1
|
Ettienne EB, Grant-Kels JM, Striano P, Russo E, Neubauer D, Rose K. Melanoma and pediatric drug development: clinical progress vs. regulatory activism in minors - a narrative review. Expert Opin Pharmacother 2025; 26:595-603. [PMID: 40035212 DOI: 10.1080/14656566.2025.2475184] [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: 10/07/2024] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Melanoma is the malignancy where, in recent years, drug treatment has massively improved prognosis and quality of life. Based on genetic analysis, we differentiate today melanomas caused by cumulative solar damage (CSD) from others. AREAS COVERED Conventional CSD-caused melanomas affect predominantly adults and occasionally adolescents. Spitz melanoma and melanoma arising in congenital nevi, the two other pediatric melanoma types, are not CSD-caused, are genetically different, have different prognoses, and need different treatment. In contrast to the improved situation in adult melanoma, regulatory demand for pediatric labels in minors has resulted in pointless and harmful studies and has obfuscated diagnosis and treatment. Modern communication facilitates a worldwide analysis of extremely rare diseases such as melanoma in minors. Regulatory demand for on-label treatment only is demonstrably wrong for pediatric melanomas. They are too rare for randomized controlled trials but nevertheless deserve effective treatment. EXPERT OPINION Adolescents with conventional, CSD-caused melanoma should be treated as adults. Their bodies are already mature. For other childhood melanomas, registries and consultations with clinical specialists are better options than dogmatically demanded regulatory studies.
Collapse
Affiliation(s)
| | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT, USA
| | | | - Emilio Russo
- Pharmacology, University of Magna Graecia, Catanzaro, Italy
| | - David Neubauer
- Department of Child, Adolescent & Developmental Neurology, University Childrens' Hospital, Ljubljana, Slovenia
| | - Klaus Rose
- klausrose Consulting, Riehen, Switzerland
| |
Collapse
|
2
|
Chen D. Targeted Therapy Evolution from Defining a Sub-population to Crossing Multi-indications. Adv Pharm Bull 2024; 14:737-744. [PMID: 40190666 PMCID: PMC11970494 DOI: 10.34172/apb.43306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/12/2024] [Accepted: 09/08/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose It tends not only to shed lights on an emerging classification framework of disease according to the shared molecular pathogenesis across various organs/tissues, but also to inspire more efficient paradigms of pharmaceutic innovation in a broader medical perspective. Methods Literature review and re-thinking. Results This article has sorted out an updated profile of the outstanding targeted medications with an extending list of clinical indications in oncology and beyond. Conclusion Pharmaceutic development can be processed in a less risky and more affordable manner through drug repurpose or tissue agnostic approval.
Collapse
Affiliation(s)
- Daohong Chen
- Research Institute, Changshan Biochemical Pharmaceutical, North Head of Yinchuan Street, Zhengding New District, Shijiazhuang, Hebei, China, 050800
| |
Collapse
|
3
|
Valenti F, Ganci F, Sacconi A, Lo Sardo F, D'Andrea M, Sanguineti G, Di Agostino S. Polo-like kinase 2 targeting as novel strategy to sensitize mutant p53-expressing tumor cells to anticancer treatments. J Mol Med (Berl) 2024; 102:1485-1501. [PMID: 39480521 DOI: 10.1007/s00109-024-02499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Polo-like kinase 2 (Plk2) belongs to a family of serine/threonine kinases, and it is involved in tumorigenesis of diverse kind of tissues. We previously reported that Plk2 gene was a transcriptional target of the mutant p53/NF-Y oncogenic complex. Plk2 protein can bind to and phosphorylate mutant p53 triggering an oncogenic autoregulatory feedback loop involved in cancer cell proliferation and chemoresistance. In this study, we aimed to assess whether the specific inhibition of Plk2 kinase activity by the selective TC-S 7005 inhibitor could decrease cell proliferation and migration inhibiting mutant p53 phosphorylation, thus disarming its oncogenic potential. We found that the Plk2 inhibitor treatment sensitized the cells to the irradiation and chemotherapy drugs, thereby overcoming the mutant p53-dependent chemoresistance. Taken together, we provided results that Plk2 could be considered a tractable pharmacological target for cancers expressing mutant p53 proteins. The combined treatment with conventional chemotherapeutic drugs and Plk2 inhibitors may represent a new candidate intervention approach, which may be considered for improving tumor cell sensitivity to DNA damaging drugs. KEY MESSAGES : Missense mutations are present in the TP53 gene in about half of all human cancers and correlate with poor patient outcome. Mutant p53 proteins exert gain of function (GOF) activities in tumor cells such as increased proliferation, genomic instability and resistance to therapies. Polo-like kinase 2 (PLK2) binds and phosphorylates mutant p53 protein strengthening its GOF activities. Pharmacologically targeting PLK2 weakens mutant p53 proteins and sensitizes tumor cells to therapeutic treatments.
Collapse
Affiliation(s)
- Fabio Valenti
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Federica Ganci
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Andrea Sacconi
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Federica Lo Sardo
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Marco D'Andrea
- Laboratory of Medical Physics and Expert Systems, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Di Agostino
- Department of Health Sciences, Magna Græcia University of Catanzaro, 88100, Catanzaro, Italy.
| |
Collapse
|
4
|
Bazhenova L, Kim DW, Cho BC, Goel S, Heist R, Werner TL, Eaton KD, Wang JS, Pant S, Adkins DR, Blakely CM, Yan X, Neuteboom S, Christensen JG, Chao R, Bauer T. Sitravatinib in patients with solid tumors selected by molecular alterations: results from a Phase Ib study. Future Oncol 2024; 20:3213-3227. [PMID: 39513224 DOI: 10.1080/14796694.2024.2418285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Aim: We report clinical activity and safety of sitravatinib in patients with advanced cancer from basket cohorts with specific molecular alterations, in a Phase Ib study.Materials & methods: Patients with advanced solid tumors harboring amplification, mutation, or rearrangement of MET, AXL, RET, NTRK, DDR2, KDR, PDGFRA, KIT or CBL received sitravatinib once daily. Primary end point was confirmed objective response rate (ORR).Results: In total, 113 patients were enrolled following a median of 3 (range 1-18) prior systemic regimens. Altered RET (n = 31), CBL (n = 31) and MET (n = 17) were most frequent cohorts. Overall, 68.9% had reduced tumor volume and most (61.5%) had a best objective response of stable disease. ORR was highest in patients with RET-rearranged non-small cell lung cancer (21.1%) but did not differ significantly from the null hypothesis (ORR ≤15%; p = 0.316). Median progression-free survival and overall survival (5.7 and 24.2 months, respectively) were also longest in the RET-rearranged non-small cell lung cancer cohort. Diarrhea (61.1%), fatigue (50.4%) and hypertension (46.9%) were the most frequent treatment-emergent adverse events. Most treatment-emergent adverse events were mild-to-moderate in severity. The study closed before the planned number of patients were enrolled in all cohorts.Conclusion: Sitravatinib had a manageable safety profile with modest signals of clinical activity in patients with molecularly selected solid tumors.Clinical trial registration: www.clinicaltrials.gov identifier is NCT02219711.
Collapse
Affiliation(s)
| | - Dong-Wan Kim
- Seoul National University College of Medicine & Seoul National University Hospital, Seoul, Republic of Korea
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sanjay Goel
- Montefiore Medical Center, Bronx, NY 10467, USA
| | - Rebecca Heist
- Massachusetts General Hospital, Boston, MA 02114, USA
| | - Theresa L Werner
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Keith D Eaton
- University of Washington & Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Judy S Wang
- Florida Cancer Specialists & Research Institute, Sarasota, FL 34232, USA
| | - Shubham Pant
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Douglas R Adkins
- Washington University School of Medicine in St Louis, MO 63110, USA
| | | | - Xiaohong Yan
- Mirati Therapeutics Inc., San Diego, CA 92121, USA
| | | | | | - Richard Chao
- Mirati Therapeutics Inc., San Diego, CA 92121, USA
| | - Todd Bauer
- Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN 37203, USA
| |
Collapse
|
5
|
Gonzalez R, Saha A, Campbell CJ, Nejat P, Lokker C, Norgan AP. Seeing the random forest through the decision trees. Supporting learning health systems from histopathology with machine learning models: Challenges and opportunities. J Pathol Inform 2024; 15:100347. [PMID: 38162950 PMCID: PMC10755052 DOI: 10.1016/j.jpi.2023.100347] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 01/03/2024] Open
Abstract
This paper discusses some overlooked challenges faced when working with machine learning models for histopathology and presents a novel opportunity to support "Learning Health Systems" with them. Initially, the authors elaborate on these challenges after separating them according to their mitigation strategies: those that need innovative approaches, time, or future technological capabilities and those that require a conceptual reappraisal from a critical perspective. Then, a novel opportunity to support "Learning Health Systems" by integrating hidden information extracted by ML models from digitalized histopathology slides with other healthcare big data is presented.
Collapse
Affiliation(s)
- Ricardo Gonzalez
- DeGroote School of Business, McMaster University, Hamilton, Ontario, Canada
- Division of Computational Pathology and Artificial Intelligence, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Ashirbani Saha
- Department of Oncology, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Escarpment Cancer Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Clinton J.V. Campbell
- William Osler Health System, Brampton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Peyman Nejat
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Cynthia Lokker
- Health Information Research Unit, Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Andrew P. Norgan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Nguyen K, Fama K, Mercado G, Myat Y, Thein K. Histology Agnostic Drug Development: An Updated Review. Cancers (Basel) 2024; 16:3642. [PMID: 39518080 PMCID: PMC11544807 DOI: 10.3390/cancers16213642] [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: 09/07/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Recent advancements in oncology have led to the development of histology-agnostic therapies, which target genetic alterations irrespective of the tumor's tissue of origin. This review aimed to provide a comprehensive update on the current state of histology-agnostic drug development, focusing on key therapies, including pembrolizumab, larotrectinib, entrectinib, dostarlimab, dabrafenib plus trametinib, selpercatinib, trastuzumab deruxtecan, and reprotrectinib. We performed a detailed analysis of each therapy's mechanism of action, clinical trial outcomes, and associated biomarkers. The review further explores challenges in drug resistance, such as adaptive signaling pathways and neoantigen variability, as well as diagnostic limitations in identifying optimal patient populations. While these therapies have demonstrated efficacy in various malignancies, significant hurdles remain, including intratumoral heterogeneity and resistance mechanisms that diminish treatment effectiveness. We propose considerations for refining trial designs and emerging biomarkers, such as tumor neoantigen burden, to enhance patient selection. These findings illustrate the transformative potential of histology-agnostic therapies in precision oncology but highlight the need for continued research to optimize their use and overcome existing barriers.
Collapse
Affiliation(s)
- Kevin Nguyen
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Karina Fama
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Guadalupe Mercado
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
| | - Yin Myat
- University College Dublin School of Medicine, Belfield, D04 V1W8 Dublin, Ireland
- One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA
| | - Kyaw Thein
- Touro University Nevada College of Osteopathic Medicine, 874 American Pacific Dr, Henderson, NV 89014, USA; (K.N.)
- Comprehensive Cancer Centers of Nevada—Central Valley, 3730 S Eastern Ave, Las Vegas, NV 89169, USA
- Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
| |
Collapse
|
7
|
Thein KZ, Myat YM, Park BS, Panigrahi K, Kummar S. Target-Driven Tissue-Agnostic Drug Approvals-A New Path of Drug Development. Cancers (Basel) 2024; 16:2529. [PMID: 39061168 PMCID: PMC11274498 DOI: 10.3390/cancers16142529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The regulatory approvals of tumor-agnostic therapies have led to the re-evaluation of the drug development process. The conventional models of drug development are histology-based. On the other hand, the tumor-agnostic drug development of a new drug (or combination) focuses on targeting a common genomic biomarker in multiple cancers, regardless of histology. The basket-like clinical trials with multiple cohorts allow clinicians to evaluate pan-cancer efficacy and toxicity. There are currently eight tumor agnostic approvals granted by the Food and Drug Administration (FDA). This includes two immune checkpoint inhibitors, and five targeted therapy agents. Pembrolizumab is an anti-programmed cell death protein-1 (PD-1) antibody that was the first FDA-approved tumor-agnostic treatment for unresectable or metastatic microsatellite instability-high (MSI-H) or deficient mismatch repair (dMMR) solid tumors in 2017. It was later approved for tumor mutational burden-high (TMB-H) solid tumors, although the TMB cut-off used is still debated. Subsequently, in 2021, another anti-PD-1 antibody, dostarlimab, was also approved for dMMR solid tumors in the refractory setting. Patients with fusion-positive cancers are typically difficult to treat due to their rare prevalence and distribution. Gene rearrangements or fusions are present in a variety of tumors. Neurotrophic tyrosine kinase (NTRK) fusions are present in a range of pediatric and adult solid tumors in varying frequency. Larotrectinib and entrectinib were approved for neurotrophic tyrosine kinase (NTRK) fusion-positive cancers. Similarly, selpercatinib was approved for rearranged during transfection (RET) fusion-positive solid tumors. The FDA approved the first combination therapy of dabrafenib, a B-Raf proto-oncogene serine/threonine kinase (BRAF) inhibitor, plus trametinib, a mitogen-activated protein kinase (MEK) inhibitor for patients 6 months or older with unresectable or metastatic tumors (except colorectal cancer) carrying a BRAFV600E mutation. The most recent FDA tumor-agnostic approval is of fam-trastuzumab deruxtecan-nxki (T-Dxd) for HER2-positive solid tumors. It is important to identify and expeditiously develop drugs that have the potential to provide clinical benefit across tumor types.
Collapse
Affiliation(s)
- Kyaw Z. Thein
- Division of Hematology and Medical Oncology, Comprehensive Cancer Centers of Nevada—Central Valley, 3730 S Eastern Ave, Las Vegas, NV 89169, USA
- Department of Medicine, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), 4505 S, Maryland Pkwy, Las Vegas, NV 89154, USA
- College of Osteopathic Medicine, Touro University Nevada, Touro College and University System, 874 American Pacific Dr, Henderson, NV 89014, USA
| | - Yin M. Myat
- Belfield Campus, University College Dublin (UCD) School of Medicine, D04 V1W8 Dublin, Ireland;
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Byung S. Park
- OHSU-PSU School of Public Health, Portland, OR 97201, USA;
- Biostatistics Shared Resource, OHSU Knight Cancer Institute, OHSU School of Medicine, Portland, OR 97239, USA
| | - Kalpana Panigrahi
- Department of Internal Medicine, One Brooklyn Health—Interfaith Medical Center Campus, 1545, Atlantic Avenue, Brooklyn, NY 11213, USA;
| | - Shivaani Kummar
- Division of Hematology & Medical Oncology, Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA;
| |
Collapse
|
8
|
Sugden BM, Grimm SE, Wolff R, Armstrong N, Otten T, Abu-Zahra T, Perry M, Patel M, Chen J, Noake C, Joore M, Witlox WJA. Bayesian Hierarchical Modelling for Histology-Independent Time-to-Event Outcomes in the NICE Single Technology Appraisal of Pembrolizumab for Solid Tumours with MSI-H/dMMR: External Assessment Group Perspective. PHARMACOECONOMICS 2024; 42:615-618. [PMID: 38713424 PMCID: PMC11126505 DOI: 10.1007/s40273-024-01381-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/07/2024] [Indexed: 05/08/2024]
Affiliation(s)
- Bradley M Sugden
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands.
| | - Sabine E Grimm
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| | | | | | - Thomas Otten
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| | - Teebah Abu-Zahra
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| | - Mark Perry
- Kleijnen Systematic Reviews Limited, York, UK
| | | | | | - Caro Noake
- Kleijnen Systematic Reviews Limited, York, UK
| | - Manuela Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| | - Willem J A Witlox
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+ (MUMC+), Maastricht, The Netherlands
| |
Collapse
|
9
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [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: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
10
|
Russo GL, Bironzo P, Bennati C, Bonanno L, Catino A, Metro G, Petrini I, Russano M, Passaro A. Clinical evidence and adverse event management update of patients with RET- rearranged advanced non-small-cell lung cancer (NSCLC) treated with pralsetinib. Crit Rev Oncol Hematol 2024; 194:104243. [PMID: 38135019 DOI: 10.1016/j.critrevonc.2023.104243] [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: 10/11/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023] Open
Abstract
Current non-small cell lung cancer (NSCLC) management relies on genome-driven precision oncology thus shifting treatment paradigm towards biomarker-guided tumor-agnostic approaches. Recently, rearranged during transfection (RET) has been endorsed as tissue-agnostic target with sensitivity to RET inhibition. There are currently two selective RET tyrosine kinase inhibitors, pralsetinib and selpercatinib. The recent introduction of pralsetinib in the treatment algorithm of RET-rearranged tumor along with the mounting clinical evidence of pralsetinib durable activity from both randomized and observational studies holds the potential to disclose new avenues in the management of RET fusion positive NSCLC patients. Our narrative review aims to discuss the available clinical evidence on pralsetinib efficacy, particularly on brain metastases, and tolerability profile. In addition, our work explores the relevance of detecting RET fusions upfront in the disease history of patients with NSCLC.
Collapse
Affiliation(s)
- Giuseppe Lo Russo
- Medical Oncology Department, Thoracic Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Bironzo
- Department of Oncology, University of Torino, Ospedale San Luigi Gonzaga, Orbassano, TO, Italy
| | - Chiara Bennati
- Department of Onco-Hematology, AUSL della Romagna, Ravenna, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - Annamaria Catino
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera Perugia, Italy
| | - Iacopo Petrini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marco Russano
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milano, Italy.
| |
Collapse
|
11
|
Bekaii-Saab TS, Yaeger R, Spira AI, Pelster MS, Sabari JK, Hafez N, Barve M, Velastegui K, Yan X, Shetty A, Der-Torossian H, Pant S. Adagrasib in Advanced Solid Tumors Harboring a KRASG12C Mutation. J Clin Oncol 2023; 41:4097-4106. [PMID: 37099736 PMCID: PMC10852394 DOI: 10.1200/jco.23.00434] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
PURPOSE Adagrasib, a KRASG12C inhibitor, has demonstrated clinical activity in patients with KRASG12C-mutated non-small-cell lung cancer (NSCLC) and colorectal cancer (CRC). KRASG12C mutations occur rarely in other solid tumor types. We report evaluation of the clinical activity and safety of adagrasib in patients with other solid tumors harboring a KRASG12C mutation. METHODS In this phase II cohort of the KRYSTAL-1 study (ClinicalTrials.gov identifier: NCT03785249; phase Ib cohort), we evaluated adagrasib (600 mg orally twice daily) in patients with KRASG12C-mutated advanced solid tumors (excluding NSCLC and CRC). The primary end point was objective response rate. Secondary end points included duration of response, progression-free survival (PFS), overall survival, and safety. RESULTS As of October 1, 2022, 64 patients with KRASG12C-mutated solid tumors were enrolled and 63 patients treated (median follow-up, 16.8 months). The median number of prior lines of systemic therapy was 2. Among 57 patients with measurable disease at baseline, objective responses were observed in 20 (35.1%) patients (all partial responses), including 7/21 (33.3%) responses in pancreatic and 5/12 (41.7%) in biliary tract cancers. The median duration of response was 5.3 months (95% CI, 2.8 to 7.3) and median PFS was 7.4 months (95% CI, 5.3 to 8.6). Treatment-related adverse events (TRAEs) of any grade were observed in 96.8% of patients and grade 3-4 in 27.0%; there were no grade 5 TRAEs. TRAEs did not lead to treatment discontinuation in any patients. CONCLUSION Adagrasib demonstrates encouraging clinical activity and is well tolerated in this rare cohort of pretreated patients with KRASG12C-mutated solid tumors.
Collapse
Affiliation(s)
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander I. Spira
- Virginia Cancer Specialists, Fairfax, VA
- NEXT Oncology, Fairfax, VA
- US Oncology Research, The Woodlands, TX
| | | | - Joshua K. Sabari
- Perlmutter Cancer Center, New York University Langone Health, New York, NY
| | | | | | | | | | | | | | - Shubham Pant
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
12
|
Vinnat V, Chiche JD, Demoule A, Chevret S. Simulation study for evaluating an adaptive-randomisation Bayesian hybrid trial design with enrichment. Contemp Clin Trials Commun 2023; 33:101141. [PMID: 37397429 PMCID: PMC10313856 DOI: 10.1016/j.conctc.2023.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/22/2023] [Accepted: 04/12/2023] [Indexed: 07/04/2023] Open
Abstract
Background As we enter the era of precision medicine, the role of adaptive designs, such as response-adaptive randomisation or enrichment designs in drug discovery and development, has become increasingly important to identify the treatment given to a patient based on one or more biomarkers. Tailoring the ventilation supply technique according to the responsiveness of patients to positive end-expiratory pressure is a suitable setting for such a design. Methods In the setting of marker-strategy design, we propose a Bayesian response-adaptive randomisation with enrichment design based on group sequential analyses. This design combines the elements of enrichment design and response-adaptive randomisation. Concerning the enrichment strategy, Bayesian treatment-by-subset interaction measures were used to adaptively enrich the patients most likely to benefit from an experimental treatment while controlling the false-positive rate.The operating characteristics of the design were assessed by simulation and compared to those of alternate designs. Results The results obtained allowed the detection of the superiority of one treatment over another and the presence of a treatment-by-subgroup interaction while keeping the false-positive rate at approximately 5\% and reducing the average number of included patients. In addition, simulation studies identified that the number of interim analyses and the burn-in period may have an impact on the performance of the scheme. Conclusion The proposed design highlights important objectives of precision medicine, such as determining whether the experimental treatment is superior to another and identifying wheter such an efficacy could depend on patient profile.
Collapse
Affiliation(s)
- Valentin Vinnat
- ECSTRRA team, INSERM U1153, Université Paris Cité, Paris, France
| | - Jean-Daniel Chiche
- Service de médecine intensive adulte, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Alexandre Demoule
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique, France
- AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Service de Médecine Intensive et Réanimation (Département R3S), Paris, France
| | - Sylvie Chevret
- ECSTRRA team, INSERM U1153, Université Paris Cité, Paris, France
| |
Collapse
|