1
|
Trin K, Dalleau C, Mathoulin-Pelissier S, Le Tourneau C, Dinart D, Bellera C. The Growth Modulation Index (GMI) as an Efficacy Outcome in Cancer Clinical Trials: A Scoping Review with Suggested Reporting Guidelines. Curr Oncol Rep 2025:10.1007/s11912-025-01667-1. [PMID: 40156702 DOI: 10.1007/s11912-025-01667-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE OF REVIEW The growth modulation index (GMI) is defined as the ratio between the time to progression of a new line of treatment and the previous line. This ratio can be used to determine whether the new line of treatment brings a clinical benefit. It has been proposed as an outcome in trials evaluating non-cytotoxic drugs. Its interest lies in the intra-patient comparison. The terminology employed to refer to the GMI, as well as its definitions, are highly variable in the literature. Some uses of the GMI are arbitrary and not based on any scientific rationale. Our aim is to describe how the GMI is reported in the scientific literature. RECENT FINDINGS We carried out a scoping review using PubMed, Scopus, Web of Science and BASE (Bielefeld Academic Search Engine). The algorithm was composed of the terms "growth modulation index", "time to progression ratio" and "progression-free survival ratio". Documents in English, with full-text available, published up to 2023, were included. Among 227 included documents, 166 of which discussed GMI specifically. On these 166 documents, 76 reported on observational studies, 62 on interventional studies and 17 on methodological or statistical developments pertaining to the GMI. All were about oncology. Our review highlights significant variability in the reporting and use of the GMI. To address this, we propose standardized reporting guidelines. Additionally, we emphasize the need for methodological and statistical developments to improve the use of the GMI and to develop novel GMI-based trial designs.
Collapse
Affiliation(s)
- Kilian Trin
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France.
- Medical Science Faculty, University of Bordeaux, Bordeaux, France.
| | - Cynthia Dalleau
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Simone Mathoulin-Pelissier
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris, France
- INSERM U900 Research Unit, Institut Curie, Paris, France
- Paris-Saclay University, Paris, France
| | - Derek Dinart
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| | - Carine Bellera
- INSERM CIC-1401, Clinical and Epidemiological Research Unit, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- ISPED, Centre INSERM U1219 Bordeaux Population Health, Epicene Team, University of Bordeaux, Bordeaux, France
| |
Collapse
|
2
|
Tang J, Karbhari N, Campian JL. Therapeutic Targets in Glioblastoma: Molecular Pathways, Emerging Strategies, and Future Directions. Cells 2025; 14:494. [PMID: 40214448 PMCID: PMC11988183 DOI: 10.3390/cells14070494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, characterized by rapid growth, invasive infiltration into surrounding brain tissue, and resistance to conventional therapies. Despite advancements in surgery, radiotherapy, and chemotherapy, median survival remains approximately 15 months, underscoring the urgent need for innovative treatments. Key considerations informing treatment development include oncogenic genetic and epigenetic alterations that may dually serve as therapeutic targets and facilitate treatment resistance. Various immunotherapeutic strategies have been explored and continue to be refined for their anti-tumor potential. Technical aspects of drug delivery and blood-brain barrier (BBB) penetration have been addressed through novel vehicles and techniques including the incorporation of nanotechnology. Molecular profiling has emerged as an important tool to individualize treatment where applicable, and to identify patient populations with the most drug sensitivity. The goal of this review is to describe the spectrum of potential GBM therapeutic targets, and to provide an overview of key trial outcomes. Altogether, the progress of clinical and preclinical work must be critically evaluated in order to develop therapies for GBM with the strongest therapeutic efficacy.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Nishika Karbhari
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (N.K.); (J.L.C.)
| |
Collapse
|
3
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
4
|
Čerina Pavlinović D, Šuto Pavičić J, Njavro A, Librenjak N, Tomaš I, Šeparović R, Pleština S, Bajić Ž, Dedić Plavetić N, Vrdoljak E. Precision Oncology in Clinical Practice: Two Years of Comprehensive Genomic Profiling in Croatia. J Pers Med 2025; 15:59. [PMID: 39997336 PMCID: PMC11856208 DOI: 10.3390/jpm15020059] [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: 01/07/2025] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Background: The widespread adoption of precision medicine in routine cancer care remains a critical challenge, even as advanced technologies expand and personalized therapies demonstrate remarkable success in certain cancer types. While breakthrough innovations in targeted treatments have revolutionized outcomes for specific cancers, translating these scientific advances into standard clinical practice continues to be an evolving and complex endeavor. Croatia has a nationwide project of precision oncology through the comprehensive genomic profiling (CGP) analysis. Since collecting and analyzing real-world data is crucial for clinical research and defining the value of CGP in precision oncology, we aimed to present the data from everyday clinical practice given the opportunities and challenges we faced. Methods: This was a retrospective observational study conducted at the national level in all patients whose tumor samples were subjected to CGP between 1 January 2020 and 31 December 2021. Results: In total, 481 patients with CGP results were included in this study. Gastrointestinal and reproductive malignancies were the most common, accounting for 29.1% and 28.9% of all tested tumors, respectively. Specifically, colorectal tumors made up 19.1% of cases, while uterine tumors represented 11.2%. At least one clinically relevant genomic alteration was found in 76.7% of patients, with the KRAS mutation (27.2%) being the most common. During the two-year study period, 26,709 individuals lost their lives to cancer in Croatia. Combining this with the CGP selection criteria valid at the time, there was an estimated population of approximately 13,350 potentially eligible patients for the CGP analysis, meaning that only 3.6% of potentially eligible patients were tested. Conclusions: The analysis identified clinically actionable genomic alterations in approximately 80% of the evaluated patients, suggesting they could be candidates for targeted therapeutic interventions. The adoption of CGP remains limited, with estimates indicating that under 5% of metastatic cancer patients received testing in the initial two-year implementation period, despite established national insurance coverage guidelines. This low utilization rate suggests a significant gap in access to genomic testing, leaving many eligible cancer patients without the potential benefits of this diagnostic approach.
Collapse
Affiliation(s)
- Dora Čerina Pavlinović
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| | - Jelena Šuto Pavičić
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
| | - Antonela Njavro
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, 10 000 Zagreb, Croatia;
| | - Nikša Librenjak
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
| | - Ilijan Tomaš
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31 000 Osijek, Croatia;
| | - Robert Šeparović
- Department of Medical Oncology, University Hospital for Tumor, Sestre Milosrdnice, 10 000 Zagreb, Croatia;
| | - Stjepko Pleština
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Žarko Bajić
- Research Unit “Dr. Mirko Grmek”, Psychiatric Clinic Sveti Ivan, 10 000 Zagreb, Croatia;
| | - Natalija Dedić Plavetić
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| |
Collapse
|
5
|
Mapendano CK, Nøhr AK, Sønderkær M, Pagh A, Carus A, Lörincz T, Haslund CA, Poulsen LØ, Ernst A, Bødker JS, Dahl SC, Sunde L, Brügmann AH, Vesteghem C, Pedersen IS, Ladekarl M. Longer survival with precision medicine in late-stage cancer patients. ESMO Open 2025; 10:104089. [PMID: 39754975 PMCID: PMC11758131 DOI: 10.1016/j.esmoop.2024.104089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND In a per-protocol analysis of molecularly profiled patients with treatment-refractory, end-stage cancer discussed at the National Molecular Tumor Board (NMTB), we aimed to assess the overall survival (OS) outcome of targeted treatment compared with no targeted treatment. MATERIALS AND METHODS Patients were prospectively included at a single oncological center. Whole exome and RNA sequencing (tumor-normal) were carried out, and cases were presented at the NMTB for discussion of targeted treatment. Treatment was available through a basket trial, by compassionate use or in early clinical trials. RESULTS One hundred and ninety-six patients were included from 2020 to 2023. In all but three patients a driver variant was disclosed, while 42% had simultaneous affection of more than three oncogenic pathways. In 42% of patients a druggable target was identified but two-thirds did not receive the suggested treatment. The fraction of patients initiating treatment yearly rose from 8% to 22%. For patients treated (N = 30), the clinical benefit rate was 44% and median time on treatment was 3.5 months. Druggable targets were enriched in lung cancers, while patients receiving or not receiving targeted treatment had similar clinical characteristics. The median OS was longer for patients receiving targeted treatment (15 months), but similar for patients with no druggable target and suggested targeted treatment not initiated (5 and 6 months, respectively) (P = 0.004). In multivariate analysis, targeted treatment (hazard ratio 0.43, confidence interval 0.25-0.72), few metastatic sites, and adenocarcinoma histology were predictive of improved OS while alterations of the RTK/RAS pathway were prognostically unfavorable. CONCLUSIONS Tissue-agnostic targeted treatment based on molecular tumor profiling is possible in an increasing fraction of end-stage cancer patients. In those who receive targeted treatment, results strongly suggest a significant survival benefit.
Collapse
Affiliation(s)
- C K Mapendano
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A K Nøhr
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | - M Sønderkær
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - A Pagh
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A Carus
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - T Lörincz
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - C A Haslund
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - L Ø Poulsen
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - A Ernst
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - J S Bødker
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - S C Dahl
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark
| | - L Sunde
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Clinical Genetics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - A H Brügmann
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - C Vesteghem
- Center for Clinical Data Science, Aalborg University and Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - I S Pedersen
- Molecular Diagnostics and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - M Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
6
|
Chhabra R. Molecular and modular intricacies of precision oncology. Front Immunol 2024; 15:1476494. [PMID: 39507541 PMCID: PMC11537923 DOI: 10.3389/fimmu.2024.1476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Precision medicine is revolutionizing the world in combating different disease modalities, including cancer. The concept of personalized treatments is not new, but modeling it into a reality has faced various limitations. The last decade has seen significant improvements in incorporating several novel tools, scientific innovations and governmental support in precision oncology. However, the socio-economic factors and risk-benefit analyses are important considerations. This mini review includes a summary of some commendable milestones, which are not just a series of successes, but also a cautious outlook to the challenges and practical implications of the advancing techno-medical era.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Business Department, Biocytogen Boston Corporation, Waltham, MA, United States
| |
Collapse
|
7
|
Widmer N, Guidi M, Buclin T. Population Pharmacokinetics in Oncology and Its Clinical Applications. Pharmaceutics 2024; 16:711. [PMID: 38931835 PMCID: PMC11207012 DOI: 10.3390/pharmaceutics16060711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Most traditional cytotoxic drugs are characterized by steep dose-response relationships and narrow therapeutic windows [...].
Collapse
Affiliation(s)
- Nicolas Widmer
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (M.G.); (T.B.)
- Pharmacy of the Eastern Vaud Hospitals, 1847 Rennaz, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, 1205 Geneva, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (M.G.); (T.B.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, 1205 Geneva, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Thierry Buclin
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland; (M.G.); (T.B.)
| |
Collapse
|
8
|
Hashimoto T, Nakamura Y, Oki E, Kobayashi S, Yuda J, Shibuki T, Bando H, Yoshino T. Bridging horizons beyond CIRCULATE-Japan: a new paradigm in molecular residual disease detection via whole genome sequencing-based circulating tumor DNA assay. Int J Clin Oncol 2024; 29:495-511. [PMID: 38551727 PMCID: PMC11043144 DOI: 10.1007/s10147-024-02493-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/26/2024]
Abstract
Circulating tumor DNA (ctDNA) is the fraction of cell-free DNA in patient blood that originates from a tumor. Advances in DNA sequencing technologies and our understanding of the molecular biology of tumors have increased interest in exploiting ctDNA to facilitate detection of molecular residual disease (MRD). Analysis of ctDNA as a promising MRD biomarker of solid malignancies has a central role in precision medicine initiatives exemplified by our CIRCULATE-Japan project involving patients with resectable colorectal cancer. Notably, the project underscores the prognostic significance of the ctDNA status at 4 weeks post-surgery and its correlation to adjuvant therapy efficacy at interim analysis. This substantiates the hypothesis that MRD is a critical prognostic indicator of relapse in patients with colorectal cancer. Despite remarkable advancements, challenges endure, primarily attributable to the exceedingly low ctDNA concentration in peripheral blood, particularly in scenarios involving low tumor shedding and the intrinsic error rates of current sequencing technologies. These complications necessitate more sensitive and sophisticated assays to verify the clinical utility of MRD across all solid tumors. Whole genome sequencing (WGS)-based tumor-informed MRD assays have recently demonstrated the ability to detect ctDNA in the parts-per-million range. This review delineates the current landscape of MRD assays, highlighting WGS-based approaches as the forefront technique in ctDNA analysis. Additionally, it introduces our upcoming endeavor, WGS-based pan-cancer MRD detection via ctDNA, in our forthcoming project, SCRUM-Japan MONSTAR-SCREEN-3.
Collapse
Affiliation(s)
- Tadayoshi Hashimoto
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Taro Shibuki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
9
|
Fountzilas E, Tsimberidou AM, Hiep Vo H, Kurzrock R. Tumor-agnostic baskets to N-of-1 platform trials and real-world data: Transforming precision oncology clinical trial design. Cancer Treat Rev 2024; 125:102703. [PMID: 38484408 DOI: 10.1016/j.ctrv.2024.102703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
Choosing the right drug(s) for the right patient via advanced genomic sequencing and multi-omic interrogation is the sine qua non of precision cancer medicine. Traditional cancer clinical trial designs follow well-defined protocols to evaluate the efficacy of new therapies in patient groups, usually identified by their histology/tissue of origin of their malignancy. In contrast, precision medicine seeks to optimize benefit in individual patients, i.e., to define who benefits rather than determine whether the overall group benefits. Since cancer is a disease driven by molecular alterations, innovative trial designs, including biomarker-defined tumor-agnostic basket trials, are driving ground-breaking regulatory approvals and deployment of gene- and immune-targeted drugs. Molecular interrogation further reveals the disruptive reality that advanced cancers are extraordinarily complex and individually distinct. Therefore, optimized treatment often requires drug combinations and N-of-1 customization, addressed by a new generation of N-of-1 trials. Real-world data and structured master registry trials are also providing massive datasets that are further fueling a transformation in oncology. Finally, machine learning is facilitating rapid discovery, and it is plausible that high-throughput computing, in silico modeling, and 3-dimensional printing may be exploitable in the near future to discover and design customized drugs in real time.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, St Luke's Clinic, Thessaloniki, Greece; European University Cyprus, German Oncology Center, Nicosia, Cyprus
| | - Apostolia-Maria Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA.
| | - Henry Hiep Vo
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| | - Razelle Kurzrock
- WIN Consortium for Precision Medicine, France; Medical College of Wisconsin, USA
| |
Collapse
|
10
|
de Jager VD, Timens W, Bayle A, Botling J, Brcic L, Büttner R, Fernandes MGO, Havel L, Hochmair M, Hofman P, Janssens A, van Kempen L, Kern I, Machado JC, Mohorčič K, Popat S, Ryška A, Wolf J, Schuuring E, van der Wekken AJ. Future perspective for the application of predictive biomarker testing in advanced stage non-small cell lung cancer. THE LANCET REGIONAL HEALTH. EUROPE 2024; 38:100839. [PMID: 38476751 PMCID: PMC10928270 DOI: 10.1016/j.lanepe.2024.100839] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/16/2023] [Accepted: 01/08/2024] [Indexed: 03/14/2024]
Abstract
For patients with advanced stage non-small cell lung cancer (NSCLC), treatment strategies have changed significantly due to the introduction of targeted therapies and immunotherapy. In the last few years, we have seen an explosive growth of newly introduced targeted therapies in oncology and this development is expected to continue in the future. Besides primary targetable aberrations, emerging diagnostic biomarkers also include relevant co-occurring mutations and resistance mechanisms involved in disease progression, that have impact on optimal treatment management. To accommodate testing of pending biomarkers, it is necessary to establish routine large-panel next-generation sequencing (NGS) for all patients with advanced stage NSCLC. For cost-effectiveness and accessibility, it is recommended to implement predictive molecular testing using large-panel NGS in a dedicated, centralized expert laboratory within a regional oncology network. The central molecular testing center should host a regional Molecular Tumor Board and function as a hub for interpretation of rare and complex testing results and clinical decision-making.
Collapse
Affiliation(s)
- Vincent D. de Jager
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arnaud Bayle
- Oncostat U1018, Inserm, Paris-Saclay University, Gustave Roussy, Villejuif, France
| | - Johan Botling
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy of University of Gothenburg, Gothenburg, Sweden
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Reinhard Büttner
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | | | - Libor Havel
- Charles University and Thomayer Hospital, Prague, Czech Republic
| | - Maximilian Hochmair
- Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Vienna, Austria
| | - Paul Hofman
- IHU RespirERA, FHU OncoAge, Nice University Hospital, Côte d’Azur University, Nice, France
| | - Annelies Janssens
- Department of Oncology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Léon van Kempen
- Department of Pathology, University Hospital Antwerp, University of Antwerp, Edegem, Belgium
| | - Izidor Kern
- Laboratory for Cytology and Pathology, University Clinic Golnik, Golnik, Slovenia
| | - José Carlos Machado
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Faculty of Medicine of the University of Porto, Portugal
- Institute for Research and Innovation in Health (i3S), Porto, Portugal
| | - Katja Mohorčič
- University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - Sanjay Popat
- Lung Unit, Royal Marsden NHS Trust, London, England, UK
| | - Aleš Ryška
- The Fingerland Department of Pathology, Charles University Medical Faculty and University Hospital, Czech Republic
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Cologne/Bonn, University Hospital Cologne, Cologne, Germany
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anthonie J. van der Wekken
- Department of Pulmonary Diseases and Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
11
|
Ahmed J, Das B, Shin S, Chen A. Challenges and Future Directions in the Management of Tumor Mutational Burden-High (TMB-H) Advanced Solid Malignancies. Cancers (Basel) 2023; 15:5841. [PMID: 38136385 PMCID: PMC10741991 DOI: 10.3390/cancers15245841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
A standardized assessment of Tumor Mutational Burden (TMB) poses challenges across diverse tumor histologies, treatment modalities, and testing platforms, requiring careful consideration to ensure consistency and reproducibility. Despite clinical trials demonstrating favorable responses to immune checkpoint inhibitors (ICIs), not all patients with elevated TMB exhibit benefits, and certain tumors with a normal TMB may respond to ICIs. Therefore, a comprehensive understanding of the intricate interplay between TMB and the tumor microenvironment, as well as genomic features, is crucial to refine its predictive value. Bioinformatics advancements hold potential to improve the precision and cost-effectiveness of TMB assessments, addressing existing challenges. Similarly, integrating TMB with other biomarkers and employing comprehensive, multiomics approaches could further enhance its predictive value. Ongoing collaborative endeavors in research, standardization, and clinical validation are pivotal in harnessing the full potential of TMB as a biomarker in the clinic settings.
Collapse
Affiliation(s)
- Jibran Ahmed
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Sarah Shin
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Alice Chen
- Developmental Therapeutics Clinic (DTC), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Tsimberidou AM, Kahle M, Vo HH, Baysal MA, Johnson A, Meric-Bernstam F. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol 2023; 20:843-863. [PMID: 37845306 DOI: 10.1038/s41571-023-00824-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Over the past 15 years, rapid progress has been made in developmental therapeutics, especially regarding the use of matched targeted therapies against specific oncogenic molecular alterations across cancer types. Molecular tumour boards (MTBs) are panels of expert physicians, scientists, health-care providers and patient advocates who review and interpret molecular-profiling results for individual patients with cancer and match each patient to available therapies, which can include investigational drugs. Interpretation of the molecular alterations found in each patient is a complicated task that requires an understanding of their contextual functional effects and their correlations with sensitivity or resistance to specific treatments. The criteria for determining the actionability of molecular alterations and selecting matched treatments are constantly evolving. Therefore, MTBs have an increasingly necessary role in optimizing the allocation of biomarker-directed therapies and the implementation of precision oncology. Ultimately, increased MTB availability, accessibility and performance are likely to improve patient care. The challenges faced by MTBs are increasing, owing to the plethora of identifiable molecular alterations and immune markers in tumours of individual patients and their evolving clinical significance as more and more data on patient outcomes and results from clinical trials become available. Beyond next-generation sequencing, broader biomarker analyses can provide useful information. However, greater funding, resources and expertise are needed to ensure the sustainability of MTBs and expand their outreach to underserved populations. Harmonization between practice and policy will be required to optimally implement precision oncology. Herein, we discuss the evolving role of MTBs and current and future considerations for their use in precision oncology.
Collapse
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michael Kahle
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Henry Hiep Vo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber Johnson
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Kokkali S, Georgaki E, Mandrakis G, Valverde C, Theocharis S. Genomic Profiling and Clinical Outcomes of Targeted Therapies in Adult Patients with Soft Tissue Sarcomas. Cells 2023; 12:2632. [PMID: 37998367 PMCID: PMC10670373 DOI: 10.3390/cells12222632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Genomic profiling has improved our understanding of the pathogenesis of different cancers and led to the development of several targeted therapies, especially in epithelial tumors. In this review, we focus on the clinical utility of next-generation sequencing (NGS) to inform therapeutics in soft tissue sarcoma (STS). The role of NGS is still controversial in patients with sarcoma, given the low mutational burden and the lack of recurrent targetable alterations in most of the sarcoma histotypes. The clinical impact of genomic profiling in STS has not been investigated prospectively. A limited number of retrospective, mainly single-institution, studies have addressed this issue using various NGS technologies and platforms and a variety of criteria to define a genomic alteration as actionable. Despite the detailed reports on the different gene mutations, fusions, or amplifications that were detected, data on the use and efficacy of targeted treatment are very scarce at present. With the exception of gastrointestinal stromal tumors (GISTs), these targeted therapies are administered either through off-label prescription of an approved drug or enrollment in a matched clinical trial. Based mainly on anecdotal reports, the outcome of targeted therapies in the different STS histotypes is discussed. Prospective studies are warranted to assess whether genomic profiling improves the management of STS patients.
Collapse
Affiliation(s)
- Stefania Kokkali
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
- Oncology Unit, 2nd Department of Medicine, Medical School, Hippocratio General Hospital of Athens, National and Kapodistrian University of Athens, V. Sofias 114, 11527 Athens, Greece;
| | - Eleni Georgaki
- Oncology Unit, 2nd Department of Medicine, Medical School, Hippocratio General Hospital of Athens, National and Kapodistrian University of Athens, V. Sofias 114, 11527 Athens, Greece;
| | - Georgios Mandrakis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| | - Claudia Valverde
- Medical Oncology Department, Vall d’Hebron University Hospital, Pg. Vall d’Hebron 119-12, 08035 Barcelona, Spain;
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece;
| |
Collapse
|