as various repurposed or targeted agents (HR 0.15 for deaths, p = 0.004) in a real-life scenario. However, targeted agents given on the basis of limited biological information were not beneficial. Conclusions: Comparative patient series analysis provides institutional-level evidence for treatment individualization in high-risk pediatric malignancies. Our findings emphasize the need for a comprehensive, multi omics assessment of the tumor and the host as well whenever molecularly driven targeted therapies are being considered. Low-dose metronomic chemotherapy or local control of the disease may be a more rational option in situations where targeted treatment cannot be justified by robust evidence and comprehensive biological information. “Targeted drugs” may be given empirically with a realistic benefit expectation when based on robust rationale.
Collapse
Affiliation(s)
- Michal Kyr
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Kristyna Polaskova
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Zuzana Kuttnerova
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Tomas Merta
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| | - Jakub Neradil
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia.,Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czechia
| | - Jitka Berkovcova
- Laboratory of Molecular Pathology, Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ondrej Horky
- Laboratory of Molecular Pathology, Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia
| | - Renata Veselska
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia.,Laboratory of Tumor Biology, Department of Experimental Biology, School of Science, Masaryk University, Brno, Czechia
| | - Giannoula Lakka Klement
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,CSTS Health Care Inc., Toronto, ON, Canada
| | - Dalibor Valik
- Department of Laboratory Medicine, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and School of Medicine, Masaryk University, Brno, Czechia.,International Clinical Research Centre, St. Anne's University Hospital Brno, Brno, Czechia
| |
Collapse
|
49
|
Locati LD, Piovesan A, Durante C, Bregni M, Castagna MG, Zovato S, Giusti M, Ibrahim T, Puxeddu E, Fedele G, Pellegriti G, Rinaldi G, Giuffrida D, Verderame F, Bertolini F, Bergamini C, Nervo A, Grani G, Rizzati S, Morelli S, Puliafito I, Elisei R. Real-world efficacy and safety of lenvatinib: data from a compassionate use in the treatment of radioactive iodine-refractory differentiated thyroid cancer patients in Italy. Eur J Cancer 2019; 118:35-40. [PMID: 31299580 DOI: 10.1016/j.ejca.2019.05.031] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/22/2019] [Accepted: 05/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Lenvatinib is a multi-kinase inhibitor approved for patients with radioactive iodine (RAI)-resistant differentiated thyroid cancer (DTC). Before the drug approval from the Italian National Regulatory Agency, a compassionate use programme has been run in Italy. This retrospective study aimed to analyse data from the first series of patients treated with lenvatinib in Italy. METHODS The primary aim was to assess the response rate (RR) and progression-free survival (PFS). Secondary end-points include overall survival (OS) and toxicity data. RESULTS From November 2014 to September 2016, 94 patients were treated in 16 Italian sites. Seventeen percent of patients had one or more comorbidities, hypertension being the most common (60%). Ninety-eight percent of patients were treated by surgery, followed by RAI in 98% of cases. Sixty-four percent of patients received a previous systemic treatment. Lenvatinib was started at 24 mg in 64 subjects. Partial response and stable disease were observed in 36% and in 41% of subjects, respectively; progression was recorded in 14% of patients. Drug-related side-effects were common; the most common were fatigue (13.6%) and hypertension (11.6%). Overall, median PFS and OS were 10.8 months (95% confidence interval [CI], 7.7-12.6) and 23.8 months (95% CI, 19.7-25.0) respectively. CONCLUSION Lenvatinib is active and safe in unselected, RAI-refractory, progressive DTC patients in real-life setting. RR and PFS seem to be less favourable than those observed in the SELECT trial, likely due to a negative selection that included heavily pretreated patients or with poor performance status.
Collapse
Affiliation(s)
- L D Locati
- Head and Neck Medical Oncology; Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - A Piovesan
- Dept. Oncology, Oncological Endocrinology, A.O.U. Città della Salute e della Scienza, Ospedale Molinette, Torino, Italy.
| | - C Durante
- Dept Internal Medicine and Medical Specialties, Policlinico Umberto I, Roma, Italy.
| | - M Bregni
- Dept Medical Oncology, Ospedale Busto Arsizio-ASST Valle Olona, Busto Arsizio, Italy.
| | - M G Castagna
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy.
| | - S Zovato
- Familial Cancer Clinic and Oncoendocrinology Veneto Institute of Oncology IOV- IRCCS, Padua, Italy.
| | - M Giusti
- Dept Internal Medicine and Medical Specialties, Clinical Endocrinology, IRCCS San Martino Hospital, Genova, Italy.
| | - T Ibrahim
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy.
| | - E Puxeddu
- Department of Medicine, University of Perugia, Perugia, Italy
| | - G Fedele
- High Research Srl, Milano, Italy.
| | - G Pellegriti
- Endocrinology Division, Garibaldi Nesima Hospital, Catania, Italy.
| | - G Rinaldi
- Dept Surgical and Oncological Sciences, Policlinico Paolo Giaccone, Palermo, Italy.
| | - D Giuffrida
- Dept Medical Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Italy.
| | - F Verderame
- Dept Hematology and Oncology, Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy.
| | - F Bertolini
- Dept Oncology and Haematology, Modena University Hospital, Modena, Italy.
| | - C Bergamini
- Head and Neck Medical Oncology; Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - A Nervo
- Dept. Oncology, Oncological Endocrinology, A.O.U. Città della Salute e della Scienza, Ospedale Molinette, Torino, Italy.
| | - G Grani
- Dept Internal Medicine and Medical Specialties, Policlinico Umberto I, Roma, Italy.
| | - S Rizzati
- Familial Cancer Clinic and Oncoendocrinology Veneto Institute of Oncology IOV- IRCCS, Padua, Italy.
| | - S Morelli
- Department of Medicine, University of Perugia, Perugia, Italy.
| | - I Puliafito
- Dept Medical Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Italy.
| | - R Elisei
- Dept Clinical and Experimental Medicine, A.O Universitaria Pisana, Pisa, Italy.
| |
Collapse
|
50
|
Cave A, Kurz X, Arlett P. Real-World Data for Regulatory Decision Making: Challenges and Possible Solutions for Europe. Clin Pharmacol Ther 2019; 106:36-39. [PMID: 30970161 PMCID: PMC6617710 DOI: 10.1002/cpt.1426] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Alison Cave
- Pharmacovigilance and Epidemiology DepartmentEuropean Medicines AgencyAmsterdamThe Netherlands
| | - Xavier Kurz
- Pharmacovigilance and Epidemiology DepartmentEuropean Medicines AgencyAmsterdamThe Netherlands
| | - Peter Arlett
- Pharmacovigilance and Epidemiology DepartmentEuropean Medicines AgencyAmsterdamThe Netherlands
| |
Collapse
|